MODULATION OF GLUCAGON RECEPTOR EXPRESSION

Compounds, compositions and methods are provided for modulating the expression of glucagon receptor. The compositions comprise oligonucleotides, targeted to nucleic acid encoding glucagon receptor. Methods of using these compounds for modulation of glucagon receptor expression and for diagnosis and treatment of disease associated with expression of glucagon receptor are provided.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
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 BIOL0007USD3SEQ.txt, created on Apr. 5, 2007 which is 208 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 compositions and methods for modulating the expression of glucagon receptor. In particular, this invention relates to compounds, particularly oligonucleotide compounds, which, in preferred embodiments, hybridize with nucleic acid molecules encoding glucagon receptor. Such compounds are shown herein to modulate the expression of glucagon receptor.

BACKGROUND OF THE INVENTION

The maintenance of normal glycemia is a carefully regulated metabolic event. Glucagon, the 29-amino acid peptide responsible for maintaining blood glucose levels in the postabsorbative state, increases glucose release from the liver by activating hepatic glycogenolysis, gluconeogenesis, stimulating lipolysis in adipose tissue, and stimulating insulin secretion. During high blood glucose levels, insulin reverses the glucagon-mediated enhancement of glycogenolysis and gluconeogenesis. In patients with diabetes, insulin is either not available or not fully effective. While treatment for diabetes has traditionally focused on increasing insulin levels, antagonism of glucagon function has been considered as an alternative therapy. As glucagon exerts its physiological effects by signaling through the glucagon receptor, the glucagon receptor has been proposed as a potential therapeutic target for diabetes (Madsen et al., Curr. Pharm. Des., 1999, 5, 683-691).

Glucagon receptor is belongs to the superfamily of G-protein-coupled receptors having seven transmembrane domains. It is also a member of the smaller sub-family of homologous receptors which bind peptides that are structurally similar to glucagon. The gene encoding human glucagon receptor was cloned in 1994 and analysis of the genomic sequence revealed multiple introns and an 82% identity to the rat glucagon receptor gene (Lok et al., Gene, 1994, 140, 203-209.; MacNeil et al., Biochem. Biophys. Res. Commun., 1994, 198, 328-334). Cloning of the rat glucagon receptor gene also led to the description of multiple alternative splice variants (Maget et al., FEBS Lett., 1994, 351, 271-275). Disclosed and claimed in U.S. Pat. No. 5,776,725 is an isolated nucleic acid sequence encoding a human or rat glucagon receptor (Kindsvogel et al., 1998). The human glucagon receptor gene is localized to chromosome 17q25 (Menzel et al., Genomics, 1994, 20, 327-328). A missense mutation of Gly to Ser at codon 40 in the glucagon receptor gene leads to a 3-fold lower affinity for glucagon (Fujisawa et al., Diabetologia, 1995, 38, 983-985) and this mutation has been linked to several disease states, including non-insulin-dependent diabetes mellitus (Fujisawa et al., Diabetologia, 1995, 38, 983-985), hypertension (Chambers and Morris, Nat. Genet., 1996, 12, 122), and central adiposity (Siani et al., Obes. Res., 2001, 9, 722-726).

Inhibiting glucagon function by antagonizing the glucagon receptor has been proposed as a therapeutic target for diabetes. Currently, there are no known therapeutic agents which effectively inhibit the synthesis of glucagon receptor and to date, investigative strategies aimed at modulating glucagon receptor function have involved the use of antibodies, peptidyl antagonists, and small molecules. In addition, targeted disruption of the glucagon receptor gene in mice has shown that, despite a total absence of glucagon receptors and elevated plasma glucagon levels, the mice maintain near-normal glycemia and lipidemia (Parker et al., Biochem. Biophys. Res. Commun., 2002, 290, 839-843). Patent application WO 02/45494 (Allen et al.) discloses transgenic mice comprising mutations in a glucagon receptor gene. Also claimed are agonists or antagonists of glucagon receptor, agents that modulate the function, expression or activity of a glucagon receptor gene, methods of identifying such agents, methods of ameliorating conditions associated with impaired glucose tolerance, methods of identifying agents that affect obesity, weight gain, diabetes, methods of treating obesity or diabetic conditions, and phenotypic data associated with a transgenic mouse comprising a mutation in a glucagon receptor gene.

A glucagon-neutralizing monoclonal antibody has been described that antagonizes glucagon-stimulated signal transduction in part by binding to the glucagon binding site of the glucagon receptor (Buggy et al., Horm. Metab. Res., 1996, 28, 215-219). An antibody which specifically binds to the amino acid sequence of a glucagon receptor has been disclosed and claimed in U.S. Pat. No. 5,770,445 (Kindsvogel et al., 1998).

Several peptidyl antagonists of glucagon receptor have been reported in the art. Six glucagon analogs with N-terminal modifications were designed to have a higher affinity than glucagon for the glucagon receptor (Zechel et al., Int. J. Pept. Protein Res., 1991, 38, 131-138). Two somatostatin analogs have been reported to be inhibitors of glucagon secretion (Rossowski and Coy, Biochem. Biophys. Res. Commun., 1994, 205, 341-346).

Many small molecules have been examined as glucagon receptor antagonists including: [(+)-3,5 diisopropyl-2-(1-hydroxyethyl)-6-propyl-4′-fluoro-1,1′-biphenyl (Bay27-9955) (Petersen and Sullivan, Diabetologia, 2001, 44, 2018-2024), a series of alkylidene hydrazides (Ling et al., Bioorg. Med. Chem. Lett., 2002, 12, 663-666), a series of 4-aryl-pyridines containing both a 3-[(1R)-hydroxyethyl] and a 2′-hydroxy group (Ladouceur et al., Bioorg. Med. Chem. Lett., 2002, 12, 3421-3424), a series of 5-hydroxyalkyl-4-phenylpyridines (Ladouceur et al., Bioorg. Med. Chem. Lett., 2002, 12, 461-464), a series of triarylimidazoles (Chang et al., Bioorg. Med. Chem. Lett., 2001, 11, 2549-2553), a series of 2-pyridyl-3,5-diaryl pyrroles (de Laszlo et al., Bioorg. Med. Chem. Lett., 1999, 9, 641-646), several substituted benzimidazoles (Madsen et al., J. Med. Chem., 1998, 41, 5150-5157), and a series of pyrrolo[1,2-a]quinoxalines (Guillon et al., Eur. J. Med. Chem., 1998, 33, 293-308).

There remains a long felt need for additional agents capable of effectively inhibiting glucagon receptor function. Antisense technology is an effective means for reducing the expression of specific gene products and has proven to be uniquely useful in a number of therapeutic, diagnostic, and research applications. The present invention provides compositions and methods for modulating glucagon receptor expression.

SUMMARY OF THE INVENTION

The present invention is directed to compounds, especially nucleic acid and nucleic acid-like oligomers, which are targeted to a nucleic acid encoding glucagon receptor, and which modulate the expression of glucagon receptor. Pharmaceutical and other compositions comprising the compounds of the invention are also provided. Further provided are methods of screening for modulators of glucagon receptor and methods of modulating the expression of glucagon receptor in cells, tissues or animals comprising contacting said cells, tissues or animals with one or more of the compounds or compositions of the invention. Methods of treating an animal, particularly a human, suspected of having or being prone to a disease or condition associated with expression of glucagon receptor are also set forth herein. Such methods comprise administering a therapeutically or prophylactically effective amount of one or more of the compounds or compositions of the invention to the person in need of treatment.

DETAILED DESCRIPTION OF THE INVENTION

A. Overview of the Invention

The present invention employs compounds, preferably oligonucleotides and similar species for use in modulating the function or effect of nucleic acid molecules encoding glucagon receptor. This is accomplished by providing oligonucleotides which specifically hybridize with one or more nucleic acid molecules encoding glucagon receptor. As used herein, the terms “target nucleic acid” and “nucleic acid molecule encoding glucagon receptor” have been used for convenience to encompass DNA encoding glucagon receptor, RNA (including pre-mRNA and mRNA or portions thereof) transcribed from such DNA, and also cDNA derived from such RNA. The hybridization of a compound of this invention with its target nucleic acid is generally referred to as “antisense”. Consequently, the preferred mechanism believed to be included in the practice of some preferred embodiments of the invention is referred to herein as “antisense inhibition.” Such antisense inhibition is typically based upon hydrogen bonding-based hybridization of oligonucleotide strands or segments such that at least one strand or segment is cleaved, degraded, or otherwise rendered inoperable. In this regard, it is presently preferred to target specific nucleic acid molecules and their functions for such antisense inhibition.

The functions of DNA to be interfered with can include replication and transcription. Replication and transcription, for example, can be from an endogenous cellular template, a vector, a plasmid construct or otherwise. The functions of RNA to be interfered with can include functions such as translocation of the RNA to a site of protein translation, translocation of the RNA to sites within the cell which are distant from the site of RNA synthesis, translation of protein from the RNA, splicing of the RNA to yield one or more RNA species, and catalytic activity or complex formation involving the RNA which may be engaged in or facilitated by the RNA. One preferred result of such interference with target nucleic acid function is modulation of the expression of glucagon receptor. In the context of the present invention, “modulation” and “modulation of expression” mean either an increase (stimulation) or a decrease (inhibition) in the amount or levels of a nucleic acid molecule encoding the gene, e.g., DNA or RNA. Inhibition is often the preferred form of modulation of expression and mRNA is often a preferred target nucleic acid.

In the context of this invention, “hybridization” means the pairing of complementary strands of oligomeric compounds. In the present invention, the preferred mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases) of the strands of oligomeric compounds. For example, adenine and thymine are complementary nucleobases which pair through the formation of hydrogen bonds. Hybridization can occur under varying circumstances.

An antisense compound is specifically hybridizable when binding of the compound to the target nucleic acid interferes with the normal function of the target nucleic acid to cause a loss of activity, and there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-target nucleic acid sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assays are performed in the case of in vitro assays.

In the present invention the phrase “stringent hybridization conditions” or “stringent conditions” refers to conditions under which a compound of the invention will hybridize to its target sequence, but to a minimal number of other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances and in the context of this invention, “stringent conditions” under which oligomeric compounds hybridize to a target sequence are determined by the nature and composition of the oligomeric compounds and the assays in which they are being investigated.

“Complementary,” as used herein, refers to the capacity for precise pairing between two nucleobases of an oligomeric compound. For example, if a nucleobase at a certain position of an oligonucleotide (an oligomeric compound), is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, said target nucleic acid being a DNA, RNA, or oligonucleotide molecule, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be a complementary position. The oligonucleotide and the further DNA, RNA, or oligonucleotide molecule are complementary to each other when a sufficient number of complementary positions in each molecule are occupied by nucleobases which can hydrogen bond with each other. Thus, “specifically hybridizable” and “complementary” are terms which are used to indicate a sufficient degree of precise pairing or complementarity over a sufficient number of nucleobases such that stable and specific binding occurs between the oligonucleotide and a target nucleic acid.

It is understood in the art that the sequence of an antisense compound need not be 100% complementary to that of its target nucleic acid to be specifically hybridizable. Moreover, an oligonucleotide may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (e.g., a loop structure or hairpin structure). It is preferred that the antisense compounds of the present invention comprise at least 70% sequence complementarity to a target region within the target nucleic acid, more preferably that they comprise 90% sequence complementarity and even more preferably comprise 95% sequence complementarity to the target region within the target nucleic acid sequence to which they are targeted. For example, an antisense compound in which 18 of 20 nucleobases of the antisense compound are complementary to a target region, and would therefore specifically hybridize, would represent 90 percent complementarity. In this example, the remaining noncomplementary nucleobases may be clustered or interspersed with complementary nucleobases and need not be contiguous to each other or to complementary nucleobases. As such, an antisense compound which is 18 nucleobases in length having 4 (four) noncomplementary nucleobases which are flanked by two regions of complete complementarity with the target nucleic acid would have 77.8% overall complementarity with the target nucleic acid and would thus fall within the scope of the present invention. Percent complementarity of an antisense compound with a region of a target nucleic acid can be determined routinely using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al., J. Mol. Biol., 1990, 215, 403-410; Zhang and Madden, Genome Res., 1997, 7, 649-656).

B. Compounds of the Invention

According to the present invention, compounds include antisense oligomeric compounds, antisense oligonucleotides, ribozymes, external guide sequence (EGS) oligonucleotides, alternate splicers, primers, probes, and other oligomeric compounds which hybridize to at least a portion of the target nucleic acid. As such, these compounds may be introduced in the form of single-stranded, double-stranded, circular or hairpin oligomeric compounds and may contain structural elements such as internal or terminal bulges or loops. Once introduced to a system, the compounds of the invention may elicit the action of one or more enzymes or structural proteins to effect modification of the target nucleic acid.

One non-limiting example of such an enzyme is RNAse H, a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. It is known in the art that single-stranded antisense compounds which are “DNA-like” elicit RNAse H. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of oligonucleotide-mediated inhibition of gene expression. Similar roles have been postulated for other ribonucleases such as those in the RNase III and ribonuclease L family of enzymes.

While the preferred form of antisense compound is a single-stranded antisense oligonucleotide, in many species the introduction of double-stranded structures, such as double-stranded RNA (dsRNA) molecules, has been shown to induce potent and specific antisense-mediated reduction of the function of a gene or its associated gene products. This phenomenon occurs in both plants and animals and is believed to have an evolutionary connection to viral defense and transposon silencing.

The first evidence that dsRNA could lead to gene silencing in animals came in 1995 from work in the nematode, Caenorhabditis elegans (Guo and Kempheus, Cell, 1995, 81, 611-620).

Montgomery et al. have shown that the primary interference effects of dsRNA are posttranscriptional (Montgomery et al., Proc. Natl. Acad. Sci. USA, 1998, 95, 15502-15507). The posttranscriptional antisense mechanism defined in Caenorhabditis elegans resulting from exposure to double-stranded RNA (dsRNA) has since been designated RNA interference (RNAi). This term has been generalized to mean antisense-mediated gene silencing involving the introduction of dsRNA leading to the sequence-specific reduction of endogenous targeted mRNA levels (Fire et al., Nature, 1998, 391, 806-811). Recently, it has been shown that it is, in fact, the single-stranded RNA oligomers of antisense polarity of the dsRNAs which are the potent inducers of RNAi (Tijsterman et al., Science, 2002, 295, 694-697).

In the context of this invention, the term “oligomeric compound” refers to a polymer or oligomer comprising a plurality of monomeric units. In the context of this invention, the term “oligonucleotide” refers to an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA) or mimetics, chimeras, analogs and homologs thereof. This term includes oligonucleotides composed of naturally occurring nucleobases, sugars and covalent internucleoside (backbone) linkages as well as oligonucleotides having non-naturally occurring portions which function similarly. Such modified or substituted oligonucleotides are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for a target nucleic acid and increased stability in the presence of nucleases.

While oligonucleotides are a preferred form of the compounds of this invention, the present invention comprehends other families of compounds as well, including but not limited to oligonucleotide analogs and mimetics such as those described herein.

The compounds in accordance with this invention preferably comprise from about 8 to about 80 nucleobases (i.e. from about 8 to about 80 linked nucleosides). One of ordinary skill in the art will appreciate that the invention embodies compounds of 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80 nucleobases in length.

In one preferred embodiment, the compounds of the invention are 12 to 50 nucleobases in length. One having ordinary skill in the art will appreciate that this embodies compounds of 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, or 50 nucleobases in length.

In another preferred embodiment, the compounds of the invention are 15 to 30 nucleobases in length. One having ordinary skill in the art will appreciate that this embodies compounds of 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleobases in length.

Particularly preferred compounds are oligonucleotides from about 12 to about 50 nucleobases, even more preferably those comprising from about 15 to about 30 nucleobases.

Antisense compounds 8-80 nucleobases in length comprising a stretch of at least eight (8) consecutive nucleobases selected from within the illustrative antisense compounds are considered to be suitable antisense compounds as well.

Exemplary preferred antisense compounds include oligonucleotide sequences that comprise at least the 8 consecutive nucleobases from the 5′-terminus of one of the illustrative preferred antisense compounds (the remaining nucleobases being a consecutive stretch of the same oligonucleotide beginning immediately upstream of the 5′-terminus of the antisense compound which is specifically hybridizable to the target nucleic acid and continuing until the oligonucleotide contains about 8 to about 80 nucleobases). Similarly preferred antisense compounds are represented by oligonucleotide sequences that comprise at least the 8 consecutive nucleobases from the 3′-terminus of one of the illustrative preferred antisense compounds (the remaining nucleobases being a consecutive stretch of the same oligonucleotide beginning immediately downstream of the 3′-terminus of the antisense compound which is specifically hybridizable to the target nucleic acid and continuing until the oligonucleotide contains about 8 to about 80 nucleobases). One having skill in the art armed with the preferred antisense compounds illustrated herein will be able, without undue experimentation, to identify further preferred antisense compounds.

C. Targets of the Invention

“Targeting” an antisense compound to a particular nucleic acid molecule, in the context of this invention, can be a multistep process. The process usually begins with the identification of a target nucleic acid whose function is to be modulated. This target nucleic acid may be, for example, a cellular gene (or mRNA transcribed from the gene) whose expression is associated with a particular disorder or disease state, or a nucleic acid molecule from an infectious agent. In the present invention, the target nucleic acid encodes glucagon receptor.

The targeting process usually also includes determination of at least one target region, segment, or site within the target nucleic acid for the antisense interaction to occur such that the desired effect, e.g., modulation of expression, will result. Within the context of the present invention, the term “region” is defined as a portion of the target nucleic acid having at least one identifiable structure, function, or characteristic. Within regions of target nucleic acids are segments. “Segments” are defined as smaller or sub-portions of regions within a target nucleic acid. “Sites,” as used in the present invention, are defined as positions within a target nucleic acid.

Since, as is known in the art, the translation initiation codon is typically 5′-AUG (in transcribed mRNA molecules; 5′-ATG in the corresponding DNA molecule), the translation initiation codon is also referred to as the “AUG codon,” the “start codon” or the “AUG start codon”. A minority of genes have a translation initiation codon having the RNA sequence 5′-GUG, 5′-UUG or 5′-CUG, and 5′-AUA, 5′-ACG and 5′-CUG have been shown to function in vivo. Thus, the terms “translation initiation codon” and “start codon” can encompass many codon sequences, even though the initiator amino acid in each instance is typically methionine (in eukaryotes) or formylmethionine (in prokaryotes). It is also known in the art that eukaryotic and prokaryotic genes may have two or more alternative start codons, any one of which may be preferentially utilized for translation initiation in a particular cell type or tissue, or under a particular set of conditions. In the context of the invention, “start codon” and “translation initiation codon” refer to the codon or codons that are used in vivo to initiate translation of an mRNA transcribed from a gene encoding glucagon receptor, regardless of the sequence(s) of such codons. It is also known in the art that a translation termination codon (or “stop codon”) of a gene may have one of three sequences, i.e., 5′-UAA, 5′-UAG and 5′-UGA (the corresponding DNA sequences are 5′-TAA, 5′-TAG and 5′-TGA, respectively).

The terms “start codon region” and “translation initiation codon region” refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation initiation codon. Similarly, the terms “stop codon region” and “translation termination codon region” refer to a portion of such an mRNA or gene that encompasses from about 25 to about 50 contiguous nucleotides in either direction (i.e., 5′ or 3′) from a translation termination codon. Consequently, the “start codon region” (or “translation initiation codon region”) and the “stop codon region” (or “translation termination codon region”) are all regions which may be targeted effectively with the antisense compounds of the present invention.

The open reading frame (ORF) or “coding region,” which is known in the art to refer to the region between the translation initiation codon and the translation termination codon, is also a region which may be targeted effectively. Within the context of the present invention, a preferred region is the intragenic region encompassing the translation initiation or termination codon of the open reading frame (ORF) of a gene.

Other target regions include the 5′ untranslated region (5′UTR), known in the art to refer to the portion of an mRNA in the 5′ direction from the translation initiation codon, and thus including nucleotides between the 5′ cap site and the translation initiation codon of an mRNA (or corresponding nucleotides on the gene), and the 3′ untranslated region (3′UTR), known in the art to refer to the portion of an mRNA in the 3′ direction from the translation termination codon, and thus including nucleotides between the translation termination codon and 3′ end of an mRNA (or corresponding nucleotides on the gene). The 5′ cap site of an mRNA comprises an N7-methylated guanosine residue joined to the 5′-most residue of the mRNA via a 5′-5′ triphosphate linkage. The 5′ cap region of an mRNA is considered to include the 5′ cap structure itself as well as the first 50 nucleotides adjacent to the cap site. It is also preferred to target the 5′ cap region.

Although some eukaryotic mRNA transcripts are directly translated, many contain one or more regions, known as “introns,” which are excised from a transcript before it is translated. The remaining (and therefore translated) regions are known as “exons” and are spliced together to form a continuous mRNA sequence. Targeting splice sites, i.e., intron-exon junctions or exon-intron junctions, may also be particularly useful in situations where aberrant splicing is implicated in disease, or where an overproduction of a particular splice product is implicated in disease. Aberrant fusion junctions due to rearrangements or deletions are also preferred target sites. mRNA transcripts produced via the process of splicing of two (or more) mRNAs from different gene sources are known as “fusion transcripts”. It is also known that introns can be effectively targeted using antisense compounds targeted to, for example, DNA or pre-mRNA.

It is also known in the art that alternative RNA transcripts can be produced from the same genomic region of DNA. These alternative transcripts are generally known as “variants”. More specifically, “pre-mRNA variants” are transcripts produced from the same genomic DNA that differ from other transcripts produced from the same genomic DNA in either their start or stop position and contain both intronic and exonic sequence.

Upon excision of one or more exon or intron regions, or portions thereof during splicing, pre-mRNA variants produce smaller “mRNA variants”. Consequently, mRNA variants are processed pre-mRNA variants and each unique pre-mRNA variant must always produce a unique mRNA variant as a result of splicing. These mRNA variants are also known as “alternative splice variants”. If no splicing of the pre-mRNA variant occurs then the pre-mRNA variant is identical to the mRNA variant.

It is also known in the art that variants can be produced through the use of alternative signals to start or stop transcription and that pre-mRNAs and mRNAs can possess more that one start codon or stop codon. Variants that originate from a pre-mRNA or mRNA that use alternative start codons are known as “alternative start variants” of that pre-mRNA or mRNA. Those transcripts that use an alternative stop codon are known as “alternative stop variants” of that pre-mRNA or mRNA. One specific type of alternative stop variant is the “polyA variant” in which the multiple transcripts produced result from the alternative selection of one of the “polyA stop signals” by the transcription machinery, thereby producing transcripts that terminate at unique polyA sites. Within the context of the invention, the types of variants described herein are also preferred target nucleic acids.

The locations on the target nucleic acid to which the preferred antisense compounds hybridize are hereinbelow referred to as “preferred target segments.” As used herein the term “preferred target segment” is defined as at least an 8-nucleobase portion of a target region to which an active antisense compound is targeted. While not wishing to be bound by theory, it is presently believed that these target segments represent portions of the target nucleic acid which are accessible for hybridization.

While the specific sequences of certain preferred target segments are set forth herein, one of skill in the art will recognize that these serve to illustrate and describe particular embodiments within the scope of the present invention. Additional preferred target segments may be identified by one having ordinary skill.

Target segments 8-80 nucleobases in length comprising a stretch of at least eight (8) consecutive nucleobases selected from within the illustrative preferred target segments are considered to be suitable for targeting as well.

Target segments can include DNA or RNA sequences that comprise at least the 8 consecutive nucleobases from the 5′-terminus of one of the illustrative preferred target segments (the remaining nucleobases being a consecutive stretch of the same DNA or RNA beginning immediately upstream of the 5′-terminus of the target segment and continuing until the DNA or RNA contains about 8 to about 80 nucleobases). Similarly preferred target segments are represented by DNA or RNA sequences that comprise at least the 8 consecutive nucleobases from the 3′-terminus of one of the illustrative preferred target segments (the remaining nucleobases being a consecutive stretch of the same DNA or RNA beginning immediately downstream of the 3′-terminus of the target segment and continuing until the DNA or RNA contains about 8 to about 80 nucleobases). One having skill in the art armed with the preferred target segments illustrated herein will be able, without undue experimentation, to identify further preferred target segments.

Once one or more target regions, segments or sites have been identified, antisense compounds are chosen which are sufficiently complementary to the target, i.e., hybridize sufficiently well and with sufficient specificity, to give the desired effect.

D. Screening and Target Validation

In a further embodiment, the “preferred target segments” identified herein may be employed in a screen for additional compounds that modulate the expression of glucagon receptor. “Modulators” are those compounds that decrease or increase the expression of a nucleic acid molecule encoding glucagon receptor and which comprise at least an 8-nucleobase portion which is complementary to a preferred target segment. The screening method comprises the steps of contacting a preferred target segment of a nucleic acid molecule encoding glucagon receptor with one or more candidate modulators, and selecting for one or more candidate modulators which decrease or increase the expression of a nucleic acid molecule encoding glucagon receptor. Once it is shown that the candidate modulator or modulators are capable of modulating (e.g. either decreasing or increasing) the expression of a nucleic acid molecule encoding glucagon receptor, the modulator may then be employed in further investigative studies of the function of glucagon receptor, or for use as a research, diagnostic, or therapeutic agent in accordance with the present invention.

The preferred target segments of the present invention may be also be combined with their respective complementary antisense compounds of the present invention to form stabilized double-stranded (duplexed) oligonucleotides.

Such double stranded oligonucleotide moieties have been shown in the art to modulate target expression and regulate translation as well as RNA processsing via an antisense mechanism. Moreover, the double-stranded moieties may be subject to chemical modifications (Fire et al., Nature, 1998, 391, 806-811; Timmons and Fire, Nature 1998, 395, 854; Timmons et al., Gene, 2001, 263, 103-112; Tabara et al., Science, 1998, 282, 430-431; Montgomery et al., Proc. Natl. Acad. Sci. USA, 1998, 95, 15502-15507; Tuschl et al., Genes Dev., 1999, 13, 3191-3197; Elbashir et al., Nature, 2001, 411, 494-498; Elbashir et al., Genes Dev. 2001, 15, 188-200). For example, such double-stranded moieties have been shown to inhibit the target by the classical hybridization of antisense strand of the duplex to the target, thereby triggering enzymatic degradation of the target (Tijsterman et al., Science, 2002, 295, 694-697).

The compounds of the present invention can also be applied in the areas of drug discovery and target validation. The present invention comprehends the use of the compounds and preferred target segments identified herein in drug discovery efforts to elucidate relationships that exist between glucagon receptor and a disease state, phenotype, or condition. These methods include detecting or modulating glucagon receptor comprising contacting a sample, tissue, cell, or organism with the compounds of the present invention, measuring the nucleic acid or protein level of glucagon receptor and/or a related phenotypic or chemical endpoint at some time after treatment, and optionally comparing the measured value to a non-treated sample or sample treated with a further compound of the invention. These methods can also be performed in parallel or in combination with other experiments to determine the function of unknown genes for the process of target validation or to determine the validity of a particular gene product as a target for treatment or prevention of a particular disease, condition, or phenotype.

E. Kits, Research Reagents, Diagnostics, and Therapeutics

The compounds of the present invention can be utilized for diagnostics, therapeutics (including prophylaxis) and as research reagents and kits. Furthermore, antisense oligonucleotides, which are able to inhibit gene expression with exquisite specificity, are often used by those of ordinary skill to elucidate the function of particular genes or to distinguish between functions of various members of a biological pathway.

For use in kits and diagnostics, the compounds of the present invention, either alone or in combination with other compounds or therapeutics, can be used as tools in differential and/or combinatorial analyses to elucidate expression patterns of a portion or the entire complement of genes expressed within cells and tissues.

As one nonlimiting example, expression patterns within cells or tissues treated with one or more antisense compounds are compared to control cells or tissues not treated with antisense compounds and the patterns produced are analyzed for differential levels of gene expression as they pertain, for example, to disease association, signaling pathway, cellular localization, expression level, size, structure or function of the genes examined. These analyses can be performed on stimulated or unstimulated cells and in the presence or absence of other compounds which affect expression patterns.

Examples of methods of gene expression analysis known in the art include DNA arrays or microarrays (Brazma and Vilo, FEBS Lett., 2000, 480, 17-24; Celis, et al., FEBS Lett., 2000, 480, 2-16), SAGE (serial analysis of gene expression)(Madden, et al., Drug Discov. Today, 2000, 5, 415-425), READS (restriction enzyme amplification of digested cDNAs) (Prashar and Weissman, Methods Enzymol., 1999, 303, 258-72), TOGA (total gene expression analysis) (Sutcliffe, et al., Proc. Natl. Acad. Sci. U.S.A., 2000, 97, 1976-81), protein arrays and proteomics (Celis, et al., FEBS Lett., 2000, 480, 2-16; Jungblut, et al., Electrophoresis, 1999, 20, 2100-10), expressed sequence tag (EST) sequencing (Celis, et al., FEBS Lett., 2000, 480, 2-16; Larsson, et al., J. Biotechnol., 2000, 80, 143-57), subtractive RNA fingerprinting (SuRF) (Fuchs, et al., Anal. Biochem., 2000, 286, 91-98; Larson, et al., Cytometry, 2000, 41, 203-208), subtractive cloning, differential display (DD) (Jurecic and Belmont, Curr. Opin. Microbiol., 2000, 3, 316-21), comparative genomic hybridization (Carulli, et al., J. Cell Biochem. Suppl., 1998, 31, 286-96), FISH (fluorescent in situ hybridization) techniques (Going and Gusterson, Eur. J. Cancer, 1999, 35, 1895-904) and mass spectrometry methods (To, Comb. Chem. High Throughput Screen, 2000, 3, 235-41).

The compounds of the invention are useful for research and diagnostics, because these compounds hybridize to nucleic acids encoding glucagon receptor. For example, oligonucleotides that are shown to hybridize with such efficiency and under such conditions as disclosed herein as to be effective glucagon receptor inhibitors will also be effective primers or probes under conditions favoring gene amplification or detection, respectively. These primers and probes are useful in methods requiring the specific detection of nucleic acid molecules encoding glucagon receptor and in the amplification of said nucleic acid molecules for detection or for use in further studies of glucagon receptor. Hybridization of the antisense oligonucleotides, particularly the primers and probes, of the invention with a nucleic acid encoding glucagon receptor can be detected by means known in the art. Such means may include conjugation of an enzyme to the oligonucleotide, radiolabelling of the oligonucleotide or any other suitable detection means. Kits using such detection means for detecting the level of glucagon receptor in a sample may also be prepared.

The specificity and sensitivity of antisense is also harnessed by those of skill in the art for therapeutic uses. Antisense compounds have been employed as therapeutic moieties in the treatment of disease states in animals, including humans. Antisense oligonucleotide drugs, including ribozymes, have been safely and effectively administered to humans and numerous clinical trials are presently underway. It is thus established that antisense compounds can be useful therapeutic modalities that can be configured to be useful in treatment regimes for the treatment of cells, tissues and animals, especially humans.

For therapeutics, an animal, preferably a human, suspected of having a disease or disorder which can be treated by modulating the expression of glucagon receptor is treated by administering antisense compounds in accordance with this invention. For example, in one non-limiting embodiment, the methods comprise the step of administering to an animal a therapeutically effective amount of a glucagon receptor inhibitor. The glucagon receptor inhibitors of the present invention effectively inhibit the activity of the glucagon receptor protein or inhibit the expression of the glucagon receptor protein. In one embodiment, the activity or expression of glucagon receptor in an animal is inhibited by about 10%. Preferably, the activity or expression of glucagon receptor in an animal is inhibited by about 30%. More preferably, the activity or expression of glucagon receptor in an animal is inhibited by 50% or more. Because the compounds herein are inhibitors of glucagon receptor, they are believed to be useful in lowering blood glucose, for example, and in treating conditions associated with glucagon receptor activity, such as high blood glucose and other metabolic conditions such as diabetes (including Type 2 diabetes), obesity, and insulin resistance.

The reduction of the expression of glucagon receptor may be measured, for example, in blood, plasma, serum, adipose tissue, liver or any other body fluid, tissue or organ of the animal. Preferably, the cells contained within said fluids, tissues or organs being analyzed contain a nucleic acid molecule encoding glucagon receptor protein and/or the glucagon receptor protein itself.

The compounds of the invention can be utilized in pharmaceutical compositions by adding an effective amount of a compound to a suitable pharmaceutically acceptable diluent or carrier. Use of the compounds and methods of the invention may also be useful prophylactically.

F. Modifications

As is known in the art, a nucleoside is a base-sugar combination. The base portion of the nucleoside is normally a heterocyclic base. The two most common classes of such heterocyclic bases are the purines and the pyrimidines. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to either the 2′, 3′ or 5′ hydroxyl moiety of the sugar. In forming oligonucleotides, the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound. In turn, the respective ends of this linear polymeric compound can be further joined to form a circular compound, however, linear compounds are generally preferred. In addition, linear compounds may have internal nucleobase complementarity and may therefore fold in a manner as to produce a fully or partially double-stranded compound. Within oligonucleotides, the phosphate groups are commonly referred to as forming the internucleoside backbone of the oligonucleotide. The normal linkage or backbone of RNA and DNA is a 3′ to 5′ phosphodiester linkage.

Modified Internucleoside Linkages (Backbones)

Specific examples of preferred antisense compounds useful in this invention include oligonucleotides containing modified backbones or non-natural internucleoside linkages. As defined in this specification, oligonucleotides having modified backbones include those that retain a phosphorus atom in the backbone and those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified oligonucleotides that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides.

Preferred modified oligonucleotide backbones containing a phosphorus atom therein include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates, 5′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, selenophosphates and boranophosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein one or more internucleotide linkages is a 3′ to 3′, 5′ to 5′ or 2′ to 2′ linkage. Preferred oligonucleotides having inverted polarity comprise a single 3′ to 3′ linkage at the 3′-most internucleotide linkage i.e. a single inverted nucleoside residue which may be abasic (the nucleobase is missing or has a hydroxyl group in place thereof). Various salts, mixed salts and free acid forms are also included.

Representative United States patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos.: 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455,233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,194,599; 5,565,555; 5,527,899; 5,721,218; 5,672,697 and 5,625,050, certain of which are commonly owned with this application, and each of which is herein incorporated by reference.

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

Representative United States patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos.: 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264,562; 5,264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; 5,792,608; 5,646,269 and 5,677,439, certain of which are commonly owned with this application, and each of which is herein incorporated by reference.

Modified Sugar and Internucleoside Linkages-Mimetics

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

Preferred embodiments of the invention are oligonucleotides with phosphorothioate backbones. Also preferred are oligonucleosides with heteroatom backbones, and in particular—CH2—NH—O—CH2—, —CH2—N(CH3)—O—CH2— [known as a methylene (methylimino) or MMI backbone], —CH2—O—N(CH3)—CH2—, —CH2—N(CH3)—N(CH3)—CH2— and —O—N(CH3)—CH2—CH2— [wherein the native phosphodiester backbone is represented as —O—P—O—CH2—] of the above referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above referenced U.S. Pat. No. 5,602,240. Also preferred are oligonucleotides having morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.

Modified Sugars

Modified oligonucleotides may also contain one or more substituted sugar moieties. Preferred oligonucleotides comprise one of the following at the 2′ position: OH; F; O—, S—, or N-alkyl; O—, S—, or N-alkenyl; O—, S—or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl. Particularly preferred are O[(CH2)nO]mCH3, O(CH2)nOCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, and O(CH2)nON[(CH2)nCH3]2, where n and m are from 1 to about 10. Other preferred oligonucleotides comprise one of the following at the 2′ position: C1 to C10 lower alkyl, substituted lower alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ON2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmacokinetic properties of an oligonucleotide, or a group for improving the pharmacodynamic properties of an oligonucleotide, and other substituents having similar properties. A preferred modification includes 2′-methoxyethoxy (2′-O—CH2CH2OCH3, also known as 2′-O-(2-methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim. Acta, 1995, 78, 486-504) i.e., an alkoxyalkoxy group. A further preferred modification includes 2′-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2′-DMAOE, as described in examples hereinbelow, and 2′-dimethylamino-ethoxyethoxy (also known in the art as 2′-O-dimethyl-amino-ethoxy-ethyl or 2′-DMAEOE), i.e., 2′-O—CH2—O—CH2—N(CH3)2, also described in examples hereinbelow.

Other preferred modifications include 2′-methoxy (2′-O—CH3), 2′-aminopropoxy (2′-OCH2CH2CH2NH2), 2′-allyl (2′-CH2—CH═CH2), 2′-O-allyl (2′-O—CH2—CH═CH2) and 2′-fluoro (2′-F). The 2′-modification may be in the arabino (up) position or ribo (down) position. A preferred 2′-arabino modification is 2′-F. Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3′ position of the sugar on the 3′ terminal nucleotide or in 2′-5′ linked oligonucleotides and the 5′ position of 5′ terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative United States patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos.: 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; 5,792,747; and 5,700,920, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference in its entirety.

A further preferred modification of the sugar includes Locked Nucleic Acids (LNAs) in which the 2′-hydroxyl group is linked to the 3′ or 4′ carbon atom of the sugar ring, thereby forming a bicyclic sugar moiety. The linkage is preferably a methylene (—CH2—)n group bridging the 2′ oxygen atom and the 4′ carbon atom wherein n is 1 or 2. LNAs and preparation thereof are described in WO 98/39352 and WO 99/14226.

Natural and Modified Nucleobases

Oligonucleotides may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (—C═C—CH3) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. Further modified nucleobases include tricyclic pyrimidines such as phenoxazine cytidine(1H-pyrimido[5,4-b][1,4]benzoxazin-2(3H)-one), phenothiazine cytidine (1H-pyrimido[5,4-b][1,4]benzothiazin-2(3H)-one), G-clamps such as a substituted phenoxazine cytidine (e.g. 9-(2-aminoethoxy)-H-pyrimido[5,4-b][1,4]benzoxazin-2(3H)-one), carbazole cytidine (2H-pyrimido[4,5-b]indol-2-one), pyridoindole cytidine (H-pyrido[3′, 2′:4,5]pyrrolo[2,3-d]pyrimidin-2-one). Modified nucleobases may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. I., ed. John Wiley & Sons, 1990, those disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y. S., Chapter 15, Antisense Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B., ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. and are presently preferred base substitutions.

Representative United States patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat. No. 3,687,808, as well as U.S. Pat. Nos.: 4,845,205; 5,130,302; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,645,985; 5,830,653; 5,763,588; 6,005,096; and 5,681,941, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference, and U.S. Pat. No. 5,750,692, which is commonly owned with the instant application and also herein incorporated by reference.

Conjugates

Another modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or more moieties or conjugates which enhance the activity, cellular distribution or cellular uptake of the oligonucleotide. These moieties or conjugates can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups. Conjugate groups of the invention include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmaco-dynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers. Typical conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of this invention, include groups that improve uptake, enhance resistance to degradation, and/or strengthen sequence-specific hybridization with the target nucleic acid. Groups that enhance the pharmaco-kinetic properties, in the context of this invention, include groups that improve uptake, distribution, metabolism or excretion of the compounds of the present invention. Representative conjugate groups are disclosed in International Patent Application PCT/US92/09196, filed Oct. 23, 1992, and U.S. Pat. No. 6,287,860, the entire disclosure of which are incorporated herein by reference. Conjugate moieties include but are not limited to lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-S-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety. Oligonucleotides of the invention may also be conjugated to active drug substances, for example, aspirin, warfarin, phenylbutazone, ibuprofen, suprofen, fenbufen, ketoprofen, (S)-(+)-pranoprofen, carprofen, dansylsarcosine, 2,3,5-triiodo-benzoic acid, flufenamic acid, folinic acid, a benzothiadiazide, chlorothiazide, a diazepine, indo-methicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibacterial or an antibiotic. Oligonucleotide-drug conjugates and their preparation are described in U.S. patent application Ser. No. 09/334,130 (filed Jun. 15, 1999) which is incorporated herein by reference in its entirety.

Representative United States patents that teach the preparation of such oligonucleotide conjugates include, but are not limited to, U.S. Pat. Nos.: 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference.

Chimeric Compounds

It is not necessary for all positions in a given compound to be uniformly modified, and in fact more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an oligonucleotide.

The present invention also includes 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 compound. 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 composite structures 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, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference in its entirety.

Salts

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. The term “pharmaceutically acceptable salts” refers to physiologically and pharmaceutically acceptable salts of the compounds of the invention: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. For oligonucleotides, preferred examples of pharmaceutically acceptable salts and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety. Sodium salts are especially suitable salts of the compounds of the present invention.

G. Formulations

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. Representative United States patents that teach the preparation of such uptake, distribution and/or absorption-assisting formulations include, but are not limited to, U.S. Pat. Nos.: 5,108,921; 5,354,844; 5,416,016; 5,459,127; 5,521,291; 5,543,158; 5,547,932; 5,583,020; 5,591,721; 4,426,330; 4,534,899; 5,013,556; 5,108,921; 5,213,804; 5,227,170; 5,264,221; 5,356,633; 5,395,619; 5,416,016; 5,417,978; 5,462,854; 5,469,854; 5,512,295; 5,527,528; 5,534,259; 5,543,152; 5,556,948; 5,580,575; and 5,595,756, each of which is herein incorporated by reference.

The present invention also includes pharmaceutical compositions and formulations which include the antisense compounds of the invention. The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic and to mucous membranes including vaginal and rectal delivery), pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal), oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; or intracranial, e.g., intrathecal or intraventricular, administration. Oligonucleotides with at least one 2′-O-methoxyethyl modification are believed to be particularly useful for oral administration. Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful.

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

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

Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, foams and liposome-containing formulations. The pharmaceutical compositions and formulations of the present invention may comprise one or more penetration enhancers, carriers, excipients or other active or inactive ingredients.

Emulsions are typically heterogenous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1 μm in diameter. Emulsions may contain additional components in addition to the dispersed phases, and the active drug which may be present as a solution in either the aqueous phase, oily phase or itself as a separate phase. Microemulsions are included as an embodiment of the present invention. Emulsions and their uses are well known in the art and are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

Formulations of the present invention include liposomal formulations. As used in the present invention, the term “liposome” means a vesicle composed of amphiphilic lipids arranged in a spherical bilayer or bilayers. Liposomes are unilamellar or multilamellar vesicles which have a membrane formed from a lipophilic material and an aqueous interior that contains the composition to be delivered. Cationic liposomes are positively charged liposomes which are believed to interact with negatively charged DNA molecules to form a stable complex. Liposomes that are pH-sensitive or negatively-charged are believed to entrap DNA rather than complex with it. Both cationic and noncationic liposomes have been used to deliver DNA to cells.

Liposomes also include “sterically stabilized” liposomes, a term which, as used herein, refers to liposomes comprising one or more specialized lipids that, when incorporated into liposomes, result in enhanced circulation lifetimes relative to liposomes lacking such specialized lipids. Examples of sterically stabilized liposomes are those in which part of the vesicle-forming lipid portion of the liposome comprises one or more glycolipids or is derivatized with one or more hydrophilic polymers, such as a polyethylene glycol (PEG) moiety. Liposomes and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

The pharmaceutical formulations and compositions of the present invention may also include surfactants. The use of surfactants in drug products, formulations and in emulsions is well known in the art. Surfactants and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

In one embodiment, the present invention employs various penetration enhancers to effect the efficient delivery of nucleic acids, particularly oligonucleotides. In addition to aiding the diffusion of non-lipophilic drugs across cell membranes, penetration enhancers also enhance the permeability of lipophilic drugs. Penetration enhancers may be classified as belonging to one of five broad categories, i.e., surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants. Penetration enhancers and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety.

One of skill in the art will recognize that formulations are routinely designed according to their intended use, i.e. route of administration.

Preferred formulations for topical administration include those in which the oligonucleotides of the invention are in admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Preferred lipids and liposomes include neutral (e.g. dioleoylphosphatidyl DOPE ethanolamine, dimyristoylphosphatidyl choline DMPC, distearolyphosphatidyl choline) negative (e.g. dimyristoylphosphatidyl glycerol DMPG) and cationic (e.g. dioleoyltetramethylaminopropyl DOTAP and dioleoylphosphatidyl ethanolamine DOTMA).

For topical or other administration, oligonucleotides of the invention may be encapsulated within liposomes or may form complexes thereto, in particular to cationic liposomes. Alternatively, oligonucleotides may be complexed to lipids, in particular to cationic lipids. Preferred fatty acids and esters, pharmaceutically acceptable salts thereof, and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety. Topical formulations are described in detail in U.S. patent application Ser. No. 09/315,298 filed on May 20, 1999, which is incorporated herein by reference in its entirety.

Compositions and formulations for oral administration include powders or granules, microparticulates, nanoparticulates, suspensions or solutions in water or non-aqueous media, capsules, gel capsules, sachets, tablets or minitablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable. Preferred oral formulations are those in which oligonucleotides of the invention are administered in conjunction with one or more penetration enhancers surfactants and chelators. Preferred surfactants include fatty acids and/or esters or salts thereof, bile acids and/or salts thereof. Preferred bile acids/salts and fatty acids and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety. Also preferred are combinations of penetration enhancers, for example, fatty acids/salts in combination with bile acids/salts. A particularly preferred combination is the sodium salt of lauric acid, capric acid and UDCA. Further penetration enhancers include polyoxyethylene-9-lauryl ether, polyoxyethylene-20-cetyl ether. Oligonucleotides of the invention may be delivered orally, in granular form including sprayed dried particles, or complexed to form micro or nanoparticles. Oligonucleotide complexing agents and their uses are further described in U.S. Pat. No. 6,287,860, which is incorporated herein in its entirety. Oral formulations for oligonucleotides and their preparation are described in detail in U.S. applications Ser. No. 09/108,673 (filed Jul. 1, 1998), Ser. No. 09/315,298 (filed May 20, 1999) and Ser. No. 10/071,822, filed Feb. 8, 2002, each of which is incorporated herein by reference in their entirety.

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

Certain embodiments of the invention provide pharmaceutical compositions containing one or more oligomeric compounds and one or more other pharmaceutical agents which function by a non-antisense mechanism. Examples of such pharmaceutical agents include but are not limited to cancer chemotherapeutic drugs, anti-inflammatory drugs, anti-viral drugs, and compounds for treatment of metabolic diseases such as diabetes, high blood sugar or obesity, or cardiovascular conditions such as elevated blood cholesterol or blood pressure. Combinations of antisense compounds and other non-antisense drugs are also within the scope of this invention. Two or more combined compounds may be used together or sequentially. When used with the compounds of the invention, such pharmaceutical agents may be used individually (e.g., rosiglitazone and oligonucleotide), sequentially (e.g., 5-fluorouracil and oligonucleotide for a period of time followed by methotrexate and oligonucleotide), or in combination with one or more other treatments (e.g., 5-fluorouracil, methotrexate and oligonucleotide, or 5-fluorouracil, radiotherapy and oligonucleotide).

In another related embodiment, compositions of the invention may contain one or more antisense compounds, particularly oligonucleotides, targeted to a first nucleic acid and one or more additional antisense compounds targeted to a second nucleic acid target. Alternatively, compositions of the invention may contain two or more antisense compounds targeted to different regions of the same nucleic acid target. Numerous examples of antisense compounds are known in the art. Two or more combined compounds may be used together or sequentially.

H. Dosing

The formulation of therapeutic compositions and their subsequent administration (dosing) is believed to be within the skill of those in the art. Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual oligonucleotides, and can generally be estimated based on EC50s found to be effective in in vitro and in vivo animal models. In general, dosage is from 0.01 ug to 100 g per kg of body weight, and may be given once or more daily, weekly, monthly or yearly, or even once every 2 to 20 years. Persons of ordinary skill in the art can easily estimate repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the oligonucleotide is administered in maintenance doses, ranging from 0.01 ug to 100 g per kg of body weight, once or more daily, to once every 20 years.

While the present invention has been described with specificity in accordance with certain of its preferred embodiments, the following examples serve only to illustrate the invention and are not intended to limit the same.

EXAMPLES Example 1 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′-O-Dimethoxytrityl-thymidine intermediate for 5-methyl dC amidite, 5′-O-Dimethoxytrityl-2′-deoxy-5-methylcytidine intermediate for 5-methyl-dC amidite, 5′-O-Dimethoxytrityl-2′-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-benzoyl-5-methylcytidin-3′-O-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite (MOE 5-Me-C amidite), [5′-O-(4,4′-Dimethoxytriphenylmethyl)-2′-O-(2-methoxyethyl)-N6-benzoyladenosin-3′-O-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite (MOE A amdite), [5′-O-(4,4′-Dimethoxytriphenylmethyl)-2′-O-(2-methoxyethyl)-N4-isobutyrylguanosin-3′-yl]-2-cyanoethyl-N,N-diisopropylphosphoramidite (MOE G amidite), 2′-O-(Aminooxyethyl) nucleoside amidites and 2′-O-(dimethylaminooxy-ethyl) nucleoside amidites, 2′-(Dimethylaminooxyethoxy) nucleoside amidites, 5′-O-tert-Butyldiphenylsilyl-O2-2′-anhydro-5-methyluridine, 5′-O-tert-Butyldiphenylsilyl-2′-O-(2-hydroxyethyl)-5-methyluridine, 2′-O-([2-phthalimidoxy)ethyl]-5′-t-butyldiphenylsilyl-5-methyluridine, 5′-O-tert-butyldiphenylsilyl-2′-O-[(2-formadoximinooxy)ethyl]-5-methyluridine, 5′-O-tert-Butyldiphenylsilyl-2′-O-[N,N dimethylaminooxyethyl]-5-methyluridine, 2′-O-(dimethylaminooxyethyl)-5-methyluridine, 5′-O-DMT-2′-O-(dimethylaminooxyethyl)-5-methyluridine, 5′-O-DMT-2′-O-(2-N,N-dimethylaminooxyethyl)-5-methyluridine-3′-[(2-cyanoethyl)-N,N-diisopropylphosphoramidite], 2′-(Aminooxyethoxy) nucleoside amidites, N2-isobutyryl-6-O-diphenylcarbamoyl-2′-O-(2-ethylacetyl)-5′-O-(4,4′-dimethoxytrityl)guanosine-3′-[(2-cyanoethyl)-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.

Example 2 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 methylenecarbonylamino linked oligonucleosides, also identified as amide-3 linked oligonucleosides, and methyleneaminocarbonyl linked oligonucleosides, also identified as amide-4 linked oligonucleo-sides, 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.

Example 3 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 support-bound 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., 1998, 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 μl 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.

Example 4 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′-deoxyl-[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′-deoxyl-[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′-O-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.

Example 5

Design and Screening of Duplexed Antisense Compounds Targeting Glucagon Receptor

In accordance with the present invention, a series of nucleic acid duplexes comprising the antisense compounds of the present invention and their complements can be designed to target glucagon receptor. The nucleobase sequence of the antisense strand of the duplex comprises at least an 8-nucleobase portion of an oligonucleotide in Table 1. The ends of the strands may be modified by the addition of one or more natural or modified nucleobases to form an overhang. The sense strand of the dsRNA is then designed and synthesized as the complement of the antisense strand and may also contain modifications or additions to either terminus. For example, in one embodiment, both strands of the dsRNA duplex would be complementary over the central nucleobases, each having overhangs at one or both termini.

For example, a duplex comprising an antisense strand having the sequence CGAGAGGCGGACGGGACCG (SEQ ID NO.: 824) and having a two-nucleobase overhang of deoxythymidine(dT) would have the following structure:

  cgagaggcggacgggaccgTT Antisense Strand   ||||||||||||||||||| (SEQ ID NO. 825) TTgctctccgcctgccctggc Complement (SEQ ID NO. 826)

In another embodiment, a duplex comprising an antisense strand having the same sequence CGAGAGGCGGACGGGACCG (SEQ ID NO: 824) may be prepared with blunt ends (no single strand overhang) as shown:

cgagaggcggacgggaccg Antisense Strand ||||||||||||||||||| (SEQ ID NO: 824) gctctccgcctgccctggc Complement (SEQ ID NO: 827)

The RNA duplex can be unimolecular or biomolecular; i.e., the two strands can be part of a single molecule or may be separate molecules. RNA strands of the duplex can be synthesized by methods disclosed herein or purchased from Dharmacon Research Inc., (Lafayette, Colo.). Once synthesized, the complementary strands are annealed. The single strands are aliquoted and diluted to a concentration of 50 uM. Once diluted, 30 uL of each strand is combined with 15 uL of a 5× solution of annealing buffer. The final concentration of said buffer is 100 mM potassium acetate, 30 mM HEPES-KOH pH 7.4, and 2 mM magnesium acetate. The final volume is 75 uL. This solution is incubated for 1 minute at 90° C. and then centrifuged for 15 seconds. The tube is allowed to sit for 1 hour at 37° C. at which time the dsRNA duplexes are used in experimentation. The final concentration of the dsRNA duplex is 20 uM. This solution can be stored frozen (−20° C.) and freeze-thawed up to 5 times.

Once prepared, the duplexed antisense compounds are evaluated for their ability to modulate glucagon receptor expression.

When cells reached 80% confluency, they are treated with duplexed antisense compounds of the invention. For cells grown in 96-well plates, wells are washed once with 200 μL OPTI-MEM-1 reduced-serum medium (Gibco BRL) and then treated with 130 μL of OPTI-MEM-1 containing 12 μg/mL LIPOFECTIN (Gibco BRL) and the desired duplex antisense compound at a final concentration of 200 nM. After 5 hours of treatment, the medium is replaced with fresh medium. Cells are harvested 16 hours after treatment, at which time RNA is isolated and target reduction measured by RT-PCR.

Example 6

Oligonucleotide Isolation

After cleavage from the controlled pore glass solid support and deblocking in concentrated ammonium hydroxide at 55° C. for 12-16 hours, the oligonucleotides or oligonucleosides are recovered by precipitation out of 1 M NH4OAc with >3 volumes of ethanol. Synthesized oligonucleotides are analyzed by electrospray mass spectroscopy (molecular weight determination) and by capillary gel electrophoresis and judged to be at least 70% fall length material. The relative amounts of phosphorothioate and phosphodiester linkages obtained in the synthesis is determined by the ratio of correct molecular weight relative to the—16 amu product (+/−32 +/−48). For some studies oligonucleotides are purified by HPLC, as described by Chiang et al., J. Biol. Chem. 1991, 266, 18162-18171. Results obtained with HPLC-purified material are similar to those obtained with non-HPLC purified material.

Example 7

Oligonucleotide Synthesis—96 Well Plate Format

Oligonucleotides are synthesized via solid phase P(III) phosphoramidite chemistry on an automated synthesizer capable of assembling 96 sequences simultaneously in a 96-well format. Phosphodiester internucleotide linkages are afforded by oxidation with aqueous iodine. Phosphorothioate internucleotide linkages are generated by sulfurization utilizing 3,H-1,2 benzodithiole-3-one 1,1 dioxide (Beaucage Reagent) in anhydrous acetonitrile. Standard base-protected beta-cyanoethyl-diiso-propyl phosphoramidites are purchased from commercial vendors (e.g. PE-Applied Biosystems, Foster City, Calif., or Pharmacia, Piscataway, N.J.). Non-standard nucleosides are synthesized as per standard or patented methods. They are utilized as base protected beta-cyanoethyldiisopropyl phosphoramidites.

Oligonucleotides are cleaved from support and deprotected with concentrated NH4OH at elevated temperature (55-60° C.) for 12-16 hours and the released product then dried in vacuo. The dried product is then re-suspended in sterile water to afford a master plate from which all analytical and test plate samples are then diluted utilizing robotic pipettors.

Example 8

Oligonucleotide Analysis—96-Well Plate Format

The concentration of oligonucleotide in each well is assessed by dilution of samples and UV absorption spectroscopy. The full-length integrity of the individual products is evaluated by capillary electrophoresis (CE) in either the 96-well format (Beckman P/ACE™ MDQ) or, for individually prepared samples, on a commercial CE apparatus (e.g., Beckman P/ACE™ 5000, ABI 270). Base and backbone composition is confirmed by mass analysis of the compounds utilizing electrospray-mass spectroscopy. All assay test plates are diluted from the master plate using single and multi-channel robotic pipettors. Plates are judged to be acceptable if at least 85% of the compounds on the plate are at least 85% full length.

Example 9

Cell Culture and Oligonucleotide Treatment

The effect of antisense compounds 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. This can be routinely determined using, for example, PCR or Northern blot analysis. The following cell types are provided for illustrative purposes, but other cell types can be routinely used, provided that the target is expressed in the cell type chosen. This can be readily determined by methods routine in the art, for example Northern blot analysis, ribonuclease protection assays, or RT-PCR.

T-24 Cells:

The human transitional cell bladder carcinoma cell line T-24 is obtained from the American Type Culture Collection (ATCC) (Manassas, Va.). T-24 cells are routinely cultured in complete McCoy's 5A basal media (Invitrogen Corporation, Carlsbad, Calif.) supplemented with 10% fetal calf serum (Invitrogen Corporation, Carlsbad, Calif.), penicillin 100 units per mL, and streptomycin 100 micrograms per mL (Invitrogen Corporation, Carlsbad, Calif.). Cells are routinely passaged by trypsinization and dilution when they reached 90% confluence. Cells are seeded into 96-well plates (Falcon-Primaria #353872) at a density of 7000 cells/well for use in RT-PCR analysis.

For Northern blotting or other analysis, cells may be seeded onto 100 mm or other standard tissue culture plates and treated similarly, using appropriate volumes of medium and oligonucleotide.

A549 Cells:

The human lung carcinoma cell line A549 is obtained from the American Type Culture Collection (ATCC) (Manassas, Va.). A549 cells are routinely cultured in DMEM basal media (Invitrogen Corporation, Carlsbad, Calif.) supplemented with 10% fetal calf serum (Invitrogen Corporation, Carlsbad, Calif.), penicillin 100 units per mL, and streptomycin 100 micrograms per mL (Invitrogen Corporation, Carlsbad, Calif.). Cells are routinely passaged by trypsinization and dilution when they reach 90% confluence.

NHDF Cells:

Human neonatal dermal fibroblast (NHDF) are obtained from the Clonetics Corporation (Walkersville, Md.). NHDFs are routinely maintained in Fibroblast Growth Medium (Clonetics Corporation, Walkersville, Md.) supplemented as recommended by the supplier. Cells are maintained for up to 10 passages as recommended by the supplier.

HEK Cells:

Human embryonic keratinocytes (HEK) are obtained from the Clonetics Corporation (Walkersville, Md.). HEKs are routinely maintained in Keratinocyte Growth Medium (Clonetics Corporation, Walkersville, Md.) formulated as recommended by the supplier. Cells are routinely maintained for up to 10 passages as recommended by the supplier.

HepG2 Cells:

The human hepatoblastoma cell line HepG2 is obtained from the American Type Culture Collection (Manassas, Va.). HepG2 cells are routinely cultured in Eagle's MEM supplemented with 10% fetal calf serum, non-essential amino acids, and 1 mM sodium pyruvate (Gibco/Life Technologies, Gaithersburg, Md.). Cells are routinely passaged by trypsinization and dilution when they reach 90% confluence. Cells are seeded into 96-well plates (Falcon-Primaria #3872) at a density of 7000 cells/well for use in RT-PCR analysis.

For Northern blotting or other analyses, cells may be seeded onto 100 mm or other standard tissue culture plates and treated similarly, using appropriate volumes of medium and oligonucleotide.

Primary Mouse Hepatocytes

Primary mouse hepatocytes are prepared from CD-1 mice purchased from Charles River Labs. Primary mouse hepatocytes are routinely cultured in Hepatocyte Attachment Media (Gibco) supplemented with 10% Fetal Bovine Serum (Gibco/Life Technologies, Gaithersburg, Md.), 250 nM dexamethasone (Sigma), 10 nM bovine insulin (Sigma). Cells are seeded into 96-well plates (Falcon-Primaria #3872) at a density of 10000 cells/well for use in RT-PCR analysis.

For Northern blotting or other analyses, cells may be seeded onto 100 mm or other standard tissue culture plates and treated similarly, using appropriate volumes of medium and oligonucleotide.

Treatment with Antisense Compounds:

When cells reached 65-75% confluency, they are treated with oligonucleotide. For cells grown in 96-well plates, wells are washed once with 100 μL OPTI-MEM™-1 reduced-serum medium (Invitrogen Corporation, Carlsbad, Calif.) and then treated with 130 μL of OPTI-MEM™-1 containing 3.75 μg/mL LIPOFECTIN™ (Invitrogen Corporation, Carlsbad, Calif.) and the desired concentration of oligonucleotide. Cells are treated and data are obtained in triplicate. After 4-7 hours of treatment at 37° C., the medium is replaced with fresh medium. Cells are harvested 16-24 hours after oligonucleotide treatment.

The concentration of oligonucleotide used varies from cell line to cell line. To determine the optimal oligonucleotide concentration for a particular cell line, the cells are treated with a positive control oligonucleotide at a range of concentrations. For human cells the positive control oligonucleotide is selected from either ISIS 13920 (TCCGTCATCGCTCCTCAGGG, SEQ ID NO: 1) which is targeted to human H-ras, or ISIS 18078, (GTGCGCGCGAGCCCGAAATC, SEQ ID NO: 2) which is targeted to human Jun-N-terminal kinase-2 (JNK2). Both controls are 2′-O-methoxyethyl gapmers (2′-O-methoxyethyls shown in bold) with a phosphorothioate backbone. For mouse or rat cells the positive control oligonucleotide is ISIS 15770, ATGCATTCTGCCCCCAAGGA, SEQ ID NO: 3, a 2′-O-methoxyethyl gapmer (2′-O-methoxyethyls shown in bold) with a phosphorothioate backbone which is targeted to both mouse and rat c-raf. The concentration of positive control oligonucleotide that results in 80% inhibition of c-H-ras (for ISIS 13920), JNK2 (for ISIS 18078) or c-raf (for ISIS 15770) mRNA is then utilized as the screening concentration for new oligonucleotides in subsequent experiments for that cell line. If 80% inhibition is not achieved, the lowest concentration of positive control oligonucleotide that results in 60% inhibition of c-H-ras, JNK2 or c-raf mRNA is then utilized as the oligonucleotide screening concentration in subsequent experiments for that cell line. If 60% inhibition is not achieved, that particular cell line is deemed as unsuitable for oligonucleotide transfection experiments. The concentrations of antisense oligonucleotides used herein are from 50 nM to 300 nM.

Example 10

Analysis of Oligonucleotide Inhibition of Glucagon Receptor Expression

Antisense modulation of glucagon receptor expression can be assayed in a variety of ways known in the art. For example, glucagon receptor mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR (RT-PCR). Real-time quantitative PCR is presently preferred. RNA analysis can be performed on total cellular RNA or poly(A)+mRNA. The preferred method of RNA analysis of the present invention is the use of total cellular RNA as described in other examples herein. Methods of RNA isolation are well known in the art. Northern blot analysis is also routine in the art. Real-time quantitative (PCR) can be conveniently accomplished using the commercially available ABI PRISM™ 7600, 7700, or 7900 Sequence Detection System, available from PE-Applied Biosystems, Foster City, Calif. and used according to manufacturer's instructions.

Protein levels of glucagon receptor can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), enzyme-linked immunosorbent assay (ELISA) or fluorescence-activated cell sorting (FACS). Antibodies directed to glucagon receptor 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 monoclonal or polyclonal antibody generation methods well known in the art.

Example 11

Design of Phenotypic Assays and in vivo Studies for the Use of Glucagon Receptor Inhibitors

Phenotypic Assays

Once glucagon receptor inhibitors have been identified by the methods disclosed herein, the compounds are further investigated in one or more phenotypic assays, each having measurable endpoints predictive of efficacy in the treatment of a particular disease state or condition.

  • Phenotypic assays, kits and reagents for their use are well known to those skilled in the art and are herein used to investigate the role and/or association of glucagon receptor in health and disease. Representative phenotypic assays, which can be purchased from any one of several commercial vendors, include those for determining cell viability, cytotoxicity, proliferation or cell survival (Molecular Probes, Eugene, Oreg.; PerkinElmer, Boston, Mass.), protein-based assays including enzymatic assays (Panvera, LLC, Madison, Wis.; BD Biosciences, Franklin Lakes, N.J.;
  • Oncogene Research Products, San Diego, Calif.), cell regulation, signal transduction, inflammation, oxidative processes and apoptosis (Assay Designs Inc., Ann Arbor, Mich.), triglyceride accumulation (Sigrna-Aldrich, St. Louis, Mo.), angiogenesis assays, tube formation assays, cytokine and hormone assays and metabolic assays (Chemicon International Inc., Temecula, Calif.; Amersham Biosciences, Piscataway, N.J.).

In one non-limiting example, cells determined to be appropriate for a particular phenotypic assay (i.e., MCF-7 cells selected for breast cancer studies; adipocytes for obesity studies) are treated with glucagon receptor inhibitors identified from the in vitro studies as well as control compounds at optimal concentrations which are determined by the methods described above. At the end of the treatment period, treated and untreated cells are analyzed by one or more methods specific for the assay to determine phenotypic outcomes and endpoints.

Phenotypic endpoints include changes in cell morphology over time or treatment dose as well as changes in levels of cellular components such as proteins, lipids, nucleic acids, hormones, saccharides or metals. Measurements of cellular status which include pH, stage of the cell cycle, intake or excretion of biological indicators by the cell, are also endpoints of interest.

Analysis of the geneotype of the cell (measurement of the expression of one or more of the genes of the cell) after treatment is also used as an indicator of the efficacy or potency of the glucagon receptor inhibitors. Hallmark genes, or those genes suspected to be associated with a specific disease state, condition, or phenotype, are measured in both treated and untreated cells.

In vivo Studies

The individual subjects of the in vivo studies described herein are warm-blooded vertebrate animals, which includes humans.

The clinical trial is subjected to rigorous controls to ensure that individuals are not unnecessarily put at risk and that they are fully informed about their role in the study. To account for the psychological effects of receiving treatments, volunteers are randomly given placebo or glucagon receptor inhibitor. Furthermore, to prevent the doctors from being biased in treatments, they may not be informed as to whether the medication they are administering is a glucagon receptor inhibitor or a placebo. Using this randomization approach, each volunteer has the same chance of being given either the new treatment or the placebo.

Volunteers may receive either the glucagon receptor inhibitor or placebo for eight week period with biological parameters associated with the indicated disease state or condition being measured at the beginning (baseline measurements before any treatment), end (after the final treatment), and at regular intervals during the study period. Such measurements may include the levels of nucleic acid molecules encoding glucagon receptor or glucagon receptor protein levels in body fluids, tissues or organs compared to pre-treatment levels. Other measurements may include, but are not limited to, indices of the disease state or condition being treated, body weight, blood pressure, serum titers of pharmacologic indicators of disease or toxicity as well as ADME (absorption, distribution, metabolism and excretion) measurements.

Information recorded for each patient may include age (years), gender, height (cm), family history of disease state or condition (yes/no), motivation rating (some/moderate/great) and number and type of previous treatment regimens for the indicated disease or condition.

Volunteers taking part in this study are healthy adults (age 18 to 65 years) and, typically, roughly an equal number of males and females participate in the study. Volunteers with certain characteristics are equally distributed for placebo and glucagon receptor inhibitor treatment. In general, the volunteers treated with placebo have little or no response to treatment, whereas the volunteers treated with the glucagon receptor inhibitor show positive trends in their disease state or condition index at the conclusion of the study.

One of ordinary skill will know how to conduct an appropriate clinical trial and will recognize that this is just one of many protocols which may be appropriately used.

Example 12

RNA Isolation

Poly(A)+mRNA Isolation

Poly(A)+mRNA was isolated according to Miura et al., (Clin. Chem., 1996, 42, 1758-1764). Other methods for poly(A)+mRNA isolation are routine in the art. Briefly, for cells grown on 96-well plates, growth medium was removed from the cells and each well was washed with 200 μL cold PBS. 60 μL lysis buffer (10 mM Tris-HCl, pH 7.6, 1 mM EDTA, 0.5 M NaCl, 0.5% NP-40, 20 mM vanadyl-ribonucleoside complex) was added to each well, the plate was gently agitated and then incubated at room temperature for five minutes. 55 μL of lysate was transferred to Oligo d(T) coated 96-well plates (AGCT Inc., Irvine Calif.). Plates were incubated for 60 minutes at room temperature, washed 3 times with 200 μL of wash buffer (10 mM Tris-HCl pH 7.6, 1 mM EDTA, 0.3 M NaCl). After the final wash, the plate was blotted on paper towels to remove excess wash buffer and then air-dried for 5 minutes. 60 μL of elution buffer (5 mM Tris-HCl pH 7.6), preheated to 70° C., was added to each well, the plate was incubated on a 90° C. hot plate for 5 minutes, and the eluate was then transferred to a fresh 96-well plate.

Cells grown on 100 mm or other standard plates may be treated similarly, using appropriate volumes of all solutions.

Total RNA Isolation

Total RNA was isolated using an RNEASY 96™ kit and buffers purchased from Qiagen Inc. (Valencia, Calif.) following the manufacturer's recommended procedures. Briefly, for cells grown on 96-well plates, growth medium was removed from the cells and each well was washed with 200 μL cold PBS. 150 μL Buffer RLT was added to each well and the plate vigorously agitated for 20 seconds. 150 μL of 70% ethanol was then added to each well and the contents mixed by pipetting three times up and down. The samples were then transferred to the RNEASY 96™ well plate attached to a QIAVAC™ manifold fitted with a waste collection tray and attached to a vacuum source. Vacuum was applied for 1 minute. 500 μL of Buffer RW1 was added to each well of the RNEASY 96™ plate and incubated for 15 minutes and the vacuum was again applied for 1 minute. An additional 500 μL of Buffer RW1 was added to each well of the RNEASY 96™ plate and the vacuum was applied for 2 minutes. 1 mL of Buffer RPE was then added to each well of the RNEASY 96™ plate and the vacuum applied for a period of 90 seconds. The Buffer RPE wash was then repeated and the vacuum was applied for an additional 3 minutes. The plate was then removed from the QIAVAC™ manifold and blotted dry on paper towels. The plate was then re-attached to the QIAVAC™ manifold fitted with a collection tube rack containing 1.2 mL collection tubes. RNA was then eluted by pipetting 140 μL of RNAse free water into each well, incubating 1 minute, and then applying the vacuum for 3 minutes.

The repetitive pipetting and elution steps may be automated using a QIAGEN Bio-Robot 9604 (Qiagen, Inc., Valencia Calif.). Essentially, after lysing of the cells on the culture plate, the plate is transferred to the robot deck where the pipetting, DNase treatment and elution steps are carried out.

Example 13

Real-time Quantitative PCR Analysis of Glucagon Receptor mRNA Levels

Quantitation of glucagon receptor 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. This is a closed-tube, non-gel-based, fluorescence detection system which allows high-throughput quantitation of polymerase chain reaction (PCR) products in real-time. As opposed to standard PCR in which amplification products are quantitated after the PCR is completed, products in real-time quantitative PCR are quantitated as they accumulate. This is accomplished by including in the PCR reaction an oligonucleotide probe that anneals specifically between the forward and reverse PCR primers, and contains two fluorescent dyes. A reporter dye (e.g., FAM or JOE, obtained from either PE-Applied Biosystems, Foster City, Calif., Operon Technologies Inc., Alameda, Calif. or Integrated DNA Technologies Inc., Coralville, Iowa) is attached to the 5′ end of the probe and a quencher dye (e.g., TAMRA, obtained from either PE-Applied Biosystems, Foster City, Calif., Operon Technologies Inc., Alameda, Calif. or Integrated DNA Technologies Inc., Coralville, Iowa) is attached to the 3′ end of the probe. When the probe and dyes are intact, reporter dye emission is quenched by the proximity of the 3′ quencher dye. During amplification, annealing of the probe to the target sequence creates a substrate that can be cleaved by the 5′-exonuclease activity of Taq polymerase. During the extension phase of the PCR amplification cycle, cleavage of the probe by Taq polymerase releases the reporter dye from the remainder of the probe (and hence from the quencher moiety) and a sequence-specific fluorescent signal is generated. With each cycle, additional reporter dye molecules are cleaved from their respective probes, and the fluorescence intensity is monitored at regular intervals by laser optics built into the ABI PRISM™ Sequence Detection System. In each assay, a series of parallel reactions containing serial dilutions of mRNA from untreated control samples generates a standard curve that is used to quantitate the percent inhibition after antisense oligonucleotide treatment of test samples.

Prior to quantitative PCR analysis, primer-probe sets specific to the target gene being measured are evaluated for their ability to be “multiplexed” with a GAPDH amplification reaction. In multiplexing, both the target gene and the internal standard gene GAPDH are amplified concurrently in a single sample. In this analysis, mRNA isolated from untreated cells is serially diluted. Each dilution is amplified in the presence of primer-probe sets specific for GAPDH only, target gene only (“single-plexing”), or both (multiplexing). Following PCR amplification, standard curves of GAPDH and target mRNA signal as a function of dilution are generated from both the single-plexed and multiplexed samples. If both the slope and correlation coefficient of the GAPDH and target signals generated from the multiplexed samples fall within 10% of their corresponding values generated from the single-plexed samples, the primer-probe set specific for that target is deemed multiplexable. Other methods of PCR are also known in the art.

PCR reagents were obtained from Invitrogen Corporation, (Carlsbad, Calif.). RT-PCR reactions were carried out 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).

Gene target quantities obtained by real time RT-PCR are 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.). GAPDH expression is quantified by real time RT-PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using RiboGreen™ RNA quantification reagent (Molecular Probes, Inc. Eugene, Oreg.). Methods of RNA quantification by RiboGreen™ are taught in Jones, L. J., et al, (Analytical Biochemistry, 1998, 265, 368-374).

In this assay, 170 μL of RiboGreen™ working reagent (RiboGreen™ reagent diluted 1:350 in 10 mM Tris-HCl, 1 mM EDTA, pH 7.5) is pipetted into a 96-well plate containing 30 μL purified, cellular RNA. The plate is read in a CytoFluor 4000 (PE Applied Biosystems) with excitation at 485 nm and emission at 530 nm.

Probes and primers to human glucagon receptor were designed to hybridize to a human glucagon receptor sequence, using published sequence information (GenBank accession number NM000160.1, incorporated herein as SEQ ID NO:4). For human glucagon receptor the PCR primers were:

  • forward primer: GACACCCCCGCCAATACC (SEQ ID NO: 5)
  • reverse primer: CCGCATCTCTTGAACACGAA (SEQ ID NO: 6) and the PCR probe was:
  • FAM-TTGGCACCACAAAGT-TAMRA (SEQ ID NO: 7) where FAM is the fluorescent dye and TAMRA is the quencher dye. For human GAPDH the PCR primers were:
  • forward primer: GAAGGTGAAGGTCGGAGTC(SEQ ID NO:8)
  • reverse primer: GAAGATGGTGATGGGATTTC (SEQ ID NO:9) and the PCR probe was: 5′
  • JOE-CAAGCTTCCCGTTCTCAGCC-TAMRA 3′ (SEQ ID NO: 10) where JOE is the fluorescent reporter dye and TAMRA is the quencher dye.

Probes and primers to mouse glucagon receptor were designed to hybridize to a mouse glucagon receptor sequence, using published sequence information (GenBank accession number NM008101.1, incorporated herein as SEQ ID NO: 11). For mouse glucagon receptor the PCR primers were:

  • forward primer: ATTTCCTGCCCCTGGTACCT (SEQ ID NO:12)
  • reverse primer: CGGGCCCACACCTCTTG (SEQ ID NO: 13) and the PCR probe was: FAM-CCACAAAGTGCAGCACCGCCTAGTGT-TAMRA
  • (SEQ ID NO: 14) where FAM is the fluorescent reporter dye and TAMRA is the quencher dye.
  • For mouse GAPDH the PCR primers were:
  • forward primer: GGCAAATTCAACGGCACAGT(SEQ ID NO:15)
  • reverse primer: GGGTCTCGCTCCTGGAAGAT(SEQ ID NO:16) and the PCR probe was: 5′
  • JOE-AAGGCCGAGAATGGGAAGCTTGTCATC- TAMRA 3′ (SEQ ID NO: 17) where JOE is the fluorescent reporter dye and TAMRA is the quencher dye.

Example 14

Northern Blot Analysis of Glucagon Receptor MRNA Levels

Eighteen hours after antisense treatment, cell monolayers were washed twice with cold PBS and lysed in 1 mL RNAZOL™ (TEL-TEST “B” Inc., Friendswood, Tex.). Total RNA was prepared following manufacturer's recommended protocols. Twenty micrograms of total RNA was fractionated by electrophoresis through 1.2% agarose gels containing 1.1% formaldehyde using a MOPS buffer system (AMRESCO, Inc. Solon, Ohio). RNA was transferred from the gel to HYBOND™ -N+ nylon membranes (Amersham Pharmacia Biotech, Piscataway, N.J.) by overnight capillary transfer using a Northern/Southern Transfer buffer system (TEL-TEST “B” Inc., Friendswood, Tex.). RNA transfer was confirmed by UV visualization. Membranes were fixed by UV cross-linking using a STRATALINKER™ UV Crosslinker 2400 (Stratagene, Inc, La Jolla, Calif.) and then probed using QUICKHYB™ hybridization solution (Stratagene, La Jolla, Calif.) using manufacturer's recommendations for stringent conditions.

To detect human glucagon receptor, a human glucagon receptor specific probe was prepared by PCR using the forward primer GACACCCCCGCCAATACC (SEQ ID NO: 5) and the reverse primer CCGCATCTCTTGAACACGAA (SEQ ID NO: 6). To normalize for variations in loading and transfer efficiency membranes were stripped and probed for human glyceraldehyde-3-phosphate dehydrogenase (GAPDH) RNA (Clontech, Palo Alto, Calif.).

To detect mouse glucagon receptor, a mouse glucagon receptor specific probe was prepared by PCR using the forward primer ATTTCCTGCCCCTGGTACCT (SEQ ID NO: 12) and the reverse primer CGGGCCCACACCTCTTG (SEQ ID NO: 13). To normalize for variations in loading and transfer efficiency membranes were stripped and probed for mouse glyceraldehyde-3-phosphate dehydrogenase (GAPDH) RNA (Clontech, Palo Alto, Calif.).

Hybridized membranes were visualized and quantitated using a PHOSPHORIMAGER™ and IMAGEQUANT™ Software V3.3 (Molecular Dynamics, Sunnyvale, Calif.). Data was normalized to GAPDH levels in untreated controls.

Example 15

Antisense Inhibition of Human Glucagon Receptor Expression by Chimeric Phosphorothioate Oligonucleotides Having 2′-MOE Wings and a Deoxy Gap

In accordance with the present invention, a series of antisense compounds were designed to target different regions of the human glucagon receptor RNA, using published sequences (GenBank accession number NM000160.1, incorporated herein as SEQ ID NO: 4, a concatenation of three contigs from GenBank accession number AC069004.2, incorporated herein as SEQ ID NO: 18, and GenBank accession number AJ245489.1, incorporated herein as SEQ ID NO: 19). The compounds are shown in Table 1. “Target site” indicates the first (5′-most) nucleotide number on the particular target sequence to which the compound binds. All compounds in Table 1 are chimeric oligonucleotides (“gapmers”) 20 nucleotides in length, composed of a central “gap” region consisting of ten 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′ directions) by five-nucleotide “wings”. The wings are composed of 2′-methoxyethyl (2′-MOE)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P═S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines. The compounds were analyzed for their effect on human glucagon receptor mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments in which HepG2 cells were treated with the antisense oligonucleotides of the present invention. The positive control for each datapoint is identified in the table by sequence ID number. If present, “N.D.” indicates “no data”.

TABLE 1 Inhibition of human glucagon receptor mRNA levels by chimeric phosphorothioate oligonucleotides having 2′-MOE wings and a deoxy gap TARGET TARGET SEQ ID ISIS # REGION SEQ ID NO SITE SEQUENCE % INHIB NO 310462 Coding 4 560 ccgcatctcttgaacacgaa 61 20 299881 5′UTR 4 97 ttgagcctcagggcccgcgc 56 21 299882 5′UTR 4 121 gtgtcctcccctgaagctgc 68 22 299883 5′UTR 4 163 gagtggcagagcagcagagc 38 23 299884 5′UTR 4 192 tgtgtgtgtacgctcctccg 76 24 299885 5′UTR 4 198 tcctggtgtgtgtgtacgct 67 25 299886 5′UTR 4 205 aatgcagtcctggtgtgtgt 30 26 299887 5′UTR 4 254 ctgggcagctagctgcctcc 38 27 299888 Start Codon 4 263 ggcatgcctctgggcagcta 73 28 299889 Coding 4 462 ccagcaggaatacttgtcga 43 29 299890 Coding 4 328 ggacctgtggctggcaggcc 72 30 299891 Coding 4 350 aagtccatcacctgagcgga 39 31 299892 Coding 4 361 tctcaaacaggaagtccatc 37 32 299893 Coding 4 366 ccacttctcaaacaggaagt 24 33 299894 Coding 4 386 cactggtcaccgtagagctt 31 34 299895 Coding 4 391 ggtgacactggtcaccgtag 40 35 299896 Coding 4 431 cacaccagctccgtgggagg 35 36 299897 Coding 4 442 aggttctgttgcacaccagc 76 37 299898 Coding 4 453 atacttgtcgaaggttctgt 28 38 299899 Coding 4 539 cggtgttgcactttgtggtg 85 39 299900 5′UTR 19 546 ccctggcagagacagcggca 79 40 299901 Coding 4 552 cttgaacacgaagcggtgtt 83 41 299902 Coding 4 564 gggcccgcatctcttgaaca 87 42 299903 intron:exon 18 15279 cttctgcgagttacagtggc 58 43 junction 299904 Coding 4 651 ctggacctcaatctcctcgc 43 44 299905 Coding 4 656 tccttctggacctcaatctc 5 45 299906 Coding 4 663 ggccacctccttctggacct 80 46 299907 Coding 4 669 catcttggccacctccttct 39 47 299908 Coding 4 681 gaagctgctgtacatcttgg 71 48 299909 Coding 4 751 cccccaggatggccaaggcg 69 49 299910 Coding 4 830 acggagctggctttcagcac 49 50 299911 Coding 4 866 ctgtagcgggtcctgagcag 48 51 299912 Coding 4 872 ttctggctgtagcgggtcct 54 52 299913 Coding 4 879 gccaattttctggctgtagc 61 53 299914 Coding 4 889 tgaggtcgtcgccaattttc 56 54 299915 Coding 4 898 tgctgacactgaggtcgtcg 63 55 299916 Coding 4 904 gccaggtgctgacactgagg 67 56 299917 Coding 4 966 cacgatgccatattgcatga 59 57 299918 Coding 4 1028 gtggccaggcccagcaggtt 52 58 299919 Coding 4 1122 cagacacttgaccactgccc 40 59 299920 Coding 4 1182 ccgcaggatccaccagaagc 46 60 299921 Coding 4 1210 tgatcaggatggccaggaag 42 61 299922 Coding 4 1228 ggacgaagatgaagaagttg 44 62 299923 Coding 4 1259 cgcagcttggccacgagcag 8 63 299924 Coding 4 1274 tgcatctgccgtgcccgcag 58 64 299925 Coding 4 1291 acttgtagtctgtgtggtgc 34 65 299926 Coding 4 1415 aggtcgaagaagagcttggc 38 66 299927 Coding 4 1528 gcactttgcccaggcgccag 78 67 299928 Coding 4 1539 ctcctcccatagcactttgc 40 68 299929 Coding 4 1608 aaactgcagctccttgctgg 46 69 299930 Coding 4 1636 atgaatcctggctgccacca 70 70 299931 Coding 4 1670 ctagggaggccaccagccaa 49 71 299932 Coding 4 1681 tctcagccaatctagggagg 63 72 299933 Stop Codon 4 1704 tcccagcagggttcagaagg 30 73 299934 5′UTR 19 1747 ttcctgcaggtgacccaatg 50 74 299935 3′UTR 4 1841 tctcgcagacagccacactg 43 75 299936 3′UTR 4 1854 agaggaggcccaatctcgca 79 76 299937 3′UTR 4 1881 tgcaccagggacaaggcagg 0 77 299938 3′UTR 4 1901 tggactcctctgctcacctc 58 78 299939 3′UTR 4 1938 tggcacgcagttcacggcac 54 79 299940 3′UTR 4 1969 acatgggacgtgccgacata 63 80 299941 3′UTR 4 1978 tttccatgcacatgggacgt 63 81 299942 3′UTR 4 1989 gttggaggacatttccatgc 79 82 299943 3′UTR 4 2015 cacggtgaccacttgagctc 18 83 299944 intron 18 11002 agatgtccgtgtttgtcagc 9 84 299945 intron 18 11557 taataactttttaaagaagg 17 85 299946 intron 18 12295 tactacgttgctcgggctgg 23 86 299947 intron 18 14121 agctctgtggctcagttacc 74 87 299948 intron:exon 18 15467 gtgcagcttgctgtggcaca 47 88 junction 299949 intron 18 16094 cagcaaccgcttggtacagg 100 89 299950 intron:exon 18 17017 agaagttgatctgtgtgaga 29 90 junction 299951 intron:exon 18 17456 ccagcaggccctggagagac 53 91 junction 304471 5′UTR 4 100 cctttgagcctcagggcccg 42 92 304472 5′UTR 4 103 gcccctttgagcctcagggc 25 93 304473 5′UTR 4 167 agctgagtggcagagcagca 76 94 304474 5′UTR 4 169 gcagctgagtggcagagcag 75 95 304475 5′UTR 4 190 tgtgtgtacgctcctccgag 73 96 304476 5′UTR 4 194 ggtgtgtgtgtacgctcctc 72 97 304477 5′UTR 4 196 ctggtgtgtgtgtacgctcc 71 98 304478 5′UTR 4 209 gggcaatgcagtcctggtgt 65 99 304479 5′UTR 4 246 ctagctgcctcccacatctg 54 100 304480 5′UTR 4 249 cagctagctgcctcccacat 85 101 304481 5′UTR 4 257 cctctgggcagctagctgcc 44 102 304482 Start Codon 4 262 gcatgcctctgggcagctag 62 103 304483 Coding 4 325 cctgtggctggcaggccagc 68 104 304484 Coding 4 368 ttccacttctcaaacaggaa 24 105 304485 Coding 4 370 gcttccacttctcaaacagg 49 106 304486 Coding 4 375 gtagagcttccacttctcaa 41 107 304487 Coding 4 376 cgtagagcttccacttctca 38 108 304488 Coding 4 395 ttgtggtgacactggtcacc 24 109 304489 Coding 4 407 agcaggctcaggttgtggtg 52 110 304490 Coding 4 534 ttgcactttgtggtgccaag 61 111 304491 Coding 4 535 gttgcactttgtggtgccaa 57 112 304492 Coding 4 536 tgttgcactttgtggtgcca 67 113 304493 Coding 4 537 gtgttgcactttgtggtgcc 75 114 304494 Coding 4 563 ggcccgcatctcttgaacac 87 115 304495 Coding 4 567 gtcgggcccgcatctcttga 81 116 304496 Coding 4 617 tgggaggcatcacgccaagg 60 117 304497 Coding 4 627 catctggcactgggaggcat 48 118 304498 Coding 4 666 cttggccacctccttctgga 74 119 304499 Coding 4 671 tacatcttggccacctcctt 24 120 304500 Coding 4 685 cctggaagctgctgtacatc 71 121 304501 Coding 4 795 attcgcgtggatggcattgc 53 122 304502 Coding 4 848 agcccatcaatgaccagcac 31 123 304503 Coding 4 861 gcgggtcctgagcagcccat 42 124 304504 Coding 4 886 ggtcgtcgccaattttctgg 50 125 304505 Coding 4 893 acactgaggtcgtcgccaat 22 126 304506 Coding 4 900 ggtgctgacactgaggtcgt 60 127 304507 Coding 4 962 atgccatattgcatgaacac 27 128 304508 Coding 4 1032 gagggtggccaggcccagca 56 129 304509 Coding 4 1124 aacagacacttgaccactgc 13 130 304510 Coding 4 1125 gaacagacacttgaccactg 8 131 304511 Coding 4 1158 gttgtcattgctggtccagc 65 132 304512 Coding 4 1168 agaagcccatgttgtcattg 44 133 304513 Coding 4 1187 gggaaccgcaggatccacca 42 134 304514 Coding 4 1230 gcggacgaagatgaagaagt 54 135 304515 Coding 4 1638 agatgaatcctggctgccac 53 136 304516 3′UTR 4 1727 ccagagtccagccctagctg 41 137 304517 3′UTR 4 1732 gggtgccagagtccagccct 48 138 304518 3′UTR 4 1735 tctgggtgccagagtccagc 65 139 304519 3′UTR 4 1736 ctctgggtgccagagtccag 75 140 304520 3′UTR 4 1737 cctctgggtgccagagtcca 74 141 304521 3′UTR 4 1740 acgcctctgggtgccagagt 55 142 304522 3′UTR 4 1760 cagttctgggttgtccagcg 52 143 304523 3′UTR 4 1849 aggcccaatctcgcagacag 74 144 304524 3′UTR 4 1850 gaggcccaatctcgcagaca 80 145 304525 3′UTR 4 1856 ggagaggaggcccaatctcg 66 146 304526 3′UTR 4 1861 tgcagggagaggaggcccaa 63 147 304527 3′UTR 4 1883 tctgcaccagggacaaggca 50 148 304528 3′UTR 4 1891 tgctcacctctgcaccaggg 66 149 304529 3′UTR 4 1893 tctgctcacctctgcaccag 32 150 304530 3′UTR 4 1899 gactcctctgctcacctctg 31 151 304531 3′UTR 4 1905 gccctggactcctctgctca 69 152 304532 3′UTR 4 1932 gcagttcacggcacagcccc 53 153 304533 3′UTR 4 1933 cgcagttcacggcacagccc 30 154 304534 3′UTR 4 1945 gggacactggcacgcagttc 61 155 304535 3′UTR 4 1971 gcacatgggacgtgccgaca 83 156 304536 3′UTR 4 1984 aggacatttccatgcacatg 61 157 304537 3′UTR 4 1986 ggaggacatttccatgcaca 69 158 304538 3′UTR 4 1999 gctctttattgttggaggac 66 159 304539 3′UTR 4 2001 gagctctttattgttggagg 68 160 304540 3′UTR 4 2008 accacttgagctctttattg 40 161 304541 intron 18 3174 ggcagttttggcgtccccag 67 162 304542 intron 18 6670 gagcttcctgcctcttcacg 39 163 304543 intron 18 7544 ggataggatgtgcgtgtcta 42 164 304544 intron 18 7975 ctctctgcctccgatttctt 12 165 304545 intron:exon 18 14888 acaccagctctgcagggtag 75 166 junction 304546 intron:exon 18 15285 cacctccttctgcgagttac 33 167 junction 310441 Start Codon 4 258 gcctctgggcagctagctgc 64 168 310442 Coding 4 317 tggcaggccagcagcagcag 87 169 310443 Coding 4 321 tggctggcaggccagcagca 88 170 310444 Coding 4 347 tccatcacctgagcggaggg 55 171 310445 Coding 4 351 gaagtccatcacctgagcgg 36 172 310446 Coding 4 355 acaggaagtccatcacctga 28 173 310447 Coding 4 365 cacttctcaaacaggaagtc 59 174 310448 Coding 4 389 tgacactggtcaccgtagag 18 175 310449 Coding 4 393 gtggtgacactggtcaccgt 12 176 310450 Coding 4 397 ggttgtggtgacactggtca 72 177 310451 Coding 4 403 ggctcaggttgtggtgacac 62 178 310452 Coding 4 452 tacttgtcgaaggttctgtt 44 179 310453 Coding 4 458 caggaatacttgtcgaaggt 40 180 310454 Coding 4 493 tgttggccgtggtattggcg 90 181 310455 Coding 4 497 gagatgttggccgtggtatt 87 182 310456 Coding 4 500 caggagatgttggccgtggt 95 183 310457 Coding 4 532 gcactttgtggtgccaaggc 96 184 310458 Coding 4 540 gcggtgttgcactttgtggt 92 185 310459 Coding 4 544 cgaagcggtgttgcactttg 50 186 310460 Coding 4 548 aacacgaagcggtgttgcac 87 187 310461 Coding 4 556 atctcttgaacacgaagcgg 65 188 310463 Coding 4 588 gggtccacgcacccactgac 50 189 310464 Coding 4 606 acgccaaggctgcccccggg 71 190 310465 Coding 4 660 cacctccttctggacctcaa 31 191 310466 Coding 4 683 tggaagctgctgtacatctt 57 192 310467 Coding 4 687 cacctggaagctgctgtaca 60 193 310468 Coding 4 691 acatcacctggaagctgctg 73 194 310469 Coding 4 695 gtgtacatcacctggaagct 79 195 310470 Coding 4 720 ccccagggacaggctgtagc 86 196 310471 Coding 4 723 ggcccccagggacaggctgt 62 197 310472 Coding 4 860 cgggtcctgagcagcccatc 48 198 310473 Coding 4 864 gtagcgggtcctgagcagcc 58 199 310474 Coding 4 868 ggctgtagcgggtcctgagc 48 200 310475 Coding 4 919 ccgctccatcactgagccag 52 201 310476 Coding 4 923 gccaccgctccatcactgag 41 202 310477 Coding 4 951 catgaacaccgcggccacac 63 203 310478 Coding 4 955 attgcatgaacaccgcggcc 76 204 310479 Coding 4 960 gccatattgcatgaacaccg 66 205 310480 Coding 4 1019 cccagcaggttgtgcaggta 58 206 310481 Coding 4 1025 gccaggcccagcaggttgtg 72 207 310482 Coding 4 1029 ggtggccaggcccagcaggt 83 208 310483 Coding 4 1055 aggctgaagaagctcctctc 71 209 310484 Coding 4 1059 gtagaggctgaagaagctcc 46 210 310485 Coding 4 1063 ccaggtagaggctgaagaag 25 211 310486 Coding 4 1068 gatgcccaggtagaggctga 51 212 310487 Coding 4 1072 agccgatgcccaggtagagg 70 213 310488 Coding 4 1156 tgtcattgctggtccagcac 83 214 310489 Coding 4 1160 atgttgtcattgctggtcca 53 215 310490 Coding 4 1167 gaagcccatgttgtcattgc 45 216 310491 Coding 4 1173 ccaccagaagcccatgttgt 50 2i7 310492 Coding 4 1176 gatccaccagaagcccatgt 53 218 310493 Coding 4 1185 gaaccgcaggatccaccaga 47 219 310494 Coding 4 1206 caggatggccaggaagacgg 39 220 310495 Coding 4 1209 gatcaggatggccaggaaga 67 221 310496 Coding 4 1219 tgaagaagttgatcaggatg 10 222 310497 Coding 4 1222 agatgaagaagttgatcagg 20 223 310498 Coding 4 1287 gtagtctgtgtggtgcatct 35 224 310499 Coding 4 1290 cttgtagtctgtgtggtgca 63 225 310500 Coding 4 1293 gaacttgtagtctgtgtggt 27 226 310501 Coding 4 1414 ggtcgaagaagagcttggcg 46 227 310502 Coding 4 1417 agaggtcgaagaagagcttg 26 228 310503 Coding 4 1423 tgaggaagaggtcgaagaag 17 229 310504 Coding 4 1669 tagggaggccaccagccaag 53 230 315163 Coding 4 686 acctggaagctgctgtacat 75 231 315164 Coding 4 409 gcagcaggctcaggttgtgg 24 232 315165 Coding 4 1424 ctgaggaagaggtcgaagaa 42 233 315166 Coding 4 398 aggttgtggtgacactggtc 34 234 315167 Coding 4 1212 gttgatcaggatggccagga 47 235 315168 Coding 4 1062 caggtagaggctgaagaagc 40 236 315169 Coding 4 559 cgcatctcttgaacacgaag 48 237 315170 Coding 4 543 gaagcggtgttgcactttgt 61 238 315171 Coding 4 454 aatacttgtcgaaggttctg 16 239 315172 Coding 4 1026 ggccaggcccagcaggttgt 72 240 315173 Coding 4 1070 ccgatgcccaggtagaggct 59 241 315174 Coding 4 496 agatgttggccgtggtattg 79 242 315175 Coding 4 399 caggttgtggtgacactggt 58 243 315176 Coding 4 1420 ggaagaggtcgaagaagagc 26 244 315177 Coding 4 392 tggtgacactggtcaccgta 49 245 315178 Coding 4 402 gctcaggttgtggtgacact 62 246 315179 Coding 4 533 tgcactttgtggtgccaagg 75 247 315180 Coding 4 689 atcacctggaagctgctgta 45 248 315181 Coding 4 956 tattgcatgaacaccgcggc 78 249 315182 Coding 4 1208 atcaggatggccaggaagac 36 250 315183 Coding 4 555 tctcttgaacacgaagcggt 71 251 315184 Coding 4 553 tcttgaacacgaagcggtgt 87 252 315185 Coding 4 1027 tggccaggcccagcaggttg 61 253 315186 Coding 4 871 tctggctgtagcgggtcctg 73 254 315187 Coding 4 498 ggagatgttggccgtggtat 93 255 315188 Start Codon 4 259 tgcctctgggcagctagctg 70 256 315189 Coding 4 1058 tagaggctgaagaagctcct 54 257 315190 Coding 4 348 gtccatcacctgagcggagg 68 258 315191 Coding 4 1292 aacttgtagtctgtgtggtg 39 259 315192 Stop Codon 4 1705 gtcccagcagggttcagaag 31 260 315193 Coding 4 953 tgcatgaacaccgcggccac 73 261 315194 Coding 4 1024 ccaggcccagcaggttgtgc 73 262 315195 Coding 4 1061 aggtagaggctgaagaagct 57 263 315196 Coding 4 1169 cagaagcccatgttgtcatt 47 264 315197 Coding 4 1161 catgttgtcattgctggtcc 0 265 315198 Coding 4 1021 ggcccagcaggttgtgcagg 84 266 315199 Coding 4 400 tcaggttgtggtgacactgg 42 267 315200 Coding 4 1165 agcccatgttgtcattgctg 45 268 315201 Coding 4 363 cttctcaaacaggaagtcca 47 269 315202 Coding 4 550 tgaacacgaagcggtgttgc 83 270 315203 Coding 4 367 tccacttctcaaacaggaag 69 271 315204 Coding 4 353 aggaagtccatcacctgagc 26 272 315205 Coding 4 1071 gccgatgcccaggtagaggc 82 273 315206 Coding 4 1186 ggaaccgcaggatccaccag 36 274 315207 Coding 4 349 agtccatcacctgagcggag 63 275 315208 Coding 4 1221 gatgaagaagttgatcagga 28 276 315209 Coding 4 461 cagcaggaatacttgtcgaa 27 277 315210 Coding 4 463 gccagcaggaatacttgtcg 41 278 315211 Coding 4 320 ggctggcaggccagcagcag 72 279 315212 Coding 4 1183 accgcaggatccaocagaag 59 280 315213 Coding 4 862 agcgggtcctgagcagccca 68 281 315214 Coding 4 565 cgggcccgcatctcttgaac 88 282 315215 Coding 4 1295 cggaacttgtagtctgtgtg 29 283 315216 Coding 4 1177 ggatccaccagaagcccatg 58 284 315217 Stop Codon 4 1706 ggtcccagcagggttcagaa 34 285 315218 Coding 4 1184 aaccgcaggatccacoagaa 55 286 315219 Coding 4 410 ggcagcaggctcaggttgtg 50 287 315220 Coding 4 495 gatgttggccgtggtattgg 86 288 315221 Coding 4 455 gaatacttgtcgaaggttct 37 289 315222 Coding 4 1215 gaagttgatcaggatggcca 39 290 315223 Coding 4 688 tcacctggaagctgctgtac 48 291 315224 Coding 4 959 ccatattgcatgaaoaccgc 20 292 315225 Coding 4 863 tagcgggtcctgagcagccc 61 293 315226 5′UTR 4 256 ctctgggcagctagctgcct 28 294 315227 Coding 4 359 tcaaacaggaagtccatcac 17 295 315228 Coding 4 1172 caccagaagcccatgttgtc 15 296 315229 Coding 4 694 tgtacatcacctggaagctg 67 297 315230 Coding 4 494 atgttggccgtggtattggc 52 298 315231 Coding 4 1069 cgatgcccaggtagaggctg 7 299 315232 Coding 4 1178 aggatccaccagaagcccat 83 300 315233 Coding 4 1207 tcaggatggccaggaagacg 52 301 315234 Coding 4 352 ggaagtccatcacctgagcg 60 302 315235 Start Codon 4 261 catgcctctgggcagctagc 65 303 315236 Coding 4 561 cccgcatctcttgaacacga 51 304 315237 Coding 4 323 tgtggctggcaggccagcag 60 305 315238 Coding 4 324 ctgtggctggcaggccagca 43 306 315239 Coding 4 1179 caggatccaccagaagccca 88 307 315240 Coding 4 1223 aagatgaagaagttgatcag 0 308 315241 Coding 4 1289 ttgtagtctgtgtggtgcat 66 309 315242 Coding 4 322 gtggctggcaggccagcagc 47 310 315243 Coding 4 406 gcaggctcaggttgtggtga 44 311 315244 Coding 4 870 ctggctgtagcgggtcctga 61 312 315245 5′UTR 4 255 tctgggcagctagctgcctc 24 313 315246 Coding 4 464 ggccagcaggaatacttgtc 71 314 315247 Coding 4 360 ctcaaacaggaagtccatca 13 315 315248 Coding 4 1060 ggtagaggctgaagaagctc 49 316 315249 Coding 4 1422 gaggaagaggtcgaagaaga 69 317 315250 Coding 4 1416 gaggtcgaagaagagcttgg 32 318 315251 Coding 4 1288 tgtagtctgtgtggtgcatc 30 319 315252 Coding 4 1216 agaagttgatcaggatggcc 17 320 315253 Coding 4 542 aagcggtgttgcactttgtg 55 321 315254 Coding 4 456 ggaatacttgtcgaaggttc 44 322 315255 Coding 4 1419 gaagaggtcgaagaagagct 34 323 315256 Coding 4 460 agcaggaatacttgtcgaag 10 324 315257 Coding 4 404 aggctcaggttgtggtgaca 58 325 315258 Coding 4 538 ggtgttgcactttgtggtgc 58 326 315259 Coding 4 1294 ggaacttgtagtctgtgtgg 30 327 315260 Coding 4 390 gtgacactggtcaccgtaga 19 328 315261 Coding 4 954 ttgcatgaacaccgcggcca 59 329 315262 Coding 4 684 ctggaagctgctgtacatct 61 330 315263 Coding 4 1174 tccaccagaagcccatgttg 1 331 315264 Coding 4 1214 aagttgatcaggatggccag 44 332 315265 Coding 4 1023 caggcccagcaggttgtgca 51 333 315266 Coding 4 920 accgctccatcactgagcca 38 334 315267 Coding 4 1220 atgaagaagttgatcaggat 0 335 315268 Coding 4 554 ctcttgaacacgaagcggtg 78 336 315269 Coding 4 318 ctggcaggccagcagcagca 37 337 315270 Coding 4 499 aggagatgttggccgtggta 97 338 315271 Coding 4 1164 gcccatgttgtcattgctgg 66 339 315272 Coding 4 1217 aagaagttgatcaggatggc 25 340 315273 Coding 4 1064 cccaggtagaggctgaagaa 62 341 315274 Coding 4 1163 cccatgttgtcattgctggt 55 342 315275 Coding 4 547 acacgaagcggtgttgcact 46 343 315276 Coding 4 408 cagcaggctcaggttgtggt 62 344 315277 Coding 4 394 tgtggtgacactggtcaccg 15 345 315278 Coding 4 1020 gcccagcaggttgtgcaggt 83 346 315279 Coding 4 869 tggctgtagcgggtcctgag 33 347 315280 Coding 4 562 gcccgcatctcttgaacacg 77 348 315281 Coding 4 1418 aagaggtcgaagaagagctt 40 349 315282 Coding 4 411 gggcagcaggctcaggttgt 23 350 315283 Coding 4 557 catctcttgaacacgaagcg 40 351 315284 Coding 4 1175 atccaccagaagcccatgtt 38 352 315285 Coding 4 1155 gtcattgctggtccagcact 75 353 315286 Coding 4 566 tcgggcccgcatctcttgaa 74 354 315287 Coding 4 721 cccccagggacaggctgtag 53 355 315288 Coding 4 1162 ccatgttgtcattgctggtc 43 356 315289 Coding 4 1056 gaggctgaagaagctcctct 2 357 315290 Coding 4 549 gaacacgaagcggtgttgca 88 358 315291 Coding 4 362 ttctcaaacaggaagtccat 9 359 315292 Coding 4 1159 tgttgtcattgctggtccag 47 360 315293 Coding 4 457 aggaatacttgtcgaaggtt 55 361 315294 Coding 4 405 caggctcaggttgtggtgac 38 362 315295 Coding 4 1421 aggaagaggtcgaagaagag 19 363 315296 Coding 4 1425 gctgaggaagaggtcgaaga 33 364 315297 Coding 4 546 cacgaagcggtgttgcactt 81 365 315298 Coding 4 1166 aagcccatgttgtcattgct 35 366 315299 Start Codon 4 260 atgcctctgggcagctagct 63 367 315300 Coding 4 690 catcacctggaagctgctgt 63 368 315301 Coding 4 364 acttctcaaacaggaagtcc 31 369 315302 Coding 4 558 gcatctcttgaacacgaagc 44 370 315303 Coding 4 958 catattgcatgaacaccgcg 48 371 315304 Coding 4 1170 ccagaagcccatgttgtcat 33 372 315305 Coding 4 867 gctgtagcgggtcctgagca 50 373 315306 Coding 4 865 tgtagcgggtcctgagcagc 62 374 315307 Coding 4 1022 aggcccagcaggttgtgcag 37 375 315308 Coding 4 692 tacatcacctggaagctgct 41 376 315309 Coding 4 1181 cgcaggatccaccagaagcc 49 377 315310 Coding 4 357 aaacaggaagtccatcacct 21 378 315311 Coding 4 1057 agaggctgaagaagctcctc 49 379 315312 Coding 4 1211 ttgatcaggatggccaggaa 54 380 315313 Coding 4 541 agcggtgttgcactttgtgg 81 381 315314 Coding 4 319 gctggcaggccagcagcagc 75 382 315315 Coding 4 545 acgaagcggtgttgcacttt 68 383 315316 Coding 4 952 gcatgaacaccgcggccaca 80 384 315317 Coding 4 354 caggaagtccatcacctgag 26 385 315318 Coding 4 1180 gcaggatccaccagaagccc 72 386 315319 Coding 4 1213 agttgatcaggatggccagg 33 387 315320 Coding 4 722 gcccocagggacaggctgta 51 388 315321 Coding 4 356 aacaggaagtocatcacctg 0 389 315322 Coding 4 957 atattgcatgaacaccgcgg 56 390 315323 Coding 4 459 gcaggaatacttgtcgaagg 59 391 315324 Coding 4 693 gtacatcacctggaagctgc 79 392 315325 Coding 4 1153 cattgctggtccagcactgg 61 393 315326 Coding 4 358 caaacaggaagtccatcacc 10 394 315327 Coding 4 1031 agggtggccaggcccagcag 27 395 315328 Coding 4 551 ttgaacacgaagcggtgttg 66 396 315329 Coding 4 1171 accagaagcccatgttgtca 47 397 315330 Coding 4 401 ctcaggttgtggtgacactg 14 398 315331 Coding 4 396 gttgtggtgacactggtcac 5 399

As shown in Table 1, SEQ ID NOs 20, 21, 22, 24, 25, 28, 29, 30, 35, 37, 39, 40, 41, 42, 43, 44, 46, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 64, 67, 68, 69, 70, 71, 72, 74, 75, 76, 78, 79, 80, 81, 82, 87, 88, 89, 91, 92, 94, 95, 96, 97, 98, 99, 100, 101, 102, 103, 104, 106, 107, 110, 111, 112, 113, 114, 115, 116, 117, 118, 119, 121, 122, 124, 125, 127, 129, 132, 133, 134, 135, 136, 137, 138, 139, 140, 141, 142, 143, 144, 145, 146, 147, 148, 149, 152, 153, 155, 156, 157, 158, 159, 160, 161, 162, 164, 166, 168, 169, 170, 171, 174, 177, 178, 179, 180, 181, 182, 183, 184, 185, 186, 187, 188, 189, 190, 192, 193, 194, 195, 196, 197, 198, 199, 200, 201, 202, 203, 204, 205, 206, 207, 208, 209, 210, 212, 213, 214, 215, 216, 217, 218, 219, 221, 225, 227, 230, 231, 233, 235, 236, 237, 238, 240, 241, 242, 243, 245, 246, 247, 248, 249, 251, 252, 253, 254, 255, 256, 257, 258, 261, 262, 263, 264, 266, 267, 268, 269, 270, 271, 273, 275, 278, 279, 280, 281, 282, 284, 286, 287, 288, 291, 293, 297, 298, 300, 301, 302, 303, 304, 305, 306, 307, 309, 310, 311, 312, 314, 316, 317, 321, 322, 325, 326, 329, 330, 332, 333, 336, 338, 339, 341, 342, 343, 344, 346, 348, 349, 351, 353, 354, 355, 356, 358, 360, 361, 365, 367, 368, 370, 371, 373, 374, 376, 377, 379, 380, 381, 382, 383, 384, 386, 388, 390, 391, 392, 393, 396 and 397 demonstrated at least 40% inhibition of human glucagon receptor expression in this assay and are therefore preferred. SEQ ID NO: 183, 184, 231, 249, 254, 346, 365 and 392 are presently more preferred. The target regions to which the preferred sequences are complementary are herein referred to as “preferred target segments” and are therefore preferred for targeting by compounds of the present invention. These preferred target segments are shown in Table 3. These sequences are shown to contain thymine (T) but one of skill in the art will appreciate that thymine (T) is generally replaced by uracil (U) in RNA sequences. The sequences represent the reverse complement of the preferred antisense compounds shown in Table 1. “Target site” indicates the first (5′-most) nucleotide number on the particular target nucleic acid to which the oligonucleotide binds. Also shown in Table 3 is the species in which each of the preferred target segments was found.

Example 16

Antisense Inhibition of Mouse Glucagon Receptor Expression by Chimeric Phosphorothioate Oligonucleotides Having 2′-MOE Wings and a Deoxy Gap

In accordance with the present invention, a second series of antisense compounds were designed to target different regions of the mouse glucagon receptor RNA, using published sequences (GenBank accession number NM008101.1, incorporated herein as SEQ ID NO: 11, an mRNA sequence derived from GenBank accession number AF229079.1 with an alternate promoter, incorporated herein as SEQ ID NO: 400, GenBank accession number AF229079.1, incorporated herein as SEQ ID NO: 401, a second mRNA sequence derived from GenBank accession number AF229079.1 with an alternate promoter, incorporated herein as SEQ ID NO: 402, and GenBank accession number AA920726.1, incorporated herein as SEQ ID NO: 403). The compounds are shown in Table 2. “Target site” indicates the first (5′-most) nucleotide number on the particular target nucleic acid to which the compound binds. All compounds in Table 2 are chimeric oligonucleotides (“gapmers”) 20 nucleotides in length, composed of a central “gap” region consisting of ten 2′-deoxynucleotides, which is flanked on both sides (5′ and 3′ directions) by five-nucleotide “wings”. The wings are composed of 2′-methoxyethyl (2′-MOE)nucleotides. The internucleoside (backbone) linkages are phosphorothioate (P═S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines. The compounds were analyzed for their effect on mouse glucagon receptor mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from three experiments in which mouse primary hepatocytes were treated with the antisense oligonucleotides of the present invention. The positive control for each datapoint is identified in the table by sequence ID number. If present, “N.D.” indicates “no data”.

TABLE 2 Inhibition of mouse glucagon receptor mRNA levels by chimeric phosphorothioate oligonucleotides having 2′-MOE wings and a deoxy gap TARGET CONTROL SEQ TARGET % SEQ SEQ ID ISIS # REGION ID NO SITE SEQUENCE INHIB ID NO NO 148350 5′UTR 11 57 cccacatctggcagaggttg 30 404 1 148355 coding 11 182 ttctcaaacaaaaagtccat 7 405 1 148356 coding 11 193 agagcttccacttctcaaac 63 406 1 148357 coding 11 203 tggtcactatagagcttcca 39 407 1 148359 coding 11 227 agcaggcttaggttgtggtg 39 408 1 148363 coding 11 322 ggcaggaaatgttggcagtg 52 409 1 148366 coding 11 383 ggcccacacctcttgaacac 93 410 1 148368 exon:exon 11 477 ccccttctggacctcgatct 45 411 1 junction 148371 coding 11 538 ccagggacagactgtagccc 53 412 1 148372 exon:exon 11 589 agtgcagcttcctgaggccc 52 413 1 junction 148381 coding 11 938 cacttgaccaccacccaggg 47 414 1 148382 coding 11 947 tcaaacagacacttgaccac 0 415 1 148385 coding 11 977 ttgtcattgctggtccagca 57 416 1 148387 coding 11 998 aggatccaccagaatcccat 53 417 1 148390 coding 11 1139 agggtcagcgtggacctggc 45 418 1 148393 coding 11 1226 aagagcttggtggagcgcag 26 419 1 148394 coding 11 1277 tagagaacagccaccagcag 26 420 1 148395 coding 11 1285 ggaaacagtagagaacagcc 26 421 1 148396 exon:exon 11 1299 cacctccttgttgaggaaac 0 422 1 junction 180446 5′UTR 11 7 ctcctcaggttgcaagggag 15 423 1 180447 5′UTR 11 14 tgcacctctcctcaggttgc 38 424 1 180448 5′UTR 11 25 ctcagagtgtgtgcacctct 54 425 1 180449 5′UTR 11 30 aggtcctcagagtgtgtgca 55 426 1 180450 5′UTR 11 48 ggcagaggttgcacacctag 39 427 1 180451 Start Codon 11 80 ggcatgcctctgggtagcca 40 428 1 180452 Coding 11 141 tggcagacatgacagcacca 5 429 1 180453 Coding 11 192 gagcttccacttctcaaaca 37 430 1 180454 Coding 11 251 cagaccagctcagtaggtgg 45 431 1 180455 Coding 11 291 ggtgtcaggccagcaggagt 58 432 1 180456 Coding 11 359 cggtgctgcactttgtggca 68 433 1 180457 Coding 11 371 ttgaacactaggcggtgctg 69 434 1 180458 Coding 11 410 cgtggccctcgaacccactg 39 435 1 180459 Coding 11 545 aaggcccccagggacagact 56 436 1 180460 Coding 11 572 cccagcaggatgaccagcgc 59 437 1 180461 Coding 11 582 cttcctgaggcccagcagga 40 438 1 180462 Coding 11 650 acagagccagccttgagcac 47 439 1 180463 Coding 11 764 actgtggccactctgcagcc 43 440 1 180464 Coding 11 775 actgcatgatcactgtggcc 60 441 1 180465 Coding 11 785 atgatgccgtactgcatgat 58 442 1 180466 Coding 11 836 agcaggctgtacaggtacac 36 443 1 180467 Coding 11 974 tcattgctggtccagcactg 62 444 1 180468 Coding 11 1011 gacaggaatacgcaggatcc 0 445 1 180469 Coding 11 1079 cgcagcttggccacaagaag 56 446 1 180470 Coding 11 1090 tctgatgggcacgcagcttg 8 447 1 180471 Coding 11 1100 gcatagtgcatctgatgggc 45 448 1 180472 Coding 11 1110 cttgtaatcagcatagtgca 14 449 1 180473 Coding 11 1256 ccctggaaggagctgaggaa 45 450 1 180474 Coding 11 1292 ttgttgaggaaacagtagag 47 451 1 180475 Coding 11 1348 gagctttgccttcttgccat 64 452 1 180476 Coding 11 1360 tttcctcctgaagagctttg 56 453 1 180477 Coding 11 1388 atgtggctgccatggctgct 64 454 1 180478 Coding 11 1435 gctgaagtttctcacaggga 56 455 1 180479 Coding 11 1450 tgcctgcactcataagctga 48 456 1 180480 Coding 11 1470 acagccagtcccactgctgc 41 457 1 180481 Coding 11 1512 ccttgggagactactggcca 56 458 1 180482 Stop Codon 11 1544 caagtggagattcaggtggg 47 459 1 180483 3′UTR 11 1567 ttgaacacaacctgcctagg 9 460 1 180484 3′UTR 11 1575 gccctttcttgaacacaacc 34 461 1 180485 3′UTR 11 1600 atctggctctgggttgtcct 59 462 1 180486 3′UTR 11 1610 ttggccgggcatctggctct 53 463 1 180487 3′UTR 11 1620 ctcttcaaccttggccgggc 66 464 1 180488 3′UTR 11 1646 tacaagctgctgtcttgctg 54 465 1 180489 3′UTR 11 1687 ggcctgtgccaggctaggac 47 466 1 180490 3′UTR 11 1724 gcttctccatcatatccaac 47 467 1 180491 3′UTR 11 1750 aacactcagagttcatagat 51 468 1 180492 3′UTR 11 1756 catgggaacactcagagttc 49 469 1 180493 3′UTR 11 1795 ctgaaggacatatctgggta 51 470 1 180494 intron 401 3953 gtaacaaaggcgagaccaag 36 471 1 180495 intron 401 5396 gaggaagtgtcaccattagg 23 472 1 180496 intron:exon 401 7321 cagaccagctctgtgaaggt 32 473 1 junction 180497 intron:exon 401 7505 cggtgctgcactgggcatgg 77 474 1 junction 180498 exon:intron 401 8075 ctgggctcaccccgtcactg 27 475 1 junction 180499 intron 401 8766 ccaaggatgggcaacctgac 33 476 1 180500 exon:intron 401 9005 ccttaccaaccggaacttgt 2 477 1 junction 180501 genomic 402 128 cctctcctcaggtgtgctca 3 478 1 180502 genomic 400 10 ccaagcccaaggcctcatga 40 479 1 180503 genomic 400 85 ctcaggctgcagaggaccag 39 480 1 180504 genomic 403 40 taggtctcttccctccactc 4 481 1

As shown in Table 2, SEQ ID NOs 404, 406, 407, 408, 409, 410, 411, 412, 413, 414, 416, 417, 418, 424, 425, 426, 427, 428, 430, 431, 432, 433, 434, 435, 436, 437, 438, 439, 440, 441, 442, 443, 444, 446, 448, 450, 451, 452, 453, 454, 455, 456, 457, 458, 459, 461, 462, 463, 464, 465, 466, 467, 468, 469, 470, 471, 473, 474, 476, 479 and 480 demonstrated at least 30% inhibition of mouse glucagon receptor expression in this experiment and are therefore preferred. The target regions 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. These preferred target segments are shown in Table 3. These sequences are shown to contain thymnine (T) but one of skill in the art will appreciate that thymnine (T) is generally replaced by uracil (U) in RNA sequences. The sequences represent the reverse complement of the preferred antisense compounds shown in Tables 1 and 2. “Target site” indicates the first (5′-most) nucleotide number on the particular target nucleic acid to which the oligonucleotide binds. Also shown in Table 3 is the species in which each of the preferred target segments was found.

TABLE 3 Sequence and position of preferred target segments identified in glucagon receptor. TARGET SEQ TARGET REV COMP SEQ ID SITE ID ID NO SITE SEQUENCE OF SEQ ID ACTIVE IN NO 215734 4 97 gcgcgggccctgaggctcaa 21 H. sapiens 484 215735 4 121 gcagcttcaggggaggacac 22 H. sapiens 485 215737 4 192 cggaggagcgtacacacaca 24 H. sapiens 486 215738 4 198 agcgtacacacacaccagga 25 H. sapiens 487 110316 4 263 tagctgcccagaggcatgcc 28 H. sapiens 488 215800 4 462 tcgacaagtattcctgctgg 29 H. sapiens 489 215741 4 328 ggcctgccagccacaggtcc 30 H. sapiens 490 215746 4 391 ctacggtgaccagtgtcacc 35 H. sapiens 491 215748 4 442 gctggtgtgcaacagaacct 37 H. sapiens 492 110318 4 539 caccacaaagtgcaacaccg 39 H. sapiens 493 215798 19 546 tgccgctgtctctgccaggg 40 H. sapiens 494 215750 4 552 aacaccgcttcgtgttcaag 41 H. sapiens 495 215751 4 564 tgttcaagagatgcgggccc 42 H. sapiens 496 215801 18 15279 gccactgtaactcgcagaag 43 H. sapiens 497 215752 4 651 gcgaggagattgaggtccag 44 H. sapiens 498 215754 4 663 aggtccagaaggaggtggcc 46 H. sapiens 499 215756 4 681 ccaagatgtacagcagcttc 48 H. sapiens 500 215757 4 751 cgccttggccatcctggggg 49 H. sapiens 501 215758 4 830 gtgctgaaagccagctccgt 50 H. sapiens 502 215759 4 866 ctgctcaggacccgctacag 51 H. sapiens 503 215760 4 872 aggacccgctacagccagaa 52 H. sapiens 504 215761 4 879 gctacagccagaaaattggc 53 H. sapiens 505 215762 4 889 gaaaattggcgacgacctca 54 H. sapiens 506 215763 4 898 cgacgacctcagtgtcagca 55 H. sapiens 507 215764 4 904 cctcagtgtcagcacctggc 56 H. sapiens 508 215765 4 966 tcatgcaatatggcatcgtg 57 H. sapiens 509 215766 4 1028 aacctgctgggcctggccac 58 H. sapiens 510 215767 4 1122 gggcagtggtcaagtgtctg 59 H. sapiens 511 215768 4 1182 gcttctggtggatcctgcgg 60 H. sapiens 512 215769 4 1210 cttcctggccatcctgatca 61 H. sapiens 513 215770 4 1228 caacttcttcatcttcgtcc 62 H. sapiens 514 215771 4 1274 ctgcgggcacggcagatgca 64 H. sapiens 515 215774 4 1528 ctggcgcctgggcaaagtgc 67 H. sapiens 516 215775 4 1539 gcaaagtgctatgggaggag 68 H. sapiens 517 215776 4 1608 ccagcaaggagctgcagttt 69 H. sapiens 518 215777 4 1636 tggtggcagccaggattcat 70 H. sapiens 519 215778 4 1670 ttggctggtggcctccctag 71 H. sapiens 520 215779 4 1681 cctccctagattggctgaga 72 H. sapiens 521 215799 19 1747 cattgggtcacctgcaggaa 74 H. sapiens 522 215781 4 1841 cagtgtggctgtctgcgaga 75 H. sapiens 523 215782 4 1854 tgcgagattgggcctcctct 76 H. sapiens 524 215784 4 1901 gaggtgagcagaggagtcca 78 H. sapiens 525 215785 4 1938 gtgccgtgaactgcgtgcca 79 H. sapiens 526 215786 4 1969 tatgtcggcacgtcccatgt 80 H. sapiens 527 215787 4 1978 acgtcccatgtgcatggaaa 81 H. sapiens 528 215788 4 1989 gcatggaaatgtcctccaac 82 H. sapiens 529 215793 18 14121 ggtaactgagccacagagct 87 H. sapiens 530 215794 18 15467 tgtgccacagcaagctgcac 88 H. sapiens 531 215795 18 16094 cctgtaccaagcggttgctg 89 H. sapiens 532 215797 18 17456 gtctctccagggcctgctgg 91 H. sapiens 533 220245 4 100 cgggccctgaggctcaaagg 92 H. sapiens 534 220247 4 167 tgctgctctgccactcagct 94 H. sapiens 535 220248 4 169 ctgctctgccactcagctgc 95 H. sapiens 536 220249 4 190 ctcggaggagcgtacacaca 96 H. sapiens 537 220250 4 194 gaggagcgtacacacacacc 97 H. sapiens 538 220251 4 196 ggagcgtacacacacaccag 98 H. sapiens 539 220252 4 209 acaccaggactgcattgccc 99 H. sapiens 540 220253 4 246 cagatgtgggaggcagctag 100 H. sapiens 541 220254 4 249 atgtgggaggcagctagctg 101 H. sapiens 542 220255 4 257 ggcagctagctgcccagagg 102 H. sapiens 543 220256 4 262 ctagctgcccagaggcatgc 103 H. sapiens 544 220257 4 325 gctggcctgccagccacagg 104 H. sapiens 545 220259 4 370 cctgtttgagaagtggaagc 106 H. sapiens 546 220260 4 375 ttgagaagtggaagctctac 107 H. sapiens 547 110282 4 407 caccacaacctgagcctgct 110 H. sapiens 548 220263 4 534 cttggcaccacaaagtgcaa 111 H. sapiens 549 220264 4 535 ttggcaccacaaagtgcaac 112 H. sapiens 550 220265 4 536 tggcaccacaaagtgcaaca 113 H. sapiens 551 220266 4 537 ggcaccacaaagtgcaacac 114 H. sapiens 552 110289 4 563 gtgttcaagagatgcgggcc 115 H. sapiens 553 220267 4 567 tcaagagatgcgggcccgac 116 H. sapiens 554 220268 4 617 ccttggcgtgatgcctccca 117 H. sapiens 555 220269 4 627 atgcctcccagtgccagatg 118 H. sapiens 556 220270 4 666 tccagaaggaggtggccaag 119 H. sapiens 557 220272 4 685 gatgtacagcagcttccagg 121 H. sapiens 558 220273 4 795 gcaatgccatccacgcgaat 122 H. sapiens 559 220275 4 861 atgggctgctcaggacccgc 124 H. sapiens 560 220276 4 886 ccagaaaattggcgacgacc 125 H. sapiens 561 220277 4 900 acgacctcagtgtcagcacc 127 H. sapiens 562 220279 4 1032 tgctgggcctggccaccctc 129 H. sapiens 563 220282 4 1158 gctggaccagcaatgacaac 132 H. sapiens 564 220283 4 1168 caatgacaacatgggcttct 133 H. sapiens 565 220284 4 1187 tggtggatcctgcggttccc 134 H. sapiens 566 220285 4 1230 acttcttcatcttcgtccgc 135 H. sapiens 567 220286 4 1638 gtggcagccaggattcatct 136 H. sapiens 568 220287 4 1727 cagctagggctggactctgg 137 H. sapiens 569 220288 4 1732 agggctggactctggcaccc 138 H. sapiens 570 220289 4 1735 gctggactctggcacccaga 139 H. sapiens 571 220290 4 1736 ctggactctggcacccagag 140 H. sapiens 572 220291 4 1737 tggactctggcacccagagg 141 H. sapiens 573 220292 4 1740 actctggcacccagaggcgt 142 H. sapiens 574 220293 4 1760 cgctggacaacccagaactg 143 H. sapiens 575 220294 4 1849 ctgtctgcgagattgggcct 144 H. sapiens 576 220295 4 1850 tgtctgcgagattgggcctc 145 H. sapiens 577 220296 4 1856 cgagattgggcctcctctcc 146 H. sapiens 578 220297 4 1861 ttgggcctcctctccctgca 147 H. sapiens 579 220298 4 1883 tgccttgtccctggtgcaga 148 H. sapiens 580 220299 4 1891 ccctggtgcagaggtgagca 149 H. sapiens 581 220302 4 1905 tgagcagaggagtccagggc 152 H. sapiens 582 220303 4 1932 ggggctgtgccgtgaactgc 153 H. sapiens 583 220305 4 1945 gaactgcgtgccagtgtccc 155 H. sapiens 584 220306 4 1971 tgtcggcacgtcccatgtgc 156 H. sapiens 585 220307 4 1984 catgtgcatggaaatgtcct 157 H. sapiens 586 220308 4 1986 tgtgcatggaaatgtcctcc 158 H. sapiens 587 220309 4 1999 gtcctccaacaataaagagc 159 H. sapiens 588 220310 4 2001 cctccaacaataaagagctc 160 H. sapiens 589 220311 4 2008 caataaagagctcaagtggt 161 H. sapiens 590 220312 18 3174 ctggggacgccaaaactgcc 162 H. sapiens 591 220314 18 7544 tagacacgcacatcctatcc 164 H. sapiens 592 220316 18 14888 ctaccctgcagagctggtgt 166 H. sapiens 593 226083 4 258 gcagctagctgcccagaggc 168 H. sapiens 594 226084 4 317 ctgctgctgctggcctgcca 169 H. sapiens 595 226085 4 321 tgctgctggcctgccagcca 170 H. sapiens 596 226086 4 347 ccctccgctcaggtgatgga 171 H. sapiens 597 226089 4 365 gacttcctgtttgagaagtg 174 H. sapiens 598 110281 4 397 tgaccagtgtcaccacaacc 177 H. sapiens 599 226092 4 403 gtgtcaccacaacctgagcc 178 H. sapiens 600 226093 4 452 aacagaaccttcgacaagta 179 H. sapiens 601 226094 4 458 accttcgacaagtattcctg 180 H. sapiens 602 226095 4 493 cgccaataccacggccaaca 181 H. sapiens 603 226096 4 497 aataccacggccaacatctc 182 H. sapiens 604 226097 4 500 accacggccaacatctcctg 183 H. sapiens 605 226098 4 532 gccttggcaccacaaagtgc 184 H. sapiens 606 110288 4 540 accacaaagtgcaacaccgc 185 H. sapiens 607 226099 4 544 caaagtgcaacaccgcttcg 186 H. sapiens 608 226100 4 548 gtgcaacaccgcttcgtgtt 187 H. sapiens 609 226101 4 556 ccgcttcgtgttcaagagat 188 H. sapiens 610 226103 4 588 gtcagtgggtgcgtggaccc 189 H. sapiens 611 226104 4 606 cccgggggcagccttggcgt 190 H. sapiens 612 226106 4 683 aagatgtacagcagcttcca 192 H. sapiens 613 226107 4 687 tgtacagcagcttccaggtg 193 H. sapiens 614 226108 4 691 cagcagcttccaggtgatgt 194 H. sapiens 615 226109 4 695 agcttccaggtgatgtacac 195 H. sapiens 616 226110 4 720 gctacagcctgtccctgggg 196 H. sapiens 617 226111 4 723 acagcctgtccctgggggcc 197 H. sapiens 618 226112 4 860 gatgggctgctcaggacccg 198 H. sapiens 619 226113 4 864 ggctgctcaggacccgctac 199 H. sapiens 620 226114 4 868 gctcaggacccgctacagcc 200 H. sapiens 621 226115 4 919 ctggctcagtgatggagcgg 201 H. sapiens 622 226116 4 923 ctcagtgatggagcggtggc 202 H. sapiens 623 226117 4 951 gtgtggccgcggtgttcatg 203 H. sapiens 624 226118 4 955 ggccgcggtgttcatgcaat 204 H. sapiens 625 226119 4 960 cggtgttcatgeaatatggc 205 H. sapiens 626 226120 4 1019 tacctgcacaacctgctggg 206 H. sapiens 627 226121 4 1025 cacaacctgctgggcctggc 207 H. sapiens 628 226122 4 1029 acctgctgggcctggccacc 208 H. sapiens 629 226123 4 1055 gagaggagcttcttcagcct 209 H. sapiens 630 226124 4 1059 ggagcttcttcagcctctac 210 H. sapiens 631 226126 4 1068 tcagcctctacctgggcatc 212 H. sapiens 632 110302 4 1072 cctctacctgggcatcggct 213 H. sapiens 633 226127 4 1156 gtgctggaccagcaatgaca 214 H. sapiens 634 226128 4 1160 tggaccagcaatgacaacat 215 H. sapiens 635 226129 4 1167 gcaatgacaacatgggcttc 216 H. sapiens 636 226130 4 1173 acaacatgggcttctggtgg 217 H. sapiens 637 226131 4 1176 acatgggcttctggtggatc 218 H. sapiens 638 226132 4 1185 tctggtggatcctgcggttc 219 H. sapiens 639 226134 4 1209 tcttcctggccatcctgatc 221 H. sapiens 640 226138 4 1290 tgcaccacacagactacaag 225 H. sapiens 641 226140 4 1414 cgccaagctcttcttcgacc 227 H. sapiens 642 226143 4 1669 cttggctggtggcctcccta 230 H. sapiens 643 231032 4 686 atgtacagcagcttccaggt 231 H. sapiens 644 231034 4 1424 ttcttcgacctcttcctcag 233 H. sapiens 645 231036 4 1212 tcctggccatcctgatcaac 235 H. sapiens 646 231037 4 1062 gcttcttcagcctctacctg 236 H. sapiens 647 231038 4 559 cttcgtgttcaagagatgcg 237 H. sapiens 648 231039 4 543 acaaagtgcaacaccgcttc 238 H. sapiens 649 231041 4 1026 acaacctgctgggcctggcc 240 H. sapiens 650 231042 4 1070 agcctctacctgggcatcgg 241 H. sapiens 651 231043 4 496 caataccacggccaacatct 242 H. sapiens 652 231044 4 399 accagtgtcaccacaacctg 243 H. sapiens 653 231046 4 392 tacggtgaccagtgtcacca 245 H. sapiens 654 231047 4 402 agtgtcaccacaacctgagc 246 H. sapiens 655 110287 4 533 ccttggcaccacaaagtgca 247 H. sapiens 656 231048 4 689 tacagcagcttccaggtgat 248 H. sapiens 657 231049 4 956 gccgcggtgttcatgcaata 249 H. sapiens 658 231051 4 555 accgcttcgtgttcaagaga 251 H. sapiens 659 231052 4 553 acaccgcttcgtgttcaaga 252 H. sapiens 660 231053 4 1027 caacctgctgggcctggcca 253 H. sapiens 661 231054 4 871 caggacccgctacagccaga 254 H. sapiens 662 231055 4 498 ataccacggccaacatctcc 255 H. sapiens 663 231056 4 259 cagctagctgcccagaggca 256 H. sapiens 664 231057 4 1058 aggagcttcttcagcctcta 257 H. sapiens 665 231058 4 348 cctccgctcaggtgatggac 258 H. sapiens 666 231061 4 953 gtggccgcggtgttcatgca 261 H. sapiens 667 231062 4 1024 gcacaacctgctgggcctgg 262 H. sapiens 668 231063 4 1061 agcttcttcagcctctacct 263 H. sapiens 669 231064 4 1169 aatgacaacatgggcttctg 264 H. sapiens 670 231066 4 1021 cctgcacaacctgctgggcc 266 H. sapiens 671 231067 4 400 ccagtgtcaccacaacctga 267 H. sapiens 672 231068 4 1165 cagcaatgacaacatgggct 268 H. sapiens 673 231069 4 363 tggacttcctgtttgagaag 269 H. sapiens 674 231070 4 550 gcaacaccgcttcgtgttca 270 H. sapiens 675 231071 4 367 cttcctgtttgagaagtgga 271 H. sapiens 676 231073 4 1071 gcctctacctgggcatcggc 273 H. sapiens 677 231075 4 349 ctccgctcaggtgatggact 275 H. sapiens 678 231077 4 463 cgacaagtattcctgctggc 278 H. sapiens 679 231078 4 320 ctgctgctggcctgccagcc 279 H. sapiens 680 231079 4 1183 cttctggtggatcctgcggt 280 H. sapiens 681 231080 4 862 tgggctgctcaggacccgct 281 H. sapiens 682 231081 4 565 gttcaagagatgcgggcccg 282 H. sapiens 683 231083 4 1177 catgggcttctggtggatcc 284 H. sapiens 684 231085 4 1184 ttctggtggatcctgcggtt 286 H. sapiens 685 231086 4 410 cacaacctgagcctgctgcc 287 H. sapiens 686 231087 4 495 ccaataccacggccaacatc 288 H. sapiens 687 231090 4 688 gtacagcagcttccaggtga 291 H. sapiens 688 231092 4 863 gggctgctcaggacccgcta 293 H. sapiens 689 231096 4 694 cagcttccaggtgatgtaca 297 H. sapiens 690 231097 4 494 gccaataccacggccaacat 298 H. sapiens 691 110307 4 1178 atgggcttctggtggatcct 300 H. sapiens 692 231099 4 1207 cgtcttcctggccatcctga 301 H. sapiens 693 231100 4 352 cgctcaggtgatggacttcc 302 H. sapiens 694 231101 4 261 gctagctgcccagaggcatg 303 H. sapiens 695 231102 4 561 tcgtgttcaagagatgcggg 304 H. sapiens 696 231103 4 323 ctgctggcctgccagccaca 305 H. sapiens 697 231104 4 324 tgctggcctgccagccacag 306 H. sapiens 698 231105 4 1179 tgggcttctggtggatcctg 307 H. sapiens 699 231107 4 1289 atgcaccacacagactacaa 309 H. sapiens 700 231108 4 322 gctgctggcctgccagccac 310 H. sapiens 701 231109 4 406 tcaccacaacctgagcctgc 311 H. sapiens 702 231110 4 870 tcaggacccgctacagccag 312 H. sapiens 703 231112 4 464 gacaagtattcctgctggcc 314 H. sapiens 704 231114 4 1060 gagcttcttcagcctctacc 316 H. sapiens 705 231115 4 1422 tcttcttcgacctcttcctc 317 H. sapiens 706 231118 4 542 cacaaagtgcaacaccgctt 321 H. sapiens 707 231119 4 456 gaaccttcgacaagtattcc 322 H. sapiens 708 231122 4 404 tgtcaccacaacctgagcct 325 H. sapiens 709 231123 4 538 gcaccacaaagtgcaacacc 326 H. sapiens 710 231126 4 954 tggccgcggtgttcatgcaa 329 H. sapiens 711 231127 4 684 agatgtacagcagcttccag 330 H. sapiens 712 231129 4 1214 ctggccatcctgatcaactt 332 H. sapiens 713 231130 4 1023 tgcacaacctgctgggcctg 333 H. sapiens 714 231133 4 554 caccgcttcgtgttcaagag 336 H. sapiens 715 231135 4 499 taccacggccaacatctcct 338 H. sapiens 716 231136 4 1164 ccagcaatgacaacatgggc 339 H. sapiens 717 231138 4 1064 ttcttcagcctctacctggg 341 H. sapiens 718 231139 4 1163 accagcaatgacaacatggg 342 H. sapiens 719 231140 4 547 agtgcaacaccgcttcgtgt 343 H. sapiens 720 231141 4 408 accacaacctgagcctgctg 344 H. sapiens 721 231143 4 1020 acctgcacaacctgctgggc 346 H. sapiens 722 231145 4 562 cgtgttcaagagatgcgggc 348 H. sapiens 723 231146 4 1418 aagctcttcttcgacctctt 349 H. sapiens 724 231148 4 557 cgcttcgtgttcaagagatg 351 H. sapiens 725 231150 4 1155 agtgctggaccagcaatgac 353 H. sapiens 726 231151 4 566 ttcaagagatgcgggcccga 354 H. sapiens 727 231152 4 721 ctacagcctgtccctggggg 355 H. sapiens 728 110306 4 1162 gaccagcaatgacaacatgg 356 H. sapiens 729 231154 4 549 tgcaacaccgcttcgtgttc 358 H. sapiens 730 231155 4 1159 ctggaccagcaatgacaaca 360 H. sapiens 731 231156 4 457 aaccttcgacaagtattcct 361 H. sapiens 732 231160 4 546 aagtgcaacaccgcttcgtg 365 H. sapiens 733 231162 4 260 agctagctgcccagaggcat 367 H. sapiens 734 231163 4 690 acagcagcttccaggtgatg 368 H. sapiens 735 231165 4 558 gcttcgtgttcaagagatgc 370 H. sapiens 736 231166 4 958 cgcggtgttcatgcaatatg 371 H. sapiens 737 231168 4 867 tgctcaggacccgctacagc 373 H. sapiens 738 231169 4 865 gctgctcaggacccgctaca 374 H. sapiens 739 231171 4 692 agcagcttccaggtgatgta 376 H. sapiens 740 231172 4 1181 ggcttctggtggatcctgcg 377 H. sapiens 741 231174 4 1057 gaggagcttcttcagcctct 379 H. sapiens 742 231175 4 1211 ttcctggccatcctgatcaa 380 H. sapiens 743 231176 4 541 ccacaaagtgcaacaccgct 381 H. sapiens 744 231177 4 319 gctgctgctggcctgccagc 382 H. sapiens 745 231178 4 545 aaagtgcaacaccgcttcgt 383 H. sapiens 746 231179 4 952 tgtggccgcggtgttcatgc 384 H. sapiens 747 231181 4 1180 gggcttctggtggatcctgc 386 H. sapiens 748 231183 4 722 tacagcctgtccctgggggc 388 H. sapiens 749 231185 4 957 ccgcggtgttcatgcaatat 390 H. sapiens 750 231186 4 459 ccttcgacaagtattcctgc 391 H. sapiens 751 110293 4 693 gcagcttccaggtgatgtac 392 H. sapiens 752 231187 4 1153 ccagtgctggaccagcaatg 393 H. sapiens 753 110319 4 551 caacaccgcttcgtgttcaa 396 H. sapiens 754 231190 4 1171 tgacaacatgggcttctggt 397 H. sapiens 755 63771 11 138 caacctctgccagatgtggg 404 M. musculus 756 63777 11 274 gtttgagaagtggaagctct 406 M. musculus 757 63778 11 284 tggaagctctatagtgacca 407 M. musculus 758 63780 11 308 caccacaacctaagcctgct 408 M. musculus 759 63784 11 403 cactgccaacatttcctgcc 409 M. musculus 760 63787 11 464 gtgttcaagaggtgtgggcc 410 M. musculus 761 63789 11 558 agatcgaggtccagaagggg 411 M. musculus 762 63792 11 619 gggctacagtctgtccctgg 412 M. musculus 763 63793 11 670 gggcctcaggaagctgcact 413 M. musculus 764 63802 11 1019 ccctgggtggtggtcaagtg 414 M. musculus 765 63806 11 1058 tgctggaccagcaatgacaa 416 M. musculus 766 63808 11 1079 atgggattctggtggatcct 417 M. musculus 767 63811 11 1220 gccaggtccacgctgaccct 418 M. musculus 768 95505 400 14 gcaacctgaggagaggtgca 424 M. musculus 769 95506 400 25 agaggtgcacacactctgag 425 M. musculus 770 95507 400 30 tgcacacactctgaggacct 426 M. musculus 771 95508 400 48 ctaggtgtgcaacctctgcc 427 M. musculus 772 95509 400 80 tggctacccagaggcatgcc 428 M. musculus 773 95511 400 192 tgtttgagaagtggaagctc 430 M. musculus 774 95512 400 251 ccacctactgagctggtctg 431 M. musculus 775 95513 400 291 actcctgctggcctgacacc 432 M. musculus 776 95514 400 359 tgccacaaagtgcagcaccg 433 M. musculus 777 95515 400 371 cagcaccgcctagtgttcaa 434 M. musculus 778 95516 400 410 cagtgggttcgagggccacg 435 M. musculus 779 95517 400 545 agtctgtccctgggggcctt 436 M. musculus 780 95518 400 572 gcgctggtcatcctgctggg 437 M. musculus 781 95519 400 582 tcctgctgggcctcaggaag 438 M. musculus 782 95520 400 650 gtgctcaaggctggctctgt 439 M. musculus 783 95521 400 764 ggctgcagagtggccacagt 440 M. musculus 784 95522 400 775 ggccacagtgatcatgcagt 441 M. musculus 785 95523 400 785 atcatgcagtacggcatcat 442 M. musculus 786 95524 400 836 gtgtacctgtacagcctgct 443 M. musculus 787 95525 400 974 cagtgctggaccagcaatga 444 M. musculus 788 95527 400 1079 cttcttgtggccaagctgcg 446 M. musculus 789 95529 400 1100 gcccatcagatgcactatgc 448 M. musculus 790 95531 400 1256 ttcctcagctccttccaggg 450 M. musculus 791 95532 400 1292 ctctactgtttcctcaacaa 451 M. musculus 792 95533 400 1348 atggcaagaaggcaaagctc 452 M. musculus 793 95534 400 1360 caaagctcttcaggaggaaa 453 M. musculus 794 95535 400 1388 agcagccatggcagccacat 454 M. musculus 795 95536 400 1435 tccctgtgagaaacttcagc 455 M. musculus 796 95537 400 1450 tcagcttatgagtgcaggca 456 M. musculus 797 95538 400 1470 gcagcagtgggactggctgt 457 M. musculus 798 95539 400 1512 tggccagtagtctcccaagg 458 M. musculus 799 95540 400 1544 cccacctgaatctccacttg 459 M. musculus 800 95542 400 1575 ggttgtgttcaagaaagggc 461 M. musculus 801 95543 400 1600 aggacaacccagagccagat 462 M. musculus 802 95544 400 1610 agagccagatgcccggccaa 463 M. musculus 803 95545 400 1620 gcccggccaaggttgaagag 464 M. musculus 804 95546 400 1646 cagcaagacagcagcttgta 465 M. musculus 805 95547 400 1687 gtcctagcctggcacaggcc 466 M. musculus 806 95548 400 1724 gttggatatgatggagaagc 467 M. musculus 807 95549 400 1750 atctatgaactctgagtgtt 468 M. musculus 808 95550 400 1756 gaactctgagtgttcccatg 469 M. musculus 809 95551 400 1795 tacccagatatgtccttcag 470 M. musculus 810 95552 401 3953 cttggtctcgcctttgttac 471 M. musculus 811 95554 401 7321 accttcacagagctggtctg 473 M. musculus 812 95555 401 7505 ccatgcccagtgcagcaccg 474 M. musculus 813 95557 401 8766 gtcaggttgcccatccttgg 476 M. musculus 814 95560 11 10 tcatgaggccttgggcttgg 479 M. musculus 815 95561 11 85 ctggtcctctgcagcctgag 480 M. musculus 816

As these “preferred target segments” have been found by experimentation to be open to, and accessible for, hybridization with the antisense compounds of the present invention, one of skill in the art will recognize or be able to ascertain, using no more than routine experimentation, further embodiments of the invention that encompass other compounds that specifically hybridize to these preferred target segments and consequently inhibit the expression of glucagon receptor.

According to the present invention, antisense compounds include antisense oligomeric compounds, antisense oligonucleotides, ribozymes, external guide sequence (EGS) oligonucleotides, alternate splicers, primers, probes, and other short oligomeric compounds which hybridize to at least a portion of the target nucleic acid.

Example 17

Western Blot Analysis of Glucagon Receptor Protein Levels

Western blot analysis (immunoblot analysis) is carried out using standard methods. Cells are harvested 16-20 h after oligonucleotide treatment, washed once with PBS, suspended in Laemmli buffer (100 ul/well), boiled for 5 minutes and loaded on a 16% SDS-PAGE gel. Gels are run for 1.5 hours at 150 V, and transferred to membrane for western blotting. Appropriate primary antibody directed to glucagon receptor is used, with a radiolabeled or fluorescently labeled secondary antibody directed against the primary antibody species. Bands are visualized using a PHOSPHORIMAGER™ (Molecular Dynamics, Sunnyvale Calif.).

Example 18

Effects of Antisense Inhibition of Glucagon Receptor in Mice on Plasma Glucose Levels and Glucagon Receptor mRNA Reduction: Lean Animals, db/db Mice and ob/ob Mice

In accordance with the present invention, two antisense oligonucleotides targeted to the mouse glucagon receptor, ISIS 148359 (agcaggctta ggttgtggtg, SEQ ID NO: 408) and ISIS 180475 (gagctttgcc ttcttgccat, SEQ ID NO: 452), were evaluated for therapeutic efficacy in art-accepted mouse models of obesity and diabetes. Ob/ob mice have mutations in the leptin gene and are leptin-deficient, while db/db mice have mutations in the leptin receptor gene. The two strains exhibit obesity and diabetes strongly resembling Type 2 diabetes in humans. Tsang, S. H., 1998, P & S Medical Review, Vol. 5, No. 1.

Db/db and ob/ob mice were evaluated over the course of 4 weeks for the effects of ISIS 148359 and ISIS 180475 on serum glucose levels and glucagon receptor mRNA levels, while normoglycemic mice were evaluated for 2 weeks. Control animals received saline treatment (50 mg/kg). The normoglycemic mice were dosed subcutaneously twice a week for 2 weeks with 50 mg/kg of ISIS 148359, ISIS 180475 or saline. The db/db and ob/ob mice were dosed subcutaneously twice a week for 6 weeks with 25 mg/kg of ISIS 148359, ISIS 180475, saline, the positive control oligonucleotide ISIS 116847 (ctgctagcc tctggatttga, SEQ ID NO: 817) or the negative control oligonucleotide ISIS 141923 (ccttccctga aggttcctcc, SEQ ID NO: 818). The mice were monitored weekly for fed or fasted plasma glucose levels (fasted glucose measured 16 hr after last feeding) and upon termination of the experiment the level of glucagon receptor mRNA in the liver was determined. The data are summarized in Table 4.

TABLE 4 Effects of ISIS 148359 and ISIS 180475 treatment on fed and fasting glucose levels and glucagon receptor mRNA levels in normoglycemic mice, db/db mice, and ob/ob mice Biological ISIS # Antisense Marker mice (time day of Oligonucleotides Controls Measured course of study) treatment 148359 180475 saline 116847 141923 fed lean mice −6 221 216 210 N.D. N.D. plasma (2 week) 15 151 130 181 N.D. N.D. glucose db/db mice −1 294 295 296 295 304 mg/dL (4 weeks)  5 361 329 460 355 408 12 375 303 510 303 425 26 314 222 495 354 493 ob/ob mice −1 338 342 343 337 361 (4 weeks) 12 245 180 426 227 476 27 168 145 394 205 431 fasted db/db mice 19 336 232 320 321 298 plasma (4 weeks) 29 254 150 302 193 262 glucose ob/ob mice 19 205 132 317 167 245 mg/dL (4 weeks) 29 178 117 322 189 340 glucagon lean mice end 82 93 0 N.D. N.D. receptor (2 week) % mRNA db/db mice end 74 96 0  0  0 reduction (4 weeks) ob/ob mice end 86 97 0  10  0 (4 weeks)

These data demonstrate that the antisense oligonucleotides ISIS 148359 and ISIS 180475 targeted to glucagon receptor mRNA are capable of decreasing levels of glucagon receptor mRNA in mouse liver. These data further demonstrate that reduction of glucagon receptor expression is accompanied by a decrease in plasma glucose levels in normoglycemic mice, db/db mice and ob/ob mice. It is important to note that the treated mice become normoglycemic and do not become hypoglycemic. Antisense inhibitors of glucagon receptor are thus believed to be useful therapeutic modalities for treatment of hyperglycemia.

Example 19

Glucagon Receptor Antisense Ligonucleotides Lower Plasma Glucose in ob/ob Diabetic Mice—4 Week Study

C57Bl/eOlaHsd-Lepob (ob/ob) male mice were purchased from Harlan (Indianapolis, Ind., USA). Animals were acclimated for one week prior to study initiation. Mice were housed five per cage in polycarbonate cages with filter tops. Animals were maintained on a 12:12 hr light-dark cycle (lights on at 6:00 AM) at 21° C. All animals received de-ionized water ad libitum. ob/ob mice received Purina Diet 5015 ad libitum. Antisense compounds were prepared in normal saline, and the solution was sterilized through a 0.2 ,μm filter. Animals were dosed with antisense compound solutions or vehicle (saline) twice per week (separated by 3.5 days) via subcutaneous injection. Before the initiation of each study and once weekly during the study, blood was collected by tail clip without anesthesia into EDTA plasma tubes containing trasylol (Serologicals Proteins, Kankakee, Ill., USA) and dipeptidyl peptidase (DPP)-IV inhibitor (Linco Diagnostic Services, St. Charles, Mo., USA). Food intake and body weights were measured weekly. Plasma levels of glucose and triglycerides were determined on the Hitachi 912 clinical chemistry analyzer (Roche, Indianapolis, Ind., USA).

To test the efficacy of antisense inhibitors of glucagon receptor to treat hyperglycemia, 7-8 week-old ob/ob mice were dosed two times per week with antisense inhibitors of glucagon receptor [ISIS 148359 (SEQ ID NO: 408) or ISIS 180475(SEQ ID NO: 452)], a generic control oligonucleotide (ISIS 141923; SEQ ID NO: 818) whose sequence does not match any known transcripts in the mouse or rat genomes, a mismatch oligonucleotide (ISIS 298682; GCGATTTCCCGTTTTGACCT; SEQ ID NO: 819) whose sequence is identical to ISIS 180475 except for 7 internal bases, or saline twice a week (every 3.5 days) for 4 weeks. All oligonucleotides were administered at 25 mg/kg. Data are the mean values (±SEM where shown) of 8 mice per treatment group. Plasma glucose levels in all mice were approximately 330-370 mg/dl day zero. Whereas hyperglycemia worsened over time in saline- and control oligonucleotide-treated ob/ob mice, animals treated with glucagon receptor antisense compounds showed a dramatic reduction in plasma glucose. At day 12, plasma glucose levels in ob/ob mice treated with control oligonucleotide (ISIS 141923) and saline were approximately 472 and 425 mg/dl, respectively. Plasma glucose levels in mice treated with antisense oligonucleotides ISIS 148359 and ISIS 180475 were 240 and 180 mg/dl, respectively. At day 27, plasma glucose levels in ob/ob mice treated with control oligonucleotide (ISIS 141923) and saline were approximately 435 and 390 mg/dl, respectively. Plasma glucose levels in mice treated with antisense oligonucleotides ISIS 148359 and ISIS 180475 were 165 and 130 mg/dl, respectively. The latter is in the normal range.

A separate study, also using ob/ob mice (as well as db/db mice, lean mice, ZDF rats and lean rats) was also performed in which animals were also dosed subcutaneously every 3.5 days for a total of 9 doses of glucagon receptor antisense compound ISIS 180475 and one or more controls (unrelated control oligonucleotide ISIS 141923, mismatch control oligonucleotide ISIS 298682, and/or saline). The results of this study are shown in Table 5.

At the end of the 4-week treatment period, liver glucagon receptor mRNA was measured (normalized to total RNA in the same samples using Ribogreen) and was found to be reduced by 85-95%. Data are mean values of four mice per treatment group (P<0.05 using Student's t-test).

TABLE 5 Effects of antisense inhibition of glucagon receptor in rodents Plasma Plasma Plasma Glucose Triglycerides Plasma Insulin Glucagon Body Weight (g) (mg/dl) (mg/dl) (ng/ml) (pg/ml) ob/ob mice Saline 56.5 ± 1.5  564 ± 118 163 ± 25 35.9 ± 13.8 n.d. ISIS 180475 54.1 ± 1.6 122 ± 6* 129 ± 7  19.8 ± 9.5  n.d. db/db mice ISIS 141923 46.0 ± 0.7 571 ± 29 412 ± 33 n.d. 117 ± 12  ISIS 298682 43.9 ± 1.3 577 ± 65 448 ± 40 n.d. 131 ± 20  ISIS 180475 45.6 ± 0.6  241 ± 37*  121 ± 12* n.d. 3765 ± 952* db+/? lean mice ISIS 141923 28.0 ± 1.0 196 ± 12 121 ± 7  n.d. 80 ± 1  ISIS 180475 27.6 ± 1.0 164 ± 4*  83 ± 6* n.d. 362 ± 40* ZDF rats ISIS 141923 403 ± 12 417 ± 38  640 ± 105 5.0 ± 1.9 136 ± 7  ISIS 180475 404 ± 8   143 ± 15*  250 ± 25* 4.4 ± 0.5 548 ± 20* SD lean rats Saline 344 ± 4  116 ± 3  106 ± 26 2.7 ± 0.3 56 ± 11 ISIS 180475 327 ± 5  104 ± 7  139 ± 58  1.3 ± 0.2*  855 ± 122*
*P < 0.05.

n.d., not determined

Example 20

Glucagon Receptor Antisense Oligonucleotides Lower Plasma Glucose in db/db Diabetic Mice—4 Week Study

C57B1/KsOlaHsd-Lepdb (db/db) and lean (db+/+) male mice were purchased from Harlan (Indianapolis, Ind., USA). Animals were acclimated for one week prior to study initiation. Mice were housed five per cage in polycarbonate cages with filter tops. Animals were maintained on a 12:12 hr light-dark cycle (lights on at 6:00 AM) at 21° C. All animals received de-ionized water ad libitum. db/db mice received Purina Diet 5008 ad libitum. Antisense compounds were prepared in normal saline, and the solution was sterilized through a 0.2 μm filter. Animals were dosed with antisense compound solutions or vehicle (saline) twice per week (separated by 3.5 days) via subcutaneous injection. Before the initiation of each study and once weekly during the study, blood was collected by tail clip without anesthesia into EDTA plasma tubes containing trasylol (Serologicals Proteins, Kankakee, Ill., USA) and dipeptidyl peptidase (DPP)-IV inhibitor (Linco Diagnostic Services, St. Charles, Mo., USA). Food intake and body weights were measured weekly. Plasma levels of glucose and triglycerides were determined on the Hitachi 912 clinical chemistry analyzer (Roche, Indianapolis, Ind., USA.

To test the efficacy of antisense inhibitors of glucagon receptor to treat hyperglycemia, 7-8 week-old db/db mice were dosed two times per week with antisense inhibitors of glucagon receptor [ISIS 148359 (SEQ ID NO: 408) or ISIS 180475(SEQ ID NO: 452)], a generic control oligonucleotide (ISIS 141923) whose sequence does not match any known transcripts in the mouse or rat genomes, a mismatch oligonucleotide (ISIS 298682; SEQ ID NO: 819) whose sequence is identical to ISIS 180475 except for 7 internal bases, or saline for 4 weeks.

Glucose lowering efficacy and target reduction in db/db mice undergoing glucagon receptor antisense treatment were similar to those observed in similarly treated ob/ob mice; furthermore, plasma triglycerides were lowered from 412±33 to 121±12 mg/dl following glucagon receptor antisense treatment (Table 5). These results in db/db mice are similar to those reported in preliminary studies testing glucagon receptor antisense compound ISIS 148359 for 3 weeks [Osborne et al., 2003, Diabetes 52, A129 (abstract)].

Example 21

Glucagon Receptor Antisense Oligonucleotides Lower Plasma Glucose in ZDF Rats

ZDF/GmiCrl-fa/fa (ZDF) male rats were purchased from Charles River Laboratories (Wilmington, Mass., USA). Animals were acclimated for one week prior to study initiation. Rats were housed one per cage in polycarbonate cages with filter tops. Animals were maintained on a 12:12 hr light-dark cycle (lights on at 6:00 AM) at 21° C. All animals received de-ionized water ad libitum. ZDF rats received Purina Diet 5008 ad libitum. Antisense compounds were prepared in normal saline, and the solution was sterilized through a 0.2 μm filter. Seven-week old animals were dosed with antisense compound solutions or vehicle (saline) twice per week (separated by 3.5 days) via subcutaneous injection, for a total of 9 doses (last treatment on day 28), followed by a washout period of equal duration. Oligonucleotide concentration was 25 mg/kg of either glucagon receptor antisense oligonucleotide ISIS 180475 (SEQ ID NO: 452) or negative control oligonucleotide ISIS 141923 (SEQ ID NO: 818). Before the initiation of each study and once weekly during the study, blood was collected by tail clip without anesthesia into EDTA plasma tubes containing trasylol (Serologicals Proteins, Kankakee, Ill., USA) and dipeptidyl peptidase (DPP)-IV inhibitor (Linco Diagnostic Services, St. Charles, Mo., USA). Food intake and body weights were measured weekly. Glucagon receptor mRNA (target) was measured by real-time quantitative RT-PCR from livers of five animals removed from the study at each time point. Rat 36B4 ribosomal phosphoprotein mRNA (“18S RNA”) was measured and used to normalize RNA input. Data are the mean values of five rats per treatment group. In overall comparisons during the treatment period, target reduction by glucagon receptor antisense compound ISIS 180475 was significantly different when compared to control oligonucleotide-treated animals (P<0.05 adjusted using the Tukey method). Liver glucagon receptor mRNA decreased dramatically to 50% of controls within 24 hours after the first dose of ISIS 180475 and to 30% of controls 48 hr following the seventh dose.

For non-fasted plasma glucose levels, rats were treated as described above. Data are the mean values of five rats per treatment group. In overall comparisons during the treatment period, glucose lowering by the glucagon receptor antisense compound ISIS 180475 showed significant difference when compared to control oligonucleotide-treated animals. (P<0.05 adjusted using the Tukey method). The drop in plasma glucose paralleled the drop in glucagon receptor mRNA levels; there was a significant drop in plasma glucose within 48 hours after the initial glucagon receptor antisense dose. After 9 doses, the control oligonucleotide (ISIS 141923) treated rats had plasma glucose levels averaging approximately 417 mg/dl and antisense (ISIS 180475) treated rats had plasma glucose levels averaging approximately 143 mg/dl.

During the washout phase, hyperglycemia and glucagon receptor expression in liver began to rebound within 10 days, but even one month after the final dose, efficacy was still observed as plasma glucose and target mRNA levels in washout animals remained below pre-treatment levels. Glucose lowering achieved by the twice per week dosing schedule and the gradula rebound of glucagon receptor mRNA during the washout period are both consistent with the extended half lives of 2′-methoxyethoxy modified phosphorothioate oligonucleotides (typically ranging from 9 to 19 days according to published reports).

Non-fasted plasma insulin levels were also determined for rats treated as described above. Data are the mean values of five rats per treatment group. No significant changes were observed during the treatment period; however, individual comparisons between glucagon receptor antisense and control oligonucleotide treated animals on day 38 and 56 (washout period) were significant (P<0.05). Plasma insulin levels declined during the treatment phase in both control oligonucleotide and antisense-treated animals. During the washout phase of the control oligonucleotide treated group, insulin levels continued to decline as hyperglycemia progressed. This result is expected since beta-cell failure typically occurs in ZDF rats between 8 and 12 weeks of age. Interestingly, the mild elevation of glucose in glucagon receptor antisense-treated animals during the washout period resulted in a robust rise in plasma insulin to levels nearly as high as at start of study. This is consistent with evidence of preserved beta-cell function.

Example 22

Glucagon Receptor Antisense Oligonucleotides Do Not Cause Hyperglycemia or Hypoglycemia, in Spite of Hyperglucagonemia

In addition to effects on blood glucose, treatment with the antisense inhibitor of glucagon receptor (ISIS 180475; SEQ ID NO: 452) resulted in marked (and reversible) hyperglucagonemia in both normal and diabetic rodents (Table 5). This level of hyperglucagonemia is similar to that observed in glucagon receptor knockout mice (Parker et al., 2002, Biochem Biophys Res Commun. 290, 839-843; Gelling et al., 2003, Proc. Natl. Acad. Sci. USA., 100, 1438-1443. Because of these high levels of serum glucagon, it was important to determine whether the antisense inhibitors of glucagon receptor might induce hyperglycemia, particularly as hepatic glucagon receptor levels gradually return to normal following treatment withdrawal. It is therefore significant that at no time during the treatment or washout periods did animals with hyperglucagonemia exhibit hyperglycemia. In fact, glucagon receptor antisense-treated animals showed a moderate decrease in fed plasma glucose at all time points tested.

It was also confirmed that antisense treatment also did not cause hypoglycemia. After 4 weeks of antisense treatment, by which time maximum reduction of glucagon receptor expression had been achieved, db+/? lean mice were subjected to periods of fasting of up to 24 hours. Although the glucagon receptor antisense-treated mice displayed a 10-30% reduction in plasma glucose, at no time did the animals reach adverse levels of hypoglycemia. This is in contrast to Gcgr knockout mice, which become hypoglycemic during periods of fasting. Gelling et al., 2003, Proc Natl. Acad. Sci. U.S.A. 100, 1438-1443.

Example 23

Glucagon Receptor mRNA is Reduced in Islets of Antisense-treated db/db Mice

Pancreatic islets were isolated from 12-week-old male db/db mice (n=5-6 per treatment group) that had been treated twice per week (every 3.5 days) by subcutaneous injection with saline or glucagon receptor antisense oligonucleotide ISIS 180475 (SEQ ID NO: 452) at 25 mg/kg for a total of 9 doses. Mice were sacrificed by cervical dislocation. The common bile duct was cannulated with a 27-gauge needle and the pancreas was distended with 3 ml of Hank's buffer (Sigma, Taufkirchen, Germany) containing 2% bovine serum albumin (Applichem, Darmstadt, Germany) and 1 mg/ml collagenase (Serva, Heidelberg, Germany). Subsequently, the pancreas was removed and digested in Hank's buffer at 37° C. Islets were purified on a Histopaque-1077™ (Sigma) gradient for 15 min at 750×g. Islets were cultured overnight in RPMI-1640 medium containing 10% FBS, 100 U/ml penicillin, and 100 μg/ml streptomycin (Invitrogen, Karlsruhe, Germany). 200 islets from 3 individuals were pooled to give one sample for RNA extraction. Real-time quantitative RT-PCR was used to profile gene expression. Islet glucagon receptor mRNA levels were decreased by approximately 75% in antisense-treated animals compared to saline-treated controls. It should be noted that, in addition to pharmacologic effect of the antisense compound, a compensatory response to hyperglucagonemia or the increased alpha-cell populations in treated animals could contribute to the results observed.

Example 24

Glucagon Receptor Antisense Oligonucleotides Decrease the Number of Functional Glucagon Receptors

To assess whether the reduction in glucagon receptor mRNA correlates with a reduction in functional glucagon receptor number, a homologous competition assay was performed using hepatocyte membranes prepared from mice treated with control or glucagon receptor antisense compounds. 125I-glucagon binding was effectively competed by increasing concentrations of unlabeled glucagon in control membrane samples. 15-20 μg of membrane from control oligonucleotide- or glucagon receptor oligonucleotide (ISIS 180475; SEQ ID NO: 452)-treated db/db mice were incubated with 0.1 nM 125I-glucagon (2000 Ci/mmol, PerkinElmer, Boston, Mass., USA) and the indicated concentrations of unlabeled glucagon (Eli Lilly and Company, Indianapolis, Ind., USA) in buffer containing 50 mM Hepes, 1 mM MgCl2, 5 mM EGTA, 0.005% Tween 20, 0.1% BSA, and EDTA-free protease inhibitor cocktail (Roche). Assays were performed under steady state conditions in the presence of excess labeled ligand on 96-well MultiScreen-HV 0.45 ,μm filter plates (Millipore, Bedford, Mass., USA). Following incubation for 2 hrs at room temperature, plates were rapidly washed by filtration with ice-cold buffer (20 mM Tris, pH 7.4) and dried for 45 min at 50° C. Following the addition of Optiphase Supermix (PerkinElmer), plates were counted on a Wallac Microbeta scintillation counter. Data analyses were performed using GraphPad Prism software and expressed as mean +/−SEM. Data obtained for samples from animals treated with glucagon receptor oligonucleotide neared the limits of detection for the assay and curve-fitting parameters. In order to derive a numerical value for apparent Bmax, the Kd was fixed at the average value (0.69+/−0.2 nM) obtained from the samples from the control antisense-treated animals.

Functional GCGR expression was found to be decreased approximately 85% by glucagon receptor antisense treatment and is in accord with quantitative RT-PCR results.

Example 25

Antisense Inhibitors of Human and Monkey Glucagon Receptor—Dose Response

Based on the screen in Example 15 above, a subset of human glucagon receptor antisense oligonucleotides were chosen for further study. Dose-response studies were conducted for ISIS 315186, 310457, 315324, 315278, 315181, 315297, 315163 and 310456 in both human HepG2 cell cultures and in cynomolgus monkey primary hepatocytes. These six compounds are homologous to both human and cynomolgus monkey glucagon receptor nucleic acid targets. The universal control ISIS 29848 (NNNNNNNNNNNNNNNNNNN; SEQ ID NO: 820, where N is an equimolar mixture of A, C, G and T, a chimeric 2′ MOE gapmer with a phosphorothioate backbone and with MOEs at positions 1-5 & 16-20) was used as negative control.

The human hepatoblastoma cell line HepG2 was obtained from the American Type Culture Collection (Manassas, Va.). HepG2 cells are routinely cultured in Eagle's MEM supplemented with 10% fetal calf serum, non-essential amino acids, and 1 mM sodium pyruvate (Gibco/Life Technologies, Gaithersburg, Md.). Cells are routinely passaged by trypsinization and dilution when they reach 90% confluence.

Primary cynomolgus monkey hepatocytes were obtained from CellzDirect (Los Angeles) and plated onto collagen-coated 24-well plates (Costar) at a density of 75,000 cells/well. The culturing medium for these hepatocytes was William's E media (Invitrogen) supplemented with 10% FBS (Invitrogen). Cells were allowed to attach overnight and were then treated with oligonucleotide-Lipofectin mixture for 4 hours. The oligonucleotide-Lipofectin mixture was washed off and then cells were incubated in normal medium.

Cells were treated with oligonucleotide for 20 hours at doses of 1, 5, 10, 25, 50, 100 nM for HepG2 cells and 5, 10, 25, 50, 100 and 200 nM for primary monkey hepatocytes (n=3). RNA was analyzed by RT-PCR to determine percent inhibition of glucagon receptor expression compared to control (ISIS 29848), at each oligonucleotide concentration. The results were plotted to give the IC50, the dose of oligonucleotide which results in 50% reduction of glucagon receptor mRNA levels. Results are shown in Table 6.

TABLE 6 IC50s of glucagon receptor antisense oligonucleotides in human HepG2 cells and in cynomolgus monkey primary hepatocytes (in nM) SEQ ID IC50 in human IC50 in monkey ISIS # NO: HepG2 cells (nM) hepatocytes (nM) 315186 254 5 30 310457 184 7 10 315324 392 6 19 315278 346 8 25 315181 249 9 32 315297 365 11 11 315163 231 19 37 310456 183 25 20

Based on these results, three compounds (ISIS 315297, 310457 and 315163) were chosen for study in monkeys.

Example 26

Dose-ranging Study of Antisense Inhibition of Glucagon Receptor Expression in Cynomolgus Monkeys

A monkey study was performed at SNBL USA, Ltd., Everett, Wash. Forty mature (6-8 years old) male Macaca fascicularis (purpose-bred cynomolgus monkeys) weighing approximately 5-10 kg at the initiation of dosing were used. The animals were individually housed in primary climate-controlled enclosures conforming to the Animal Welfare Act. Animals were offered Purina Mills Laboratory Profiled Fiber Plus® Monkey Diet (Animal Specialties, Hubbard, Oreg.). Occasional fresh fruit and vegetable treats were also offered. Fresh drinking water was available to all animals, ad libitum. Before fasting measurements, food was removed from enclosures between 1630 and 1700 on the afternoon before the scheduled blood draw. After the animals were fasted for at least 16 hours, blood samples for plasma analysis were collected BEFORE dosing or feeding. For fasting analysis, approximately 2.3-2.5 mL of blood was drawn from a peripheral vein. Approximately 1.8 to 2.0 mL was be deposited into a K2-EDTA tube containing DPP-IV inhibitor at 10 μL/mL of blood and trasylol at 250 KIU/mL of blood. Approximately 0.5 mL was deposited into a lithium heparin tube. Once the blood was been deposited into the EDTA plus additives tube, it was inverted to mix and placed on ice within 5 minutes. The blood in the lithium heparin tube was also placed on ice within 5 minutes. Blood samples were centrifuged (2000×g, 15 minutes at 4 to 8° C.) to obtain plasma within 30 minutes of sample collection. The plasma was frozen at or below −70° C. Samples were shipped on dry ice via overnight courier as described below for subsequent analysis. For non-fasted analysis, the animals were given their AM feeding (between 0830 and 0930) on the day of the blood draw. Ninety minutes after feeding, blood was drawn. The number of biscuits remaining were counted at the time of the blood draw. Samples are prepared and shipped as above.

Monkeys were dosed subcutaneously for 10 weeks with ISIS 315297, ISIS 310457 or ISIS 315163 at three concentrations. In week 1, oligonucleotides were given at 2.0, 5.0 and 20 mg/kg/dose (Day 1, 3 and 5); in week 2 through 10, oligonucleotides were given at 1.0, 2.5 and 10 mg/kg/dose (twice weekly starting at Day 8) The larger dose (6, 15 or 60 mg/kg/week, i.e. 3 injections of 2, 5 or 20 mg/kg) was given in week 1 in order to rapidly achieve the desired steady state oligonucleotide concentration. In week 1, compounds were administered 3 times, every other day; for weeks 2-10, compounds were administered twice per week, with at least 2 days between dosings.

Oligonucleotides were given by subcutaneous (SC) injection, using volumes of 0.1-0.3 ml/kg). For each dosing of each animal, the appropriate volume of the relevant ASO solution or of the control/vehicle article was administered subcutaneously using a syringe and needle (27G). The total volume of the relevant dosing solution or the control article was calculated on the basis of the animal's most recent body weight. Multiple injection sites on the upper back (intra-scapular region) of each monkey were employed. During acclimation the skin of the upper back was be shaved and a clock-like grid (points at 12, 3, 6, and 9 o'clock) was tattooed on each animal. Injection points were a minimum of 5 cm apart. The injection sites were rotated so that each site was used for fourth dose, starting with 12 o'clock and rotating clockwise. The needle was inserted away from the dot and angled so that the dose was deposited underneath the dot.

After the 10 week study (approx. 2 days after last dose), animals were euthanized and three 1 to 4 gram samples of liver tissue were removed and individually snap frozen over liquid nitrogen; alternatively, biopsies could be taken from living animals and frozen. Frozen tissues were homogenized in 4M guanidinium isothiocyanate solution and subjected to CsCl centrifugation (150,000×g for 16 hr at 18° C.). The supernatant was removed and the RNA pellet was resuspended in water, following which it was applied to RNEASY mini columns (Qiagen, Valencia Calif.). After purification and quantitation, the tissues were subjected to RT-PCR analysis as described in previous examples using the following primers and probe:

Forward primer- ACTGCACCCGCAACGC (SEQ ID NO: 821) Reverse primer- CACGGAGCTGGCCTTCAG (SEQ ID NO: 822) Probe- ATCCACGCGAACCTGTTTGTGTCCTT (SEQ ID NO: 823)

RNA amounts were normalized to 18S RNA levels in the tissue. Results are shown in Table 7, as percent reduction in glucagon receptor mRNA in antisense-treated monkeys compared to saline-treated monkeys.

TABLE 7 Glucagon receptor mRNA reduction in monkey liver after treatment with antisense inhibitors of glucagon receptor - RT-PCR expt 1 SEQ % reduction % reduction % reduction ISIS # ID NO: at 2 mg/kg At 5 mg/kg at 20 mg/kg 310457 184 17 31 64 315297 365 2 21 49 315163 231 22 18 47

RNA analysis of the same tissue samples by RT-PCR was repeated independently using the same primer-probe set as above. Results are shown in Table 8 as percent reduction in glucagon receptor mRNA in antisense-treated monkeys compared to saline-treated monkeys.

TABLE 8 Glucagon receptor mRNA reduction in monkey liver after treatment with antisense inhibitors of glucagon receptor -RT-PCR expt 2 SEQ ID % reduction % reduction % reduction ISIS # NO: at 2 mg/kg At 5 mg/kg at 20 mg/kg 310457 184 25 23 63 315297 365 18 21 56 315163 231 25 29 44

The results obtained by RT-PCR were confirmed by Northern blot analysis according to standard methods (Example 14). The cDNA probe that was used for northern blots was a 900-base fragment of monkey GCGR generated by RT-PCR from cynomolgus monkey liver. Results are shown in Table 9.

TABLE 9 Glucagon receptor mRNA reduction in monkey liver after treatment with antisense inhibitors of glucagon receptor - Northern blot SEQ ID % reduction % reduction % reduction ISIS # NO: at 2 mg/kg At 5 mg/kg at 20 mg/kg 310457 184 8 16 65 315297 365 0 10 38 315163 231 8 30 27

Blood glucose levels were measured in monkeys after treatment with antisense inhibitors of glucagon receptor. Glucose readings were performed using a drop of blood from the blood samples collected as above and read on a One Touch Profile® (Lifescan Inc., a Johnson and Johnson Company). Because normoglycemic (nondiabetic) monkeys were used in this study, no significant changes in blood glucose levels were expected or observed. At no point did animals become hypoglycemic after antisense treatment.

Glucagon levels were measured in plasma of fasted monkeys before (baseline) and after treatment for 5 weeks or 10 weeks with antisense inhibitors of glucagon receptor. Monkeys were anesthetized prior to blood collection to avoid artifacts due to stress. Glucagon levels were determined by radioimmunoassay, ELISA and/or Luminex immunoassay by contract laboratory (Linco, St. Charles Mo.). Results are shown in Table 10.

TABLE 10 Fasted glucagon levels in monkey liver after treatment with antisense inhibitors of glucagon receptor Glucagon Glucagon SEQ Antisense Glucagon (pg/ml) (pg/ml) ID Dose (pg/ml) Week 5 Week 10 ISIS # NO: (mg/kg) Baseline fasted fasted Saline 155 ± 31 150 ± 19  250 ± 141 310457 184 2  487 ± 123 278 ± 54 189 ± 36 5 211 ± 25 179 ± 55 169 ± 26 20 308 ± 77  580 ± 247 1247 ± 451 315297 365 2 410 ± 94 140 ± 42 133 ± 26 5 519 ± 58 193 ± 31 204 ± 30 20 375 ± 87 209 ± 23 276 ± 67 315163 231 2 176 ± 42 152 ± 33 143 ± 26 5 262 ± 76 203 ± 74 225 ± 95 20 251 ± 40 257 ± 76  421 ± 197

Glucagon-like peptide 1 (GLP-1) levels were measured in plasma of fasted monkeys before (baseline) and after treatment for 5 weeks or 10 weeks with antisense inhibitors of glucagon receptor. Monkeys were anesthetized prior to blood collection to avoid artifacts due to stress. GLP-1 levels were determined by radioimmunoassay, ELISA and/or Luminex immunoassay by contract laboratory (Linco, St. Charles Mo.). Results are shown in Table 11.

TABLE 11 Fasted GLP-1 levels in monkey liver after treatment with antisense inhibitors of glucagon receptor SEQ Antisense GLP-1 ID Dose (pM) GLP-1 (pM) GLP-1 (pM) ISIS # NO: (mg/kg) Baseline Week 5 fasted Week 10 fasted Saline 4 ± 2 4 ± 1 5 ± 1 310457 184 2 4 ± 1 3 ± 1   3 ± .41 5 4 ± 1   4 ± .48 3 ± 1 20 8 ± 3 17 ± 6  30 ± 15 315297 365 2 3 ± 1 3 ± 1   3 ± .29 5 4 ± 1 4 ± 1 4 ± 2 20 11 ± 8  9 ± 4 7 ± 5 315163 231 2 4 ± 1 4 ± 1 4 ± 1 5 3 ± 1 5 ± 2 3 ± 1 20 2 ± 0   4 ± .48 5 ± 1

Claims

1. A compound comprising 8 to 80 nucleobases targeted to the region comprising nucleotides 545 to 576, 681 to 714, or 860 to 891 of SEQ ID NO: 4.

2. The compound of claim 1, wherein the region comprises nucleotides 545 to 576 and wherein the compound comprises at least an 8-nucleobase portion of SEQ ID NO: 383, 365, 343, 187, 358, 270, 396, 41, 252, 336, 251, 188 or 351.

3. The compound of claim 1, wherein the region comprises nucleotides 545 to 576 and wherein the compound consists of SEQ ID NO: 383, 365, 343, 187, 358, 270, 396, 41, 252, 336, 251, 188 or 351.

4. The compound of claim 1, wherein the region comprises nucleotides 681 to 714 and wherein the compound comprises at least an 8-nucleobase portion of SEQ ID NO: 48, 192, 330, 121, 231, 193, 291, 248, 368, 194, 376, 392, 297 or 195.

5. The compound of claim 1, wherein the region comprises nucleotides 681 to 714 and wherein the compound consists of SEQ ID NO: 48, 192, 330, 121, 231, 193, 291, 248, 368, 194, 376, 392, 297 or 195.

6. The compound of claim 1, wherein the region comprises nucleotides 860 to 891 and wherein the compound comprises at least an 8-nucleobase portion of SEQ ID NO: 198, 124, 281, 293, 199, 374, 51, 373, 200, 347, 312, 254, 52.

7. The compound of claim 1, wherein the region comprises nucleotides 860 to 891 and wherein the compound consists of SEQ ID NO: 198, 124, 281, 293, 199, 374, 51, 373, 200, 347, 312, 254, 52.

8. The compound of claim 1 which is at least 70% complementary to SEQ ID NO: 4.

9. The compound of claim 1 which is at least 80% complementary to SEQ ID NO: 4.

10. The compound of claim 1 which is at least 90% complementary SEQ ID NO: 4.

11. The compound of claim 1 which is at least 95% complementary to SEQ ID NO: 4.

12. The compound of claim 1 which is 12 to 50 nucleobases in length.

13. The compound of claim 1 which is 15 to 30 nucleobases in length.

14. The compound of claim 1 which is 20 nucleobases in length.

15. The compound of claim 1 comprising an oligonucleotide.

16. The compound of claim 15, wherein the oligonucleotide is DNA.

17. The compound of claim 15, wherein the oligonucleotide is RNA.

18. The compound of claim 15, wherein the oligonucleotide is chimeric.

19. The compound of claim 15 wherein at least a portion of said compound hybridizes with RNA to form an oligonucleotide-RNA duplex.

20. The compound of claim 15, wherein the oligonucleotide is single-stranded.

21. The compound of claim 1 comprising at least one chemical modification.

22. The compound of claim 21 comprising at least one modified internucleoside linkage, sugar moiety, or nucleobase.

23. The compound of claim 21 comprising at least one 2′-O-methoxyethyl sugar moiety.

24. The compound of claim 21 comprising at least one phosphorothioate internucleoside linkage.

25. The compound of claim 21 comprising at least one 5-methylcytosine.

26. The compound of claim 18, wherein every internucleoside linkage is a phosphorothioate linkage, nucleobases 1-5 and 16-20 comprise a 2′-O-methoxyethyl modification and every cytidine residue comprises a 5-methyl modification.

27. The compound of claim 1, wherein the compound is a sodium salt.

28. A pharmaceutical composition comprising the compound of claim 1 and a pharmaceutical carrier, diluent or excipient.

29. The composition of claim 28 comprising a colloidal dispersion system.

30. A kit or assay device comprising the compound of claim 1.

31. A method of inhibiting the expression of human glucagon receptor in cells or tissues comprising contacting said cells or tissues with the compound of claim 1 so that expression of human glucagon receptor is inhibited.

32. A method of treating or delaying the onset of a disease or condition associated with glucagon receptor in a human comprising administering to said human a therapeutically or prophylactically effective amount of the compound of claim 1 so that expression of human glucagon receptor is inhibited.

33. The method of claim 28 wherein the disease or condition is a metabolic disease or condition.

34. The method of claim 29, wherein the disease or condition is diabetes, obesity primary hyperglucagonemia, insulin deficiency or insulin resistance.

35. The method of claim 30 wherein the disease or condition is Type 2 diabetes.

36. The method of claim 29, wherein the disease or condition is associated with elevated blood glucose levels, elevated blood triclyceride levels or elevated blood cholesterol levels.

37. A method of decreasing blood glucose levels in a human comprising administering to said human the compound of claim 1.

38. The method of claim 31 wherein the blood glucose levels are plasma glucose levels.

39. The method of claim 31 wherein the human has diabetes or is obese.

40. A method of preventing or delaying the onset of an increase in blood glucose levels in a human comprising administering to said human the compound of claim 1.

41. The method of claim 33 wherein the human suffers from diabetes, obesity or insulin resistance.

42. The method of claim 33 wherein the blood glucose levels are plasma glucose levels.

Patent History
Publication number: 20070238688
Type: Application
Filed: Apr 5, 2007
Publication Date: Oct 11, 2007
Inventors: Sanjay Bhanot (Carlsbad, CA), Susan Freier (San Diego, CA), Kenneth Dobie (Del Mar, CA), Robert McKay (Poway, CA)
Application Number: 11/697,270
Classifications
Current U.S. Class: 514/44.000; 536/23.100
International Classification: A61K 31/711 (20060101); A61K 31/7105 (20060101); A61K 31/7125 (20060101); C07H 21/02 (20060101); C07H 21/04 (20060101);