WO2007131232A2 - Compositions and their uses directed to ptpr alpha - Google Patents

Compositions and their uses directed to ptpr alpha Download PDF

Info

Publication number
WO2007131232A2
WO2007131232A2 PCT/US2007/068390 US2007068390W WO2007131232A2 WO 2007131232 A2 WO2007131232 A2 WO 2007131232A2 US 2007068390 W US2007068390 W US 2007068390W WO 2007131232 A2 WO2007131232 A2 WO 2007131232A2
Authority
WO
WIPO (PCT)
Prior art keywords
compound
ptpr
alpha
levels
antisense
Prior art date
Application number
PCT/US2007/068390
Other languages
French (fr)
Other versions
WO2007131232A8 (en
WO2007131232A3 (en
Inventor
Sanjay Bhanot
Susan F. Murray
Original Assignee
Isis Pharmaceuticals, Inc.
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Isis Pharmaceuticals, Inc. filed Critical Isis Pharmaceuticals, Inc.
Priority to EP07797356A priority Critical patent/EP2023936A4/en
Priority to US12/299,610 priority patent/US8158598B2/en
Publication of WO2007131232A2 publication Critical patent/WO2007131232A2/en
Publication of WO2007131232A3 publication Critical patent/WO2007131232A3/en
Publication of WO2007131232A8 publication Critical patent/WO2007131232A8/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • C12N15/1138Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing against receptors or cell surface proteins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/16Drugs for disorders of the alimentary tract or the digestive system for liver or gallbladder disorders, e.g. hepatoprotective agents, cholagogues, litholytics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P3/00Drugs for disorders of the metabolism
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/12Antidiuretics, e.g. drugs for diabetes insipidus
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/31Chemical structure of the backbone
    • C12N2310/315Phosphorothioates
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3212'-O-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/3222'-R Modification
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/32Chemical structure of the sugar
    • C12N2310/323Chemical structure of the sugar modified ring structure
    • C12N2310/3231Chemical structure of the sugar modified ring structure having an additional ring, e.g. LNA, ENA
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/33Chemical structure of the base
    • C12N2310/334Modified C
    • C12N2310/33415-Methylcytosine
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/341Gapmers, i.e. of the type ===---===
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/30Chemical structure
    • C12N2310/34Spatial arrangement of the modifications
    • C12N2310/345Spatial arrangement of the modifications having at least two different backbone modifications

Definitions

  • proteins can be phosphorylated on serine, threonine or tyrosine residues and the extent of phosphorylation is regulated by the opposing action of phosphatases, which remove the phosphate moieties. .
  • PTPR.alpha also known as protein tyrosine phosphatase receptor type alpha, LCA- related phosphatase; LRP, HLPR, HPTPA, PTPRA, PTPA, PTP-R alpha, PTPRL2 and RPTPA
  • LRP protein tyrosine phosphatase receptor type alpha, LCA- related phosphatase
  • LRP protein tyrosine phosphatase receptor type alpha, LCA- related phosphatase
  • LRP HLPR
  • HPTPA protein tyrosine phosphatase receptor type alpha, LCA- related phosphatase
  • PTPRA protein tyrosine phosphatase receptor type alpha, LCA- related phosphatase
  • PTPA protein tyrosine phosphatase receptor type alpha, LCA- related phosphatase
  • PTPRL2 and RPTPA receptor-like phosphatases
  • chromosome 20pl 3 a chromosomal region involved in translocations and deletions in myeloid disorders and neoplasms (Jink et al., Cytogenet. Cell Genet., 1992, 60, 117-118; Kaplan et al., Proc. Natl. Acad. Sci. U. S. A., 1990, 87, 7000-7004).
  • RNA transcripts of approximately 4.3 and 6.3 kb (Kaplan et al., Proc. Natl. Acad. Sci. U. S. A., 1990, 87, 7000- 7004) whose proteins differ in a stretch of 9 amino acid residues situated in the external juxtamembrane segment (Daum et al., J. Biol. Chem., 1994, 269, 10524-10528).
  • PTPR.alpha. has been identified as a negative regulator of insulin receptor signaling indicating that it may play a key role in insulin action and in the pathophysiology of non-insulin-dependent diabetes (Moeller et al., J. Biol. Chem., 1995, 270, 23126-23131). However, the importance of PTPR.alpha.'s role in insulin signaling is considered unclear. (Cheng, A., et al, Eur. J. Biochem. (2002) 269: 1050-1059).
  • antisense compounds particularly oligomeric compounds, especially nucleic acid and nucleic acid-like oligomers, which are targeted to a nucleic acid encoding PTPR.alpha.
  • the antisense compounds are antisense oligonucleotides targeted to PTPR.alpha., particularly human PTPR.alpha., and are capable of modulating the expression of PTPR.alpha..
  • Prefered compounds comprise at least an 8 nucleobase portion, preferably a 12 nucleobase portion, more preferably at least a 15 nucleobase portion, of the sequences listed in Table 3 or 4, or are at least 80% identical to the complement of validated target segments, or to the antisense oligonucleotide sequences listed in Table 3 or 4.
  • PTPR.alpha. in cells or tissues comprising contacting the cells with at least one antisense compound, as described, and analyzing the cells for indicators of a decrease in expression of PTPR.alpha. rnRNA and/or protein by direct measurement of rnRNA and/or protein levels, and/or indirect indicators of a disease or condition such as measuring plasma triglyceride levels, plasma glucose levels, plasma transaminase levels, plasma insulin levels, hepatic triglyceride levels or similar indicators of type 2 diabetes, fatty liver, obesity or metabolic syndrome.
  • Provided are methods for the prevention, amelioration, and/or treatment of type 2 diabetes, fatty liver disease or metabolic syndrome comprising administering at least one antisense compound of this invention to an individual in need of such intervention.
  • a medicament for the prevention, amelioration, and/or treatment of a disease or condition, especially a those associated with at least one indicator of type 2 diabetes, fatty liver disease, obesity or metabolic syndrome.
  • Any one of the antisense compounds described herein can be targeted to at least two or more nucleic acid molecules encoding human PTPR.alpha.
  • any one of the compounds described herein wherein the at least one modified sugar moiety comprises a 2'-MOE , a LNA, a 2'-OMe, an ENA, a 2'-F or combinations thereof.
  • a pharmaceutical composition comprising one or more compounds described herein and a pharmaceutically acceptable penetration enhancer, carrier, or diluent.
  • a method for the prevention, amelioration, or treatment of a disease or condition wherein indicators of the disease or condition comprise increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels, increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereof comprising administration of one or more compounds described herein to an individual in need of such intervention.
  • the methods described herein wherein the disease or condition is type-2 diabetes and wherein administration of one or more compounds described herein results in a decrease in plasma glucose levels, an improvement in insulin sensitivity or combinations thereof.
  • the methods described herein wherein the disease or condition is metabolic syndrome and wherein administration of one or more compounds described herein results in a decrease in plasma glucose levels, a decrease in plasma lipid levels, a decrease in hepatic triglyceride levels, an improvement
  • I condition comprise increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels , increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereofwherein the medicament reduces the expression of a nucleic acid molecule encoding PTPR.alpha..
  • antisense compounds optionally further comprises a complementary strand 15 to 35 nucleobases in length.
  • Figures Ia and Ib illustrate that administration of the disclosed antisense compounds is useful for clearing fat from the liver.
  • Figure Ia is an oil red stained liver section of an ob/ob mouse treated with saline and showing no clearing.
  • Figure Ib is an oil red stained liver section of an ob/ob mouse treated with an antisense inhibitor of PTPR.alpha and showing clearing of fat from the liver.
  • Figure 2 is a graph illustrating weekly body weights for DIO mice receiving treatment oligo, control oligo or saline.
  • Figure 3 is a graph illustrating food intake for DIO receiving treatment oligo, control oligo or saline.
  • antisense compounds for the prevention, amelioration, and /or treatment of diseases or conditions associated with increases in plasma triglyceride levels, plasma glucose levels, plasma transaminase levels, plasma insulin levels hepatic triglyceride levels and further associated with PTPR.alpha activity.
  • prevention means to delay or forestall onset or development of a condition or disease for a period of time from hours to days, preferably weeks to months.
  • amelioration means a lessening of at least one indicator of the severity of a condition or disease. The severity of indicators may be determined by subjective or objective measures which are known to those skilled in the art.
  • treatment means to administer an antisense composition to effect an alteration or improvement of the disease or condition.
  • Prevention, amelioration, and/or treatment may require administration of multiple doses at regular intervals, or prior to exposure to an agent to alter the course of the condition or disease.
  • a single agent may be used in a single individual for each prevention, amelioration, and treatment of a condition or disease sequentially, or concurrently.
  • antisense compounds including antisense oligonucleotides and other antisense compounds for use in modulating the expression of nucleic acid molecules encoding PTPR.alpha.. This is accomplished by providing antisense compounds that hybridize with one or more target nucleic acid molecules encoding PTPR.alpha..
  • target nucleic acid and “nucleic acid molecule encoding PTPR.alpha. " have been used for convenience to encompass RNA (including pre-mRNA and mRNA or portions thereof) transcribed from DNA encoding PTPR.alpha., and also cDNA derived from such RNA.
  • the target nucleic acid is an mRNA encoding human PTPR.alpha.. Target Nucleic Acids
  • Targeting an antisense compound to a particular target nucleic acid molecule can be a multistep process. The process usually begins with the identification of a target nucleic acid whose expression is to be modulated.
  • the target nucleic acid can be 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.
  • the target nucleic acid encodes PTPR.alpha..
  • 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. Variants can result in mRNA variants including, but not limited to, those with alternate splice junctions, or alternate initiation and termination codons. Variants in genomic and mRNA sequences can result in disease. . Target Names, Synonyms, Features
  • Modulation of expression of a target nucleic acid can be achieved through alteration of any number of nucleic acid (DNA or RNA) functions.
  • “Modulation” or “Modulation of Expression” means a perturbation of function, for example, either an increase (stimulation or induction) or a decrease
  • modulation of expression can include perturbing splice site selection of pre-mRNA processing. "Expression” includes all the functions by which a gene's coded information is converted into structures present and operating in a cell. These structures include the products of transcription and translation.
  • the functions of RNA to be modulated can include translocation functions, which include, but are not limited to, 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, and translation of protein from the RNA.
  • RNA processing functions that can be modulated include, but are not limited to, splicing of the RNA to yield one or more RNA species, capping of the RNA, 3' maturation of the RNA and catalytic activity or complex formation involving the RNA which may be engaged in or facilitated by the RNA.
  • Modulation of expression can result in the increased level of one or more nucleic acid species or the decreased level of one or more nucleic acid species, either temporally or by net steady state level.
  • modulation of expression can mean increase or decrease in target RNA or protein levels.
  • modulation of expression can mean an increase or decrease of one or more RNA splice products, or a change in the ratio of two or more splice products.
  • 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 and can be routinely determined using, for example, PCR or Northern blot analysis.
  • Cell lines are derived from both normal tissues and cell types and from cells associated with various disorders (e.g. hyperproliferative disorders). Cell lines derived from multiple tissues and species can be obtained from American Type Culture Collection (ATCC, Manassas, VA) and other public sources.
  • Primary cells, or those cells which are isolated from an animal and not subjected to continuous culture can be prepared according to methods known in the art, or obtained from various commercial suppliers. Additionally, primary cells include those obtained from donor human subjects in a clinical setting (i.e. blood donors, surgical patients). These techniques are well known to those skilled in the art. Assaying Modulation of Expression
  • PTPR.alpha. mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR.
  • PCR competitive polymerase chain reaction
  • RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA by methods known in the art. Methods of RNA isolation are taught in, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.1.1-4.2.9 and 4.5.1-4.5.3, John Wiley & Sons, Inc., 1993.
  • RNA blot analysis is routine in the art and is taught in, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.2.1-4.2.9, John Wiley & Sons, Inc., 1996.
  • Real-time quantitative (PCR) can be conveniently accomplished using the commercially available ABI PRISMTM 7700 Sequence Detection System, available from PE-Applied Biosystems, Foster City, CA and used according to manufacturer's instructions.
  • the method of analysis of modulation of RNA levels is not a limitation of the instant invention.
  • Levels of a protein encoded by PTPR.alpha. can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), ELISA or fluorescence-activated cell sorting (FACS).
  • Antibodies directed to a protein encoded by PTPR.alpha. can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, MI), or can be prepared via conventional antibody generation methods. Methods for preparation of polyclonal antisera are taught in, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 2, pp.
  • Immunoprecipitation methods are standard in the art and can be found at, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.16.1-10.16.11, John Wiley & Sons, Inc., 1998.
  • Western blot (immunoblot) analysis is standard in the art and can be found at, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.8.1-10.8.21, John Wiley & Sons, Inc., 1997. Active Target Segments
  • active target segments The locations on the target nucleic acid defined by having one or more active antisense compounds targeted thereto are referred to as "active target segments.” There may be substantial variation in activity (e.g., as defined by percent inhibition) of the antisense compounds within an active target segment.
  • Active antisense compounds are those that are determined to modulate the expression of their target RNA.
  • active antisense compounds inhibit expression of their target RNA at least about 50%, more preferably at least about 70% and most preferably at least about 80%.
  • the level of inhibition required to define an active antisense compound is defined based on the results from the screen used to define the active target segments. Those skilled in the art understand that the percent inhibition by an antisense compound on a target mRNA will vary between assays due to factors relating to assay conditions. Hybridization
  • hybridization means the pairing of complementary strands of antisense compounds to their target sequence. While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases).
  • nucleobases complementary nucleoside or nucleotide bases
  • the natural base adenine is complementary to the natural nucleobases thymidine and uracil which pair through the formation of hydrogen bonds.
  • the natural base guanine is complementary to the natural base 5-methyl cytosine and the artificial base known as a G-clamp. Hybridization can occur under varying circumstances.
  • An antisense compound is specifically hybridizable when 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.
  • stringent hybridization conditions or “stringent conditions” refers to conditions under which an antisense compound 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 “stringent conditions” under which antisense compounds hybridize to a target sequence are determined by the nature and composition of the antisense compounds and the assays in which they are being investigated. Complementarity
  • “Complementarity,” as used herein, refers to the capacity for precise pairing between two nucleobases on either two oligomeric compound strands or an antisense compound with its target nucleic acid. For example, if a nucleobase at a certain position of an antisense compound is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be a complementary position.
  • the antisense compound and the further DNA or RNA 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.
  • Antisense compounds, or a portion thereof, may have a defined percent identity to a SEQ ID NO, or a compound having a specific Isis number.
  • a sequence is identical to the sequence disclosed herein if it has the same nucleobase pairing ability.
  • a RNA which contains uracil in place of thymidine in the disclosed sequences would be considered identical as they both pair with adenine.
  • a G-clamp modified heterocyclic base would be considered identical to a cytosine or a 5-Me cytosine in the sequences of the instant application as it pairs with a guanine.
  • This identity may be over the entire length of the oligomeric compound, or in a portion of the antisense compound (e.g., nucleobases 1-20 of a 27-mer may be compared to a 20-mer to determine percent identity of the oligomeric compound to the SEQ DD NO.) It is understood by those skilled in the art that an antisense compound need not have an identical sequence to those described herein to function similarly to the antisense compound described herein. Shortened versions of antisense compound taught herein, or non-identical versions of the antisense compound taught herein fall within the scope of this disclosure. Non-identical versions are those wherein each base does not have the same pairing activity as the antisense compounds disclosed herein.
  • Bases do not have the same pairing activity by being shorter or having at least one abasic site.
  • a non-identical version can include at least one base replaced with a different base with different pairing activity (e.g., G can be replaced by C, A, or T). Percent identity is calculated according to the number of bases that have identical base pairing corresponding to the SEQ ID NO or antisense compound to which it is being compared.
  • the non-identical bases may be adjacent to each other, dispersed through out the oligonucleotide, or both.
  • a 16-mer having the same sequence as nucleobases 2-17 of a 20-mer is 80% identical to the 20-mer.
  • a 20-mer containing four nucleobases not identical to the 20-mer is also 80% identical to the 20-mer.
  • a 14-mer having the same sequence as nucleobases 1-14 of an 18-mer is 78% identical to the 18-mer.
  • the percent identity is based on the percent of nucleobases in the original sequence present in a portion of the modified sequence. Therefore, a 30 nucleobase antisense compound comprising the full sequence of the complement of a 20 nucleobase active target segment would have a portion of 100% identity with the complement of the 20 nucleobase active target segment, while further comprising an additional 10 nucleobase portion.
  • the complement of an active target segment may constitute a single portion.
  • the oligonucleotides are at least about 80%, more preferably at least about 85%, even more preferably at least about 90%, most preferably at least 95% identical to at least a portion of the complement of the active target segments presented herein.
  • target specific cleavage was achieved using a 13 nucleobase ASOs, including those with 1 or 3 mismatches.
  • Maher and Dolnick (Nuc. Acid. Res. 16:3341-3358,1988, incorporated herein by reference) tested a series of tandem 14 nucleobase ASOs, and a 28 and 42 nucleobase ASOs comprised of the sequence of two or three of the tandem ASOs, respectively, for their ability to arrest translation of human DHFR in a rabbit reticulocyte assay.
  • Each of the three 14 nucleobase ASOs alone were able to inhibit translation, albeit at a more modest level than the 28 or 42 nucleobase ASOs.
  • Antisense compounds having a contiguous nucleobase composition that is shorter or longer or that comprises mismatches are contemplated in this invention so long as the antisense oligonucleotide activity is maintained. Therapeutics
  • Antisense compounds can be used to modulate the expression of PTPR.alpha. in an animal, such as a human.
  • the methods comprise the step of administering to said animal in need of therapy for a disease or condition associated with, increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels , increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereof, an effective amount of an antisense compound that inhibits expression of PTPR.alpha..
  • the antisense compounds effectively inhibit the levels or function of PTPR.alpha. RNA. Because reduction in PTPR.alpha. mRNA levels can lead to alteration in PTPR.alpha. protein products of expression as well, such resultant alterations can also be measured. Antisense compounds that effectively inhibit the level or function of PTPR.alpha. RNA or protein products of expression are considered an active antisense compounds. In one embodiment, the antisense compounds inhibit the expression of PTPR.alpha.
  • RNA causing a reduction of RNA by at least 10%, by at least 20%, by at least 25%, by at least 30%, by at least 40%, by at least 50%, by at least 60%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, by at least 95%, by at least 98%, by at least 99%, or by 100%.
  • the reduction of the expression of PTPR.alpha. can be measured in a bodily fluid, tissue or organ of the animal.
  • samples for analysis such as body fluids (e.g., plasma), tissues (e.g., biopsy), or organs, and methods of preparation of the samples to allow for analysis are well known to those skilled in the art.
  • Methods for analysis of RNA and protein levels are discussed above and are well known to those skilled in the art.
  • the effects of treatment can be assessed by measuring biomarkers associated with the PTPR.alpha. expression in the aforementioned fluids, tissues or organs, collected from an animal contacted with one or more compounds, by routine clinical methods known in the art.
  • biomarkers include but are not limited to: liver transaminases, body weight, organ weight, percent fat, lipids, non-esterif ⁇ ed fatty acids, glucose, insulin and other markers of diabetes, fatty liver and metabolic syndrome.
  • the antisense compounds can be utilized in pharmaceutical compositions by adding an effective amount of a compound to a suitable pharmaceutically acceptable diluent or carrier.
  • Acceptable carriers and diluents are well known to those skilled in the art. Selection of a diluent or carrier is based on a number of factors, including, but not limited to, the solubility of the compound and the desired route of administration. Such considerations are well understood by those skilled in the art.
  • the compounds inhibit the expression of PTPR.alpha..
  • the antisense compounds can also be used in the manufacture of a medicament for the treatment of diseases and conditions related to PTPR.alpha. expression.
  • Bodily fluids, organs or tissues can be contacted with one or more of the compounds disclosed herein resulting in modulation of PTPR.alpha. expression in the cells of bodily fluids, organs or tissues.
  • an isolated single- or double-stranded antisense compound targeted to PTPR.alpha. in the manufacture of a medicament for the treatment of a disease or disorder by means of the method described above.
  • the antisense compound is a single stranded antisense compound. Kits, Research Reagents, and Diagnostics
  • the antisense compounds disclosed herein can be utilized for diagnostics, and as research reagents and kits.
  • antisense compounds, which are able to inhibit gene expression with 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.
  • the antisense compounds 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. Methods of gene expression analysis are well known to those skilled in the art.
  • oligomeric compound refers to a polymeric structure capable of hybridizing to a region of a nucleic acid molecule.
  • oligomeric compounds comprise a plurality of monomelic subunits linked together by internucleoside linking groups and/or internucleoside linkage mimetics.
  • Each of the monomelic subunits comprises a sugar, abasic sugar, modified sugar, or a sugar mimetic, and except for the abasic sugar includes a nucleobase, modified nucleobase or a nucleobase mimetic.
  • Preferred monomeric subunits comprise nucleosides and modified nucleosides.
  • an “antisense compound” or “antisense oligomeric compound” refers to an oligomeric compound that is at least partially complementary to the region of a target nucleic acid molecule to which it hybridizes and which modulates (increases or decreases) its expression. This term includes oligonucleotides, oligonucleosides, oligonucleotide analogs, oligonucleotide mimetics, antisense compounds, antisense oligomeric compounds, and chimeric combinations of these. Consequently, while all antisense compounds can be said to be oligomeric compounds, not all oligomeric compounds are antisense compounds.
  • an "antisense oligonucleotide” is an antisense compound that is a nucleic acid- based oligomer.
  • An antisense oligonucleotide can, in some cases, include one or more chemical modifications to the sugar, base, and/or internucleoside linkages.
  • Nonlimiting examples of antisense compounds include primers, probes, antisense compounds, antisense oligonucleotides, external guide sequence (EGS) oligonucleotides, alternate splicers, and siRNAs. As such, these compounds can be introduced in the form of single-stranded, double-stranded, circular, branched or hairpins and can contain structural elements such as internal or terminal bulges or loops.
  • Antisense double-stranded compounds can be two strands hybridized to form double-stranded compounds or a single strand with sufficient self complementarity to allow for hybridization and formation of a fully or partially double-stranded compound.
  • the compounds are not auto-catalytic.
  • auto-catalytic means a compound has the ability to promote cleavage of the target RNA in the absence of accessory factors, e.g. proteins.
  • the antisense compound comprises a single stranded oligonucleotide. In some embodiments the antisense compound contains chemical modifications. In a preferred embodiment, the antisense compound is a single stranded, chimeric oligonucleotide wherein the modifications of sugars, bases, and internucleoside linkages are independently selected.
  • the antisense compounds disclosed herein may comprise an antisense compound from about 12 to about 35 nucleobases (i.e. from about 12 to about 35 linked nucleosides).
  • a single-stranded compound comprises from about 12 to about 35 nucleobases
  • a double-stranded antisense compound (such as a siRNA, for example) comprises two strands, each of which is independently from about 12 to about 35 nucleobases. This includes oligonucleotides 15 to 35 and 16 to 35 nucleobases in length. Contained within the antisense compounds (whether single or double stranded and on at least one strand) are antisense portions.
  • the "antisense portion” is that part of the antisense compound that is designed to work by one of the aforementioned antisense mechanisms.
  • about 12 to about 35 nucleobases includes 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleobases.
  • Antisense compounds about 12 to 35 nucleobases in length, preferably about 15 to 35 nucleobases in length, comprising a complementary stretch of at least 8, preferably at least 12, more preferably at least 15 consecutive nucleobases selected from within the active target regions, are considered to be suitable antisense compounds as well.
  • Modifications can be made to the antisense compounds and may include conjugate groups attached to one of the termini, selected nucleobase positions, sugar positions or to one of the internucleoside linkages. Possible modifications include, but are not limited to, 2'-fluoro (2'-F), T- OMethyl (2'-OMe), 2'-Methoxy ethoxy (2'-MOE) sugar modifications, inverted abasic caps, deoxynucleobases, and bicyclice nucleobase analogs such as locked nucleic acids (including LNA) and ENA.
  • double-stranded antisense compounds encompass short interfering RNAs (siRNAs).
  • siRNA short interfering RNAs
  • the term "siRNA” is defined as a double-stranded compound having a first and second strand, each strand having a central portion and two independent terminal portions.
  • the central portion of the first strand is complementary to the central portion of the second strand, allowing hybridization of the strands.
  • the terminal portions are independently, optionally complementary to the corresponding terminal portion of the complementary strand.
  • the ends of the strands may be modified by the addition of one or more natural or modified nucleobases to form an overhang
  • Each strand of the siRNA duplex may be from about 12 to about 35 nucleobases. In a preferred embodiment, each strand of the siRNA duplex is about 17 to about 25 nucleobases.
  • the two strands may be fully complementary (i.e., form a blunt ended compound), or include a 5' or 3' overhang on one or both strands.
  • Double-stranded compounds can be made to include chemical modifications as discussed herein. Chemical Modifications
  • nucleoside is a base-sugar combination.
  • the base portion of the nucleoside is normally a heterocyclic base (sometimes referred to as a "nucleobase” or simply a “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 the 2', 3' or 5' hydroxyl moiety of the sugar.
  • the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound.
  • 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. It is often preferable to include chemical modifications in oligonucleotides to alter their activity.
  • Chemical modifications can alter oligonucleotide activity by, for example: increasing affinity of an antisense oligonucleotide for its target RNA, increasing nuclease resistance, and/or altering the pharmacokinetics of the oligonucleotide.
  • the use of chemistries that increase the affinity of an oligonucleotide for its target can allow for the use of shorter oligonucleotide compounds.
  • nucleobase refers to the heterocyclic base portion of a nucleoside.
  • a nucleobase is any group that contains one or more atom or groups of atoms capable of hydrogen bonding to a base of another nucleic acid.
  • nucleobases such as the purine nucleobases adenine (A) and guanine (G), and the pyrimidine nucleobases thymine (T), cytosine (C) and uracil (U)
  • A purine
  • G guanine
  • T cytosine
  • C cytosine
  • U uracil
  • modified nucleobase and nucleobase mimetic can overlap but generally a modified nucleobase refers to a nucleobase that is fairly similar in structure to the parent nucleobase, such as for example a 7- deaza purine or a 5-methyl cytosine, whereas a nucleobase mimetic would include more complicated structures, such as for example a tricyclic phenoxazine nucleobase mimetic. Methods for preparation of the above noted modified nucleobases are well known to those skilled in the art.
  • Antisense compounds disclosed herein may also contain one or more nucleosides having modified sugar moieties.
  • the furanosyl sugar ring of a nucleoside can be modified in a number of ways including, but not limited to, addition of a substituent group, bridging of two non-geminal ring atoms to form a bicyclic nucleic acid (BNA) and substitution of an atom or group such as -S-, -N(R)- or - C(R.sub.l)(R.sub.2) for the ring oxygen at the 4'-position.
  • BNA bicyclic nucleic acid
  • Modified sugar moieties are well known and can be used to alter, typically increase, the affinity of the antisense compound for its target and/or increase nuclease resistance.
  • a representative list of preferred modified sugars includes but is not limited to bicyclic modified sugars (BNA's), including LNA and ENA (4'-(CH.sub.2).sub.2-O-2' bridge); and substituted sugars, especially 2'-substituted sugars having a 2'-F, 2'-OCH.sub.2 or a 2'-O(CH.sub.2).sub.2- OCH 3 substituent group.
  • BNA's bicyclic modified sugars
  • LNA LNA
  • ENA 4'-(CH.sub.2).sub.2-O-2' bridge
  • substituted sugars especially 2'-substituted sugars having a 2'-F, 2'-OCH.sub.2 or a 2'-O(CH.sub.2).sub.2- OCH 3 substituent group.
  • Sugars can also be replaced with sugar mimetic groups among others. Methods for the preparations of modified sugars are well known to those skilled in the art.
  • internucleoside linking groups that link the nucleosides or otherwise modified monomer units together thereby forming an antisense compound.
  • the two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom.
  • Representative phosphorus containing internucleoside linkages include, but are not limited to, phosphodiesters, phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates.
  • Non-phosphorus containing internucleoside linking groups include, but are not limited to, methylenemethylimino (-CH.sub.2-N(CH.sub.3)-O-CH.sub.2-), thiodiester (-O-C(O)-S-), thionocarbamate (-0-C(O)(NH)-S-); siloxane (-0-Si(H)2-0-); and N,N'-dimethylhydrazine (-CH.sub.2- N(CH.sub.3)-N(CH.sub.3)-).
  • Antisense compounds having non-phosphorus internucleoside linking groups are referred to as oligonucleosides.
  • Modified internucleoside linkages can be used to alter, typically increase, nuclease resistance of the antisense compound.
  • Internucleoside linkages having a chiral atom can be prepared racemic, chiral, or as a mixture.
  • Representative chiral internucleoside linkages include, but are not limited to, alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous- containing linkages are well known to those skilled in the art.
  • mimetic refers to groups that are substituted for a sugar, a nucleobase, and/ or internucleoside linkage. Generally, a mimetic is used in place of the sugar or sugar- internucleoside linkage combination, and the nucleobase is maintained for hybridization to a selected target.
  • Representative examples of a sugar mimetic include, but are not limited to, cyclohexenyl or morpholino.
  • Representative examples of a mimetic for a sugar-internucleoside linkage combination include, but are not limited to, peptide nucleic acids (PNA) and morpholino groups linked by uncharged achiral linkages.
  • PNA peptide nucleic acids
  • nucleobase mimetics are well known in the art and include, but are not limited to, tricyclic phenoxazine analogs and universal bases (Berger et al., Nuc Acid Res. 2000, 28:2911-14, incorporated herein by reference). Methods of synthesis of sugar, nucleoside and nucleobase mimetics are well known to those skilled in the art.
  • nucleoside includes, nucleosides, abasic nucleosides, modified nucleosides, and nucleosides having mimetic bases and/or sugar groups.
  • oligonucleotide refers to an oligomeric compound which is an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA). This term includes oligonucleotides composed of naturally- and non-naturally-occurring nucleobases, sugars and covalent internucleoside linkages, possibly further including non-nucleic acid conjugates.
  • internucleoside linkages including for example phosphodiester and phosphorothioate internucleoside linkages.
  • Methods of preparation and/or purification of precursors or antisense compounds are not a limitation of the compositions or methods disclosed herein. Methods for synthesis and purification of DNA, RNA, and the antisense compounds are well known to those skilled in the art.
  • chimeric antisense compound refers to an antisense compound, having at least one sugar, nucleobase and/or internucleoside linkage that is differentially modified as compared to the other sugars, nucleobases and internucleoside linkages within the same oligomeric compound. The remainder of the sugars, nucleobases and internucleoside linkages can be independently modified or unmodified.
  • a chimeric oligomeric compound will have modified nucleosides that can be in isolated positions or grouped together in regions that will define a particular motif. Any combination of modifications and or mimetic groups can comprise a chimeric oligomeric compound.
  • Chimeric oligomeric compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid.
  • An additional region of the oligomeric compound may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNAiRNA hybrids.
  • RNase H is a cellular endonuclease that 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 inhibition of gene expression.
  • RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
  • chimeric as well as non-chimeric oligomeric compounds can be further described as having a particular motif.
  • motif refers to the orientation of modified sugar moieties and/or sugar mimetic groups in an antisense compound relative to like or differentially modified or unmodified nucleosides.
  • sucgars “sugar moieties” and “sugar mimetic groups' are used interchangeably.
  • Such motifs include, but are not limited to, gapped motifs, alternating motifs, fully modified motifs, hemimer motifs, blockmer motifs, and positionally modified motifs. The sequence and the structure of the nucleobases and type of internucleoside linkage is not a factor in determining the motif of an antisense compound.
  • the term "gapped motif” refers to an antisense compound comprising a contiguous sequence of nucleosides that is divided into 3 regions, an internal region (gap) flanked by two external regions (wings).
  • the regions are differentiated from each other at least by having differentially modified sugar groups that comprise the nucleosides.
  • each modified region is uniformly modified (e.g. the modified sugar groups in a given region are identical); however, other motifs can be applied to regions.
  • the wings in a gapmer could have an alternating motif.
  • the nucleosides located in the gap of a gapped antisense compound have sugar moieties that are different than the modified sugar moieties in each of the wings.
  • alternating motif refers to an antisense compound comprising a contiguous sequence of nucleosides comprising two differentially sugar modified nucleosides that alternate for essentially the entire sequence of the antisense compound, or for essentially the entire sequence of a region of an antisense compound.
  • the term "fully modified motif” refers to an antisense compound comprising a contiguous sequence of nucleosides wherein essentially each nucleoside is a sugar modified nucleoside having uniform modification.
  • hemimer motif refers to a sequence of nucleosides that have uniform sugar moieties (identical sugars, modified or unmodified) and wherein one of the 5 '-end or the 3'- end has a sequence of from 2 to 12 nucleosides that are sugar modified nucleosides that are different from the other nucleosides in the hemimer modified antisense compound.
  • blockmer motif refers to a sequence of nucleosides that have uniform sugars (identical sugars, modified or unmodified) that is internally interrupted by a block of sugar modified nucleosides that are uniformly modified and wherein the modification is different from the other nucleosides.
  • Methods of preparation of chimeric oligonucleotide compounds are well known to those skilled in the art.
  • positionally modified motif comprises all other motifs. Methods of preparation of positionally modified oligonucleotide compounds are well known to those skilled in the art.
  • the compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), alpha or beta, or as (D) or (L) such as for amino acids et al. All such possible isomers, as well as their racemic and optically pure forms, are included herein.
  • antisense compounds are modified by covalent attachment of one or more conjugate groups.
  • Conjugate groups may be attached by reversible or irreversible attachments.
  • Conjugate groups may be attached directly to antisense compounds or by use of a linker.
  • Linkers may be mono- or bifunctional linkers. Such attachment methods and linkers are well known to those skilled in the art.
  • conjugate groups are attached to antisense compounds to modify one or more properties. Such considerations are well known to those skilled in the art. Oligomer Synthesis
  • Oligomerization of modified and unmodified nucleosides can be routinely performed according to literature procedures for DNA (Protocols for Oligonucleotides and Analogs, Ed. Agrawal (1993), Humana Press) and/or RNA (Scaringe, Methods (2001), 23, 206-217. Gait et al., Applications of Chemically synthesized RNA in RNA: Protein Interactions, Ed. Smith (1998), 1-36. Gallo et al., Tetrahedron (2001), 57, 5707-5713).
  • Antisense compounds can 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, CA). 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. The invention is not limited by the method of antisense compound synthesis. Oligomer Purification and Analysis
  • oligonucleotide purification and analysis are known to those skilled in the art. Analysis methods include capillary electrophoresis (CE) and electrospray-mass spectroscopy. Such synthesis and analysis methods can be performed in multi-well plates. The method of the invention is not limited by the method of oligomer purification. Salts, prodrugs and bioequivalents
  • the antisense compounds disclosed herein may comprise any pharmaceutically acceptable salts, esters, or salts of such esters, or any other functional chemical equivalent which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the antisense compounds, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
  • prodrug indicates a therapeutic agent that is prepared in an inactive or less active form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes, chemicals, and/or conditions.
  • active form i.e., drug
  • prodrug versions of the oligonucleotides are prepared as SATE ((S-acetyl-2-thioethyl) phosphate) derivatives according to the methods disclosed in WO 93/24510 or WO 94/26764.
  • Prodrugs can also include antisense compounds wherein one or both ends comprise nucleobases that are cleaved (e.g., by incorporating phosphodiester backbone linkages at the ends) to produce the active compound.
  • pharmaceutically acceptable salts refers to physiologically and pharmaceutically acceptable salts of the compounds: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto.
  • Sodium salts of antisense oligonucleotides are useful and are well accepted for therapeutic administration to humans.
  • sodium salts of dsRNA compounds are also provided.
  • the antisense compounds may also be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds.
  • compositions and formulations which include the antisense compounds .
  • the pharmaceutical compositions may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated
  • the pharmaceutical formulations 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, finely divided solid carriers, or both, and then, if necessary, shaping the product (e.g., into a specific particle size for delivery).
  • a “pharmaceutical carrier” or “excipient” can be a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal and are known in the art.
  • the excipient may be liquid or solid and is selected, with the planned manner of administration in mind, so as to provide for the desired bulk, consistency, etc., when combined with a nucleic acid and the other components of a given pharmaceutical composition.
  • compositions may comprise two or more antisense compounds.
  • pharmaceutical compositions may comprise 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.
  • pharmaceutical compositions may comprise two or more antisense compounds targeted to different regions of the same nucleic acid target. Two or more combined compounds may be used together or sequentially.
  • Pharmaceutical compositions may comprise an antisense compound combined with other non-antisense compound therapeutic agents.
  • Example 1- Cell types- The effect of oligomeric compounds on target nucleic acid expression was tested in one or more of the following cell types.
  • T-24 cells The human transitional cell bladder carcinoma cell line T-24 was obtained from the American Type Culture Collection (ATCC) (Manassas, VA). T-24 cells were routinely cultured in complete McCoy's 5 A basal media (Invitrogen Corporation, Carlsbad, CA) supplemented with 10% fetal calf serum (Invitrogen Corporation, Carlsbad, CA), penicillin 100 units per mL, and streptomycin 100 micrograms per mL (Invitrogen Corporation, Carlsbad, CA). Cells were routinely passaged by trypsinization and dilution when they reached 90% confluence.
  • ATCC American Type Culture Collection
  • b.END cells The mouse brain endothelial cell line b.END was obtained from Dr. Werner Risau at the Max Plank Institute (Bad Nauheim, Germany). b.END cells were routinely cultured in DMEM, high glucose (Gibco/Life Technologies, Gaithersburg, MD) supplemented with 10% fetal calf serum (Gibco/Life Technologies, Gaithersburg, MD). Cells were routinely passaged by trypsinization and dilution when they reached 90% confluence. Cells were seeded into 96-well plates (Falcon-Primaria #3872) at a density of 3000 cells/well for use in RT-PCR analysis.
  • cells may be seeded onto 100 mm or other standard tissue culture plates and treated similarly, using appropriate volumes of medium and oligonucleotide.
  • NHDF cells Human neonatal dermal fibroblast (NHDF) were obtained from the Clonetics Corporation (Walkersville, MD). NHDFs were routinely maintained in Fibroblast Growth Medium (Clonetics Corporation, Walkersville, MD) supplemented as recommended by the supplier. Cells were maintained for up to 10 passages as recommended by the supplier.
  • HEK cells Human embryonic keratinocytes (HEK) were obtained from the Clonetics Corporation (Walkersville, MD). HEKs were routinely maintained in Keratinocyte Growth Medium (Clonetics Corporation, Walkersville, MD) formulated as recommended by the supplier. Cells were routinely maintained for up to 10 passages as recommended by the supplier.
  • oligomeric compounds When cells reach appropriate confluency, they are treated with oligonucleotide using a transfection method as described. LipofectinTM When cells reached 65-75% confluency, they were treated with oligonucleotide. Oligonucleotide was mixed with LIPOFECTINTM Invitrogen Life Technologies, Carlsbad, CA) in Opti-MEMTM-1 reduced serum medium (Invitrogen Life Technologies, Carlsbad, CA) to achieve the desired concentration of oligonucleotide and a LIPOFECTIN TM concentration of 2.5 or 3 .micro.g/mL per 100 nM oligonucleotide. This transfection mixture was incubated at room temperature for approximately 0.5 hours.
  • Control oligonucleotides are used to determine the optimal oligomeric compound concentration for a particular cell line. Furthermore, when oligomeric compounds are tested in oligomeric compound screening experiments or phenotypic assays, control oligonucleotides are tested in parallel.
  • 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. The concentration of positive control oligonucleotide that results in 80% inhibition of the target 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 the target mRNA is then utilized as the oligonucleotide screening concentration in subsequent experiments for that cell line.
  • the concentrations of antisense oligonucleotides used herein are from 50 nM to 300 nM when the antisense oligonucleotide is transfected using a liposome reagent and 1 ⁇ M to 40 ⁇ M when the antisense oligonucleotide is transfected by electroporation.
  • Example 2 Real-time Quantitative PCR Analysis of PTPR.alpha. mRNA Levels
  • Quantitation of PTPR.alpha. mRNA levels was accomplished by real-time quantitative PCR using the ABI PRISMTM 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, CA) according to manufacturer's instructions.
  • primer-probe sets specific to the PTPR.alpha. being measured were evaluated for their ability to be "multiplexed” with a GAPDH amplification reaction. After isolation the RNA is subjected to sequential reverse transcriptase (RT) reaction and real-time PCR, both of which are performed in the same well.
  • RT and PCR reagents were obtained from Invitrogen Life Technologies (Carlsbad, CA).
  • RT real-time PCR was carried out in the same by adding 20 .micro.L PCR cocktail (2.5x PCR buffer minus MgCl.sub.2, 6.6 mM MgCl.sub.2, 375 .micro.M 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 2.5x ROX dye) to 96- well plates containing 30 .micro.L total RNA solution (20-200 ng). The RT reaction was carried out by incubation for 30 minutes at 48.deg.C.
  • 20 .micro.L PCR cocktail 2.5x PCR buffer minus MgCl.sub.2, 6.6 mM MgCl.sub.2, 375 .micro.M each of dATP, dCTP, dCTP and dGTP, 375
  • Gene target quantities obtained by RT, real-time PCR were normalized using either the expression level of GAPDH, a gene whose expression is constant, or by quantifying total RNA using RiboGreenTM (Molecular Probes, Inc. Eugene, OR).
  • GAPDH expression was quantified by RT, real-time PCR, by being run simultaneously with the target, multiplexing, or separately.
  • Total RNA was quantified using RiboGreenTM RNA quantification reagent (Molecular Probes, Inc. Eugene, OR).
  • the GAPDH PCR probes have JOE covalently linked to the 5' end and TAMRA or MGB covalently linked to the 3' end, where JOE is the fluorescent reporter dye and TAMRA or MGB is the quencher dye.
  • primers and probe designed to a GAPDH sequence from a different species are used to measure GAPDH expression.
  • a human GAPDH primer and probe set is used to measure GAPDH expression in monkey-derived cells and cell lines.
  • Probes and primers for use in real-time PCR were designed to hybridize to target-specific sequences.
  • the primers and probes and the target nucleic acid sequences to which they hybridize are presented in Table 2.
  • the target-specific PCR probes have FAM covalently linked to the 5' end and TAMRA or MGB covalently linked to the 3' end, where FAM is the fluorescent dye and TAMRA or MGB is the quencher dye.
  • Example 3 Antisense inhibition of human PTPR.alpha. expression by oligomeric compounds
  • RNA RNA sequences
  • GenBank accession number NM 080841.1 representing a human PTPR.alpha. variant herein designated hPTPRA-3, incorporated herein as SEQ ID NO: 4
  • GenBank accession number M34668.1 representing the main mRNA of human PTPR.alpha., herein designated hPTPRA, incorporated herein as SEQ ID NO: 18
  • GenBank accession number AL161656.15 and AL121905.23 representing a genomic sequence of human PTPR.alpha., incorporated herein as SEQ ID NO: 19
  • GenBank accession number X54890.1 representing a human PTPR.alpha.
  • hPTPRA-4 incorporated herein as SEQ ID NO: 20
  • GenBank accession number X53364.1 representing a human PTPR.alpha. variant herein designated hPTPRA-5, incorporated herein as SEQ ID NO: 21
  • GenBank accession number AF121 183.1 representing a 5'-extension of SEQ ID NO: 18, incorporated herein as SEQ ID NO: 22
  • the complement of GenBank accession number BEl 68541.1 representing a 5'-extenstion of SEQ ID NO: 22 incorporated herein as SEQ DD NO: 23
  • the complement of GenBank accession number AW024120.1 representing a 5'-extension of SEQ ID NO: 20, incorporated herein as SEQ ID NO: 24
  • the complement of GenBank accession number AA903762.1 representing a 3 '-extension of SEQ ID NO: 18, incorporated herein as SEQ ID NO: 25
  • hPTPRA-7 incorporated herein as SEQ ID NO: 26
  • the oligonucleotides are shown in Table 3. "Target site” indicates the first (5 '-most) nucleotide number on the particular target sequence to which the oligonucleotide binds.
  • All compounds in Table 3 are chimeric oligonucleotides ("gapmers") 20 nucleotides in length, composed of a central "gap" region consisting often 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.
  • All cytidine residues are 5-methylcytidines. The compounds were analyzed for their effect on human PTPR.alpha.
  • mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from two experiments in which T-24 cells were treated with the antisense oligonucleotides. The positive control for each data point is identified in the table by sequence ID number. If present, "N.D.” indicates "no data”.
  • the target sites to which these preferred sequences are complementary are herein referred to as "preferred target sites" and are therefore preferred sites for targeting by antisense compounds. These preferred target sites are shown in Table 5.
  • Example 4 Antisense inhibition of mouse PTPR.alpha. expression by oligomeric compounds
  • RNA using published sequences (GenBank accession number NM_008980.1, representing a variant of mouse PTPR.alpha.
  • mPTPRA-2 incorporated herein as SEQ ID NO: 11
  • GenBank accession number AW323517.1 representing a 5'-extension of SEQ ID NO: 11, incorporated herein as SEQ ID NO: 93
  • GenBank accession numbers L13686.1, L13668.1, L13670.1, Ll 3672.1, Ll 3674.1, Ll 3675.1, Ll 3676.1 and Ll 3608.1 representing partial genomic sequences of mouse PTPR.alpha., incorporated herein as SEQ ID NOs: 94, 95, 96, 97, 98, 99, 100 and 101 respectively
  • GenBank accession number BE372094.1 representing the main mRNA of mouse PTPR.alpha., herein designated mPTPRA, incorporated herein as SEQ ED NO: 102).
  • oligonucleotides are shown in Table 4. "Target site” indicates the first (5 '-most) nucleotide number on the particular target sequence to which the oligonucleotide binds. All compounds in Table 4 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.
  • antisense oligonucleotides with mismatches or varying nucleotide lengths will have similar profiles to those described herein.
  • the compounds were analyzed for their effect on mouse PTPR.alpha. mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from two experiments. The positive control for each data point is identified in the table by sequence ID number. If present, "N.D.” indicates "no data”.
  • SEQ ID NOs 103, 106, 108, 110, 111, 112, 116, 117, 118, 119, 122, 125, 127, 128, 129, 130, 134, 135, 136, 137, 139, 145, 148 and 149 demonstrated at least 50% inhibition of mouse PTPR.alpha. expression in this experiment and are therefore preferred.
  • the target sites to which these preferred sequences are complementary are herein referred to as "preferred target sites" and are therefore preferred sites for targeting antisense oligonucleotides. These preferred target sites are shown in Table 5.
  • nucleotides 2860 to 2990 of SEQ ID NO: 11 form a region whereint eh screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 70%, and thus is designated as an active target segment (active target segment E).
  • the "preferred target region” may be employed in screening candidate antisense compounds.
  • “Candidate antisense compounds” are those that inhibit the expression of a nucleic acid molecule encoding PTPR.alpha. and which comprise at least an 8-nucleobase portion which is complementary to a preferred target region.
  • the method comprises the steps of contacting a preferred target region of a nucleic acid molecule encoding PTPR.alpha. with one or more candidate antisense compounds, and selecting for one or more candidate antisense compounds which inhibit the expression of a nucleic acid molecule encoding PTPR.alpha..
  • the candidate antisense compound may be employed as an antisense compound as described herein.
  • antisense compounds include ribozymes, external guide sequence (EGS) oligonucleotides (oligozymes), and other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and modulate its expression.
  • EGS external guide sequence
  • oligozymes oligonucleotides
  • other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and modulate its expression.
  • oligonucleotides that selectively target, hybridize to, and specifically inhibit one or more, but fewer than all of the variants of human PTPR.alpha..
  • a summary of the target sites of the PTPR.alpha. main mRNA and variants is shown in Table 6 and includes GenBank accession number NM_080841.1 , representing hPTPRA-3 , incorporated herein as SEQ ID NO: 4; GenBank accession number M34668.1, representing hPTPR.alpha.
  • GenBank accession number X54890.1 representing hPTPRA-4, incorporated herein as SEQ ID NO: 20
  • GenBank accession number X53364.1 representing hPTPRA-5, incorporated herein as SEQ ID NO: 21
  • the complement of GenBank accession number AI674319.1, representing hPTPRA-7 incorporated herein as SEQ ID NO: 26
  • GenBank accession number NM_002836.1 encodetered Mar. 24, 1999
  • GenBank accession number NM 080840.1 (entered Jan.
  • Table 6 Targeting of individual oligonucleotides to specific variants of human PTPR.alpha. Human PTPR.alpha. mRNA sequences from Table 1 were aligned and the screened human oligos were determined to have a complementary sequence to a common PTPR.alpha. mRNA, specifically (SEQ ID NO: 18). Antisense compounds targeting SEQ ID NO: 18 are shown in Table 6b. The antisense compounds were then grouped according to activity and active target segments were determined. An active target segment is used herein to describe a region of the PTPR.alpha. mRNA that has one or more active antisense compounds targeted thereto. Each of the following active target segment was targeted by multiple, active antisense oligonucleotides.
  • These regions include nucleotides 716 to 894, nucleotides 1063 to 1229, nucleotides 1818 to 2045 and nucleotides 2328 to 2779 of SEQ ID NO: 18.
  • Each of the oligonucleotides tested within each of these regions inhibited expression of human PTPR.alpha. by at least 60% in this assay.
  • active target segment A nucleotides 716 to 894
  • the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 85%.
  • active target segment B In active target segment B (nucleotides 1063 to 1229) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 75%.
  • active target segment C In active target segment C (nucleotides 1818 to 2045) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 90%. In active target segment D (nucleotides 2328 to 2779) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 80%. In active target segment F (nucleotides 1210-1905) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 87%. In active target segment G (nucleotides 2982-3211) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 73%.Identification of these regions allows for the design of antisense oligonucleotides that modulate the expression of PTPR.alpha..
  • Table 6b Exemplary human PTPR.alpha. oligos targeting SEQ ID NO: 18
  • Example 6 Targeting of individual oligonucleotides to specific variants of mouse PTPR.aIpha.
  • Oligonucleotides are provided that selectively target, hybridize to, and specifically inhibit one or more, but fewer than all of the variants of mouse PTPR.aIpha..
  • a summary of the target sites of the PTPR.alpha. main mRNA and variants is shown in Table 7 and includes GenBank accession number NM_008980.1, representing mPTPRA-2, incorporated herein as SEQ ID NO: 11 and GenBank accession number BE372094.1, representing mPTPR.alpha. main mRNA, incorporated herein as SEQ ID NO: 102.
  • ob/ob mice Effect of antisense inhibitors of PTPR.alpha. in ob/ob mice: a model of obesity and diabetes.
  • ob/ob mice are used as a model of obesity and of type-2 diabetes. These mice have a deficiency in the Leptin hormone produced by fat that regulates appetite. Deficiencies in this hormone in both humans and non-human animals lead to obesity.
  • the ob/ob mice have a mutation in the leptin gene which results in obesity and hyperglycemia, ob/ob mice have higher circulating levels of insulin and are less hyperglycemic than db/db mice, which harbor a mutation in the leptin receptor.
  • ISIS 147299 SEQ ID NO: 135) was tested in the ob/ob model of obesity and diabetes.
  • mice were further evaluated throughout the treatment period and at the end of the treatment period for body weight, percent fat from body weight, organ weight, chemistries, including serum transaminase, cholesterol, triglycerides and non-esterified fatty acids, liver triglycerides, plasma glucose levels, fed plasma insulin levels and glucose tolerance.
  • chemistries including serum transaminase, cholesterol, triglycerides and non-esterified fatty acids, liver triglycerides, plasma glucose levels, fed plasma insulin levels and glucose tolerance.
  • Triglycerides, lipoproteins, cholesterol and transaminases were measured by routine clinical analyzer instruments (e.g. Olympus Clinical Analyzer, Melville, NY).
  • Serum free fatty acids were measured using a Wako Chemicals kit for non-esterif ⁇ ed free fatty acids (Richmond, VA).
  • Serum apolipoproteins were measured using apolipoprotein-specific ELISA or by protein immunoblot with apolipoprotein-specific antibodies.
  • Tissue triglyceride levels were measured using a Triglyceride GPO Assay from Roche Diagnostics (Indianapolis, IN). Liver triglyceride levels were used to assess hepatic steatosis, or clearing of lipids from the liver. Hepatic steatosis was also assessed by routine histological analysis of frozen liver tissue sections stained with oil red O stain.
  • fed plasma glucose levels were approximately 403.3 mg/dl at base line, 376.7 mg/dl at week 2, 330.0 mg/dl at week 3 and 295.7 mg/dl at sacrifice.
  • Fed plasma insulin levels were approximately 21.57 ng/ml at baseline, 31.37 at week 2, 25.08 at week 3 and 15.2 at week 4.5.
  • plasma glucose levels are reduced in animals following treatment with an antisense oligonucleotide that inhibit PTPR.alpha..
  • Plasma lipids in ob/ob mice were also measured throughout the treatment period and at the end of the treatment period. Table 10. Cholesterol levels were approximately 179.3 mg/dl at baseline, 240.3 mg/dl after 2 weeks, 242.9 mg.dl after 3 weeks and 247.3 mg/dl at sacrifice for the ice receiving an antisense inhibitor of PTPR.alpha.. There was no significant change in plasma cholesterol levels for mice receiving an antisense inhibitor of PTPR.alpha.. Triglycerides were approximately 165.7 mg/dl at base line, 107.9 mg/dl after 2 weeks, 87.4 mg/dl after 3 weeks and 88.9 mg/dl at sacrifice for ISIS 147299-treated mice. For mice receiving treatment with an antisense inhibitor of PTPR.alpha. there was a decrease in fed plasma triglyceride levels.
  • Plasma Aspartate Aminotransferase (AST) levels were approximately 71.1 RJ/L at base line, 128.4 KJ/L after 2 weeks, 144.0 KJ/L after 3 weeks and 134.4 IU/L at sacrifice following treatment. Fasted transaminase levels were determined after 4 weeks of treatment and were approximately 246.9 R7/L for ALT and 139.1 for AST. Table 12. These values show that treatment with an antisense inhibitor of PTPR.alpha. reduced hepatic and plasma triglyceride levels and improved hepatic function compared to the saline control group.
  • antisense inhibition of PTPR.alpha. expression improves plasma glucose levels, glucose tolerance and plasma and hepatic triglycerides levels in the ob/ob mouse model of diabetes and obesity. Consequently, antisense inhibitors of PTPR.alpha. are useful for preventing, ameliorating or treating conditions and disorders associated with these factors.
  • Antisense inhibitors of PTPR.alpha. expression are expected to reduce obesity or weight gain. This is tested in the high fat diet (HFD) model, also known as the DIO (diet-induced obesity) model.
  • HFD high fat diet
  • DIO diet-induced obesity
  • mice Four week old male C57bl/6 mice were fed a high fat diet (60% kCal) ad libitum beginning at 4 weeks of age and continuing for 10 weeks. After the 10 weeks the mice were maintained on the diet as described and were treated with either an antisense inhibitor of PTPR.alpha., a control oligonucleotide having similar chemistry to the treatment oligonucleotide but not having a sequence that is complementary to the target mRNA, or saline. During this feeding schedule, animals were weighed at regular intervals and body weight and percent fat measurements were determined. At sacrifice, organ weights were determined as well.
  • a high fat diet (60% kCal) ad libitum beginning at 4 weeks of age and continuing for 10 weeks. After the 10 weeks the mice were maintained on the diet as described and were treated with either an antisense inhibitor of PTPR.alpha., a control oligonucleotide having similar chemistry to the treatment oligonucleotide but not having a sequence
  • mice in the treatment group received 25 mg/kg of ISIS 147299 (SEQ ID NO: 135) twice a week by subcutaneous injection.
  • Control group mice received a same treatment routine except using saline or, 25 mg/kg of control oligonucleotide that is not an antisense inhibitor of PTPR.alpha (CCTTCCCTGAAGGTTCCTCC - SEQ ID NO: 228).
  • a further control group received no treatment and was fed normal chow. Food consumption was determined for mice in each of the groups.
  • Mice treated with the antisense PTPR.alpha. inhibitor had reduced body weight gains and reduced food consumption.
  • Figures 2 and 3. There were no differences in organ weight and percent fat between the groups.
  • Insulin levels are presented in Table 13. For mice treated with an antisense inhibitor of PTPR.alpha. fed plasma insulin levels were approximately 2.66 ng/ml at base line, 2.96 ng/ml after 2 weeks, 2.71 ng/ml after 3 weeks and 1.95 ng/ml after 5 weeks. At week four fasted insulin levels were approximately 0.58 ng/ml for the mice receiving treatment with an antisense inhibitor of PTPR.alpha.
  • Insulin tolerance tests are a good measure of insulin sensitivity. Mice were fasted for 3 hours and then received 0.35 U/kg of humilin (Lilly, Indianapolis, IN) via gavage of a O.lU/ml solution in saline. Blood glucose was measured at time point 0 and at 15, 20 or 30 minute intervals for up to 2 hours. Glucose levels are measured using a YSI glucose analyzer (YSI Scientific, Yellow Springs, OH). Results of the insulin tolerance test are shown in Table 14.
  • Insulin levels were reduced for the animals receiving ISIS 147299 antisense inhibitor to PTPR.alpha.
  • PTPR.alpha results in a reduction of plasma glucose levels and triglyceride levels in the plasma and liver.
  • Antisense inhibition of PTPR.alpha. may provide a therapeutic strategy for conditions and diseases associated with these factors, such as type II diabetes, fatty liver disease and metabolic syndrome.

Landscapes

  • Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Genetics & Genomics (AREA)
  • Organic Chemistry (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Chemical & Material Sciences (AREA)
  • Biomedical Technology (AREA)
  • General Health & Medical Sciences (AREA)
  • General Engineering & Computer Science (AREA)
  • Molecular Biology (AREA)
  • Biotechnology (AREA)
  • Wood Science & Technology (AREA)
  • Zoology (AREA)
  • Medicinal Chemistry (AREA)
  • General Chemical & Material Sciences (AREA)
  • Veterinary Medicine (AREA)
  • Diabetes (AREA)
  • Animal Behavior & Ethology (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Public Health (AREA)
  • Biophysics (AREA)
  • Physics & Mathematics (AREA)
  • Plant Pathology (AREA)
  • Microbiology (AREA)
  • Hematology (AREA)
  • Biochemistry (AREA)
  • Obesity (AREA)
  • Gastroenterology & Hepatology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

Disclosed herein are compounds, compositions and methods for modulating the expression of PTPR.alpha. in a cell, tissue or animal. Also provided are methods of target validation. Also provided are uses of disclosed compounds and compositions in the manufacture of a medicament for treatment of diseases and disorders. Also provided are methods for the prevention, amelioration and/or treatment of airway hyperresponsiveness and pulmonary inflammation by administration of antisense compounds targeted to PTPR.alpha.

Description

COMPOSITIONS AND THEIR USES DIRECTED TO PTPR ALPHA
CROSS-REFERENCE TO RELATED APPLICATIONS
This application is related to U.S. Patent Application Serial No.: 11/502,251 , filed August 9, 2006; which is a continuation-in-part of U.S. Patent Application Serial No 11/036,095, filed on January 14, 2005, which is a continuation in part of U.S. Patent Application Serial No. 10/210,556, filed on July 31, 2002 now abandoned. The entire contents of these applications and patents are incorporated herein by reference in their entirety.
SEQUENCE LISTING
The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BIOL0084WOSEQ.TXT, created on May 7, 2007 which is 508 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.
BACKGROUND OF THE INVENTION
The process of protein phosphorylation represents one course by which intracellular signals are propagated in a cellular response. Within the cell, proteins can be phosphorylated on serine, threonine or tyrosine residues and the extent of phosphorylation is regulated by the opposing action of phosphatases, which remove the phosphate moieties. .
PTPR.alpha. (also known as protein tyrosine phosphatase receptor type alpha, LCA- related phosphatase; LRP, HLPR, HPTPA, PTPRA, PTPA, PTP-R alpha, PTPRL2 and RPTPA) is a widely expressed member of the family of receptor-like phosphatases. It was cloned from a human hepatoblastoma cell line (JMk et al., FEBS Lett., 1990, 273, 239-242) and a brain stem cell line (Kaplan et al., Proc. Natl. Acad. Sci. U. S. A., 1990, 87, 7000-7004) and was mapped to chromosome 20pl 3, a chromosomal region involved in translocations and deletions in myeloid disorders and neoplasms (Jink et al., Cytogenet. Cell Genet., 1992, 60, 117-118; Kaplan et al., Proc. Natl. Acad. Sci. U. S. A., 1990, 87, 7000-7004).
Kaplan et al. have presented evidence for the existence of two major PTPR.alpha. RNA transcripts of approximately 4.3 and 6.3 kb (Kaplan et al., Proc. Natl. Acad. Sci. U. S. A., 1990, 87, 7000- 7004) whose proteins differ in a stretch of 9 amino acid residues situated in the external juxtamembrane segment (Daum et al., J. Biol. Chem., 1994, 269, 10524-10528). The larger of the two species appears to be more prevalent in fetal tissues, suggesting that the differential expression of the two transcripts is developmentally regulated and/or a result of alternative splicing mechanisms (Kaplan et al., Proc. Natl. Acad. Sci. U. S. A., 1990, 87, 7000-7004). Four additional variants have been subsequently identified.
PTPR.alpha. has been identified as a negative regulator of insulin receptor signaling indicating that it may play a key role in insulin action and in the pathophysiology of non-insulin- dependent diabetes (Moeller et al., J. Biol. Chem., 1995, 270, 23126-23131). However, the importance of PTPR.alpha.'s role in insulin signaling is considered unclear. (Cheng, A., et al, Eur. J. Biochem. (2002) 269: 1050-1059).
There is a need in the art for compositions that modulate the activity of PTPR.alpha. and methods for their use as a treatment of prophylactic for diseases and conditions associated with alterations in plasma glucose and in insulin levels as well as insulin action, such as for type 2 diabetes.
SUMMARY OF THE INVENTION
Provided are antisense compounds, particularly oligomeric compounds, especially nucleic acid and nucleic acid-like oligomers, which are targeted to a nucleic acid encoding PTPR.alpha.. Preferably, the antisense compounds are antisense oligonucleotides targeted to PTPR.alpha., particularly human PTPR.alpha., and are capable of modulating the expression of PTPR.alpha.. Prefered compounds comprise at least an 8 nucleobase portion, preferably a 12 nucleobase portion, more preferably at least a 15 nucleobase portion, of the sequences listed in Table 3 or 4, or are at least 80% identical to the complement of validated target segments, or to the antisense oligonucleotide sequences listed in Table 3 or 4.
Provided are methods for modulating the expression of PTPR.alpha. in cells or tissues comprising contacting the cells with at least one antisense compound, as described, and analyzing the cells for indicators of a decrease in expression of PTPR.alpha. rnRNA and/or protein by direct measurement of rnRNA and/or protein levels, and/or indirect indicators of a disease or condition such as measuring plasma triglyceride levels, plasma glucose levels, plasma transaminase levels, plasma insulin levels, hepatic triglyceride levels or similar indicators of type 2 diabetes, fatty liver, obesity or metabolic syndrome.
Provided are methods for the prevention, amelioration, and/or treatment of type 2 diabetes, fatty liver disease or metabolic syndrome comprising administering at least one antisense compound of this invention to an individual in need of such intervention.
Provided are methods for using the antisense compounds disclosed herein to prepare a medicament for the prevention, amelioration, and/or treatment of a disease or condition, especially a those associated with at least one indicator of type 2 diabetes, fatty liver disease, obesity or metabolic syndrome.
Further Methods and compositions are provided as follows:
1. An antisense compound of 15 to 35 nucleobases targeted to a nucleic acid molecule encoding human PTPR.alpha.. wherein the nucleic acid molecule encoding human PTPR.alpha. is SEQ ID NO: 18 and wherein the antisense compound hybridizes with nucleotides 716 to 894, nucleotides 1063 to 1229, nucleotides 1818 to 2045, nucleotides 2328 to 2779, nucleotides 2982 to 3211, nucleotides 1210 to 1905 of SEQ E) NO: 18 or nucleotides 2860 to 2990 of SEQ ID NO: 11.
2. Any one of the antisense compounds described herein can be targeted to at least two or more nucleic acid molecules encoding human PTPR.alpha.
3. Any one of the antisense compounds described herein, wherein the compound is at least about 80% complementary to a target region of the nucleic acid encoding PTPR.alpha.
4. Any one of the antisense compounds described herein, wherein the compound optionally includes a complementary strand 15 to 35 nucleobases in length.
5. Any one of the antisense compounds described herein, wherein the compound is an antisense oligonucleotide.
6. Any one of the compounds described herein having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
7. Any one of the compounds described herein comprising a chimeric oligonucleotide.
8. Any one of the compounds described herein wherein the modified internucleoside linkage comprises a phosphorothioate linkage.
9. Any one of the compounds described herein wherein the at least one modified sugar moiety comprises a 2'-MOE , a LNA, a 2'-OMe, an ENA, a 2'-F or combinations thereof.
10. Any one of the compounds described herein wherein the modified nucleobase comprises 5-methylcytosine.
11. A pharmaceutical composition comprising one or more compounds described herein and a pharmaceutically acceptable penetration enhancer, carrier, or diluent.
12. A method for the prevention, amelioration, or treatment of a disease or condition wherein indicators of the disease or condition comprise increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels, increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereof comprising administration of one or more compounds described herein to an individual in need of such intervention.
13. The methods described herein wherein the disease or condition is fatty liver disease and wherein administration of compounds described herein results in a decrease in plasma lipid levels, a decrease in plasma transaminase levels, a decrease in hepatic triglyceride levels or combinations thereof.
14. The methods described herein wherein the disease or condition is type-2 diabetes and wherein administration of one or more compounds described herein results in a decrease in plasma glucose levels, an improvement in insulin sensitivity or combinations thereof. 15. The methods described herein wherein the disease or condition is metabolic syndrome and wherein administration of one or more compounds described herein results in a decrease in plasma glucose levels, a decrease in plasma lipid levels, a decrease in hepatic triglyceride levels, an improvement
> in insulin sensitivity, an improvement in hepatic function, a decrease in plasma transaminase levels or combinations thereof.
16. Use of an antisense compounds described herein in the manufacture of a medicament for the treatment, prevention or amelioration of a disease or condition wherein indicators of the disease or
I condition comprise increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels , increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereofwherein the medicament reduces the expression of a nucleic acid molecule encoding PTPR.alpha..
i 17. The use of the compounds described herein wherein the disease or condition comprises type-2 diabetes, fatty liver, metabolic syndrome or combinations thereof.
18. The use of the compounds described herein wherein the antisense compounds optionally further comprises a complementary strand 15 to 35 nucleobases in length.
19. A method for lowering blood glucose levels in an animal in need thereof by administering one or more compounds provided herein.
20. A method for lowering triglyceride levels in an animal in need thereof by administering one or more compounds provided herein.
21. The method of lowering triglyceride levels wherein the triglyceride levels are lowered in the plasma.
22. The method of lowering triglyceride levels wherein the triglyceride levels are lowered in the liver.
23. The methods as provided wherein the animal suffers from type-2 diabetes, fatty liver disease, obesity, metabolic syndrome of combinations thereof.
BRIEF DESCRIPTION OF THE FIGURES
Figures Ia and Ib illustrate that administration of the disclosed antisense compounds is useful for clearing fat from the liver. Figure Ia is an oil red stained liver section of an ob/ob mouse treated with saline and showing no clearing. Figure Ib is an oil red stained liver section of an ob/ob mouse treated with an antisense inhibitor of PTPR.alpha and showing clearing of fat from the liver.
Figure 2 is a graph illustrating weekly body weights for DIO mice receiving treatment oligo, control oligo or saline.
Figure 3 is a graph illustrating food intake for DIO receiving treatment oligo, control oligo or saline.
DETAILED DESCRIPTION OF THE INVENTION
Provided are antisense compounds for the prevention, amelioration, and /or treatment of diseases or conditions associated with increases in plasma triglyceride levels, plasma glucose levels, plasma transaminase levels, plasma insulin levels hepatic triglyceride levels and further associated with PTPR.alpha activity. As used herein, the term "prevention" means to delay or forestall onset or development of a condition or disease for a period of time from hours to days, preferably weeks to months. As used herein, the term "amelioration" means a lessening of at least one indicator of the severity of a condition or disease. The severity of indicators may be determined by subjective or objective measures which are known to those skilled in the art. As used herein, "treatment" means to administer an antisense composition to effect an alteration or improvement of the disease or condition. Prevention, amelioration, and/or treatment may require administration of multiple doses at regular intervals, or prior to exposure to an agent to alter the course of the condition or disease. Moreover, a single agent may be used in a single individual for each prevention, amelioration, and treatment of a condition or disease sequentially, or concurrently.
Disclosed herein are antisense compounds, including antisense oligonucleotides and other antisense compounds for use in modulating the expression of nucleic acid molecules encoding PTPR.alpha.. This is accomplished by providing antisense compounds that hybridize with one or more target nucleic acid molecules encoding PTPR.alpha.. As used herein, the terms "target nucleic acid" and "nucleic acid molecule encoding PTPR.alpha. " have been used for convenience to encompass RNA (including pre-mRNA and mRNA or portions thereof) transcribed from DNA encoding PTPR.alpha., and also cDNA derived from such RNA. hi a preferred embodiment, the target nucleic acid is an mRNA encoding human PTPR.alpha.. Target Nucleic Acids
"Targeting" an antisense compound to a particular target nucleic acid molecule can be a multistep process. The process usually begins with the identification of a target nucleic acid whose expression is to be modulated. For example, the target nucleic acid can be 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. As disclosed herein, the target nucleic acid encodes PTPR.alpha.. Variants
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. Variants can result in mRNA variants including, but not limited to, those with alternate splice junctions, or alternate initiation and termination codons. Variants in genomic and mRNA sequences can result in disease. . Target Names, Synonyms, Features
Accordingly, there are provided compositions and methods for modulating the expression of PTPR.alpha.. Table 1 lists the GenBank accession numbers of sequences corresponding to nucleic acid molecules encoding PTPR.alpha. (nt = nucleotide), the date the version of the sequence was entered in GenBank, and the corresponding SEQ ID NO in the instant application, when assigned, each of which is incorporated herein by reference.
Table 1 Gene Targets
Figure imgf000007_0001
Modulation of Target Expression
Modulation of expression of a target nucleic acid can be achieved through alteration of any number of nucleic acid (DNA or RNA) functions. "Modulation" or "Modulation of Expression" means a perturbation of function, for example, either an increase (stimulation or induction) or a decrease
(inhibition or reduction) in expression of the target mRNA.. As another example, modulation of expression can include perturbing splice site selection of pre-mRNA processing. "Expression" includes all the functions by which a gene's coded information is converted into structures present and operating in a cell. These structures include the products of transcription and translation. The functions of RNA to be modulated can include translocation functions, which include, but are not limited to, 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, and translation of protein from the RNA. RNA processing functions that can be modulated include, but are not limited to, splicing of the RNA to yield one or more RNA species, capping of the RNA, 3' maturation of the RNA and catalytic activity or complex formation involving the RNA which may be engaged in or facilitated by the RNA. Modulation of expression can result in the increased level of one or more nucleic acid species or the decreased level of one or more nucleic acid species, either temporally or by net steady state level. One result of such interference with target nucleic acid function is modulation of the expression of PTPR.alpha.. Thus, in one embodiment, modulation of expression can mean increase or decrease in target RNA or protein levels. In another embodiment modulation of expression can mean an increase or decrease of one or more RNA splice products, or a change in the ratio of two or more splice products.
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 and can be routinely determined using, for example, PCR or Northern blot analysis. Cell lines are derived from both normal tissues and cell types and from cells associated with various disorders (e.g. hyperproliferative disorders). Cell lines derived from multiple tissues and species can be obtained from American Type Culture Collection (ATCC, Manassas, VA) and other public sources. Primary cells, or those cells which are isolated from an animal and not subjected to continuous culture, can be prepared according to methods known in the art, or obtained from various commercial suppliers. Additionally, primary cells include those obtained from donor human subjects in a clinical setting (i.e. blood donors, surgical patients). These techniques are well known to those skilled in the art. Assaying Modulation of Expression
Modulation of PTPR.alpha. expression can be assayed in a variety of ways known in the art. PTPR.alpha. mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR. RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA by methods known in the art. Methods of RNA isolation are taught in, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.1.1-4.2.9 and 4.5.1-4.5.3, John Wiley & Sons, Inc., 1993.
Northern blot analysis is routine in the art and is taught in, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.2.1-4.2.9, John Wiley & Sons, Inc., 1996. Real-time quantitative (PCR) can be conveniently accomplished using the commercially available ABI PRISM™ 7700 Sequence Detection System, available from PE-Applied Biosystems, Foster City, CA and used according to manufacturer's instructions. The method of analysis of modulation of RNA levels is not a limitation of the instant invention.
Levels of a protein encoded by PTPR.alpha. can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), ELISA or fluorescence-activated cell sorting (FACS). Antibodies directed to a protein encoded by PTPR.alpha. can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, MI), or can be prepared via conventional antibody generation methods. Methods for preparation of polyclonal antisera are taught in, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.12.1 -11.12.9, John Wiley & Sons, Inc., 1997. Preparation of monoclonal antibodies is taught in, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.4.1-11.11.5, John Wiley & Sons, Inc., 1997.
Immunoprecipitation methods are standard in the art and can be found at, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.16.1-10.16.11, John Wiley & Sons, Inc., 1998. Western blot (immunoblot) analysis is standard in the art and can be found at, for example, Ausubel, F.M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.8.1-10.8.21, John Wiley & Sons, Inc., 1997. Active Target Segments
The locations on the target nucleic acid defined by having one or more active antisense compounds targeted thereto are referred to as "active target segments." There may be substantial variation in activity (e.g., as defined by percent inhibition) of the antisense compounds within an active target segment. Active antisense compounds are those that are determined to modulate the expression of their target RNA. Preferrably, active antisense compounds inhibit expression of their target RNA at least about 50%, more preferably at least about 70% and most preferably at least about 80%. In a more preferred embodiment, the level of inhibition required to define an active antisense compound is defined based on the results from the screen used to define the active target segments. Those skilled in the art understand that the percent inhibition by an antisense compound on a target mRNA will vary between assays due to factors relating to assay conditions. Hybridization
As used herein, "hybridization" means the pairing of complementary strands of antisense compounds to their target sequence. While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases). For example, the natural base adenine is complementary to the natural nucleobases thymidine and uracil which pair through the formation of hydrogen bonds. The natural base guanine is complementary to the natural base 5-methyl cytosine and the artificial base known as a G-clamp. Hybridization can occur under varying circumstances.
An antisense compound is specifically hybridizable when 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.
As used herein, "stringent hybridization conditions" or "stringent conditions" refers to conditions under which an antisense compound 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 "stringent conditions" under which antisense compounds hybridize to a target sequence are determined by the nature and composition of the antisense compounds and the assays in which they are being investigated. Complementarity
"Complementarity," as used herein, refers to the capacity for precise pairing between two nucleobases on either two oligomeric compound strands or an antisense compound with its target nucleic acid. For example, if a nucleobase at a certain position of an antisense compound is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be a complementary position. The antisense compound and the further DNA or RNA 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 antisense compound and a target nucleic acid. Identity
Antisense compounds, or a portion thereof, may have a defined percent identity to a SEQ ID NO, or a compound having a specific Isis number. As used herein, a sequence is identical to the sequence disclosed herein if it has the same nucleobase pairing ability. For example, a RNA which contains uracil in place of thymidine in the disclosed sequences would be considered identical as they both pair with adenine. Similarly, a G-clamp modified heterocyclic base would be considered identical to a cytosine or a 5-Me cytosine in the sequences of the instant application as it pairs with a guanine. This identity may be over the entire length of the oligomeric compound, or in a portion of the antisense compound (e.g., nucleobases 1-20 of a 27-mer may be compared to a 20-mer to determine percent identity of the oligomeric compound to the SEQ DD NO.) It is understood by those skilled in the art that an antisense compound need not have an identical sequence to those described herein to function similarly to the antisense compound described herein. Shortened versions of antisense compound taught herein, or non-identical versions of the antisense compound taught herein fall within the scope of this disclosure. Non-identical versions are those wherein each base does not have the same pairing activity as the antisense compounds disclosed herein. Bases do not have the same pairing activity by being shorter or having at least one abasic site. Alternatively, a non-identical version can include at least one base replaced with a different base with different pairing activity (e.g., G can be replaced by C, A, or T). Percent identity is calculated according to the number of bases that have identical base pairing corresponding to the SEQ ID NO or antisense compound to which it is being compared. The non-identical bases may be adjacent to each other, dispersed through out the oligonucleotide, or both.
For example, a 16-mer having the same sequence as nucleobases 2-17 of a 20-mer is 80% identical to the 20-mer. Alternatively, a 20-mer containing four nucleobases not identical to the 20-mer is also 80% identical to the 20-mer. A 14-mer having the same sequence as nucleobases 1-14 of an 18-mer is 78% identical to the 18-mer. Such calculations are well within the ability of those skilled in the art.
The percent identity is based on the percent of nucleobases in the original sequence present in a portion of the modified sequence. Therefore, a 30 nucleobase antisense compound comprising the full sequence of the complement of a 20 nucleobase active target segment would have a portion of 100% identity with the complement of the 20 nucleobase active target segment, while further comprising an additional 10 nucleobase portion. The complement of an active target segment may constitute a single portion. In a preferred embodiment, the oligonucleotides are at least about 80%, more preferably at least about 85%, even more preferably at least about 90%, most preferably at least 95% identical to at least a portion of the complement of the active target segments presented herein.
It is well known by those skilled in the art that it is possible to increase or decrease the length of an antisense compound and/or introduce mismatch bases without eliminating activity. For example, in Woolf et al. (Proc. Natl. Acad. Sci. USA 89:7305-7309, 1992, incorporated herein by reference), a series of ASOs 13-25 nucleobases in length were tested for their ability to induce cleavage of a target RNA. ASOs 25 nucleobases in length with 8 or 11 mismatch bases near the ends of the ASOs were able to direct specific cleavage of the target mRNA, albeit to a lesser extent than the ASOs that contained no mismatches. Similarly, target specific cleavage was achieved using a 13 nucleobase ASOs, including those with 1 or 3 mismatches. Maher and Dolnick (Nuc. Acid. Res. 16:3341-3358,1988, incorporated herein by reference) tested a series of tandem 14 nucleobase ASOs, and a 28 and 42 nucleobase ASOs comprised of the sequence of two or three of the tandem ASOs, respectively, for their ability to arrest translation of human DHFR in a rabbit reticulocyte assay. Each of the three 14 nucleobase ASOs alone were able to inhibit translation, albeit at a more modest level than the 28 or 42 nucleobase ASOs. Antisense compounds having a contiguous nucleobase composition that is shorter or longer or that comprises mismatches are contemplated in this invention so long as the antisense oligonucleotide activity is maintained. Therapeutics
Antisense compounds can be used to modulate the expression of PTPR.alpha. in an animal, such as a human. In one non-limiting embodiment, the methods comprise the step of administering to said animal in need of therapy for a disease or condition associated with, increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels , increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereof, an effective amount of an antisense compound that inhibits expression of PTPR.alpha.. A disease or condition associated with PTPR.alpha. includes, but is not limited to, , increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels , increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereof. In one embodiment, the antisense compounds effectively inhibit the levels or function of PTPR.alpha. RNA. Because reduction in PTPR.alpha. mRNA levels can lead to alteration in PTPR.alpha. protein products of expression as well, such resultant alterations can also be measured. Antisense compounds that effectively inhibit the level or function of PTPR.alpha. RNA or protein products of expression are considered an active antisense compounds. In one embodiment, the antisense compounds inhibit the expression of PTPR.alpha. causing a reduction of RNA by at least 10%, by at least 20%, by at least 25%, by at least 30%, by at least 40%, by at least 50%, by at least 60%, by at least 70%, by at least 75%, by at least 80%, by at least 85%, by at least 90%, by at least 95%, by at least 98%, by at least 99%, or by 100%.
For example, the reduction of the expression of PTPR.alpha. can be measured in a bodily fluid, tissue or organ of the animal. Methods of obtaining samples for analysis, such as body fluids (e.g., plasma), tissues (e.g., biopsy), or organs, and methods of preparation of the samples to allow for analysis are well known to those skilled in the art. Methods for analysis of RNA and protein levels are discussed above and are well known to those skilled in the art. The effects of treatment can be assessed by measuring biomarkers associated with the PTPR.alpha. expression in the aforementioned fluids, tissues or organs, collected from an animal contacted with one or more compounds, by routine clinical methods known in the art. These biomarkers include but are not limited to: liver transaminases, body weight, organ weight, percent fat, lipids, non-esterifϊed fatty acids, glucose, insulin and other markers of diabetes, fatty liver and metabolic syndrome.
The antisense compounds can be utilized in pharmaceutical compositions by adding an effective amount of a compound to a suitable pharmaceutically acceptable diluent or carrier. Acceptable carriers and diluents are well known to those skilled in the art. Selection of a diluent or carrier is based on a number of factors, including, but not limited to, the solubility of the compound and the desired route of administration. Such considerations are well understood by those skilled in the art. In one aspect, the compounds inhibit the expression of PTPR.alpha.. The antisense compounds can also be used in the manufacture of a medicament for the treatment of diseases and conditions related to PTPR.alpha. expression.
Methods whereby bodily fluids, organs or tissues are contacted with an effective amount of one or more of the antisense compounds or compositions are also contemplated. Bodily fluids, organs or tissues can be contacted with one or more of the compounds disclosed herein resulting in modulation of PTPR.alpha. expression in the cells of bodily fluids, organs or tissues.
Thus, provided herein is the use of an isolated single- or double-stranded antisense compound targeted to PTPR.alpha. in the manufacture of a medicament for the treatment of a disease or disorder by means of the method described above. In a preferred embodiment, the antisense compound is a single stranded antisense compound. Kits, Research Reagents, and Diagnostics The antisense compounds disclosed herein can be utilized for diagnostics, and as research reagents and kits. Furthermore, antisense compounds, which are able to inhibit gene expression with 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 antisense compounds, 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. Methods of gene expression analysis are well known to those skilled in the art. Compounds
The term "oligomeric compound" refers to a polymeric structure capable of hybridizing to a region of a nucleic acid molecule. Generally, oligomeric compounds comprise a plurality of monomelic subunits linked together by internucleoside linking groups and/or internucleoside linkage mimetics. Each of the monomelic subunits comprises a sugar, abasic sugar, modified sugar, or a sugar mimetic, and except for the abasic sugar includes a nucleobase, modified nucleobase or a nucleobase mimetic. Preferred monomeric subunits comprise nucleosides and modified nucleosides.
An "antisense compound" or "antisense oligomeric compound" refers to an oligomeric compound that is at least partially complementary to the region of a target nucleic acid molecule to which it hybridizes and which modulates (increases or decreases) its expression. This term includes oligonucleotides, oligonucleosides, oligonucleotide analogs, oligonucleotide mimetics, antisense compounds, antisense oligomeric compounds, and chimeric combinations of these. Consequently, while all antisense compounds can be said to be oligomeric compounds, not all oligomeric compounds are antisense compounds. An "antisense oligonucleotide" is an antisense compound that is a nucleic acid- based oligomer. An antisense oligonucleotide can, in some cases, include one or more chemical modifications to the sugar, base, and/or internucleoside linkages. Nonlimiting examples of antisense compounds include primers, probes, antisense compounds, antisense oligonucleotides, external guide sequence (EGS) oligonucleotides, alternate splicers, and siRNAs. As such, these compounds can be introduced in the form of single-stranded, double-stranded, circular, branched or hairpins and can contain structural elements such as internal or terminal bulges or loops. Antisense double-stranded compounds can be two strands hybridized to form double-stranded compounds or a single strand with sufficient self complementarity to allow for hybridization and formation of a fully or partially double-stranded compound. The compounds are not auto-catalytic. As used herein, "auto-catalytic" means a compound has the ability to promote cleavage of the target RNA in the absence of accessory factors, e.g. proteins.
In one embodiment, the antisense compound comprises a single stranded oligonucleotide. In some embodiments the antisense compound contains chemical modifications. In a preferred embodiment, the antisense compound is a single stranded, chimeric oligonucleotide wherein the modifications of sugars, bases, and internucleoside linkages are independently selected.
The antisense compounds disclosed herein may comprise an antisense compound from about 12 to about 35 nucleobases (i.e. from about 12 to about 35 linked nucleosides). In other words, a single-stranded compound comprises from about 12 to about 35 nucleobases, and a double-stranded antisense compound (such as a siRNA, for example) comprises two strands, each of which is independently from about 12 to about 35 nucleobases. This includes oligonucleotides 15 to 35 and 16 to 35 nucleobases in length. Contained within the antisense compounds (whether single or double stranded and on at least one strand) are antisense portions. The "antisense portion" is that part of the antisense compound that is designed to work by one of the aforementioned antisense mechanisms. One of ordinary skill in the art will appreciate that about 12 to about 35 nucleobases includes 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleobases.
Antisense compounds about 12 to 35 nucleobases in length, preferably about 15 to 35 nucleobases in length, comprising a complementary stretch of at least 8, preferably at least 12, more preferably at least 15 consecutive nucleobases selected from within the active target regions, are considered to be suitable antisense compounds as well.
Modifications can be made to the antisense compounds and may include conjugate groups attached to one of the termini, selected nucleobase positions, sugar positions or to one of the internucleoside linkages. Possible modifications include, but are not limited to, 2'-fluoro (2'-F), T- OMethyl (2'-OMe), 2'-Methoxy ethoxy (2'-MOE) sugar modifications, inverted abasic caps, deoxynucleobases, and bicyclice nucleobase analogs such as locked nucleic acids (including LNA) and ENA.
In one embodiment, double-stranded antisense compounds encompass short interfering RNAs (siRNAs). As used herein, the term "siRNA" is defined as a double-stranded compound having a first and second strand, each strand having a central portion and two independent terminal portions. The central portion of the first strand is complementary to the central portion of the second strand, allowing hybridization of the strands. The terminal portions are independently, optionally complementary to the corresponding terminal portion of the complementary strand. The ends of the strands may be modified by the addition of one or more natural or modified nucleobases to form an overhang
Each strand of the siRNA duplex may be from about 12 to about 35 nucleobases. In a preferred embodiment, each strand of the siRNA duplex is about 17 to about 25 nucleobases. The two strands may be fully complementary (i.e., form a blunt ended compound), or include a 5' or 3' overhang on one or both strands. Double-stranded compounds can be made to include chemical modifications as discussed herein. Chemical 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 (sometimes referred to as a "nucleobase" or simply a "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 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. 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. It is often preferable to include chemical modifications in oligonucleotides to alter their activity. Chemical modifications can alter oligonucleotide activity by, for example: increasing affinity of an antisense oligonucleotide for its target RNA, increasing nuclease resistance, and/or altering the pharmacokinetics of the oligonucleotide. The use of chemistries that increase the affinity of an oligonucleotide for its target can allow for the use of shorter oligonucleotide compounds.
The term "nucleobase" or "heterocyclic base moiety" as used herein, refers to the heterocyclic base portion of a nucleoside. In general, a nucleobase is any group that contains one or more atom or groups of atoms capable of hydrogen bonding to a base of another nucleic acid. In addition to "unmodified" or "natural" nucleobases such as the purine nucleobases adenine (A) and guanine (G), and the pyrimidine nucleobases thymine (T), cytosine (C) and uracil (U), many modified nucleobases or nucleobase mimetics known to those skilled in the art are amenable to the disclosed antisense compounds. The terms modified nucleobase and nucleobase mimetic can overlap but generally a modified nucleobase refers to a nucleobase that is fairly similar in structure to the parent nucleobase, such as for example a 7- deaza purine or a 5-methyl cytosine, whereas a nucleobase mimetic would include more complicated structures, such as for example a tricyclic phenoxazine nucleobase mimetic. Methods for preparation of the above noted modified nucleobases are well known to those skilled in the art.
Antisense compounds disclosed herein may also contain one or more nucleosides having modified sugar moieties. The furanosyl sugar ring of a nucleoside can be modified in a number of ways including, but not limited to, addition of a substituent group, bridging of two non-geminal ring atoms to form a bicyclic nucleic acid (BNA) and substitution of an atom or group such as -S-, -N(R)- or - C(R.sub.l)(R.sub.2) for the ring oxygen at the 4'-position. Modified sugar moieties are well known and can be used to alter, typically increase, the affinity of the antisense compound for its target and/or increase nuclease resistance. A representative list of preferred modified sugars includes but is not limited to bicyclic modified sugars (BNA's), including LNA and ENA (4'-(CH.sub.2).sub.2-O-2' bridge); and substituted sugars, especially 2'-substituted sugars having a 2'-F, 2'-OCH.sub.2 or a 2'-O(CH.sub.2).sub.2- OCH3 substituent group. Sugars can also be replaced with sugar mimetic groups among others. Methods for the preparations of modified sugars are well known to those skilled in the art.
Also included are internucleoside linking groups that link the nucleosides or otherwise modified monomer units together thereby forming an antisense compound. The two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom. Representative phosphorus containing internucleoside linkages include, but are not limited to, phosphodiesters, phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates. Representative non-phosphorus containing internucleoside linking groups include, but are not limited to, methylenemethylimino (-CH.sub.2-N(CH.sub.3)-O-CH.sub.2-), thiodiester (-O-C(O)-S-), thionocarbamate (-0-C(O)(NH)-S-); siloxane (-0-Si(H)2-0-); and N,N'-dimethylhydrazine (-CH.sub.2- N(CH.sub.3)-N(CH.sub.3)-). Antisense compounds having non-phosphorus internucleoside linking groups are referred to as oligonucleosides. Modified internucleoside linkages, compared to natural phosphodiester linkages, can be used to alter, typically increase, nuclease resistance of the antisense compound. Internucleoside linkages having a chiral atom can be prepared racemic, chiral, or as a mixture. Representative chiral internucleoside linkages include, but are not limited to, alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous- containing linkages are well known to those skilled in the art.
As used herein the term "mimetic" refers to groups that are substituted for a sugar, a nucleobase, and/ or internucleoside linkage. Generally, a mimetic is used in place of the sugar or sugar- internucleoside linkage combination, and the nucleobase is maintained for hybridization to a selected target. Representative examples of a sugar mimetic include, but are not limited to, cyclohexenyl or morpholino. Representative examples of a mimetic for a sugar-internucleoside linkage combination include, but are not limited to, peptide nucleic acids (PNA) and morpholino groups linked by uncharged achiral linkages. In some instances a mimetic is used in place of the nucleobase. Representative nucleobase mimetics are well known in the art and include, but are not limited to, tricyclic phenoxazine analogs and universal bases (Berger et al., Nuc Acid Res. 2000, 28:2911-14, incorporated herein by reference). Methods of synthesis of sugar, nucleoside and nucleobase mimetics are well known to those skilled in the art.
As used herein the term "nucleoside" includes, nucleosides, abasic nucleosides, modified nucleosides, and nucleosides having mimetic bases and/or sugar groups.
The term "oligonucleotide" refers to an oligomeric compound which is an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA). This term includes oligonucleotides composed of naturally- and non-naturally-occurring nucleobases, sugars and covalent internucleoside linkages, possibly further including non-nucleic acid conjugates.
Compounds having reactive phosphorus groups are used for forming internucleoside linkages including for example phosphodiester and phosphorothioate internucleoside linkages. Methods of preparation and/or purification of precursors or antisense compounds are not a limitation of the compositions or methods disclosed herein. Methods for synthesis and purification of DNA, RNA, and the antisense compounds are well known to those skilled in the art.
As used herein the term "chimeric antisense compound" refers to an antisense compound, having at least one sugar, nucleobase and/or internucleoside linkage that is differentially modified as compared to the other sugars, nucleobases and internucleoside linkages within the same oligomeric compound. The remainder of the sugars, nucleobases and internucleoside linkages can be independently modified or unmodified. In general a chimeric oligomeric compound will have modified nucleosides that can be in isolated positions or grouped together in regions that will define a particular motif. Any combination of modifications and or mimetic groups can comprise a chimeric oligomeric compound.
Chimeric oligomeric compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the oligomeric compound may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNAiRNA hybrids. By way of example, RNase H is a cellular endonuclease that 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 inhibition of gene expression. Consequently, comparable results can often be obtained with shorter oligomeric compounds when chimeras are used, compared to for example phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.
Certain chimeric as well as non-chimeric oligomeric compounds can be further described as having a particular motif. As used herein, the term "motif refers to the orientation of modified sugar moieties and/or sugar mimetic groups in an antisense compound relative to like or differentially modified or unmodified nucleosides. As used herein, the terms "sugars", "sugar moieties" and "sugar mimetic groups' are used interchangeably. Such motifs include, but are not limited to, gapped motifs, alternating motifs, fully modified motifs, hemimer motifs, blockmer motifs, and positionally modified motifs. The sequence and the structure of the nucleobases and type of internucleoside linkage is not a factor in determining the motif of an antisense compound.
As used herein, the term "gapped motif refers to an antisense compound comprising a contiguous sequence of nucleosides that is divided into 3 regions, an internal region (gap) flanked by two external regions (wings). The regions are differentiated from each other at least by having differentially modified sugar groups that comprise the nucleosides. In some embodiments, each modified region is uniformly modified (e.g. the modified sugar groups in a given region are identical); however, other motifs can be applied to regions. For example, the wings in a gapmer could have an alternating motif. The nucleosides located in the gap of a gapped antisense compound have sugar moieties that are different than the modified sugar moieties in each of the wings.
As used herein, the term "alternating motif refers to an antisense compound comprising a contiguous sequence of nucleosides comprising two differentially sugar modified nucleosides that alternate for essentially the entire sequence of the antisense compound, or for essentially the entire sequence of a region of an antisense compound.
As used herein, the term "fully modified motif refers to an antisense compound comprising a contiguous sequence of nucleosides wherein essentially each nucleoside is a sugar modified nucleoside having uniform modification.
As used herein, the term "hemimer motif refers to a sequence of nucleosides that have uniform sugar moieties (identical sugars, modified or unmodified) and wherein one of the 5 '-end or the 3'- end has a sequence of from 2 to 12 nucleosides that are sugar modified nucleosides that are different from the other nucleosides in the hemimer modified antisense compound.
As used herein, the term "blockmer motif refers to a sequence of nucleosides that have uniform sugars (identical sugars, modified or unmodified) that is internally interrupted by a block of sugar modified nucleosides that are uniformly modified and wherein the modification is different from the other nucleosides. Methods of preparation of chimeric oligonucleotide compounds are well known to those skilled in the art.
As used herein, the term "positionally modified motif comprises all other motifs. Methods of preparation of positionally modified oligonucleotide compounds are well known to those skilled in the art.
The compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), alpha or beta, or as (D) or (L) such as for amino acids et al. All such possible isomers, as well as their racemic and optically pure forms, are included herein.
In one aspect, antisense compounds are modified by covalent attachment of one or more conjugate groups. Conjugate groups may be attached by reversible or irreversible attachments. Conjugate groups may be attached directly to antisense compounds or by use of a linker. Linkers may be mono- or bifunctional linkers. Such attachment methods and linkers are well known to those skilled in the art. In general, conjugate groups are attached to antisense compounds to modify one or more properties. Such considerations are well known to those skilled in the art. Oligomer Synthesis
Oligomerization of modified and unmodified nucleosides can be routinely performed according to literature procedures for DNA (Protocols for Oligonucleotides and Analogs, Ed. Agrawal (1993), Humana Press) and/or RNA (Scaringe, Methods (2001), 23, 206-217. Gait et al., Applications of Chemically synthesized RNA in RNA: Protein Interactions, Ed. Smith (1998), 1-36. Gallo et al., Tetrahedron (2001), 57, 5707-5713).
Antisense compounds can 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, CA). 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. The invention is not limited by the method of antisense compound synthesis. Oligomer Purification and Analysis
Methods of oligonucleotide purification and analysis are known to those skilled in the art. Analysis methods include capillary electrophoresis (CE) and electrospray-mass spectroscopy. Such synthesis and analysis methods can be performed in multi-well plates. The method of the invention is not limited by the method of oligomer purification. Salts, prodrugs and bioequivalents
The antisense compounds disclosed herein may comprise any pharmaceutically acceptable salts, esters, or salts of such esters, or any other functional chemical equivalent which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the antisense compounds, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.
The term "prodrug" indicates a therapeutic agent that is prepared in an inactive or less active form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes, chemicals, and/or conditions. In particular, prodrug versions of the oligonucleotides are prepared as SATE ((S-acetyl-2-thioethyl) phosphate) derivatives according to the methods disclosed in WO 93/24510 or WO 94/26764. Prodrugs can also include antisense compounds wherein one or both ends comprise nucleobases that are cleaved (e.g., by incorporating phosphodiester backbone linkages at the ends) to produce the active compound.
The term "pharmaceutically acceptable salts" refers to physiologically and pharmaceutically acceptable salts of the compounds: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. Sodium salts of antisense oligonucleotides are useful and are well accepted for therapeutic administration to humans. In another embodiment, sodium salts of dsRNA compounds are also provided. Formulations
The antisense compounds may also be admixed, encapsulated, conjugated or otherwise associated with other molecules, molecule structures or mixtures of compounds.
Also included are pharmaceutical compositions and formulations which include the antisense compounds . The pharmaceutical compositions may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated
The pharmaceutical formulations, 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, finely divided solid carriers, or both, and then, if necessary, shaping the product (e.g., into a specific particle size for delivery).
A "pharmaceutical carrier" or "excipient" can be a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal and are known in the art. The excipient may be liquid or solid and is selected, with the planned manner of administration in mind, so as to provide for the desired bulk, consistency, etc., when combined with a nucleic acid and the other components of a given pharmaceutical composition. Combinations
Pharmaceutical compositions may comprise two or more antisense compounds. In another related embodiment, pharmaceutical compositions may comprise 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, pharmaceutical compositions may comprise two or more antisense compounds targeted to different regions of the same nucleic acid target. Two or more combined compounds may be used together or sequentially. Pharmaceutical compositions may comprise an antisense compound combined with other non-antisense compound therapeutic agents. Nonlimiting disclosure and incorporation by reference
While certain compounds, compositions and are being described herein with specificity in accordance with certain embodiments, the following examples serve only as illustrations and not as limitations. Each of the references, GenBank accession numbers, and the like recited in the present application is incorporated herein by reference in its entirety.
Example 1: Cell Types and Transfection Methods
Example 1- Cell types- The effect of oligomeric compounds on target nucleic acid expression was tested in one or more of the following cell types.
T-24 cells: The human transitional cell bladder carcinoma cell line T-24 was obtained from the American Type Culture Collection (ATCC) (Manassas, VA). T-24 cells were routinely cultured in complete McCoy's 5 A basal media (Invitrogen Corporation, Carlsbad, CA) supplemented with 10% fetal calf serum (Invitrogen Corporation, Carlsbad, CA), penicillin 100 units per mL, and streptomycin 100 micrograms per mL (Invitrogen Corporation, Carlsbad, CA). Cells were routinely passaged by trypsinization and dilution when they reached 90% confluence. Cells were seeded into 96-well plates (Falcon-Primaria #353872) at a density of 7000 cells/well for use in RT-PCR analysis. b.END cells: The mouse brain endothelial cell line b.END was obtained from Dr. Werner Risau at the Max Plank Institute (Bad Nauheim, Germany). b.END cells were routinely cultured in DMEM, high glucose (Gibco/Life Technologies, Gaithersburg, MD) supplemented with 10% fetal calf serum (Gibco/Life Technologies, Gaithersburg, MD). Cells were routinely passaged by trypsinization and dilution when they reached 90% confluence. Cells were seeded into 96-well plates (Falcon-Primaria #3872) at a density of 3000 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.
NHDF cells: Human neonatal dermal fibroblast (NHDF) were obtained from the Clonetics Corporation (Walkersville, MD). NHDFs were routinely maintained in Fibroblast Growth Medium (Clonetics Corporation, Walkersville, MD) supplemented as recommended by the supplier. Cells were maintained for up to 10 passages as recommended by the supplier.
HEK cells: Human embryonic keratinocytes (HEK) were obtained from the Clonetics Corporation (Walkersville, MD). HEKs were routinely maintained in Keratinocyte Growth Medium (Clonetics Corporation, Walkersville, MD) formulated as recommended by the supplier. Cells were routinely maintained for up to 10 passages as recommended by the supplier.
Treatment with oligomeric compounds: When cells reach appropriate confluency, they are treated with oligonucleotide using a transfection method as described. Lipofectin™ When cells reached 65-75% confluency, they were treated with oligonucleotide. Oligonucleotide was mixed with LIPOFECTIN™ Invitrogen Life Technologies, Carlsbad, CA) in Opti-MEM™-1 reduced serum medium (Invitrogen Life Technologies, Carlsbad, CA) to achieve the desired concentration of oligonucleotide and a LIPOFECTIN ™ concentration of 2.5 or 3 .micro.g/mL per 100 nM oligonucleotide. This transfection mixture was incubated at room temperature for approximately 0.5 hours. For cells grown in 96-well plates, wells were washed once with 100 .micro.L OPTI-MEM™-1 and then treated with 130 .micro.L of the transfection mixture. Cells grown in 24-well plates or other standard tissue culture plates are treated similarly, using appropriate volumes of medium and oligonucleotide. Cells are treated and data are obtained in duplicate or triplicate. After approximately 4-7 hours of treatment at 37.deg.C, the medium containing the transfection mixture was replaced with fresh culture medium. Cells were harvested 16-24 hours after oligonucleotide treatment. Control oligonucleotides
Control oligonucleotides are used to determine the optimal oligomeric compound concentration for a particular cell line. Furthermore, when oligomeric compounds are tested in oligomeric compound screening experiments or phenotypic assays, control oligonucleotides are tested in parallel.
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. The concentration of positive control oligonucleotide that results in 80% inhibition of the target 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 the target 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 when the antisense oligonucleotide is transfected using a liposome reagent and 1 μM to 40 μM when the antisense oligonucleotide is transfected by electroporation. Example 2: Real-time Quantitative PCR Analysis of PTPR.alpha. mRNA Levels
Quantitation of PTPR.alpha. 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, CA) according to manufacturer's instructions.
Prior to quantitative PCR analysis, primer-probe sets specific to the PTPR.alpha. being measured were evaluated for their ability to be "multiplexed" with a GAPDH amplification reaction. After isolation the RNA is subjected to sequential reverse transcriptase (RT) reaction and real-time PCR, both of which are performed in the same well. RT and PCR reagents were obtained from Invitrogen Life Technologies (Carlsbad, CA). RT, real-time PCR was carried out in the same by adding 20 .micro.L PCR cocktail (2.5x PCR buffer minus MgCl.sub.2, 6.6 mM MgCl.sub.2, 375 .micro.M 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 2.5x ROX dye) to 96- well plates containing 30 .micro.L total RNA solution (20-200 ng). The RT reaction was carried out by incubation for 30 minutes at 48.deg.C. Following a 10 minute incubation at 95.deg.C to activate the PLATINUM® Taq, 40 cycles of a two-step PCR protocol were carried out: 95.deg.C for 15 seconds (denaturation) followed by όO.deg.C for 1.5 minutes (annealing/extension).
Gene target quantities obtained by RT, real-time PCR were normalized using either the expression level of GAPDH, a gene whose expression is constant, or by quantifying total RNA using RiboGreen™ (Molecular Probes, Inc. Eugene, OR). GAPDH expression was quantified by RT, real-time PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA was quantified using RiboGreen™ RNA quantification reagent (Molecular Probes, Inc. Eugene, OR).
170 .micro.L of RiboGreen™ working reagent (RiboGreen™ reagent diluted 1 :350 in 1OmM Tris-HCl, 1 mM EDTA, pH 7.5) was pipetted into a 96-well plate containing 30 .micro.L purified cellular RNA. The plate was read in a CytoFluor 4000 (PE Applied Biosystems) with excitation at 485nm and emission at 530nm.
The GAPDH PCR probes have JOE covalently linked to the 5' end and TAMRA or MGB covalently linked to the 3' end, where JOE is the fluorescent reporter dye and TAMRA or MGB is the quencher dye. In some cell types, primers and probe designed to a GAPDH sequence from a different species are used to measure GAPDH expression. For example, a human GAPDH primer and probe set is used to measure GAPDH expression in monkey-derived cells and cell lines.
Probes and primers for use in real-time PCR were designed to hybridize to target-specific sequences. The primers and probes and the target nucleic acid sequences to which they hybridize are presented in Table 2. The target-specific PCR probes have FAM covalently linked to the 5' end and TAMRA or MGB covalently linked to the 3' end, where FAM is the fluorescent dye and TAMRA or MGB is the quencher dye.
Table 2 PTPR.alpha.-specific primers and probes for use in real-time PCR
Figure imgf000022_0001
Example 3: Antisense inhibition of human PTPR.alpha. expression by oligomeric compounds
Accordingly, a series of oligonucleotides were designed to target different regions of the human PTPR.alpha. RNA, using published sequences (GenBank accession number NM 080841.1, representing a human PTPR.alpha. variant herein designated hPTPRA-3, incorporated herein as SEQ ID NO: 4; GenBank accession number M34668.1, representing the main mRNA of human PTPR.alpha., herein designated hPTPRA, incorporated herein as SEQ ID NO: 18; a consensus sequence constructed from GenBank accession numbers AL161656.15 and AL121905.23, representing a genomic sequence of human PTPR.alpha., incorporated herein as SEQ ID NO: 19; GenBank accession number X54890.1, representing a human PTPR.alpha. variant herein designated hPTPRA-4, incorporated herein as SEQ ID NO: 20; GenBank accession number X53364.1 , representing a human PTPR.alpha. variant herein designated hPTPRA-5, incorporated herein as SEQ ID NO: 21; GenBank accession number AF121 183.1, representing a 5'-extension of SEQ ID NO: 18, incorporated herein as SEQ ID NO: 22; the complement of GenBank accession number BEl 68541.1, representing a 5'-extenstion of SEQ ID NO: 22 incorporated herein as SEQ DD NO: 23; the complement of GenBank accession number AW024120.1, representing a 5'-extension of SEQ ID NO: 20, incorporated herein as SEQ ID NO: 24; the complement of GenBank accession number AA903762.1, representing a 3 '-extension of SEQ ID NO: 18, incorporated herein as SEQ ID NO: 25; the complement of GenBank accession number AI674319.1, representing a variant of human PTPR.alpha. herein designated hPTPRA-7, incorporated herein as SEQ ID NO: 26, and residues 2749000-2925000 of GenBank accession number NT Ol 1387.6, representing a genomic sequence of human PRPTA, incorporated herein as SEQ ID NO: 27). The oligonucleotides are shown in Table 3. "Target site" indicates the first (5 '-most) nucleotide number on the particular target sequence to which the oligonucleotide binds. All compounds in Table 3 are chimeric oligonucleotides ("gapmers") 20 nucleotides in length, composed of a central "gap" region consisting often 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 PTPR.alpha. mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from two experiments in which T-24 cells were treated with the antisense oligonucleotides. The positive control for each data point is identified in the table by sequence ID number. If present, "N.D." indicates "no data".
Table 3
Inhibition of human PTPR.alpha. mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap
Figure imgf000023_0001
Figure imgf000024_0001
Figure imgf000025_0001
As shown in Table 3, SEQ ID NOS: 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, 57, 60, 61, 62, 63, 64, 65, 67, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 81, 90 and 92 demonstrated at least 50% inhibition of human PTPR.alpha. expression in this assay and are therefore preferred. The target sites to which these preferred sequences are complementary are herein referred to as "preferred target sites" and are therefore preferred sites for targeting by antisense compounds. These preferred target sites are shown in Table 5.
Example 4: Antisense inhibition of mouse PTPR.alpha. expression by oligomeric compounds
Accordingly, a second series of oligonucleotides were designed to target different regions of the mouse PTPR.alpha. RNA, using published sequences (GenBank accession number NM_008980.1, representing a variant of mouse PTPR.alpha. herein designated mPTPRA-2, incorporated herein as SEQ ID NO: 11; GenBank accession number AW323517.1, representing a 5'-extension of SEQ ID NO: 11, incorporated herein as SEQ ID NO: 93; GenBank accession numbers L13686.1, L13668.1, L13670.1, Ll 3672.1, Ll 3674.1, Ll 3675.1, Ll 3676.1 and Ll 3608.1, representing partial genomic sequences of mouse PTPR.alpha., incorporated herein as SEQ ID NOs: 94, 95, 96, 97, 98, 99, 100 and 101 respectively; and GenBank accession number BE372094.1, representing the main mRNA of mouse PTPR.alpha., herein designated mPTPRA, incorporated herein as SEQ ED NO: 102). The oligonucleotides are shown in Table 4. "Target site" indicates the first (5 '-most) nucleotide number on the particular target sequence to which the oligonucleotide binds. All compounds in Table 4 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. It is expected that antisense oligonucleotides with mismatches or varying nucleotide lengths will have similar profiles to those described herein. 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 PTPR.alpha. mRNA levels by quantitative real-time PCR as described in other examples herein. Data are averages from two experiments. The positive control for each data point is identified in the table by sequence ID number. If present, "N.D." indicates "no data".
Table 4
Inhibition of mouse PTPR.alpha. mRNA levels by chimeric phosphorothioate oligonucleotides having 2'-MOE wings and a deoxy gap
Figure imgf000026_0001
Figure imgf000027_0001
As shown in Table 4, SEQ ID NOs 103, 106, 108, 110, 111, 112, 116, 117, 118, 119, 122, 125, 127, 128, 129, 130, 134, 135, 136, 137, 139, 145, 148 and 149 demonstrated at least 50% inhibition of mouse PTPR.alpha. expression in this experiment and are therefore preferred. The target sites to which these preferred sequences are complementary are herein referred to as "preferred target sites" and are therefore preferred sites for targeting antisense oligonucleotides. These preferred target sites are shown in Table 5. Moreover, nucleotides 2860 to 2990 of SEQ ID NO: 11 form a region whereint eh screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 70%, and thus is designated as an active target segment (active target segment E).
Table 5 Sequence and position of preferred target sites identified in PTPR.alpha..
Figure imgf000027_0002
Figure imgf000028_0001
As these "preferred target regions" have been found by experimentation to be open to, and accessible for, hybridization with the antisense compounds, one of skill in the art will recognize or be able to ascertain, using no more than routine experimentation, further embodiments encompassing other compounds that specifically hybridize to these sites and consequently inhibit the expression of PTPR.alpha..
In one embodiment, the "preferred target region" may be employed in screening candidate antisense compounds. "Candidate antisense compounds" are those that inhibit the expression of a nucleic acid molecule encoding PTPR.alpha. and which comprise at least an 8-nucleobase portion which is complementary to a preferred target region. The method comprises the steps of contacting a preferred target region of a nucleic acid molecule encoding PTPR.alpha. with one or more candidate antisense compounds, and selecting for one or more candidate antisense compounds which inhibit the expression of a nucleic acid molecule encoding PTPR.alpha.. Once it is shown that the candidate antisense compound or compounds are capable of inhibiting the expression of a nucleic acid molecule encoding PTPR.alpha., the candidate antisense compound may be employed as an antisense compound as described herein.
Accordingly, antisense compounds include ribozymes, external guide sequence (EGS) oligonucleotides (oligozymes), and other short catalytic RNAs or catalytic oligonucleotides which hybridize to the target nucleic acid and modulate its expression.
Example 5
Targeting of individual oligonucleotides to specific variants of human PTPR.alpha.
In some instances it is advantageous to selectively inhibit the expression of one or more variants of PTPR.alpha.. Consequently, there are provided oligonucleotides that selectively target, hybridize to, and specifically inhibit one or more, but fewer than all of the variants of human PTPR.alpha.. A summary of the target sites of the PTPR.alpha. main mRNA and variants is shown in Table 6 and includes GenBank accession number NM_080841.1 , representing hPTPRA-3 , incorporated herein as SEQ ID NO: 4; GenBank accession number M34668.1, representing hPTPR.alpha. main mRNA, incorporated herein as SEQ ID NO: 18; GenBank accession number X54890.1, representing hPTPRA-4, incorporated herein as SEQ ID NO: 20; GenBank accession number X53364.1 , representing hPTPRA-5, incorporated herein as SEQ ID NO: 21; the complement of GenBank accession number AI674319.1, representing hPTPRA-7, incorporated herein as SEQ ID NO: 26; GenBank accession number NM_002836.1 (entered Mar. 24, 1999), representing hPTPRA-1, incorporated herein as SEQ ID NO: 225; GenBank accession number NM 080840.1 (entered Jan. 31, 2002), representing hPTPRA-2, incorporated herein as SEQ ID NO: 226; and GenBank accession number X54130.1 (entered Apr. 21, 1993), representing hPTPRA-6, incorporated herein as SEQ ID NO: 227.
Table 6 Targeting of individual oligonucleotides to specific variants of human PTPR.alpha.
Figure imgf000030_0001
Figure imgf000031_0001
Figure imgf000032_0001
Figure imgf000033_0001
Human PTPR.alpha. mRNA sequences from Table 1 were aligned and the screened human oligos were determined to have a complementary sequence to a common PTPR.alpha. mRNA, specifically (SEQ ID NO: 18). Antisense compounds targeting SEQ ID NO: 18 are shown in Table 6b. The antisense compounds were then grouped according to activity and active target segments were determined. An active target segment is used herein to describe a region of the PTPR.alpha. mRNA that has one or more active antisense compounds targeted thereto. Each of the following active target segment was targeted by multiple, active antisense oligonucleotides. These regions include nucleotides 716 to 894, nucleotides 1063 to 1229, nucleotides 1818 to 2045 and nucleotides 2328 to 2779 of SEQ ID NO: 18. Each of the oligonucleotides tested within each of these regions inhibited expression of human PTPR.alpha. by at least 60% in this assay. In active target segment A (nucleotides 716 to 894) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 85%. In active target segment B (nucleotides 1063 to 1229) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 75%. In active target segment C (nucleotides 1818 to 2045) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 90%. In active target segment D (nucleotides 2328 to 2779) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 80%. In active target segment F (nucleotides 1210-1905) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 87%. In active target segment G (nucleotides 2982-3211) the screened oligonucleotides inhibited expression of PTPR.alpha. by an average of about 73%.Identification of these regions allows for the design of antisense oligonucleotides that modulate the expression of PTPR.alpha..
Table 6b Exemplary human PTPR.alpha. oligos targeting SEQ ID NO: 18
Figure imgf000034_0001
Figure imgf000035_0001
Example 6: Targeting of individual oligonucleotides to specific variants of mouse PTPR.aIpha.
It is advantageous to selectively inhibit the expression of one or more variants of PTPR.aIpha. Oligonucleotides are provided that selectively target, hybridize to, and specifically inhibit one or more, but fewer than all of the variants of mouse PTPR.aIpha.. A summary of the target sites of the PTPR.alpha. main mRNA and variants is shown in Table 7 and includes GenBank accession number NM_008980.1, representing mPTPRA-2, incorporated herein as SEQ ID NO: 11 and GenBank accession number BE372094.1, representing mPTPR.alpha. main mRNA, incorporated herein as SEQ ID NO: 102.
Table 7 Targeting of individual oligonucleotides to specific variants of mouse PTPR.alpha.
Figure imgf000035_0002
Figure imgf000036_0001
Example 7
Effect of antisense inhibitors of PTPR.alpha. in ob/ob mice: a model of obesity and diabetes. ob/ob mice are used as a model of obesity and of type-2 diabetes. These mice have a deficiency in the Leptin hormone produced by fat that regulates appetite. Deficiencies in this hormone in both humans and non-human animals lead to obesity. The ob/ob mice have a mutation in the leptin gene which results in obesity and hyperglycemia, ob/ob mice have higher circulating levels of insulin and are less hyperglycemic than db/db mice, which harbor a mutation in the leptin receptor. ISIS 147299 (SEQ ID NO: 135) was tested in the ob/ob model of obesity and diabetes.
Male C57B1/6J-Lep ob/ob mice (Jackson Laboratory, Bar Harbor, ME) were fed a diet with a fat content of 10-15% and were subcutaneously injected with ISIS 147299 at a dose of 25 mg/kg two times per week for 4 weeks. The mice received a final dose at 12.5 mg/kg at day 30. Saline-injected animals served as controls. After the treatment period, mice were sacrificed and target levels were evaluated in liver and fat. RNA isolation and target mRNA expression level quantitation were performed as described by other examples herein. Animals treated with ISIS 147299 on average showed about a 75% reduction in liver PTPR.alpha. mRNA levels (n=5), and on average about a 50% reduction in fat PTPR.alpha. (n=5) compared to saline controls.
To assess the physiological effects resulting from inhibition of PTPR.alpha. mRNA, the ob/ob mice were further evaluated throughout the treatment period and at the end of the treatment period for body weight, percent fat from body weight, organ weight, chemistries, including serum transaminase, cholesterol, triglycerides and non-esterified fatty acids, liver triglycerides, plasma glucose levels, fed plasma insulin levels and glucose tolerance. Triglycerides, lipoproteins, cholesterol and transaminases were measured by routine clinical analyzer instruments (e.g. Olympus Clinical Analyzer, Melville, NY). Serum free fatty acids were measured using a Wako Chemicals kit for non-esterifϊed free fatty acids (Richmond, VA). Serum apolipoproteins were measured using apolipoprotein-specific ELISA or by protein immunoblot with apolipoprotein-specific antibodies. Tissue triglyceride levels were measured using a Triglyceride GPO Assay from Roche Diagnostics (Indianapolis, IN). Liver triglyceride levels were used to assess hepatic steatosis, or clearing of lipids from the liver. Hepatic steatosis was also assessed by routine histological analysis of frozen liver tissue sections stained with oil red O stain.
The effects of target inhibition on glucose and insulin metabolism were evaluated in the ob/ob mice treated with the oligomeric compounds. Plasma glucose was measured at the start of the treatment and after 2 weeks, 3 weeks and 4.5 weeks of treatment. Mice were sacrificed at day 33 and plasma glucose was measured. (Table 8) Plasma insulin is similarly measured at the beginning of the treatment, and at 2 weeks, at 3 weeks and at 4.5 weeks of treatment. (Table 9)
Table 8 Fed Plasma Glucose Levels
Figure imgf000037_0001
Table 9 Fed Plasma Insulin Levels
Figure imgf000037_0002
In ob/ob mice treated with ISIS 147299, fed plasma glucose levels were approximately 403.3 mg/dl at base line, 376.7 mg/dl at week 2, 330.0 mg/dl at week 3 and 295.7 mg/dl at sacrifice. Fed plasma insulin levels were approximately 21.57 ng/ml at baseline, 31.37 at week 2, 25.08 at week 3 and 15.2 at week 4.5. Compared to saline control animals, plasma glucose levels are reduced in animals following treatment with an antisense oligonucleotide that inhibit PTPR.alpha..
Plasma lipids in ob/ob mice were also measured throughout the treatment period and at the end of the treatment period. Table 10. Cholesterol levels were approximately 179.3 mg/dl at baseline, 240.3 mg/dl after 2 weeks, 242.9 mg.dl after 3 weeks and 247.3 mg/dl at sacrifice for the ice receiving an antisense inhibitor of PTPR.alpha.. There was no significant change in plasma cholesterol levels for mice receiving an antisense inhibitor of PTPR.alpha.. Triglycerides were approximately 165.7 mg/dl at base line, 107.9 mg/dl after 2 weeks, 87.4 mg/dl after 3 weeks and 88.9 mg/dl at sacrifice for ISIS 147299-treated mice. For mice receiving treatment with an antisense inhibitor of PTPR.alpha. there was a decrease in fed plasma triglyceride levels.
Table 10 Plasma Lipid Levels in ob/ob mice
Figure imgf000038_0001
Treatment of the ob/ob mice with ISIS 147299 decreased hepatic triglyceride levels and improved hepatic function. Liver triglycerides were approximately 109.71 mg/g after 4.5 weeks of treatment. (Table 11) Oil Red-0 staining of liver sections showed a marked clearing of lipid deposits for the treated group compared to control. (Figures Ia and Ib.) Plasma transaminase levels were reduced in the mice receiving treatment compared to those receiving saline. Plasma Alanine Aminotransferase (ALT) levels were approximately 86.3 IU/L at baseline, 244.1 after 2 weeks, 258.3 IU/1 after 3 weeks and 137.7 IU/L at sacrifice for the treated group. Plasma Aspartate Aminotransferase (AST) levels were approximately 71.1 RJ/L at base line, 128.4 KJ/L after 2 weeks, 144.0 KJ/L after 3 weeks and 134.4 IU/L at sacrifice following treatment. Fasted transaminase levels were determined after 4 weeks of treatment and were approximately 246.9 R7/L for ALT and 139.1 for AST. Table 12. These values show that treatment with an antisense inhibitor of PTPR.alpha. reduced hepatic and plasma triglyceride levels and improved hepatic function compared to the saline control group.
Table 11 Liver Triglyceride Levels
Figure imgf000038_0002
Table 12 Plasma Transaminase Levels
Figure imgf000038_0003
Figure imgf000039_0001
No changes in body weight, fat content, plasma cholesterol or plasma free fatty acid levels were observed for mice receiving treatment with ISIS 147299 compared to saline controls.
These results indicate that antisense inhibition of PTPR.alpha. expression improves plasma glucose levels, glucose tolerance and plasma and hepatic triglycerides levels in the ob/ob mouse model of diabetes and obesity. Consequently, antisense inhibitors of PTPR.alpha. are useful for preventing, ameliorating or treating conditions and disorders associated with these factors.
Example 8
Effect of antisense inhibitors of PTPR.alpha. on diet-induced obesity in mice
Antisense inhibitors of PTPR.alpha. expression are expected to reduce obesity or weight gain. This is tested in the high fat diet (HFD) model, also known as the DIO (diet-induced obesity) model.
Four week old male C57bl/6 mice were fed a high fat diet (60% kCal) ad libitum beginning at 4 weeks of age and continuing for 10 weeks. After the 10 weeks the mice were maintained on the diet as described and were treated with either an antisense inhibitor of PTPR.alpha., a control oligonucleotide having similar chemistry to the treatment oligonucleotide but not having a sequence that is complementary to the target mRNA, or saline. During this feeding schedule, animals were weighed at regular intervals and body weight and percent fat measurements were determined. At sacrifice, organ weights were determined as well.
Mice in the treatment group received 25 mg/kg of ISIS 147299 (SEQ ID NO: 135) twice a week by subcutaneous injection. Control group mice received a same treatment routine except using saline or, 25 mg/kg of control oligonucleotide that is not an antisense inhibitor of PTPR.alpha (CCTTCCCTGAAGGTTCCTCC - SEQ ID NO: 228). A further control group received no treatment and was fed normal chow. Food consumption was determined for mice in each of the groups. Mice treated with the antisense PTPR.alpha. inhibitor had reduced body weight gains and reduced food consumption. Figures 2 and 3. There were no differences in organ weight and percent fat between the groups.
Blood was drawn weekly and transaminase levels, plasmid glucose levels, NEFA levels, and plasma lipid levels were determined. There was a decrease in plasma lipid levels for the mice receiving an antisense inhibitor of PTPR.alpha.. After five weeks of treatment plasma triglyceride levels for mice receiving an antisense inhibitor of PTPR.alpha. were 59.9 IU/1 (sem = 3.57) compared to 75.9 IU/L (sem = 2.20) for saline, 77.2 IU/L (sem = 4.81) for control oligo and 91.80 IU/L (sem = 5.92) for normal chow.
Insulin levels are presented in Table 13. For mice treated with an antisense inhibitor of PTPR.alpha. fed plasma insulin levels were approximately 2.66 ng/ml at base line, 2.96 ng/ml after 2 weeks, 2.71 ng/ml after 3 weeks and 1.95 ng/ml after 5 weeks. At week four fasted insulin levels were approximately 0.58 ng/ml for the mice receiving treatment with an antisense inhibitor of PTPR.alpha.
Table 13 Insulin Levels in DIO mice
Figure imgf000040_0001
At day 41 of the treatment period the DIO mice were given an insulin tolerance test. Insulin tolerance tests are a good measure of insulin sensitivity. Mice were fasted for 3 hours and then received 0.35 U/kg of humilin (Lilly, Indianapolis, IN) via gavage of a O.lU/ml solution in saline. Blood glucose was measured at time point 0 and at 15, 20 or 30 minute intervals for up to 2 hours. Glucose levels are measured using a YSI glucose analyzer (YSI Scientific, Yellow Springs, OH). Results of the insulin tolerance test are shown in Table 14.
Table 14 Glucose During Insulin Tolerance Test in DIO Mice
Figure imgf000040_0002
Insulin levels were reduced for the animals receiving ISIS 147299 antisense inhibitor to PTPR.alpha. In addition, there was a reduction in plasma glucose levels seen during the insulin tolerance test for animals receiving ISIS 147229. A similar decrease was seen with control oligo, but not in the groups receiving saline.
The results from the ob/ob mouse studies and the DIO mouse studies indicate that antisense inhibition of PTPR.alpha. results in a reduction of plasma glucose levels and triglyceride levels in the plasma and liver. Antisense inhibition of PTPR.alpha. may provide a therapeutic strategy for conditions and diseases associated with these factors, such as type II diabetes, fatty liver disease and metabolic syndrome.

Claims

CLAIMSWhat is claimed is:
1. An antisense compound of 15 to 35 nucleobases targeted to a nucleic acid molecule encoding human PTPR.alpha.. wherein the nucleic acid molecule encoding human PTPR.alpha. is SEQ ID NO: 18 and wherein the antisense compound hybridizes with nucleotides 716 to 894, nucleotides 1063 to 1229, nucleotides 1818 to 2045, nucleotides 2328 to 2779, nucleotides 2982 to 3211, nucleotides 1210 to 1905 of SEQ ID NO: 18 or nucleotides 2860 to 2990 of SEQ ID NO: 11.
2. The antisense compound of claim 1 wherein the antisense compound is targeted to at least two nucleic acid molecules encoding human PTPR.alpha..
3. The antisense compound of claim 1, wherein the compound is at least about 80% complementary to a target region of the nucleic acid encoding PTPR.alpha..
4. The antisense compound of claim 1 , wherein the compound optionally includes a complementary strand 15 to 35 nucleobases in length.
5. The antisense compound of claim 1 , wherein the compound is an antisense oligonucleotide.
6. The compound of claim 5 having at least one modified internucleoside linkage, sugar moiety, or nucleobase.
7. The compound of claim 6 comprising a chimeric oligonucleotide.
8. The compound of claim 5 wherein the modified internucleoside linkage comprises a phosphorothioate linkage.
9. The compound of claim 5 wherein the at least one modified sugar moiety comprises a 2'- MOE , a LNA, a 2'-OMe, an ENA, a 2'-F or combinations thereof.
10. The compound of claim 5 wherein the modified nucleobase comprises 5-methylcytosine.
11. A pharmaceutical composition comprising a compound claim 1 and a pharmaceutically acceptable penetration enhancer, carrier, or diluent.
12. A method for the prevention, amelioration, or treatment of a disease or condition wherein indicators of the disease or condition comprise increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels, increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereof comprising administration of the compound of claim 1 to an individual in need of such intervention.
13. The method of claim 12 wherein the disease or condition is fatty liver disease and wherein administration of a compound of claim 1 results in a decrease in plasma lipid levels, a decrease in plasma transaminase levels, a decrease in hepatic triglyceride levels or combinations thereof.
14. The method of claim 12 wherein the disease or condition is type-2 diabetes and wherein administration of a compound of claim 1 results in a decrease in plasma glucose levels, an improvement in insulin sensitivity or combinations thereof.
15. The method of claim 12 wherein the disease or condition is metabolic syndrome and wherein administration of a compound of claim 1 results in a decrease in plasma glucose levels, a decrease in plasma lipid levels, a decrease in hepatic triglyceride levels, an improvement in insulin sensitivity, an improvement in hepatic function, a decrease in plasma transaminase levels or combinations thereof.
16. Use of an antisense compound of claim 1 in the manufacture of a medicament for the treatment, prevention or amelioration of a disease or condition wherein indicators of the disease or condition comprise increased plasma glucose levels, increased plasma lipid levels, increased hepatic triglyceride levels , increased plasma transaminase levels, reduced hepatic function, reduced insulin sensitivity or combinations thereofwherein the medicament reduces the expression of a nucleic acid molecule encoding PTPR.alpha..
17. The use of claim 16 wherein the disease or condition comprises type-2 diabetes, fatty liver, metabolic syndrome or combinations thereof.
18. The use of claim 16 wherein the antisense compound of claim 1 optionally further comprises a complementary strand 15 to 35 nucleobases in length.
19. A method for lowering blood glucose levels in an animal in need thereof by administering a compound of claim 1.
20. A method for lowering triglyceride levels in an animal in need thereof by administering a compound of claim 1.
21. The method of claim 20 wherein the triglyceride levels are lowered in the plasma.
22. The method of claim 20 wherein the triglyceride levels are lowered in the liver.
23. The method of claim 19, 20, 21 or 22 wherein the animal suffers from type-2 diabetes, fatty liver disease, obesity, metabolic syndrome of combinations thereof.
PCT/US2007/068390 2006-05-05 2007-05-07 Compositions and their uses directed to ptpr alpha WO2007131232A2 (en)

Priority Applications (2)

Application Number Priority Date Filing Date Title
EP07797356A EP2023936A4 (en) 2006-05-05 2007-05-07 Compositions and their uses directed to ptpr alpha
US12/299,610 US8158598B2 (en) 2006-05-05 2007-05-07 Compositions and their uses directed to PTPR alpha

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US79824606P 2006-05-05 2006-05-05
US60/798,246 2006-05-05

Publications (3)

Publication Number Publication Date
WO2007131232A2 true WO2007131232A2 (en) 2007-11-15
WO2007131232A3 WO2007131232A3 (en) 2008-08-07
WO2007131232A8 WO2007131232A8 (en) 2009-11-05

Family

ID=38668630

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/US2007/068390 WO2007131232A2 (en) 2006-05-05 2007-05-07 Compositions and their uses directed to ptpr alpha

Country Status (3)

Country Link
US (1) US8158598B2 (en)
EP (1) EP2023936A4 (en)
WO (1) WO2007131232A2 (en)

Cited By (13)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010044441A1 (en) 2008-10-17 2010-04-22 塩野義製薬株式会社 Acetic acid amide derivative having inhibitory activity on vascular endothelial lipase
WO2011074560A1 (en) 2009-12-15 2011-06-23 塩野義製薬株式会社 Oxadiazole derivative having endothelial lipase inhibitory activity
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
US9598458B2 (en) 2012-07-13 2017-03-21 Wave Life Sciences Japan, Inc. Asymmetric auxiliary group
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries

Families Citing this family (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2016040510A1 (en) * 2014-09-09 2016-03-17 La Jolla Institute For Allergy & Immunology Modulation of ptpra to treat arthritis

Family Cites Families (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6582908B2 (en) 1990-12-06 2003-06-24 Affymetrix, Inc. Oligonucleotides
FR2692265B1 (en) 1992-05-25 1996-11-08 Centre Nat Rech Scient BIOLOGICALLY ACTIVE COMPOUNDS OF THE PHOSPHOTRIESTER TYPE.
NZ255028A (en) * 1992-07-02 1997-03-24 Hybridon Inc Antisense oligonucleotides resistant to nucleolytic degradation
FR2705099B1 (en) 1993-05-12 1995-08-04 Centre Nat Rech Scient Phosphorothioate triester oligonucleotides and process for their preparation.
US5801154A (en) 1993-10-18 1998-09-01 Isis Pharmaceuticals, Inc. Antisense oligonucleotide modulation of multidrug resistance-associated protein
US20030228597A1 (en) 1998-04-13 2003-12-11 Cowsert Lex M. Identification of genetic targets for modulation by oligonucleotides and generation of oligonucleotides for gene modulation
US20040023906A1 (en) * 2002-08-01 2004-02-05 Isis Pharmaceuticals Inc. Antisense modulation of phosphotyrosyl phosphatase activator expression
US20040023904A1 (en) * 2002-07-31 2004-02-05 Isis Pharmaceuticals Inc. Antisense modulation of PTPRA expression
JP2006507841A (en) 2002-11-14 2006-03-09 ダーマコン, インコーポレイテッド Functional and ultrafunctional siRNA

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of EP2023936A4 *

Cited By (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2010044441A1 (en) 2008-10-17 2010-04-22 塩野義製薬株式会社 Acetic acid amide derivative having inhibitory activity on vascular endothelial lipase
US9394333B2 (en) 2008-12-02 2016-07-19 Wave Life Sciences Japan Method for the synthesis of phosphorus atom modified nucleic acids
US10329318B2 (en) 2008-12-02 2019-06-25 Wave Life Sciences Ltd. Method for the synthesis of phosphorus atom modified nucleic acids
US9695211B2 (en) 2008-12-02 2017-07-04 Wave Life Sciences Japan, Inc. Method for the synthesis of phosphorus atom modified nucleic acids
US10307434B2 (en) 2009-07-06 2019-06-04 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
US9744183B2 (en) 2009-07-06 2017-08-29 Wave Life Sciences Ltd. Nucleic acid prodrugs and methods of use thereof
WO2011074560A1 (en) 2009-12-15 2011-06-23 塩野義製薬株式会社 Oxadiazole derivative having endothelial lipase inhibitory activity
US8754113B2 (en) 2009-12-15 2014-06-17 Shionogi & Co., Ltd. Oxadiazole derivative having endothelial lipase inhibitory activity
US10428019B2 (en) 2010-09-24 2019-10-01 Wave Life Sciences Ltd. Chiral auxiliaries
US9605019B2 (en) 2011-07-19 2017-03-28 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US10280192B2 (en) 2011-07-19 2019-05-07 Wave Life Sciences Ltd. Methods for the synthesis of functionalized nucleic acids
US10167309B2 (en) 2012-07-13 2019-01-01 Wave Life Sciences Ltd. Asymmetric auxiliary group
US9982257B2 (en) 2012-07-13 2018-05-29 Wave Life Sciences Ltd. Chiral control
US9617547B2 (en) 2012-07-13 2017-04-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant
US9598458B2 (en) 2012-07-13 2017-03-21 Wave Life Sciences Japan, Inc. Asymmetric auxiliary group
US10590413B2 (en) 2012-07-13 2020-03-17 Wave Life Sciences Ltd. Chiral control
US10144933B2 (en) 2014-01-15 2018-12-04 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having immunity induction activity, and immunity induction activator
US10149905B2 (en) 2014-01-15 2018-12-11 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having antitumor effect and antitumor agent
US10322173B2 (en) 2014-01-15 2019-06-18 Shin Nippon Biomedical Laboratories, Ltd. Chiral nucleic acid adjuvant having anti-allergic activity, and anti-allergic agent
US10160969B2 (en) 2014-01-16 2018-12-25 Wave Life Sciences Ltd. Chiral design

Also Published As

Publication number Publication date
US8158598B2 (en) 2012-04-17
EP2023936A2 (en) 2009-02-18
EP2023936A4 (en) 2010-11-24
US20100022619A1 (en) 2010-01-28
WO2007131232A8 (en) 2009-11-05
WO2007131232A3 (en) 2008-08-07

Similar Documents

Publication Publication Date Title
US8158598B2 (en) Compositions and their uses directed to PTPR alpha
US7259150B2 (en) Modulation of apolipoprotein (a) expression
US8076306B2 (en) Compositions and their uses directed to hepcidin
AU2009201229B2 (en) Modulation of glucose-6-phosphatase translocase expression
US20090156526A1 (en) Compositions and methods for modulation of lmna expression
EP2338992A2 (en) Antisense compounds having enhanced anti-microRNA activity
JP2008513507A (en) Enhanced antisense oligonucleotides
US8202981B2 (en) Compositions and their uses directed to PTPRU
AU2017234678A1 (en) Methods of modulating KEAP1
EP1888083B1 (en) Compositions and their uses directed to lmw-ptpase
WO2007025229A2 (en) Compositions and their uses directed to hsp27
US20130109849A1 (en) Compositions and their uses directed to aceytl-coa carboxylases
EP2057177B1 (en) Compositions and their uses directed to diacylglycerol acyltransferase 1
US20140275207A1 (en) Antisense compounds and methods of use thereof

Legal Events

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

Ref document number: 07797356

Country of ref document: EP

Kind code of ref document: A2

NENP Non-entry into the national phase

Ref country code: DE

WWE Wipo information: entry into national phase

Ref document number: 2007797356

Country of ref document: EP

WWE Wipo information: entry into national phase

Ref document number: 12299610

Country of ref document: US