SELECTIVE REDUCTION OF ALLELIC VARIANTS

Disclosed herein are antisense compounds and methods for selectively reducing expression of an allelic variant of a huntingtin gene containing a single nucleotide polymorphism (SNP). Such methods, compounds, and composition are useful to treat, prevent, or ameliorate Huntington's Disease (HD).

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
SEQUENCE LISTING

The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BIOL0125USC2SEQ_ST25.txt created Dec. 18, 2017, which is 344 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.

FIELD OF THE INVENTION

Embodiments of the present invention provide methods, compounds, and compositions for selectively reducing expression of an allelic variant of a gene containing a single nucleotide polymorphism (SNP). Such methods, compounds, and compositions are useful to treat, prevent, or ameliorate Huntington's Disease.

BACKGROUND OF THE INVENTION

Genetic diseases are caused by abnormalities in genes or chromosomes. Such abnormalities may include insertions, deletions, and expansions. Huntington's Disease (HD) is one example of a genetic disease caused by an expansion. HD is a progressive neurodegenerative disorder that is inherited in a dominant fashion and results from a mutation that expands the polymorphic trinucleotide (CAG) tract in the huntingtin gene (HTT). The average CAG tract size in the general population is 17-26 repeats (wild type allele), however, in HD patients the CAG tract has expanded to 36 repeats or more (mutant allele) (Huntington's Disease Collaborative Research Group 1993. Cell 72(6):971-83). The HTT gene encodes the HTT protein and the expanded CAG tract results in a pathological increase in the polyglutamine repeats near the N-terminal of the protein. Individuals carry two copies of the HTT gene and one mutant allele is sufficient to result in HD.

HTT protein appears to have a role during development of the nervous system and a protective role in cells. In mouse models, constitutive knockout of the HTT gene is lethal during embryonic development (Nasir et al 1995. Cell 81(5):811-23), while adult inactivation of the HTT gene leads to progressive cell death in the brain and the testes (Dragatsis et al 2000. Nat. Genet 26:300-306). Reduction of huntingtin expression from the wild type allele may, therefore, have negative consequences.

Like HD, there are disorders for which a strategy of selective reduction of a mutant allele would be beneficial. Thus, there remains an unmet need to selectively reduce expression of mutant allelic variants like that of HTT, which are causative of disease, over the wild type variant, which appears to be necessary for normal cellular processes.

BRIEF DESCRIPTION OF THE FIGURE

FIGS. 1 A—EEEEEEE provide the mRNA (SEQ ID NO:2) and genomic (SEQ ID NO:1) HTT sequence showing SNP positions.

SUMMARY OF THE INVENTION

Provided herein are methods, compounds, and compositions for selectively reducing expression of an allelic variant of a gene containing a single nucleotide polymorphism (SNP). Such methods, compounds, and compositions are useful to treat, prevent, or ameliorate Huntington's Disease (HD). SNPs may be associated with a mutant allele, the expression of which causes HD.

In certain embodiments, selective reduction of mRNA and protein expression of a mutant huntingtin is achieved by targeting a SNP located on the mutant allele with an antisense compound. In certain embodiments, the antisense compound is an antisense oligonucleotide

In certain embodiments, antisense compounds designed to selectively reduce an allelic variant of a gene containing a SNP are created based on potency and selectivity of the antisense compound as well as population genetics.

DETAILED DESCRIPTION OF THE INVENTION

It is to be understood that both the foregoing general description and the following detailed description are exemplary and explanatory only and are not restrictive of the invention, as claimed. Herein, the use of the singular includes the plural unless specifically stated otherwise. As used herein, the use of “or” means “and/or” unless stated otherwise. Furthermore, the use of the term “including” as well as other forms, such as “includes” and “included”, is not limiting. Also, terms such as “element” or “component” encompass both elements and components comprising one unit and elements and components that comprise more than one subunit, unless specifically stated otherwise.

The section headings used herein are for organizational purposes only and are not to be construed as limiting the subject matter described. All documents, or portions of documents, cited in this application, including, but not limited to, patents, patent applications, articles, books, and treatises, are hereby expressly incorporated by reference for the portions of the document discussed herein, as well as in their entirety.

Definitions

Unless specific definitions are provided, the nomenclature utilized in connection with, and the procedures and techniques of, analytical chemistry, synthetic organic chemistry, and medicinal and pharmaceutical chemistry described herein are those well known and commonly used in the art. Standard techniques may be used for chemical synthesis, and chemical analysis. Where permitted, all patents, applications, published applications and other publications, GENBANK Accession Numbers and associated sequence information obtainable through databases such as National Center for Biotechnology Information (NCBI) and other data referred to throughout in the disclosure herein are incorporated by reference for the portions of the document discussed herein, as well as in their entirety.

Unless otherwise indicated, the following terms have the following meanings:

“2′-O-methoxyethyl” (also 2′-MOE and 2′-O(CH2)2—OCH3) refers to an O-methoxy-ethyl modification of the 2′ position of a furosyl ring. A 2′-O-methoxyethyl modified sugar is a modified sugar.

“2′-O-methoxyethyl nucleotide” means a nucleotide comprising a 2′-O-methoxyethyl modified sugar moiety.

“5-methylcytosine” means a cytosine modified with a methyl group attached to the 5′ position. A 5-methylcytosine is a modified nucleobase.

“Active pharmaceutical agent” means the substance or substances in a pharmaceutical composition that provide a therapeutic benefit when administered to an individual. For example, in certain embodiments an antisense oligonucleotide targeted to an allelic variant is an active pharmaceutical agent.

“Active target region” or “target region” means a region to which one or more active antisense compounds is targeted. “Active antisense compounds” means antisense compounds that reduce target nucleic acid levels or protein levels.

“Administered concomitantly” refers to the co-administration of two agents in any manner in which the pharmacological effects of both are manifest in the patient at the same time. Concomitant administration does not require that both agents be administered in a single pharmaceutical composition, in the same dosage form, or by the same route of administration. The effects of both agents need not manifest themselves at the same time. The effects need only be overlapping for a period of time and need not be coextensive.

“Administering” means providing a pharmaceutical agent to an individual, and includes, but is not limited to administering by a medical professional and self-administering.

“Allele” is one member of a pair of genes or one member of a series of different forms of a DNA sequences that can exist at a single locus or marker on a specific chromosome. For a diploid organism or cell or for autosomal chromosomes, each allelic pair will normally occupy corresponding positions (loci) on a pair of homologous chromosomes, one inherited from the mother and one inherited from the father. If these alleles are identical, the organism or cell is said to be ‘homozygous’ for that allele; if they differ, the organism or cell is said to be ‘heterozygous’ for that allele. “Major allele” refers to an allele containing the nucleotide present in a statistically significant proportion of individuals in the human population. “Minor allele” refers to an allele containing the nucleotide present in a relatively small proportion of individuals in the human population. “Wild type allele” refers to the genotype typically not associated with disease or dysfunction of the gene product. “Mutant allele” refers to the genotype associated with disease or dysfunction of the gene product.

“Allelic variant” refers to one of the pair of genes or DNA sequence existing at a single locus. For example, an allelic variant may refer to either the major allele or the minor allele.

“Amelioration” refers to a lessening of at least one indicator, sign, or symptom of an associated disease, disorder, or condition. The severity of indicators may be determined by subjective or objective measures, which are known to those skilled in the art.

“Animal” refers to a human or non-human animal, including, but not limited to, mice, rats, rabbits, dogs, cats, pigs, and non-human primates, including, but not limited to, monkeys and chimpanzees.

“Antibody” refers to a molecule characterized by reacting specifically with an antigen in some way, where the antibody and the antigen are each defined in terms of the other. Antibody may refer to a complete antibody molecule or any fragment or region thereof, such as the heavy chain, the light chain, Fab region, and Fc region.

“Antisense activity” means any detectable or measurable activity attributable to the hybridization of an antisense compound to its target nucleic acid. In certain embodiments, antisense activity is a decrease in the amount or expression of a target nucleic acid or protein encoded by such target nucleic acid.

“Antisense compound” means an oligomeric compound that is is capable of undergoing hybridization to a target nucleic acid through hydrogen bonding.

“Antisense inhibition” means reduction of target nucleic acid levels or target protein levels in the presence of an antisense compound complementary to a target nucleic acid compared to target nucleic acid levels or target protein levels in the absence of the antisense compound.

“Antisense oligonucleotide” means a single-stranded oligonucleotide having a nucleobase sequence that permits hybridization to a corresponding region or segment of a target nucleic acid.

“Bicyclic sugar” means a furosyl ring modified by the bridging of two ring atoms. A bicyclic sugar is a modified sugar.

“Bicyclic nucleoside” means a nucleoside having a sugar moiety comprising a bridge connecting two carbon atoms of the sugar ring, thereby forming a bicyclic ring system. In certain embodiments, the bridge connects the 4′-carbon and the 2′-carbon of the sugar ring.

“Cap structure” or “terminal cap moiety” means chemical modifications, which have been incorporated at either terminus of an antisense compound.

“cEt” or “constrained ethyl” means a bicyclic nucleoside having a sugar moiety comprising a bridge connecting the 4′-carbon and the 2′-carbon, wherein the bridge has the formula: 4′-CH(CH3)—O-2′.

“Chemically distinct region” refers to a region of an antisense compound that is in some way chemically different than another region of the same antisense compound. For example, a region having 2′-O-methoxyethyl nucleotides is chemically distinct from a region having nucleotides without 2′-O-methoxyethyl modifications.

“Chimeric antisense compound” means an antisense compound that has at least two chemically distinct regions.

“Co-administration” means administration of two or more pharmaceutical agents to an individual. The two or more pharmaceutical agents may be in a single pharmaceutical composition, or may be in separate pharmaceutical compositions. Each of the two or more pharmaceutical agents may be administered through the same or different routes of administration. Co-administration encompasses parallel or sequential administration.

“Complementarity” means the capacity for pairing between nucleobases of a first nucleic acid and a second nucleic acid.

“Contiguous nucleobases” means nucleobases immediately adjacent to each other.

“Differentiating polymorphism” means a variation in a nucleotide sequence that permits differentiation between a wild type and a mutant allele of a nucleic acid sequence. Differentiating polymorphisms may include insertions or deletions of one or a few nucleotides in a sequence, or changes in one or a few nucleotides in a sequence. A differentiating polymorphism or polymorphic allele can be in linkage disequilibrium with one or more other polymorphisms or polymorphic alleles.

“Diluent” means an ingredient in a composition that lacks pharmacological activity, but is pharmaceutically necessary or desirable. For example, the diluent in an injected composition may be a liquid, e.g. saline solution.

“Dose” means a specified quantity of a pharmaceutical agent provided in a single administration, or in a specified time period. In certain embodiments, a dose may be administered in one, two, or more boluses, tablets, or injections. For example, in certain embodiments where subcutaneous administration is desired, the desired dose requires a volume not easily accommodated by a single injection, therefore, two or more injections may be used to achieve the desired dose. In certain embodiments, the pharmaceutical agent is administered by infusion over an extended period of time or continuously. Doses may be stated as the amount of pharmaceutical agent per hour, day, week, or month.

“Effective amount” means the amount of active pharmaceutical agent sufficient to effectuate a desired physiological outcome in an individual in need of the agent. The effective amount may vary among individuals depending on the health and physical condition of the individual to be treated, the taxonomic group of the individuals to be treated, the formulation of the composition, assessment of the individual's medical condition, and other relevant factors.

“Fully complementary” or “100% complementary” means each nucleobase of a first nucleic acid has a complementary nucleobase in a second nucleic acid. In certain embodiments, a first nucleic acid is an antisense compound and a target nucleic acid is a second nucleic acid.

“Gapmer” means a chimeric antisense compound in which an internal region having a plurality of nucleosides that support RNase H cleavage is positioned between external regions having one or more nucleosides, wherein the nucleosides comprising the internal region are chemically distinct from the nucleoside or nucleosides comprising the external regions. The internal region may be referred to as the “gap” and the external regions may be referred to as the “wings.”

“Gap-widened” means a chimeric antisense compound having a gap segment of 12 or more contiguous 2′-deoxyribonucleosides positioned between and immediately adjacent to 5′ and 3′ wing segments having from one to six nucleosides.

“Gene product” refers to a biochemical material, such as RNA or protein, resulting from expression of a gene.

“Haplotype” means a set of alleles of closely linked loci on a chromosome that are generally inherited together. For example, a polymorphic allele at a first site in a nucleic acid sequence on the chromosome may be found to be associated with another polymorphic allele at a second site on the same chromosome, at a frequency other than would be expected for a random associate (e.g. “linkage equilibrium”). These two polymorphic alleles may be described as being in “linkage disequilibrium.” A haplotype may comprise two, three, four, or more alleles. The set of alleles in a haplotype along a given segment of a chromosome are generally transmitted to progeny together unless there has been a recombination event.

“High-affinity sugar modification” is a modified sugar moiety which when it is included in a nucleoside and said nucleoside is incorporated into an antisense oligonucleotide, the stability (as measured by Tm) of said antisense oligonucleotide: RNA duplex is increased as compared to the stability of a DNA:RNA duplex.

“High-affinity sugar-modified nucleoside” is a nucleoside comprising a modified sugar moiety that when said nucleoside is incorporated into an antisense compound, the binding affinity (as measured by Tm) of said antisense compound toward a complementary RNA molecule is increased. In certain embodiments of the invention at least one of said sugar-modified high-affinity nucleosides confers a ΔTm of at least 1 to 4 degrees per nucleoside against a complementary RNA as determined in accordance with the methodology described in Freier et al., Nucleic Acids Res., 1997, 25, 4429-4443, which is incorporated by reference in its entirety. In another aspect, at least one of the high-affinity sugar modifications confers about 2 or more, 3 or more, or 4 or more degrees per modification. In the context of the present invention, examples of sugar-modified high affinity nucleosides include, but are not limited to, (i) certain 2′-modified nucleosides, including 2′-substituted and 4′ to 2′ bicyclic nucleosides, and (ii) certain other non-ribofuranosyl nucleosides which provide a per modification increase in binding affinity such as modified tetrahydropyran and tricycloDNA nucleosides. For other modifications that are sugar-modified high-affinity nucleosides see Freier et al., Nucleic Acids Res., 1997, 25, 4429-4443.

“Hybridization” means the annealing of complementary nucleic acid molecules. In certain embodiments, complementary nucleic acid molecules include an antisense compound and a target nucleic acid.

“Immediately adjacent” means there are no intervening elements between the immediately adjacent elements.

“Individual” means a human or non-human animal selected for treatment or therapy.

“Internucleoside linkage” refers to the chemical bond between nucleosides.

“Linked nucleosides” means adjacent nucleosides which are bonded together.

“Mismatch” or “non-complementary nucleobase” refers to the case when a nucleobase of a first nucleic acid is not capable of pairing with the corresponding nucleobase of a second or target nucleic acid.

“Modified internucleoside linkage” refers to a substitution or any change from a naturally occurring internucleoside bond (i.e. a phosphodiester internucleoside bond).

“Modified nucleobase” refers to any nucleobase other than adenine, cytosine, guanine, thymidine, or uracil. An “unmodified nucleobase” means the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C), and uracil (U).

“Modified nucleotide” means a nucleotide having, independently, a modified sugar moiety, modified internucleoside linkage, or modified nucleobase. A “modified nucleoside” means a nucleoside having, independently, a modified sugar moiety or modified nucleobase.

“Modified oligonucleotide” means an oligonucleotide comprising a modified internucleoside linkage, a modified sugar, or a modified nucleobase.

“Modified sugar” refers to a substitution or change from a natural sugar.

“Motif” means the pattern of chemically distinct regions in an antisense compound.

“Naturally occurring internucleoside linkage” means a 3′ to 5′ phosphodiester linkage.

“Natural sugar moiety” means a sugar found in DNA (2′-H) or RNA (2′-OH).

“Nuclease resistant modification” means a sugar modification or modified internucleoside linkage which, when incorporated into an oligonucleotide, makes said oligonucleotide more stable to degradation under cellular nucleases (e.g. exo- or endo-nucleases). Examples of nuclease resistant modifications include, but are not limited to, phosphorothioate internucleoside linkages, bicyclic sugar modifications, 2′-modified nucleotides, or neutral internucleoside linkages.

“Nucleic acid” refers to molecules composed of monomeric nucleotides. A nucleic acid includes ribonucleic acids (RNA), deoxyribonucleic acids (DNA), single-stranded nucleic acids, double-stranded nucleic acids, small interfering ribonucleic acids (siRNA), and microRNAs (miRNA).

“Nucleobase” means a heterocyclic moiety capable of pairing with a base of another nucleic acid.

“Nucleobase sequence” means the order of contiguous nucleobases independent of any sugar, linkage, or nucleobase modification.

“Nucleoside” means a nucleobase linked to a sugar.

“Nucleoside mimetic” includes those structures used to replace the sugar or the sugar and the base and not necessarily the linkage at one or more positions of an oligomeric compound such as for example nucleoside mimetics having morpholino, cyclohexenyl, cyclohexyl, tetrahydropyranyl, bicyclo or tricyclo sugar mimetics e.g. non furanose sugar units. Nucleotide mimetic includes those structures used to replace the nucleoside and the linkage at one or more positions of an oligomeric compound such as for example peptide nucleic acids or morpholinos (morpholinos linked by —N(H)—C(═O)—O— or other non-phosphodiester linkage). Sugar surrogate overlaps with the slightly broader term nucleoside mimetic but is intended to indicate replacement of the sugar unit (furanose ring) only. The tetrahydropyranyl rings provided herein are illustrative of an example of a sugar surrogate wherein the furanose sugar group has been replaced with a tetrahydropyranyl ring system.

“Nucleotide” means a nucleoside having a phosphate group covalently linked to the sugar portion of the nucleoside.

“Oligomeric compound” or “oligomer” means a polymer of linked monomeric subunits which is capable of hybridizing to at least a region of a nucleic acid molecule.

“Oligonucleotide” means a polymer of linked nucleosides each of which can be modified or unmodified, independent one from another.

“Parenteral administration” means administration through injection or infusion. Parenteral administration includes subcutaneous administration, intravenous administration, intramuscular administration, intraarterial administration, intraperitoneal administration, or intracranial administration, e.g. intrathecal or intracerebroventricular administration.

“Peptide” means a molecule formed by linking at least two amino acids by amide bonds. Peptide refers to polypeptides and proteins.

“Pharmaceutical composition” means a mixture of substances suitable for administering to an individual. For example, a pharmaceutical composition may comprise one or more active pharmaceutical agents and a sterile aqueous solution.

“Pharmaceutically acceptable salts” means physiologically and pharmaceutically acceptable salts of antisense compounds, i.e., salts that retain the desired biological activity of the parent oligonucleotide and do not impart undesired toxicological effects thereto.

“Phosphorothioate linkage” means a linkage between nucleosides where the phosphodiester bond is modified by replacing one of the non-bridging oxygen atoms with a sulfur atom. A phosphorothioate linkage (P═S) is a modified internucleoside linkage.

“Portion” means a defined number of contiguous (i.e. linked) nucleobases of a nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of a target nucleic acid. In certain embodiments, a portion is a defined number of contiguous nucleobases of an antisense compound.

“Prevent” refers to delaying or forestalling the onset or development of a disease, disorder, or condition for a period of time from minutes to indefinitely. Prevent also means reducing risk of developing a disease, disorder, or condition.

“Prodrug” means a therapeutic agent that is prepared in an inactive form that is converted to an active form within the body or cells thereof by the action of endogenous enzymes or other chemicals or conditions.

“Selectively reducing expression of an allelic variant” means reducing expression of one allele more than the other, differing allele among a set of alleles. For example, a mutant allele containing a single nucleotide polymorphism (SNP) may be reduced more than a wild type allele not containing the SNP.

“Side effects” means physiological responses attributable to a treatment other than the desired effects. In certain embodiments, side effects include injection site reactions, liver function test abnormalities, renal function abnormalities, liver toxicity, renal toxicity, central nervous system abnormalities, myopathies, and malaise. For example, increased aminotransferase levels in serum may indicate liver toxicity or liver function abnormality. For example, increased bilirubin may indicate liver toxicity or liver function abnormality.

“Single nucleotide polymorphism” or “SNP” means a single nucleotide variation between the genomes of individuals of the same species. In some cases, a SNP may be a single nucleotide deletion or insertion. In general, SNPs occur relatively frequently in genomes and thus contribute to genetic diversity. SNPs are thought to be mutationally more stable than other polymorphisms, lending their use in association studies in which linkage disequilibrium between markers and an unknown variant is used to map disease-causing mutations. The location of a SNP is generally flanked by highly conserved sequences. An individual may be homozygous or heterozygous for an allele at each SNP site. A heterozygous SNP allele can be a differentiating polymorphism. A SNP may be targeted with an antisense oligonucleotide, meaning that the SNP anneals to (or aligns with) position 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 of the antisense oligonucleotide. The remainder of the antisense oligonucleotide bases must have sufficient complementarity to the SNP site to facilitate hybridization.

“Single nucleotide polymorphism position” or “SNP position” refers to the nucleotide position of the SNP on a reference sequence.

“Single nucleotide polymorphism site” or “SNP site” refers to the nucleotides surrounding a SNP contained in a target nucleic acid to which an antisense compound is targeted.

“Single-stranded oligonucleotide” means an oligonucleotide which is not hybridized to a complementary strand.

“Specifically hybridizable” refers to an antisense compound having a sufficient degree of complementarity between an antisense oligonucleotide and a target nucleic acid to induce a desired effect, while exhibiting minimal or no effects on non-target nucleic acids under conditions in which specific binding is desired, i.e. under physiological conditions in the case of in vivo assays and therapeutic treatments.

“Targeting” or “targeted” means the process of design and selection of an antisense compound that will specifically hybridize to a target nucleic acid and induce a desired effect.

“Target nucleic acid,” “target RNA,” and “target RNA transcript” all refer to a nucleic acid capable of being targeted by antisense compounds.

“Target segment” means the sequence of nucleotides of a target nucleic acid to which an antisense compound is targeted. For example, for the purposes of this patent application, the target segment may be within the SNP site. “5′ target site” refers to the 5′-most nucleotide of a target segment. “3′ target site” refers to the 3′-most nucleotide of a target segment.

“Therapeutically effective amount” means an amount of a pharmaceutical agent that provides a therapeutic benefit to an individual.

“Treat” refers to administering a pharmaceutical composition to effect an alteration or improvement of a disease, disorder, or condition.

“Unmodified nucleotide” means a nucleotide composed of naturally occurring nucleobases, sugar moieties, and internucleoside linkages. In certain embodiments, an unmodified nucleotide is an RNA nucleotide (i.e. β-D-ribonucleosides) or a DNA nucleotide (i.e. β-D-deoxyribonucleoside).

Certain Embodiments

Embodiments of the present invention provide methods, compounds, and compositions for selectively inhibiting mRNA and protein expression of an allelic variant of a gene or DNA sequence. In certain embodiments, the allelic variant contains a single nucleotide polymorphism (SNP). In certain embodiments, the SNP is a differentiating polymorphism. In certain embodiments, a SNP is associated with a mutant allele. In certain embodiments, a SNP is in linkage disequilibrium with another polymorphism that is associated with or is causative of disease. In certain embodiments, a mutant allele is associated with disease. In certain embodiments, mRNA and protein expression of a mutant allele is associated with disease.

In certain embodiments, the expressed gene product of a mutant allele results in aggregation of the mutant proteins causing disease. In certain embodiments, the expressed gene product of a mutant allele results in gain of function causing disease

In certain embodiments, selective reduction of mRNA and protein expression of a mutant allele is achieved by targeting a SNP located on the mutant allele with an antisense compound. In certain embodiments, the antisense compound is an antisense oligonucleotide. In certain embodiments, the antisense compound is not a ribozyme, a double stranded siRNA, or an shRNA.

In certain embodiments, the antisense oligonucleotide may have one or more modified sugar(s), nucleobase(s), or internucleoside linkage(s). In certain embodiments, the antisense oligonucleotide is complementary to the SNP site. In certain embodiments, the antisense oligonucleotide is complementary to the SNP site. In certain embodiments, the antisense oligonucleotide is at least 65%, 70%, 75%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, or 99% complementary to the SNP site. In certain embodiments, the antisense oligonucleotide is 100% complementary to the SNP site. In certain embodiments, the SNP site is 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleobases in length. In certain embodiments, the SNP anneals to position 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, or 20 of the antisense oligonucleotide.

In certain embodiments, antisense compounds designed to selectively reduce an allelic variant of a gene containing a SNP are created based on potency and selectivity of the antisense compound as well as population genetics.

In certain embodiments, selective reduction of mRNA and protein expression of an allelic variant of a gene containing a SNP occurs in a cell or tissue. In certain embodiments, the cell or tissue is in an animal. In certain embodiments, the animal is a human.

In certain embodiments, described is a method of treating Huntington's Disease, comprising selectively reducing expression of a mutant huntingtin allele in an animal in need thereof, comprising administering to the animal a compound, comprising a modified oligonucleotide consisting of 15 to 20 linked nucleosides and fully complementary to the mutant huntingtin allele, wherein position 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 of the modified oligonucleotide, as counted from the 5′ terminus of the modified oligonucleotide, aligns with any of the SNP sites in the group consisting of rs6446723, rs3856973, rs2285086, rs363092, rs916171, rs6844859, rs7691627, rs4690073, rs2024115, rs11731237, rs362296, rs10015979, rs7659144, rs363096, rs362273, rs16843804, rs362271, rs362275, rs3121419, rs362272, rs3775061, rs34315806, rs363099, rs2298967, rs363088, rs363064, rs363102, rs2798235, rs363080, rs363072, rs363125, rs362303, rs362310, rs10488840, rs362325, rs35892913, rs1936032, rs2276881, rs363070, rs12502045, rs7685686, rs3733217, rs362331, rs2857936, rs12506200, rs762855, rs4690072, rs363081, rs363075, rs3025849, rs6855981, rs363144, rs3025838, rs2298969, rs362322, rs362307, rs362306, and rs1006798.

In certain embodiments, described is a compound comprising a modified oligonucleotide 15 to 20 linked nucleosides in length, targeted to a huntingtin single nucleotide polymorphism, comprising a 12 nucleobase portion of the nucleobase sequence of any one of SEQ ID NO: 6 to 285, wherein position 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 of the modified oligonucleotide, as counted from the 5′ terminus of the modified oligonucleotide, aligns with the huntingtin single nucleotide polymorphism.

In certain embodiments, the modified oligonucleotide comprises a wing-gap-wing motif of any one of the group consisting of 5-10-5, 2-9-6, 3-9-3, 3-9-4, 3-9-5, 4-7-4, 4-9-3, 4-9-4, 4-9-5, 4-10-5, 4-11-4, 4-11-5, 5-7-5, 5-8-6, 5-9-3, 5-9-5, 5-10-4, 5-10-5, 6-7-6, 6-8-5, and 6-9-2.

In certain embodiments, at least one internucleoside linkage is a modified internucleoside linkage. In certain embodiments, each internucleoside linkage is a phosphorothioate internucleoside linkage.

In certain embodiments, at least one nucleoside comprises a modified nucleobase. In certain embodiments, the modified nucleobase is a 5′-methylcytosine.

In certain embodiments, at least one nucleoside of at least one of the wing regions comprises a modified sugar or sugar surrogate. In certain embodiments, each of the nucleosides of each wing region comprises a modified sugar or sugar surrogate. In certain embodiments, the sugar or sugar surrogate is a 2′-O-methoxyethyl modified sugar. In certain embodiments, at least one of the wing regions comprises a 4′ to 2′ bicyclic nucleoside and at least one of the remaining wing nucleosides is a non-bicyclic 2′-modified nucleoside. In certain embodiments, the non-bicyclic 2′-modified nucleoside is a 2′-O-methoxyethyl nucleoside. In certain embodiments, the 4′ to 2′ bicyclic nucleoside is 4′-CH(CH3)-O-2′ bicyclic nucleoside.

In certain embodiments, described is a method of selectively reducing mutant huntingtin, comprising administering a compound comprising a modified oligonucleotide 15 to 20 linked nucleosides in length, targeted to a huntingtin single nucleotide polymorphism, comprising a 12 nucleobase portion of the nucleobase sequence of any one of SEQ ID NO: 6 to 285, wherein position 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 of the modified oligonucleotide, as counted from the 5′ terminus of the modified oligonucleotide, aligns with the huntingtin single nucleotide polymorphism.

In certain embodiments, described is a use of a compound comprising a modified oligonucleotide 15 to 20 linked nucleosides in length, targeted to a huntingtin single nucleotide polymorphism, comprising a 12 nucleobase portion of the nucleobase sequence of any one of SEQ ID NO: 6 to 285, wherein position 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 of the modified oligonucleotide, as counted from the 5′ terminus of the modified oligonucleotide, aligns with the huntingtin single nucleotide polymorphism for the treatment of Huntington's Disease.

Single Nucleotide Polymorphisms (SNPs)

Single-nucleotide polymorphisms (SNPs) are single base-pair alterations in the DNA sequence that represent a major source of genetic heterogeneity (Gene. 1999, 234:177). SNP genotyping is an important tool with which to investigate these genetic variants (Genome Res. 2000, 10:895; Trends Biotechnol. 2000, 18:77). In certain embodiments, antisense compounds designed to selectively reduce an allelic variant of a gene containing an SNP were selected based on potency, selectivity and population genetics coverage.

Potency

In certain embodiments, antisense compounds designed to selectively reduce an allelic variant of a gene containing a SNP are created based on potency of the antisense compound. Potency generally refers to how amenable the targeted sequence area is to antisense inhibition. In certain embodiments, specific SNP sites may be particularly amenable to antisense inhibition. Certain such highly amenable SNP sites may be targeted by antisense compounds for selectively reducing an allelic variant of a gene. Potency is demonstrated by the percent inhibition of mutant mRNA achieved by the antisense oligonucleotides targeting a SNP compared to the percent inhibition of mutant mRNA achieved by the benchmark oligonucleotide.

Selectivity

In certain embodiments, antisense compounds designed to selectively reduce an allelic variant of a gene containing a SNP are created based on selectivity of the antisense compound. Selectivity generally refers to antisense compounds comprising a particular sequence, motif, and chemical modification(s) that preferentially target the one or more differentiating polymorphisms (SNPs) in the RNA encoding a mutant HTT protein compared to the RNA encoding a wild type HTT protein. In certain embodiments, specific sequences, motifs, and chemical modification(s) are particularly selective in reducing an allelic variant of a gene containing a SNP. Certain such sequences, motifs, and chemical modification(s) are utilized to selectively reduce an allelic variant of a gene. Selectivity is demonstrated by the ability of the antisense oligonucleotide targeting a SNP to inhibit expression of the major allele or mutant allele preferentially compared to the minor allele or wild type allele.

Population Genetics

In certain embodiments, antisense compounds designed to selectively reduce an allelic variant of a gene containing an SNP are created based on the population genetics of a population afflicted with disease. Population genetics means the frequency at which the SNP appears in the disease chromosome of patients afflicted with a particular disease. In certain embodiments, the disease is Huntington disease. Where potency and selectivity amongst antisense compounds is equal, SNP targets that have higher population genetics coverage are favored over SNPs that have a weaker association with disease chromosomes.

Antisense Compounds

Oligomeric compounds may include, but are not limited to, oligonucleotides, oligonucleosides, oligonucleotide analogs, oligonucleotide mimetics, antisense compounds, antisense oligonucleotides, and siRNAs. An oligomeric compound may be “antisense” to a target nucleic acid, meaning that is is capable of undergoing hybridization to a target nucleic acid through hydrogen bonding.

In certain embodiments, an antisense compound is an antisense oligonucleotide. In certain embodiments, the antisense compound is not a ribozyme, a double stranded siRNA, or an shRNA.

In certain embodiments, an antisense compound has a nucleobase sequence that, when written in the 5′ to 3′ direction, comprises the reverse complement of the target segment of a target nucleic acid to which it is targeted. In certain such embodiments, an antisense oligonucleotide has a nucleobase sequence that, when written in the 5′ to 3′ direction, comprises the reverse complement of the target segment of a target nucleic acid to which it is targeted.

In certain embodiments, antisense compounds are 12 to 30 subunits in length. In other words, such antisense compounds are from 12 to 30 linked subunits. In other embodiments, the antisense compound is 8 to 80, 12 to 50, 15 to 30, 18 to 24, 19 to 22, or 20 linked subunits. In certain such embodiments, the antisense compounds are 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 40, 41, 42, 43, 44, 45, 46, 47, 48, 49, 50, 51, 52, 53, 54, 55, 56, 57, 58, 59, 60, 61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, or 80 linked subunits in length, or a range defined by any two of the above values. In some embodiments the antisense compound is an antisense oligonucleotide, and the linked subunits are nucleosides.

In certain embodiments antisense oligonucleotides targeted to a nucleic acid may be shortened or truncated. For example, a single subunit may be deleted from the 5′ end (5′ truncation), or alternatively from the 3′ end (3′ truncation). A shortened or truncated antisense compound targeted to a nucleic acid may have two subunits deleted from the 5′ end, or alternatively may have two subunits deleted from the 3′ end, of the antisense compound. Alternatively, the deleted nucleosides may be dispersed throughout the antisense compound, for example, in an antisense compound having one nucleoside deleted from the 5′ end and one nucleoside deleted from the 3′ end.

When a single additional subunit is present in a lengthened antisense compound, the additional subunit may be located at the 5′ or 3′ end of the antisense compound. When two or more additional subunits are present, the added subunits may be adjacent to each other, for example, in an antisense compound having two subunits added to the 5′ end (5′ addition), or alternatively to the 3′ end (3′ addition), of the antisense compound. Alternatively, the added subunits may be dispersed throughout the antisense compound, for example, in an antisense compound having one subunit added to the 5′ end and one subunit added to the 3′ end.

It is possible to increase or decrease the length of an antisense compound, such as an antisense oligonucleotide, and/or introduce mismatch bases without eliminating activity.

For example, in Woolf et al. (Proc. Natl. Acad. Sci. USA 89:7305-7309, 1992), a series of antisense oligonucleotides 13-25 nucleobases in length were tested for their ability to induce cleavage of a target RNA in an oocyte injection model. Antisense oligonucleotides 25 nucleobases in length with 8 or 11 mismatch bases near the ends of the antisense oligonucleotides were able to direct specific cleavage of the target mRNA, albeit to a lesser extent than the antisense oligonucleotides that contained no mismatches. Similarly, target specific cleavage was achieved using 13 nucleobase antisense oligonucleotides, including those with 1 or 3 mismatches.

Gautschi et al (J. Natl. Cancer Inst. 93:463-471, March 2001) demonstrated the ability of an oligonucleotide having 100% complementarity to the bcl-2 mRNA and having 3 mismatches to the bcl-xL mRNA to reduce the expression of both bcl-2 and bcl-xL in vitro and in vivo. Furthermore, this oligonucleotide demonstrated potent anti-tumor activity in vivo.

Maher and Dolnick (Nuc. Acid. Res. 16:3341-3358, 1988) tested a series of tandem 14 nucleobase antisense oligonucleotides, and a 28 and 42 nucleobase antisense oligonucleotides comprised of the sequence of two or three of the tandem antisense oligonucleotides, respectively, for their ability to arrest translation of human DHFR in a rabbit reticulocyte assay. Each of the three 14 nucleobase antisense oligonucleotides alone was able to inhibit translation, albeit at a more modest level than the 28 or 42 nucleobase antisense oligonucleotides.

However, selective reduction of expression of an allelic variant is optimized when the SNP contained in the target nucleic anneals to a complementary base in the antisense compound and not a mismatched base. Moreover, selectivity in general is increased when there are fewer mismatches between the SNP site and the antisense compound. However, a certain number of mismatches may be tolerated.

Antisense Compound Motifs

In certain embodiments, antisense compounds targeted to a nucleic acid have chemically modified subunits arranged in patterns, or motifs, to confer to the antisense compounds properties such as enhanced the inhibitory activity, increased binding affinity for a target nucleic acid, or resistance to degradation by in vivo nucleases.

Chimeric antisense compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, increased binding affinity for the target nucleic acid, and/or increased inhibitory activity. A second region of a chimeric antisense compound may optionally serve as a substrate for the cellular endonuclease RNase H, which cleaves the RNA strand of an RNA:DNA duplex.

Antisense compounds having a gapmer motif are considered chimeric antisense compounds. In a gapmer an internal region having a plurality of nucleotides that supports RNaseH cleavage is positioned between external regions having a plurality of nucleotides that are chemically distinct from the nucleosides of the internal region. In the case of an antisense oligonucleotide having a gapmer motif, the gap segment generally serves as the substrate for endonuclease cleavage, while the wing segments comprise modified nucleosides. In the case of an antisense oligonucleotide for selectively reducing expression of an allelic variant of a gene containing a SNP, the SNP anneals to a nucleobase within the gap segment.

In certain embodiments, the SNP anneals or is complementary to a nucleobase at position 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 of the antisense oligonucleotide, wherein position refers to the orientation of a nucleobase within the antisense oligonucleotide counting from the 5′ terminus of the antisense oligonucleotide. For example, the 5′ most nucleobase within the antisense oligonucleotide is in the first position of the antisense oligonucleotide. In certain embodiments, the SNP anneals or is complementary to a nucleobase at position 6, 7, 8, 9, or 10 of the antisense oligonucleotide (counting from the 5′ terminus). In certain embodiments, the SNP anneals or is complementary to a nucleobase at position 9 or 10 of the antisense oligonucleotide (counting from the 5′ terminus).

In certain embodiments, the SNP anneals to a nucleobase at position 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 of the gap segment, wherein position refers to the orientation of a nucleobase within the gap segment counting from the 5′ terminus of the gap segment. For example, the 5′ most nucleobase within the gap segment is in the first position of the gap segment. In certain embodiments, the SNP anneals to a nucleobase at position 4, 5, 6, or 7 counting from the 5′ terminus of the gap segment. In certain embodiments, the SNP anneals to a nucleobase at position 4 or 5 beginning from the 5′ terminus of the gap segment.

In certain embodiments, the regions of a gapmer are differentiated by the types of sugar moieties comprising each distinct region. The types of sugar moieties that are used to differentiate the regions of a gapmer may in some embodiments include β-D-ribonucleosides, β-D-deoxyribonucleosides, 2′-modified nucleosides (such 2′-modified nucleosides may include 2′-MOE, and 2′-O—CH3, among others), and bicyclic sugar modified nucleosides (such bicyclic sugar modified nucleosides may include those having a 4′-(CH2)n-O-2′ bridge, where n=1 or n=2). The bicyclic moiety may be a cEt having the formula 4′-CH(CH3)—O-2.′

The wing-gap-wing motif is frequently described as “X-Y-Z”, where “X” represents the length of the 5′ wing region, “Y” represents the length of the gap region, and “Z” represents the length of the 3′ wing region. As used herein, a gapmer described as “X-Y-Z” has a configuration such that the gap segment is positioned immediately adjacent to each of the 5′ wing segment and the 3′ wing segment. Thus, no intervening nucleotides exist between the 5′ wing segment and gap segment, or the gap segment and the 3′ wing segment. Any of the antisense compounds described herein can have a gapmer motif. In some embodiments, X and Z are the same, in other embodiments they are different. In certain embodiments, Y is between 8 and 15 nucleotides. In certain embodiments, Y is comprised of deoxynucleotides. X, Y or Z can be any of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 25, 30 or more nucleotides. Thus, gapmers of the present invention include, but are not limited to, for example 1-10-1, 1-18-1, 2-8-2, 2-9-6, 2-10-2, 2-13-5, 2-16-2, 3-9-3, 3-9-5, 3-10-3, 3-14-3, 4-8-4, 4-9-5, 4-10-5, 4-11-4, 4-12-3, 4-12-4, 5-8-5, 5-9-5, 5-10-4, 5-10-5, or 6-8-6.

In certain embodiments, the antisense compound has a “wingmer” motif, having a wing-gap or gap-wing configuration, i.e. an X-Y or Y-Z configuration as described above for the gapmer configuration. Thus, wingmer configurations of the present invention include, but are not limited to, for example 5-10, 8-4, 4-12, 12-4, 3-14, 16-2, 18-1, 10-3, 2-10, 1-10, 8-2, 2-13, 5-13, 5-8, or 6-8.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 2-9-6 gapmer motif or a 6-9-2 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 3-9-3 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 3-9-5 gapmer motif or 5-9-3 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 4-9-5 gapmer motif or 5-9-4 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 4-10-5 gapmer motif or 5-10-4 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 4-11-4 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 5-9-5 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 5-8-6 gapmer motif or a 6-8-5 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 6-7-6 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 6-8-5 gapmer motif or a 5-8-6 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 3-9-4 gapmer motif or a 4-9-3 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 5-7-5 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 4-7-4 gapmer motif.

In certain embodiments, antisense compounds targeted to a nucleic acid possess a 5-10-5 gapmer motif.

In certain embodiments, an antisense compound targeted to a nucleic acid has a gap-widened motif.

Certain Mixed Wings

In certain embodiments, the invention provides gapmer compounds wherein at least one nucleoside of one wing is differently modified compared to at least one other nucleoside of the same wing. Such antisense compounds are referred to as mixed wing antisense compounds (see WO 2008/049085). In certain embodiments, the modifications (or no modification) of one or more nucleosides of the 3′ wing are different from those of one or more other nucleosides of the 3′ wing. Such antisense compounds may be referred to as 3′ mixed wing gapmers. In certain embodiments, the modifications (or no modification) of one or more nucleosides of the 5′ wing are different from those of one or more other nucleosides of the 5′ wing. Such antisense compounds may be referred to as 5′ mixed wing gapmers. In certain embodiments, the modifications (or no modification) of one or more nucleosides of the 3′ wing are different from those of one or more other nucleosides of the 3′ wing and the modifications (or no modification) of one or more nucleosides of the 5′ wing are different from those of one or more other nucleosides of the 5′ wing. Such antisense compounds may be referred to as 3′, 5′ mixed wing gapmers. In such embodiment, the modifications and combination of modifications at the 3′ wing and at the 5′ wing may be the same or they may be different.

In certain embodiments, mixed wing compounds have desirable properties. Certain nucleoside modifications confer on the antisense compound a desirable property, for example increased affinity for a target or nuclease resistance, but also confer an undesirable property, for example increased toxicity. Incorporation of certain other nucleoside modifications results in antisense compounds with different profiles of properties. In certain embodiments, one may combine modifications in one or both wings to optimize desirable characteristics and/or minimize undesirable characteristics. In certain embodiments, the wings of a mixed wing antisense compound comprise one or more nucleoside comprising a first modification that increases affinity of the antisense compound for a target nucleic acid compared to an antisense compound comprising unmodified nucleosides; and one or more nucleoside comprising a second modification that results in reduced toxicity compared to an antisense compound with wings comprising nucleosides that all comprise the first modification.

In certain embodiments, an antisense compound comprises at least one wing comprising at least one MOE substituted nucleoside and at least one high affinity modification. In certain such embodiments, the at least one MOE substituted nucleoside and the at least one high affinity are in the 3′ wing. In certain such embodiments, the at least one MOE substituted nucleoside and the at least one high affinity are in the 5′ wing.

In certain embodiments, an antisense compound comprises 1, 2 or 3 high affinity modifications in the 5′ and/or 3′ wings.

Target Nucleic Acids, Target Regions and Nucleotide Sequences

In certain embodiments, an allelic variant of huntingtin is selectively reduced. Nucleotide sequences that encode huntingtin include, without limitation, the following: GENBANK Accession No. NT_006081.18, truncated from nucleotides 1566000 to 1768000 (replaced by GENBANK Accession No. NT_006051), incorporated herein as SEQ ID NO: 1, and NM_002111.6, incorporated herein as SEQ ID NO: 2.

It is understood that the sequence set forth in each SEQ ID NO in the Examples contained herein is independent of any modification to a sugar moiety, an internucleoside linkage, or a nucleobase. As such, antisense compounds defined by a SEQ ID NO may comprise, independently, one or more modifications to a sugar moiety, an internucleoside linkage, or a nucleobase. Antisense compounds described by Isis Number (Isis No) indicate a combination of nucleobase sequence and motif.

In certain embodiments, a target region is a structurally defined region of the target nucleic acid. For example, a target region may encompass a 3′ UTR, a 5′ UTR, an exon, an intron, an exon/intron junction, a coding region, a translation initiation region, translation termination region, or other defined nucleic acid region. The structurally defined regions for huntingtin can be obtained by accession number from sequence databases such as NCBI and such information is incorporated herein by reference. In certain embodiments, a target region may encompass the sequence from a 5′ target site of one target segment within the target region to a 3′ target site of another target segment within the same target region.

Targeting includes determination of at least one target segment to which an antisense compound hybridizes, such that a desired effect occurs. In certain embodiments, the desired effect is a reduction in mRNA target nucleic acid levels of a particular allelic variant. In certain embodiments, the desired effect is reduction of levels of the protein encoded by the target nucleic acid or a phenotypic change associated with a particular alleleic variant.

A target region may contain one or more target segments. Multiple target segments within a target region may be overlapping. Alternatively, they may be non-overlapping. In certain embodiments, target segments within a target region are separated by no more than about 300 nucleotides. In certain embodiments, target segments within a target region are separated by a number of nucleotides that is, is about, is no more than, is no more than about, 250, 200, 150, 100, 90, 80, 70, 60, 50, 40, 30, 20, or 10 nucleotides on the target nucleic acid, or is a range defined by any two of the preceeding values. In certain embodiments, target segments within a target region are separated by no more than, or no more than about, 5 nucleotides on the target nucleic acid. In certain embodiments, target segments are contiguous. Contemplated are target regions defined by a range having a starting nucleic acid that is any of the 5′ target sites or 3′ target sites listed herein.

Suitable target segments may be found within a 5′ UTR, a coding region, a 3′ UTR, an intron, an exon, or an exon/intron junction. Target segments containing a start codon or a stop codon are also suitable target segments. A suitable target segment may specifically exclude a certain structurally defined region such as the start codon or stop codon.

The determination of suitable target segments may include a comparison of the sequence of a target nucleic acid to other sequences throughout the genome. For example, the BLAST algorithm may be used to identify regions of similarity amongst different nucleic acids. This comparison can prevent the selection of antisense compound sequences that may hybridize in a non-specific manner to sequences other than a selected target nucleic acid (i.e., non-target or off-target sequences).

Cell Lines

In certain embodiments, the GM04281, GM02171, and GM02173B cell lines are used in experiments described herein below. The GM04281 cell line has a wild-type HTT allele that contains 17 repeats and a mutant HTT allele that contains 69 repeats. The cell line was derived from a patient both of whose parents were also affected by the disease. The GM02171 cell line was chosen as a counter screen control to the GM04281. This cell line was derived from the daughter of parents, only one of whom had the disease. The daughter had not developed HD but was considered to be at risk. The GM02173B cell line was also patient-derived and was used as a haplotype test control.

Table 1 provides SNPs found in the GM04281, GM02171, and GM02173B cell lines. Also provided are the allelic variants found at each SNP position, the genotype for each of the cell lines, and the percentage of HD patients having a particular allelic variant. For example, the two allelic variants for SNP rs6446723 are T and C. The GM02171 cell line is homozygous CC, the GM02173 cell line is heterozygous TC, and the GM04281 cell line is homozygous TT. Fifty percent of HD patients have a T at SNP position rs6446723.

TABLE 1 Allelic Variations for SNPs Associated with HD SNP Variation GM02171 GM02173 GM04281 TargetPOP allele rs6446723 T/C CC TC TT 0.50 T rs3856973 A/G AA AG GG 0.50 G rs2285086 A/G GG AG AA 0.50 A rs363092 A/C AA AC CC 0.49 C rs916171 C/G GG GC CC 0.49 C rs6844859 T/C CC TC TT 0.49 T rs7691627 A/G AA AG GG 0.49 G rs4690073 A/G AA AG GG 0.49 G rs2024115 A/G GG AG AA 0.48 A rs11731237 T/C CC TC TT 0.43 T rs362296 A/C AC AC AC 0.42 C rs10015979 A/G AA AG GG 0.42 G rs7659144 C/G CG CG CC 0.41 C rs363096 T/C CC TC TT 0.40 T rs362273 A/G AG AG AA 0.39 A rs16843804 T/C TC TC CC 0.38 C rs362271 A/G AG AG GG 0.38 G rs362275 T/C TC TC CC 0.38 C rs3121419 T/C TC TC CC 0.38 C rs362272 A/G AG GG 0.38 G rs3775061 A/G AG AG AA 0.38 A rs34315806 T/C TC TC CC 0.38 C rs363099 T/C TC TC CC 0.38 C rs2298967 T/C TC TC TT 0.38 T rs363088 A/T TA TA AA 0.38 A rs363064 T/C TC TC CC 0.35 C rs363102 A/G AA AA AA 0.23 G rs2798235 A/G GG GG GG 0.21 A rs363080 T/C CC CC CC 0.21 T rs363072 A/T TA AA AA 0.13 A rs363125 A/C AC CC CC 0.12 C rs362303 T/C TC CC CC 0.12 C rs362310 T/C TC CC CC 0.12 C rs10488840 A/G AG GG GG 0.12 G rs362325 T/C TC TT TT 0.11 T rs35892913 A/G GG GG GG 0.10 A rs363102 A/G AA AA AA 0.09 A rs363096 T/C CC TC TT 0.09 C rs11731237 T/C CC TC TT 0.09 C rs10015979 A/G AA AG GG 0.08 A rs363080 T/C CC CC CC 0.07 C rs2798235 A/G GG GG GG 0.07 G rs1936032 C/G CC CC CC 0.06 C rs2276881 A/G GG GG GG 0.06 G rs363070 A/G AA AA AA 0.06 A rs35892913 A/G GG GG GG 0.04 G rs12502045 T/C CC CC CC 0.04 C rs6446723 T/C CC TC TT 0.04 C rs7685686 A/G GG AG AA 0.04 G rs3733217 T/C CC CC CC 0.03 C rs6844859 T/C CC TC TT 0.03 C rs362331 T/C CC TC TT 0.03 C

Hybridization

In some embodiments, hybridization occurs between an antisense compound disclosed herein and a SNP site. The most common mechanism of hybridization involves hydrogen bonding (e.g., Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding) between complementary nucleobases of the nucleic acid molecules.

Hybridization can occur under varying conditions. Stringent conditions are sequence-dependent and are determined by the nature and composition of the nucleic acid molecules to be hybridized.

In certain embodiments, the antisense compounds provided herein are specifically hybridizable with the nucleic acid of a particular allelic variant.

Complementarity

An antisense compound and a target nucleic acid are complementary to each other when a sufficient number of nucleobases of the antisense compound can hydrogen bond with the corresponding nucleobases of the target nucleic acid, such that a desired effect will occur (e.g., selective reduction of a gene product of an allelic variant).

Non-complementary nucleobases between an antisense compound and a target nucleic acid may be tolerated provided that the antisense compound remains able to specifically hybridize to a target nucleic acid. Moreover, an antisense compound may hybridize over one or more segments of a target nucleic acid such that intervening or adjacent segments are not involved in the hybridization event (e.g., a loop structure, mismatch or hairpin structure).

In certain embodiments, the antisense compounds provided herein, or a specified portion thereof, are, or are at least, 70%, 80%, 85%, 86%, 87%, 88%, 89%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99%, or 100% complementary to a target nucleic acid, a target region, target segment, SNP site, or specified portion thereof. Percent complementarity of an antisense compound with a target nucleic acid can be determined using routine methods.

For example, an antisense compound in which 18 of 20 nucleobases of the antisense compound are complementary to a target region, and would therefore specifically hybridize, would represent 90 percent complementarity. In this example, the remaining noncomplementary nucleobases may be clustered or interspersed with complementary nucleobases and need not be contiguous to each other or to complementary nucleobases. As such, an antisense compound which is 18 nucleobases in length having 4 (four) noncomplementary nucleobases which are flanked by two regions of complete complementarity with the target nucleic acid would have 77.8% overall complementarity with the target nucleic acid and would thus fall within the scope of the present invention. Percent complementarity of an antisense compound with a region of a target nucleic acid can be determined routinely using BLAST programs (basic local alignment search tools) and PowerBLAST programs known in the art (Altschul et al., J. Mol. Biol., 1990, 215, 403 410; Zhang and Madden, Genome Res., 1997, 7, 649 656). Percent homology, sequence identity or complementarity, can be determined by, for example, the Gap program (Wisconsin Sequence Analysis Package, Version 8 for Unix, Genetics Computer Group, University Research Park, Madison Wis.), using default settings, which uses the algorithm of Smith and Waterman (Adv. Appl. Math., 1981, 2, 482 489).

In certain embodiments, the antisense compounds provided herein, or specified portions thereof, are fully complementary (i.e. 100% complementary) to a target nucleic acid, a SNP site, target region, target segment, or specified portion thereof. As used herein, “fully complementary” means each nucleobase of an antisense compound is capable of precise base pairing with the corresponding nucleobases of a target nucleic acid. For example, a 20 nucleobase antisense compound is fully complementary to a target sequence that is 400 nucleobases long, so long as there is a corresponding 20 nucleobase portion of the target nucleic acid that is fully complementary to the antisense compound. Fully complementary can also be used in reference to a specified portion of the first and/or the second nucleic acid. For example, a 20 nucleobase portion of a 30 nucleobase antisense compound can be “fully complementary” to a target sequence that is 400 nucleobases long. The 20 nucleobase portion of the 30 nucleobase oligonucleotide is fully complementary to the target sequence if the target sequence has a corresponding 20 nucleobase portion wherein each nucleobase is complementary to the 20 nucleobase portion of the antisense compound. At the same time, the entire 30 nucleobase antisense compound may or may not be fully complementary to the target sequence, depending on whether the remaining 10 nucleobases of the antisense compound are also complementary to the target sequence.

The location of a non-complementary nucleobase may be at the 5′ end or 3′ end of the antisense compound. Alternatively, the non-complementary nucleobase or nucleobases may be at an internal position of the antisense compound. When two or more non-complementary nucleobases are present, they may be contiguous (i.e. linked) or non-contiguous. In one embodiment, a non-complementary nucleobase is located in the wing segment of a gapmer antisense oligonucleotide.

In certain embodiments, antisense compounds that are, or are up to 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleobases in length comprise no more than 6, no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 non-complementary nucleobase(s) relative to a target nucleic acid, SNP site, or specified portion thereof.

In certain embodiments, antisense oligonucleotides that are, or are up to 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, or 30 nucleobases in length comprise no more than 6, no more than 5, no more than 4, no more than 3, no more than 2, or no more than 1 non-complementary nucleobase(s) relative to a target nucleic acid, SNP site, or specified portion thereof.

The antisense compounds provided herein also include those which are complementary to a portion of a target nucleic acid. As used herein, “portion” refers to a defined number of contiguous (i.e. linked) nucleobases within a region or segment of a target nucleic acid. A “portion” can also refer to a defined number of contiguous nucleobases of an antisense compound. In certain embodiments, the antisense compounds, are complementary to at least an 8 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 12 nucleobase portion of a target segment. In certain embodiments, the antisense compounds are complementary to at least a 15 nucleobase portion of a target segment. Also contemplated are antisense compounds that are complementary to at least a 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more nucleobase portion of a target segment, or a range defined by any two of these values.

Identity

The antisense compounds provided herein may also have a defined percent identity to a particular nucleotide sequence, SEQ ID NO, or compound represented by a specific Isis number, or portion thereof. As used herein, an antisense compound 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 a disclosed DNA sequence would be considered identical to the DNA sequence since both uracil and thymidine pair with adenine. Shortened and lengthened versions of the antisense compounds described herein as well as compounds having non-identical bases relative to the antisense compounds provided herein also are contemplated. The non-identical bases may be adjacent to each other or dispersed throughout the antisense compound. Percent identity of an antisense compound is calculated according to the number of bases that have identical base pairing relative to the sequence to which it is being compared.

In certain embodiments, the antisense compounds, or portions thereof, are at least 70%, 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, 99% or 100% identical to one or more of the antisense compounds or SEQ ID NOs, or a portion thereof, disclosed herein.

In certain embodiments, a portion of the antisense compound is compared to an equal length portion of the target nucleic acid. In certain embodiments, an 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleobase portion is compared to an equal length portion of the target nucleic acid.

In certain embodiments, a portion of the antisense oligonucleotide is compared to an equal length portion of the target nucleic acid. In certain embodiments, an 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25 nucleobase portion is compared to an equal length portion of the target nucleic acid.

Modifications

A nucleoside is a base-sugar combination. The nucleobase (also known as base) portion of the nucleoside is normally a heterocyclic base moiety. 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. Oligonucleotides are formed through the covalent linkage of adjacent nucleosides to one another, to form a linear polymeric oligonucleotide. Within the oligonucleotide structure, the phosphate groups are commonly referred to as forming the internucleoside linkages of the oligonucleotide.

Modifications to antisense compounds encompass substitutions or changes to internucleoside linkages, sugar moieties, or nucleobases. Modified antisense compounds are often preferred over native forms because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for nucleic acid target, increased stability in the presence of nucleases, or increased inhibitory activity.

Chemically modified nucleosides may also be employed to increase the binding affinity of a shortened or truncated antisense oligonucleotide for its target nucleic acid. Consequently, comparable results can often be obtained with shorter antisense compounds that have such chemically modified nucleosides.

Chemically modified nucleosides may also be employed to increase selectivity in reducing expression the gene product of an allelic variant.

Modified Internucleoside Linkages

The naturally occurring internucleoside linkage of RNA and DNA is a 3′ to 5′ phosphodiester linkage. Antisense compounds having one or more modified, i.e. non-naturally occurring, internucleoside linkages are often selected over antisense compounds having naturally occurring internucleoside linkages because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for target nucleic acids, and increased stability in the presence of nucleases.

Oligonucleotides having modified internucleoside linkages include internucleoside linkages that retain a phosphorus atom as well as internucleoside linkages that do not have a phosphorus atom. Representative phosphorus containing internucleoside linkages include, but are not limited to, phosphodiesters, phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioate. Methods of preparation of phosphorous-containing and non-phosphorous-containing linkages are well known.

In certain embodiments, antisense compounds comprise one or more modified internucleoside linkages. In certain embodiments, the modified internucleoside linkages are phosphorothioate linkages. In certain embodiments, each internucleoside linkage of an antisense compound is a phosphorothioate internucleoside linkage.

Modified Sugar Moieties

Antisense compounds of the invention can optionally contain one or more nucleosides wherein the sugar group has been modified. Such sugar modified nucleosides may impart enhanced nuclease stability, increased binding affinity, increased selectivity for an allelic variant, or some other beneficial biological property to the antisense compounds. In certain embodiments, nucleosides comprise a chemically modified ribofuranose ring moieties. Examples of chemically modified ribofuranose rings include without limitation, addition of substitutent groups (including 5′ and 2′ substituent groups, bridging of non-geminal ring atoms to form bicyclic nucleic acids (BNA), replacement of the ribosyl ring oxygen atom with S, N(R), or C(R1)(R)2 (R═H, C1-C12 alkyl or a protecting group) and combinations thereof. Examples of chemically modified sugars include 2′-F-5′-methyl substituted nucleoside (see PCT International Application WO 2008/101157 Published on Aug. 21, 2008 for other disclosed 5′,2′-bis substituted nucleosides) or replacement of the ribosyl ring oxygen atom with S with further substitution at the 2′-position (see published U.S. Patent Application US2005-0130923, published on Jun. 16, 2005) or alternatively 5′-substitution of a BNA (see PCT International Application WO 2007/134181 Published on Nov. 22, 2007 wherein LNA is substituted with for example a 5′-methyl or a 5′-vinyl group).

Examples of nucleosides having modified sugar moieties include without limitation nucleosides comprising 5′-vinyl, 5′-methyl (R or S), 4′-S, 2′-F, 2′-OCH3 and 2′-O(CH2)2OCH3 substituent groups. The substituent at the 2′ position can also be selected from allyl, amino, azido, thio, O-allyl, O—C1-C10 alkyl, OCF3, O(CH2)2SCH3, O(CH2)2-O—N(Rm)(Rn), and O—CH2-C(═O)—N(Rm)(Rn), where each Rm and Rn is, independently, H or substituted or unsubstituted C1-C10 alkyl.

As used herein, “bicyclic nucleosides” refer to modified nucleosides comprising a bicyclic sugar moiety. Examples of bicyclic nucleosides include without limitation nucleosides comprising a bridge between the 4′ and the 2′ ribosyl ring atoms. In certain embodiments, antisense compounds provided herein include one or more bicyclic nucleosides wherein the bridge comprises a 4′ to 2′ bicyclic nucleoside. Examples of such 4′ to 2′ bicyclic nucleosides, include but are not limited to one of the formulae: 4′-(CH2)—O-2′ (LNA); 4′-(CH2)—S-2′; 4′-(CH2)2—O-2′ (ENA); 4′-CH(CH3)—O-2′ and 4′-CH(CH2OCH3)—O-2′ (and analogs thereof see U.S. Pat. No. 7,399,845, issued on Jul. 15, 2008); 4′-C(CH3)(CH3)—O-2′ (and analogs thereof see published International Application WO/2009/006478, published Jan. 8, 2009); 4′-CH2—N(OCH3)-2′ (and analogs thereof see published International Application WO/2008/150729, published Dec. 11, 2008); 4′-CH2—O—N(CH3)-2′ (see published U.S. Patent Application US2004-0171570, published Sep. 2, 2004); 4′-CH2—N(R)—O-2′, wherein R is H, C1-C12 alkyl, or a protecting group (see U.S. Pat. No. 7,427,672, issued on Sep. 23, 2008); 4′-CH2—C(H)(CH3)-2′ (see Chattopadhyaya, et al., J. Org. Chem., 2009, 74, 118-134); and 4′-CH2—C(═CH2)-2′ (and analogs thereof see published International Application WO 2008/154401, published on Dec. 8, 2008). See, for example: Singh et al., Chem. Commun., 1998, 4, 455-456; Koshkin et al., Tetrahedron, 1998, 54, 3607-3630; Wahlestedt et al., Proc. Natl. Acad. Sci. U.S.A, 2000, 97, 5633-5638; Kumar et al., Bioorg. Med. Chem. Lett., 1998, 8, 2219-2222; Singh et al., J. Org. Chem., 1998, 63, 10035-10039; Srivastava et al., J. Am. Chem. Soc., 129(26) 8362-8379 (Jul. 4, 2007); U.S. Pat. Nos. 7,053,207; 6,268,490; 6,770,748; 6,794,499; 7,034,133; and U.S. Pat. No. 6,525,191; Elayadi et al., Curr. Opinion Invens. Drugs, 2001, 2, 558-561; Braasch et al., Chem. Biol., 2001, 8, 1-7; and Orum et al., Curr. Opinion Mol. Ther., 2001, 3, 239-243; and U.S. Pat. No. 6,670,461; International applications WO 2004/106356; WO 94/14226; WO 2005/021570; U.S. Patent Publication Nos. US2004-0171570; US2007-0287831; US2008-0039618; U.S. Pat. No. 7,399,845; U.S. patent Ser. Nos. 12/129,154; 60/989,574; 61/026,995; 61/026,998; 61/056,564; 61/086,231; 61/097,787; 61/099,844; PCT International Applications Nos. PCT/US2008/064591; PCT/US2008/066154; PCT/US2008/068922; and Published PCT International Applications WO 2007/134181. Each of the foregoing bicyclic nucleosides can be prepared having one or more stereochemical sugar configurations including for example α-L-ribofuranose and β-D-ribofuranose (see PCT international application PCT/DK98/00393, published on Mar. 25, 1999 as WO 99/14226).

In certain embodiments, bicyclic sugar moieties of BNA nucleosides include, but are not limited to, compounds having at least one bridge between the 4′ and the 2′ position of the pentofuranosyl sugar moiety wherein such bridges independently comprises 1 or from 2 to 4 linked groups independently selected from —[C(Ra)(Rb)]n—, —C(Ra)═C(Rb)—, —C(Ra)═N—, —C(═NRa)—, —C(═O)—, —C(═S)—, —O—, —Si(Ra)2—, —S(═O)x—, and —N(Ra)—;

wherein:

x is 0, 1, or 2;

n is 1, 2, 3, or 4;

each Ra and Rb is, independently, H, a protecting group, hydroxyl, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, heterocycle radical, substituted heterocycle radical, heteroaryl, substituted heteroaryl, C5-C7 alicyclic radical, substituted C5-C7 alicyclic radical, halogen, OJ1, NJ1J2, SJ1, N3, COOJ1, acyl (C(═O)—H), substituted acyl, CN, sulfonyl (S(═O)2-J1), or sulfoxyl (S(═O)-J1); and each J1 and J2 is, independently, H, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C5-C20 aryl, substituted C5-C20 aryl, acyl (C(═O)—H), substituted acyl, a heterocycle radical, a substituted heterocycle radical, C1-C12 aminoalkyl, substituted C1-C12 aminoalkyl or a protecting group.

In certain embodiments, the bridge of a bicyclic sugar moiety is, —[C(Ra)(Rb)]n—, —[C(Ra)(Rb)]n—O—, —C(RaRb)—N(R)—O— or —C(RaRb)—O—N(R)—. In certain embodiments, the bridge is 4′-CH2-2′, 4′-(CH2)2-2′, 4′-(CH2)3-2′, 4′-CH2—O-2′, 4′-(CH2)2—O-2′, 4′-CH2—O—N(R)-2′ and 4′-CH2—N(R)—O-2′- wherein each R is, independently, H, a protecting group or C1-C12 alkyl.

In certain embodiments, bicyclic nucleosides are further defined by isomeric configuration. For example, a nucleoside comprising a 4′-2′ methylene-oxy bridge, may be in the α-L configuration or in the β-D configuration. Previously, α-L-methyleneoxy (4′-CH2—O-2′) BNA's have been incorporated into antisense oligonucleotides that showed antisense activity (Frieden et al., Nucleic Acids Research, 2003, 21, 6365-6372).

In certain embodiments, bicyclic nucleosides include, but are not limited to, (A) α-L-Methyleneoxy (4′-CH2—O-2′) BNA, (B) β-D-Methyleneoxy (4′-CH2—O-2′) BNA, (C) Ethyleneoxy (4′-(CH2)2—O-2′) BNA, (D) Aminooxy (4′-CH2—O—N(R)-2′) BNA, (E) Oxyamino (4′-CH2—N(R)—O-2′) BNA, and (F) Methyl(methyleneoxy) (4′-CH(CH3)—O-2′) BNA, (G) methylene-thio (4′-CH2—S-2′) BNA, (H) methylene-amino (4′-CH2—N(R)-2′) BNA, (I) methyl carbocyclic (4′-CH2—CH(CH3)-2′) BNA, (J) propylene carbocyclic (4′-(CH2)3-2′) BNA, and (K) ethylene carbocyclic (4′-CH2—CH2-2′) (carba LNA or “cLNA”) as depicted below.

wherein Bx is the base moiety and R is independently H, a protecting group or C1-C12 alkyl.

In certain embodiments, bicyclic nucleoside having Formula I:

wherein:

Bx is a heterocyclic base moiety;

-Qa-Qb-Qc- is —CH2—N(Rc)—CH2—, —C(═O)—N(Rc)—CH2—, —CH2—O—N(Rc)—, —CH2—N(Rc)—O— or —N(Rc)—O—CH2;

Rc is C1-C12 alkyl or an amino protecting group; and

Ta and Tb are each, independently H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety or a covalent attachment to a support medium.

In certain embodiments, bicyclic nucleoside having Formula II:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety or a covalent attachment to a support medium;

Za is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, substituted C1-C6 alkyl, substituted C2-C6 alkenyl, substituted C2-C6 alkynyl, acyl, substituted acyl, substituted amide, thiol or substituted thio.

In one embodiment, each of the substituted groups, is, independently, mono or poly substituted with substituent groups independently selected from halogen, oxo, hydroxyl, OJc, NJcJd, SJc, N3, OC(═X)Jc, and NJcC(═X)NJcJd, wherein each Jc, Jd and Je is, independently, H, C1-C6 alkyl, or substituted C1-C6 alkyl and X is O or NJc.

In certain embodiments, bicyclic nucleoside having Formula III:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety or a covalent attachment to a support medium;

Zb is C1-C6 alkyl, C2-C6 alkenyl, C2-C6 alkynyl, substituted C1-C6 alkyl, substituted C2-C6 alkenyl, substituted C2-C6 alkynyl or substituted acyl (C(═O)—).

In certain embodiments, bicyclic nucleoside having Formula IV:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety or a covalent attachment to a support medium;

Rd is C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl or substituted C2-C6 alkynyl;

each qa, qb, qc and qd is, independently, H, halogen, C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl or substituted C2-C6 alkynyl, C1-C6 alkoxyl, substituted C1-C6 alkoxyl, acyl, substituted acyl, C1-C6 aminoalkyl or substituted C1-C6 aminoalkyl;

In certain embodiments, bicyclic nucleoside having Formula V:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety or a covalent attachment to a support medium;

qa, qb, qe and qf are each, independently, hydrogen, halogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C1-C12 alkoxy, substituted C1-C12 alkoxy, OJj, SJj, SOJj, SO2Jj, NJjJk, N3, CN, C(═O)OJj, C(═O)NJjJk, C(═O)Jj, O—C(═O)NJjJk, N(H)C(═NH)NJjJk, N(H)C(═O)NJjJk or N(H)C(═S)NJjJk;

or qe and of together are ═C(qg)(qh);

qg and qh are each, independently, H, halogen, C1-C12 alkyl or substituted C1-C12 alkyl.

The synthesis and preparation of the methyleneoxy (4′-CH2—O-2′) BNA monomers adenine, cytosine, guanine, 5-methyl-cytosine, thymine and uracil, along with their oligomerization, and nucleic acid recognition properties have been described (Koshkin et al., Tetrahedron, 1998, 54, 3607-3630). BNAs and preparation thereof are also described in WO 98/39352 and WO 99/14226.

Analogs of methyleneoxy (4′-CH2—O-2′) BNA, methyleneoxy (4′-CH2—O-2′) BNA and 2′-thio-BNAs, have also been prepared (Kumar et al., Bioorg. Med. Chem. Lett., 1998, 8, 2219-2222). Preparation of locked nucleoside analogs comprising oligodeoxyribonucleotide duplexes as substrates for nucleic acid polymerases has also been described (Wengel et al., WO 99/14226). Furthermore, synthesis of 2′-amino-BNA, a novel comformationally restricted high-affinity oligonucleotide analog has been described in the art (Singh et al., J. Org. Chem., 1998, 63, 10035-10039). In addition, 2′-Amino- and 2′-methylamino-BNA's have been prepared and the thermal stability of their duplexes with complementary RNA and DNA strands has been previously reported.

In certain embodiments, bicyclic nucleoside having Formula VI:

wherein:

Bx is a heterocyclic base moiety;

Ta and Tb are each, independently H, a hydroxyl protecting group, a conjugate group, a reactive phosphorus group, a phosphorus moiety or a covalent attachment to a support medium;

each qi, qj, qk and qi is, independently, H, halogen, C1-C12 alkyl, substituted C1-C12 alkyl, C2-C12 alkenyl, substituted C2-C12 alkenyl, C2-C12 alkynyl, substituted C2-C12 alkynyl, C1-C12 alkoxyl, substituted C1-C12 alkoxyl, OJj, SJj, SOJj, SO2Jj, NJjJk, N3, CN, C(═O)OJj, C(═O)NJjJk, C(═O)Jj, O—C(═O)NJjJk, N(H)C(═NH)NJjJk, N(H)C(═O)NJjJk or N(H)C(═S)NJjJk; and

qi and qj or ql and qk together are ═C(qg)(qh), wherein qg and qh are each, independently, H, halogen, C1-C12 alkyl or substituted C1-C12 alkyl.

One carbocyclic bicyclic nucleoside having a 4′-(CH2)3-2′ bridge and the alkenyl analog bridge 4′-CH═CH—CH2-2′ have been described (Frier et al., Nucleic Acids Research, 1997, 25(22), 4429-4443 and Albaek et al., J. Org. Chem., 2006, 71, 7731-7740). The synthesis and preparation of carbocyclic bicyclic nucleosides along with their oligomerization and biochemical studies have also been described (Srivastava et al., J. Am. Chem. Soc. 2007, 129(26), 8362-8379).

As used herein, “4′-2′ bicyclic nucleoside” or “4′ to 2′ bicyclic nucleoside” refers to a bicyclic nucleoside comprising a furanose ring comprising a bridge connecting two carbon atoms of the furanose ring connects the 2′ carbon atom and the 4′ carbon atom of the sugar ring.

As used herein, “monocylic nucleosides” refer to nucleosides comprising modified sugar moieties that are not bicyclic sugar moieties. In certain embodiments, the sugar moiety, or sugar moiety analogue, of a nucleoside may be modified or substituted at any position.

As used herein, “2′-modified sugar” means a furanosyl sugar modified at the 2′ position. In certain embodiments, such modifications include substituents selected from: a halide, including, but not limited to substituted and unsubstituted alkoxy, substituted and unsubstituted thioalkyl, substituted and unsubstituted amino alkyl, substituted and unsubstituted alkyl, substituted and unsubstituted allyl, and substituted and unsubstituted alkynyl. In certain embodiments, 2′ modifications are selected from substituents including, but not limited to: O[(CH2)nO]mCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2)nONH2, OCH2C(═O)N(H)CH3, and O(CH2)nON[(CH2)nCH3]2, where n and m are from 1 to about 10. Other 2′-substituent groups can also be selected from: C1-C12 alkyl, substituted alkyl, alkenyl, alkynyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving pharmacokinetic properties, or a group for improving the pharmacodynamic properties of an antisense compound, and other substituents having similar properties. In certain embodiments, modified nucleosides comprise a 2′-MOE side chain (Baker et al., J. Biol. Chem., 1997, 272, 11944-12000). Such 2′-MOE substitution have been described as having improved binding affinity compared to unmodified nucleosides and to other modified nucleosides, such as 2′-O-methyl, O-propyl, and O-aminopropyl. Oligonucleotides having the 2′-MOE substituent also have been shown to be antisense inhibitors of gene expression with promising features for in vivo use (Martin, P., Helv. Chim. Acta, 1995, 78, 486-504; Altmann et al., Chimia, 1996, 50, 168-176; Altmann et al., Biochem. Soc. Trans., 1996, 24, 630-637; and Altmann et al., Nucleosides Nucleotides, 1997, 16, 917-926).

As used herein, a “modified tetrahydropyran nucleoside” or “modified THP nucleoside” means a nucleoside having a six-membered tetrahydropyran “sugar” substituted in for the pentofuranosyl residue in normal nucleosides (a sugar surrogate). Modified THP nucleosides include, but are not limited to, what is referred to in the art as hexitol nucleic acid (HNA), anitol nucleic acid (ANA), manitol nucleic acid (MNA) (see Leumann, C J. Bioorg. & Med. Chem. (2002) 10:841-854), fluoro HNA (F-HNA) or those compounds having Formula X:

wherein independently for each of said at least one tetrahydropyran nucleoside analog of Formula X:

Bx is a heterocyclic base moiety;

T3 and T4 are each, independently, an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound or one of T3 and T4 is an internucleoside linking group linking the tetrahydropyran nucleoside analog to the antisense compound and the other of T3 and T4 is H, a hydroxyl protecting group, a linked conjugate group or a 5′ or 3′-terminal group;

q1, q2, q3, q4, q5, q6 and q7 are each independently, H, C1-C6 alkyl, substituted C1-C6 alkyl, C2-C6 alkenyl, substituted C2-C6 alkenyl, C2-C6 alkynyl or substituted C2-C6 alkynyl; and

one of R1 and R2 is hydrogen and the other is selected from halogen, substituted or unsubstituted alkoxy, NJ1J2, SJ1, N3, OC(═X)J1, OC(═X)NJ1J2, NJ3C(═X)NJ1J2 and CN, wherein X is O, S or NJ1 and each J1, J2 and J3 is, independently, H or C1-C6 alkyl.

In certain embodiments, the modified THP nucleosides of Formula X are provided wherein qm, qn, qp, qr, qs, qt and qu are each H. In certain embodiments, at least one of qm, qn, qp, qr, qs, qt and qu is other than H. In certain embodiments, at least one of qm, qn, qp, qr, qs, qt and qu is methyl. In certain embodiments, THP nucleosides of Formula X are provided wherein one of R1 and R2 is F. In certain embodiments, R1 is fluoro and R2 is H; R1 is methoxy and R2 is H, and R1 is methoxyethoxy and R2 is H.

As used herein, “2′-modified” or “2′-substituted” refers to a nucleoside comprising a sugar comprising a substituent at the 2′ position other than H or OH. 2′-modified nucleosides, include, but are not limited to, bicyclic nucleosides wherein the bridge connecting two carbon atoms of the sugar ring connects the 2′ carbon and another carbon of the sugar ring; and nucleosides with non-bridging 2′ substituents, such as allyl, amino, azido, thio, O-allyl, O—C1-C10 alkyl, —OCF3, O—(CH2)2—O—CH3, 2′-O(CH2)2SCH3, O—(CH2)2—O—N(Rm)(Rn), or O—CH2—C(═O)—N(Rm)(Rn), where each Rm and Rn is, independently, H or substituted or unsubstituted C1-C10 alkyl. 2′-modified nucleosides may further comprise other modifications, for example at other positions of the sugar and/or at the nucleobase.

As used herein, “2′-F” refers to a nucleoside comprising a sugar comprising a fluoro group at the 2′ position.

As used herein, “2′-OMe” or “2′-OCH3” or “2′-O-methyl” each refers to a nucleoside comprising a sugar comprising an —OCH3 group at the 2′ position of the sugar ring.

As used herein, “MOE” or “2′-MOE” or “2′-OCH2CH2OCH3” or “2′-O-methoxyethyl” each refers to a nucleoside comprising a sugar comprising a —OCH2CH2OCH3 group at the 2′ position of the sugar ring.

As used herein, “oligonucleotide” refers to a compound comprising a plurality of linked nucleosides. In certain embodiments, one or more of the plurality of nucleosides is modified. In certain embodiments, an oligonucleotide comprises one or more ribonucleosides (RNA) and/or deoxyribonucleosides (DNA).

Many other bicyclo and tricyclo sugar surrogate ring systems are also know in the art that can be used to modify nucleosides for incorporation into antisense compounds (see for example review article: Leumann, J. C, Bioorganic & Medicinal Chemistry, 2002, 10, 841-854). Such ring systems can undergo various additional substitutions to enhance activity.

Methods for the preparations of modified sugars are well known to those skilled in the art.

In nucleotides having modified sugar moieties, the nucleobase moieties (natural, modified or a combination thereof) are maintained for hybridization with an appropriate nucleic acid target.

In certain embodiments, antisense compounds comprise one or more nucleotides having modified sugar moieties. In certain embodiments, the modified sugar moiety is 2′-MOE. In certain embodiments, the 2′-MOE modified nucleotides are arranged in a gapmer motif. In certain embodiments, the modified sugar moiety is a cEt. In certain embodiments, the cEt modified nucleotides are arranged throughout the wings of a gapmer motif.

Modified Nucleobases

Nucleobase (or base) modifications or substitutions are structurally distinguishable from, yet functionally interchangeable with, naturally occurring or synthetic unmodified nucleobases.

Both natural and modified nucleobases are capable of participating in hydrogen bonding. Such nucleobase modifications may impart nuclease stability, binding affinity, increased selectivity for an allelic variant, or some other beneficial biological property to antisense compounds. Modified nucleobases include synthetic and natural nucleobases such as, for example, 5-methylcytosine (5-me-C). Certain nucleobase substitutions, including 5-methylcytosine substitutions, are particularly useful for increasing the binding affinity of an antisense compound for a target nucleic acid. For example, 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278).

Additional modified nucleobases include 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl (—C≡C—CH3) uracil and cytosine and other alkynyl derivatives of pyrimidine bases, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl and other 8-substituted adenines and guanines, 5-halo particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 2-F-adenine, 2-amino-adenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine.

Heterocyclic base moieties may also include those in which the purine or pyrimidine base is replaced with other heterocycles, for example 7-deaza-adenine, 7-deazaguanosine, 2-aminopyridine and 2-pyridone. Nucleobases that are particularly useful for increasing the binding affinity of antisense compounds include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and O-6 substituted purines, including 2 aminopropyladenine, 5-propynyluracil and 5-propynylcyto sine.

In certain embodiments, antisense compounds comprise one or more modified nucleobases. In certain embodiments, gap-widened antisense oligonucleotides comprise one or more modified nucleobases. In certain embodiments, the modified nucleobase is 5-methylcytosine. In certain embodiments, each cytosine is a 5-methylcytosine.

Compositions and Methods for Formulating Pharmaceutical Compositions

Antisense oligonucleotides may be admixed with pharmaceutically acceptable active or inert substances for the preparation of pharmaceutical compositions or formulations. Compositions and methods for the formulation of pharmaceutical compositions are dependent upon a number of criteria, including, but not limited to, route of administration, extent of disease, or dose to be administered.

An antisense compound can be utilized in pharmaceutical compositions by combining the antisense compound with a suitable pharmaceutically acceptable diluent or carrier. A pharmaceutically acceptable diluent includes phosphate-buffered saline (PBS). PBS is a diluent suitable for use in compositions to be delivered parenterally. Accordingly, in one embodiment, employed in the methods described herein is a pharmaceutical composition comprising an antisense compound and a pharmaceutically acceptable diluent. In certain embodiments, the pharmaceutically acceptable diluent is PBS. In certain embodiments, the antisense compound is an antisense oligonucleotide.

Pharmaceutical compositions comprising antisense compounds encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other oligonucleotide 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 pharmaceutically acceptable salts of antisense compounds, prodrugs, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents. Suitable pharmaceutically acceptable salts include, but are not limited to, sodium and potassium salts.

A prodrug can include the incorporation of additional nucleosides at one or both ends of an antisense compound which are cleaved by endogenous nucleases within the body, to form the active anti sense compound.

Conjugated Antisense Compounds

Antisense compounds may be covalently linked to one or more moieties or conjugates which enhance the activity, cellular distribution, increased selectivity for an allelic variant, or cellular uptake of the resulting antisense oligonucleotides. Typical conjugate groups include cholesterol moieties and lipid moieties. Additional conjugate groups include carbohydrates, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes.

Antisense compounds can also be modified to have one or more stabilizing groups that are generally attached to one or both termini of antisense compounds to enhance properties such as, for example, nuclease stability. Included in stabilizing groups are cap structures. These terminal modifications protect the antisense compound having terminal nucleic acid from exonuclease degradation, and can help in delivery and/or localization within a cell. The cap can be present at the 5′-terminus (5′-cap), or at the 3′-terminus (3′-cap), or can be present on both termini. Cap structures are well known in the art and include, for example, inverted deoxy abasic caps. Further 3′ and 5′-stabilizing groups that can be used to cap one or both ends of an antisense compound to impart nuclease stability include those disclosed in WO 03/004602 published on Jan. 16, 2003.

Cell Culture and Antisense Compounds Treatment

The effects of antisense compounds on the level, activity or expression target nucleic acids can be tested in vitro in a variety of cell types. Cell types used for such analyses are available from commercial vendors (e.g. American Type Culture Collection, Manassas, Va.; Zen-Bio, Inc., Research Triangle Park, N.C.; Clonetics Corporation, Walkersville, Md.) and are cultured according to the vendor's instructions using commercially available reagents (e.g. Invitrogen Life Technologies, Carlsbad, Calif.). Illustrative cell types include, but are not limited to, HepG2 cells, Hep3B cells, and primary hepatocytes. Illustrative cell lines include GM04281, GM02171, and GM02173B cells.

In Vitro Testing of Antisense Oligonucleotides

Described herein are methods for treatment of cells with antisense oligonucleotides, which can be modified appropriately for treatment with other antisense compounds.

In general, cells are treated with antisense oligonucleotides when the cells reach approximately 60-80% confluency in culture.

One reagent commonly used to introduce antisense oligonucleotides into cultured cells includes the cationic lipid transfection reagent LIPOFECTIN (Invitrogen, Carlsbad, Calif.). Antisense oligonucleotides are mixed with LIPOFECTIN in OPTI-MEM 1 (Invitrogen, Carlsbad, Calif.) to achieve the desired final concentration of antisense oligonucleotide and a LIPOFECTIN concentration that typically ranges 2 to 12 ug/mL per 100 nM antisense oligonucleotide.

Another reagent used to introduce antisense oligonucleotides into cultured cells includes LIPOFECTAMINE (Invitrogen, Carlsbad, Calif.). Antisense oligonucleotide is mixed with LIPOFECTAMINE in OPTI-MEM 1 reduced serum medium (Invitrogen, Carlsbad, Calif.) to achieve the desired concentration of antisense oligonucleotide and a LIPOFECTAMINE concentration that typically ranges 2 to 12 ug/mL per 100 nM antisense oligonucleotide.

Another technique used to introduce antisense oligonucleotides into cultured cells includes electroporation.

Cells are treated with antisense oligonucleotides by routine methods. Cells are typically harvested 16-24 hours after antisense oligonucleotide treatment, at which time RNA or protein levels of target nucleic acids are measured by methods known in the art and described herein. In general, when treatments are performed in multiple replicates, the data are presented as the average of the replicate treatments.

The concentration of antisense oligonucleotide used varies from cell line to cell line. Methods to determine the optimal antisense oligonucleotide concentration for a particular cell line are well known in the art. Antisense oligonucleotides are typically used at concentrations ranging from 1 nM to 300 nM when transfected with LIPOFECTAMINE. Antisense oligonucleotides are used at higher concentrations ranging from 625 to 20,000 nM when transfected using electroporation.

RNA Isolation

RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA. Methods of RNA isolation are well known in the art. RNA is prepared using methods well known in the art, for example, using the TRIZOL Reagent (Invitrogen, Carlsbad, Calif.) according to the manufacturer's recommended protocols.

Analysis of Inhibition of Target Levels or Expression

Reduction, inhibition, or expression of a target nucleic acid can be assayed in a variety of ways known in the art. For example, target nucleic acid levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or quantitative real-time PCR. RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA. Methods of RNA isolation are well known in the art. Northern blot analysis is also routine in the art. Quantitative real-time PCR can be conveniently accomplished using the commercially available ABI PRISM 7600, 7700, or 7900 Sequence Detection System, available from PE-Applied Biosystems, Foster City, Calif. and used according to manufacturer's instructions.

Quantitative Real-Time PCR Analysis of Target RNA Levels

Quantitation of target RNA levels may be accomplished by quantitative real-time PCR using the ABI PRISM 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, Calif.) according to manufacturer's instructions. Methods of quantitative real-time PCR are well known in the art.

Prior to real-time PCR, the isolated RNA is subjected to a reverse transcriptase (RT) reaction, which produces complementary DNA (cDNA) that is then used as the substrate for the real-time PCR amplification. The RT and real-time PCR reactions are performed sequentially in the same sample well. RT and real-time PCR reagents are obtained from Invitrogen (Carlsbad, Calif.). RT real-time-PCR reactions are carried out by methods well known to those skilled in the art.

Gene (or RNA) target quantities obtained by real time PCR are normalized using either the expression level of a gene whose expression is constant, such as cyclophilin A, or by quantifying total RNA using RIBOGREEN (Invitrogen, Inc. Carlsbad, Calif.). Cyclophilin A expression is quantified by real time PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA is quantified using RIBOGREEN RNA quantification reagent (Invetrogen, Inc. Eugene, Oreg.). Methods of RNA quantification by RIBOGREEN are taught in Jones, L. J., et al, (Analytical Biochemistry, 1998, 265, 368-374). A CYTOFLUOR 4000 instrument (PE Applied Biosystems) is used to measure RIBOGREEN fluorescence.

Probes and primers are designed to hybridize to target nucleic acids. Methods for designing real-time PCR probes and primers are well known in the art, and may include the use of software such as PRIMER EXPRESS Software (Applied Biosystems, Foster City, Calif.).

Analysis of Protein Levels

Reduction, inhibition, or expression of target nucleic acids can be assessed by measuring target protein levels. Target protein levels can be evaluated or quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), enzyme-linked immunosorbent assay (ELISA), quantitative protein assays, protein activity assays (for example, caspase activity assays), immunohistochemistry, immunocytochemistry or fluorescence-activated cell sorting (FACS). Antibodies directed to a target can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, Mich.), or can be prepared via conventional monoclonal or polyclonal antibody generation methods well known in the art. Antibodies useful for the detection of mouse, rat, monkey, and human proteins are commercially available.

In Vivo Testing of Antisense Compounds

Antisense compounds, for example, antisense oligonucleotides, are tested in animals to assess their ability to selectively reduce or inhibit expression of target gene product and produce phenotypic changes, such as, amelioration of a disease symptom. Testing may be performed in normal animals, or in experimental disease models. For administration to animals, antisense oligonucleotides are formulated in a pharmaceutically acceptable diluent, such as phosphate-buffered saline. Administration includes parenteral routes of administration, such as intraperitoneal, intravenous, and subcutaneous. Calculation of antisense oligonucleotide dosage and dosing frequency is within the abilities of those skilled in the art, and depends upon factors such as route of administration and animal body weight. Following a period of treatment with antisense oligonucleotides, RNA or protein is isolated from tissue and changes in target nucleic acid or protein expression are measured.

Administration

In certain embodiments, the compounds and compositions described herein may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic, vaginal, rectal, intranasal), oral, pulmonary (including by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal) or parenteral, for example, by intravenous drip, intravenous injection or subcutaneous, intraperitoneal, intraocular, intravitreal, or intramuscular injection.

In certain embodiments, the compounds and compositions as described herein are administered parenterally.

In certain embodiments, parenteral administration is by infusion. Infusion can be chronic or continuous or short or intermittent. In certain embodiments, infused pharmaceutical agents are delivered with a pump. In certain embodiments, parenteral administration is by injection.

In certain embodiments, compounds and compositions are delivered to the CNS. In certain embodiments, compounds and compositions are delivered to the cerebrospinal fluid. In certain embodiments, compounds and compositions are administered to the brain parenchyma. In certain embodiments, compounds and compositions are delivered to an animal by intrathecal administration, or intracerebroventricular administration. Broad distribution of compounds and compositions, described herein, within the central nervous system may be achieved with intraparenchymal administration, intrathecal administration, or intracerebroventricular administration.

In certain embodiments, parenteral administration is by injection. The injection may be delivered with a syringe or a pump. In certain embodiments, the injection is a bolus injection. In certain embodiments, the injection is administered directly to a tissue, such as striatum, caudate, cortex, hippocampus and cerebellum.

In certain embodiments, methods of specifically localizing a pharmaceutical agent, such as by bolus injection, decreases median effective concentration (EC50) by a factor of 20, 25, 30, 35, 40, 45 or 50. In certain embodiments, the pharmaceutical agent in an antisense compound as further described herein. In certain embodiments, the targeted tissue is brain tissue. In certain embodiments the targeted tissue is striatal tissue. In certain embodiments, decreasing EC50 is desirable because it reduces the dose required to achieve a pharmacological result in a patient in need thereof.

In certain embodiments, an antisense oligonucleotide is delivered by injection or infusion once every month, every two months, every 90 days, every 3 months, every 6 months, twice a year or once a year.

Certain Compounds and Indications

Provided herein are compounds and methods that provide potent inhibition and increased selectivity for a mutant allele. Potency is demonstrated by the percent inhibition of mutant mRNA achieved by the antisense oligonucleotides targeting a SNP compared to the percent inhibition of mutant mRNA achieved by the benchmark oligonucleotide. Selectivity is demonstrated by the ability of the antisense oligonucleotide targeting a SNP to inhibit expression of the major allele or mutant allele preferentially compared to the minor allele or wild type allele. The usage of three cell lines with different genotypes at each SNP position have facilitated the determination of design rules that provide for potent and selective SNP targeting antisense oligonucleotides.

In certain embodiments, the compounds are antisense oligonucleotides as further described herein. The antisense oligonucleotides preferentially target a SNP or differentiating polymorphism. Oligonucleotides of various lengths were tested and certain lengths were determined to be beneficial for the targeting of SNPs.

In certain embodiments, the antisense oligonucleotides have a sequence that is 12-30 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 12-25 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 12-21 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 12-20 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 13-20 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 14-20 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 15-20 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 12-19 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 13-19 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 14-19 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 15-19, nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 16-19 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 17-19 nucleobases in length. In certain embodiments, the antisense oligonucleotides have a sequence that is 12, 13, 14, 15, 16, 17, 18, 19, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30 nucleobases in length.

For oligonucleotides of various lengths, the position of the nucleoside complementary to the SNP position was shifted within the gap and the wings and the effect was tested. Certain positions within the antisense oligonucleotide are shown to be beneficial for targeting SNPs.

In certain embodiments, the antisense oligonucleotide is at least 12, at least 13, at least 14, at least 15, at least 16, at least 17 at least 18 or at least 19 nucleobases in length and the SNP is complementary to positions 6-15 counting from the 5′ terminus of the antisense oligonucleotide and/or positions 1-9 counting from the 5′ end of the gap. In certain embodiments, the antisense oligonucleotide is at least 12, at least 13, at least 14, at least 15, at least 16, at least 17 at least 18 or at least 19 nucleobases in length and the SNP is complementary to positions 8-14 counting from the 5′ terminus of the antisense oligonucleotide and/or positions 1-9 counting from the 5′ end of the gap. In certain embodiments, the antisense oligonucleotide is at least 12, at least 13, at least 14, at least 15, at least 16, at least 17 at least 18 or at least 19 nucleobases in length and the SNP is complementary to positions 8-14 counting from the 5′ terminus of the antisense oligonucleotide and/or positions 4-7 counting from the 5′ end of the gap. In certain embodiments, the antisense oligonucleotide is at least 12, at least 13, at least 14, at least 15, at least 16, at least 17 at least 18 or at least 19 nucleobases in length and the SNP is complementary to positions 8-10 counting from the 5′ terminus of the antisense oligonucleotide and/or positions 4-6 counting from the 5′ end of the gap.

In certain embodiments, the SNP is complementary to position 8, 9, or 10 counting from the 5′ terminus of the oligonucleotide or position 4, 5, or 6, counting from the 5′ end of the gap. For oligonucleotides of various lengths, the effect of the length of the gap, 5′ wing, and 3′ wing was tested.

Certain wing-gap-wing combinations were shown to be beneficial for a SNP targeting antisense oligonucleotide. In certain embodiments the gap is 7-11 nucleobases in length and each wing is independently 1-6 nucleobases in length. In certain embodiments the gap is 7-11 nucleobases in length and each wing is independently 2-6 nucleobases in length. In certain embodiments the gap is 8-11 nucleobases in length and each wing is independently 2-6 nucleobases in length. In certain embodiments the gap is 9-11 nucleobases in length and each wing is independently 2-6 nucleobases in length. In certain embodiments the gap is 9 nucleobases in length and each wing is independently 2-6 nucleobases in length. In certain embodiments the gap is 10 nucleobases in length and each wing is independently 2-6 or 4-5 nucleobases in length. In certain embodiments the gap is 11 nucleobases in length and each wing is independently 2-6, or 4-5 nucleobases in length. In certain embodiments, the wing-gap-wing configuration is one of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5, 5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4, 4-10-5 and 5-10-4.

For oligonucleotides of various lengths, the effect of certain chemistries was tested. Certain chemistry modifications were shown to be beneficial for a SNP targeting antisense oligonucleotide. In certain embodiments, each nucleoside of each wing of the modified antisense oligonucleotide has a 2′-MOE modification. In certain embodiments, each nucleoside of each wing of the modified antisense oligonucleotide has a high affinity modification. In certain embodiments, the antisense oligonucleotide is a mixed wing gapmer. In such embodiment, the modifications and combination of modifications at the 3′ wing and at the 5′ wing may be the same or they may be different. In certain embodiments, the antisense oligonucleotide has one or more 2′-MOE modifications in the wings and/or one or more high affinity modifications in the wings. In certain embodiments, the high affinity modification is a cEt modification. In certain embodiments, the antisense oligonucleotide has a high affinity modification at positions 2, 3, 13, and 14 of the antisense oligonucleotide (counting from the 5′ terminus). In certain embodiments, the antisense oligonucleotide has one, two, three, or four high affinity modifications in at least one of the wings.

In certain embodiments, the antisense oligonucleotide has one, two, three, or four high affinity modifications in each of the 5′ and 3′ wings independently. In certain embodiments, the antisense oligonucleotide has a high affinity modification at positions 2 and 3 in one or both of the 5′ and 3′ wings (counting from the 5′ terminus of the 5′ wing and the 3′ terminus of the 3′wing). In certain embodiments, the antisense oligonucleotide has a high affinity modification at positions 2, 3 and 4 in one or both of the 5′ and 3′ wings (counting from the 5′ terminus of the 5′ wing and the 3′ terminus of the 3′wing,). In certain embodiments, the antisense oligonucleotide has a high affinity modification at positions 1 of the 5′ and/or 3′ wings (counting from the 5′ terminus of the 5′ wing and the 3′ terminus of the 3′wing,). In certain embodiments, the antisense oligonucleotide has a high affinity modification at positions 1 of the 5′ and 3′ wings (counting from the 5′ terminus of the 5′ wing and the 3′ terminus of the 3′wing,) and at least one other position in the wing. In certain embodiments, the antisense oligonucleotide has alternating 2′-MOE and high affinity modification in at least one of the 5′ and 3′wings.

In certain embodiments, the compound comprises an antisense oligonucleotide incorporating one or more of the design rules provided above.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 12 to 30 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 6-15 beginning from the 5′ terminus of the antisense oligonucleotide or positions 1-9 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments the single nucleotide polymorphism site contains a differentiating polymorphism. In certain embodiments, the single nucleotide polymorphism site is on a mutant allele. In certain embodiments, the mutant allele is associated with disease. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 12 to 20 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 6-15 beginning from the 5′ terminus of the antisense oligonucleotide or positions 1-9 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 12 to 20 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 8-14 beginning from the 5′ terminus of the antisense oligonucleotide or positions 1-9 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 12 to 20 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 8-14 beginning from the 5′ terminus of the antisense oligonucleotide or positions 4-7 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 12 to 20 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 8-10 beginning from the 5′ terminus of the antisense oligonucleotide or positions 4-6 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 12 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 8-10 beginning from the 5′ terminus of the antisense oligonucleotide or positions 4-6 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 13 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 8-10 beginning from the 5′ terminus of the antisense oligonucleotide or positions 4-6 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 14 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 8-10 beginning from the 5′ terminus of the antisense oligonucleotide or positions 4-6 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 6-15 beginning from the 5′ terminus of the antisense oligonucleotide or positions 1-9 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein the single nucleotide polymorphism aligns with any one of positions 8-10 beginning from the 5′ terminus of the antisense oligonucleotide or positions 4-6 beginning from the 5′ end of the gap of the modified antisense oligonucleotide; and wherein each nucleoside of each wing has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 4-7-4, 5-8-6, 6-8-5, 6-7-6, 5-7-5. 6-8-5, 5-8-6, 3-9-4, 4-9-3, 2-9-6, 6,9,2,3-9-3, 3-9-5,5-9-3, 5-9-4, 4-9-5, 5-9-5, 4-11-4,4-10-5 and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 6, 8, 9, 10, 11, or 14 beginning from the 5′ terminus of the modified antisense oligonucleotide aligns with the single nucleotide polymorphism; and wherein each nucleoside of each wing segment modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 1, 4, 5, 6, 7, or 9 of the gap segment aligns with the single nucleotide polymorphism; and wherein each nucleoside of each wing segment has a modified sugar or sugar surrogate. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 6, 7, 8, 9, 10, 11, or 12 of the modified antisense oligonucleotide aligns with the single nucleotide polymorphism; and positions 2 and 3 of the 5′ and 3′ wing segments comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides and fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 3, 4, 5, 6, 7, 8 or 9 of the gap segment aligns with the single nucleotide polymorphism; and positions 2 and 3 of the 5′ and 3′ wing segments comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

A compound comprising a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides and fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 6, 7, 8, 9, 10, 11, or 12 of the modified antisense oligonucleotide aligns with the single nucleotide polymorphism; and positions 2, 3, 13, and 14 of the antisense oligonucleotide comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

A compound comprising a modified antisense oligonucleotide consisting of 15 to 19 linked nucleosides and fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 3, 4, 5, 6, 7, 8, or 9 of the gap segment aligns with the single nucleotide polymorphism; and positions 2, 3, 13, and 14 of the antisense antisense oligonucleotide comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In certain embodiments, the compound comprise a modified antisense oligonucleotide consisting of 17 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 8, 9, or 10 of the modified antisense oligonucleotide aligns with the single nucleotide polymorphism; and wherein each nucleoside of each wing segment comprises a 2′-O-methoxyethyl sugar. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 17 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 5, 6, or 7 of the gap segment aligns with the single nucleotide polymorphism; and wherein each nucleoside of each wing segment comprises a 2′-O-methoxyethyl sugar. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 17 to 19 linked nucleosides, fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 8, 9, or 10 of the modified antisense oligonucleotide aligns with the single nucleotide polymorphism; and positions 2 and 3 of the 5′ and 3′ wing segments comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In certain embodiments, the compound comprises a modified antisense oligonucleotide consisting of 17 to 19 linked nucleosides and fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 5, 6, or 7 of the gap segment aligns with the single nucleotide polymorphism; and positions 2 and 3 of the 5′ and 3′ wing segments comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

A compound comprising a modified antisense oligonucleotide consisting of 17 to 19 linked nucleosides and fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 8, 9, or 10 of the modified oligonucleotide aligns with the single nucleotide polymorphism; and positions 2, 3, 13, and 14 of the antisense antisense oligonucleotide comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

A compound comprising a modified antisense oligonucleotide consisting of 17 to 19 linked nucleosides and fully complementary to a single nucleotide polymorphism site, wherein the modified antisense oligonucleotide comprises a wing-gap-wing motif, wherein position 5, 6, or 7 of the gap segment aligns with the single nucleotide polymorphism; and positions 2, 3, 13, and 14 of the antisense oligonucleotide comprise a 4′-CH(CH3)—O-2′ bridge. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 2-9-6, 3-9-3, 3-9-5, 4-9-5, 4-11-4, and 5-10-4.

In a certain embodiment, the antisense oligonucleotide is 11 to 20 linked nucleosides in length and has, independently, 2 to 5 linked nucleosides in the 5′ and 3′ wings and 7 to 11 linked nucleosides in the gap. The SNP is complementary to position 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 of the antisense oligonucleotide (counting from the 5′ terminus of the antisense oligonucleotide) or position 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 counting from the 5′ terminus of the gap segment.

In a certain embodiment, the antisense oligonucleotide is 15 to 19 linked nucleosides in length and has, independently, 2 to 5 linked nucleosides in the 5′ and 3′ wings and 7 to 11 linked nucleosides in the gap. The SNP is complementary to position 6, 7, 8, 9, or 10 of the antisense oligonucleotide (counting from the 5′ terminus of the antisense oligonucleotide) or position 4, 5, 6, or 7 counting from the 5′ terminus of the gap segment.

In a certain embodiment, the antisense oligonucleotide is 17 linked nucleosides in length and has, independently, 2 to 5 linked nucleosides in the 5′ and 3′ wing segments and 9 to 11 linked nucleosides in the gap segment. The SNP is complementary to position 8, 9, or 10 of the antisense oligonucleotide (counting from the 5′ terminus of the antisense oligonucleotide) or position 5, 6, or 7 (counting from the 5′ terminus of the gap segment).

In a certain embodiment, the antisense oligonucleotide is 18 linked nucleosides in length and has, independently, 2 to 5 linked nucleosides in the 5′ and 3′ wing segments and 9 to 11 linked nucleosides in the gap segment. The SNP is complementary to position 8, 9, or 10 of the antisense oligonucleotide (counting from the 5′ terminus of the antisense oligonucleotide) or position 5, 6, or 7 (counting from the 5′ terminus of the gap segment).

In a certain embodiment, the antisense oligonucleotide is 19 linked nucleosides in length and has, independently, 2 to 5 linked nucleosides in the 5′ and 3′ wing segments and 9 to 11 linked nucleosides in the gap segment. The SNP is complementary to position 8, 9, or 10 of the antisense oligonucleotide (counting from the 5′ terminus of the antisense oligonucleotide) or position 5, 6, or 7 (counting from the 5′ terminus of the gap segment).

In certain, the antisense oligonucleotide is any of those listed in the examples. In certain embodiments, they are designated by sequence and chemistry as reflected by the ISIS number. In certain embodiments, the antisense oligonucleotide is any of those listed in the examples as having at least, about, greater than about or greater than 50%, 55%, 60%, 65%, 70%, 75%, 80%, 85%, 90% or 95% inhibition of a mutant Huntingtin allele.

In certain embodiments, the antisense oligonucleotide is any of those listed in the examples as selectively inhibiting the expression of the mutant huntingtin allele at least, about, greater than about or greater than 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold or 10 fold over the expression of a wild-type huntingtin allele or by at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80% compared to expression of a wild-type huntingtin allele.

In certain embodiments, the invention provides methods of treating an individual comprising administering one or more pharmaceutical compositions described herein. In certain embodiments, the individual has an allelic variant associated with a disease or disorder. The pharmaceutical compositions provided herein preferentially target a SNP. In certain embodiments, the SNP is a differentiating polymorphism.

Methods have been described for determining whether a SNP is specific to a disease associated allele and more specifically whether a SNP variant of an allele of a heterozygous patient is on the same allele as a disease-causing mutation that is at a remote region of the gene's mRNA (WO 2008/147930 and WO 2008/143774).

In certain embodiments, the disease is a neurodegenerative disorder. In certain embodiments, the neurodegenerative disorder is Huntington's Disease. In certain embodiments, the targeted SNP is one or more of: rs6446723, rs3856973, rs2285086, rs363092, rs916171, rs6844859, rs7691627, rs4690073, rs2024115, rs11731237, rs362296, rs10015979, rs7659144, rs363096, rs362273, rs16843804, rs362271, rs362275, rs3121419, rs362272, rs3775061, rs34315806, rs363099, rs2298967, rs363088, rs363064, rs363102, rs2798235, rs363080, rs363072, rs363125, rs362303, rs362310, rs10488840, rs362325, rs35892913, rs363102, rs363096, rs11731237, rs10015979, rs363080, rs2798235, rs1936032, rs2276881, rs363070, rs35892913, rs12502045, rs6446723, rs7685686, rs3733217, rs6844859, rs362331, rs1143646, rs2285086, rs2298969, rs4690072, rs916171, rs3025849, rs7691627, rs4690073, rs3856973, rs363092, rs362310, rs362325, rs363144, rs362303, rs34315806, rs363099, rs363081, rs3775061, rs2024115, rs10488840, rs363125, rs362296, rs2298967, rs363088, rs363064, rs362275, rs3121419, rs3025849, rs363070, rs362273, rs362272, rs362306, rs362271, rs363072, rs16843804, rs7659144, rs363120, and rs12502045. In certain embodiments the compounds are ISIS 460065, ISIS 459978, ISIS 460028, ISIS 460209, ISIS 460208, and ISIS 460206.

In certain embodiments, described herein, are methods of selectively reducing the expression of a mutant huntingtin allele in a cell, tissue or animal by administering to the cell, tissue, or animal a compound as described herein. In certain embodiments, the compound comprises a modified oligonucleotide complementary to the mutant huntingtin allele. In certain embodiments, the modified oligonucleotide is complementary to the mutant huntingtin allele at a position comprising a single nucleotide polymorphism described herein. In certain embodiments, the single nucleotide polymorphism is selected from those described herein. In certain embodiments, the single nucleotide polymorphism is selected from rs362306, rs362331, rs2298969, rs7685686, rs4690072, rs2024115 or rs363088.

In certain embodiments, described herein, are methods of treating, ameliorating or slowing the onset or progression of Huntington's Disease by administering to an animal a compound described herein. In certain embodiment the compound comprises a modified oligonucleotide complementary to a mutant huntingtin allele at a position on the allele comprising a single nucleotide polymorphism described herein. In certain embodiments, the single nucleotide polymorphism is selected from rs362306, rs362331, rs2298969, rs7685686, rs4690072, rs2024115 or rs363088. In certain embodiments, the compound administered to the animal treats, ameliorates or slows the onset or progression Huntington's Disease by selectively reducing the mutant huntingtin allele.

In certain embodiments the expression of the mutant huntingtin allele is selectively reduced at least, about or greater than 2 fold, 3 fold, 4 fold, 5 fold, 6 fold, 7 fold, 8 fold, 9 fold or 10 fold over the expression of a wild-type huntingtin allele or by at least 40%, 45%, 50%, 55%, 60%, 65%, 70%, 75%, 80% compared to expression of a wild-type huntingtin allele.

In certain embodiments, one or more additional compounds comprising a modified oligonucleotide complementary to a mutant huntingtin allele at a position comprising a single nucleotide polymorphism site selected from rs362306, rs362331, rs2298969, rs7685686, rs4690072, rs2024115 or rs363088 is administered.

In certain embodiments, the modified oligonucleotide has a specific length, gap, wing, chemistry and SNP position alignment as further described herein. In certain embodiments, the modified oligonucleotide consists of 15 to 20 linked nucleosides and position 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 of the modified oligonucleotide, as counted from the 5′ terminus of the modified oligonucleotide, aligns with the single nucleotide polymorphism.

In certain embodiments, the modified oligonucleotide is 90% complementary to the mutant huntingtin allele.

The method of claim 40, wherein the modified oligonucleotide is 95% complementary to the mutant huntingtin allele.

The method of claim 40, wherein the modified oligonucleotide is 100% complementary to the mutant huntingtin allele.

In certain embodiments, described herein are methods for determining whether an agent selectively inhibits expression of a first allelic variant of a gene relative to a second allelic variant of the gene wherein the first allelic variant comprises a first nucleotide at a SNP position and the second allelic variant comprises a second nucleotide at the SNP position, comprising:

  • (i) contacting a first cell, tissue or animal with the agent at one or more concentrations, wherein the first cell, tissue or animal is homozygous for the first nucleotide at the SNP position;
  • (ii) contacting a second cell, tissue or animal with the agent at one or more concentrations, wherein the second cell line is homozygous for the second nucleotide at the SNP position or is heterozygous for the first and the second nucleotides at the SNP position;
  • (iii) measuring inhibition of expression of the allelic variant in each of the cell, tissue or animal for each of the one or more concentrations of the agent; and
  • (iv) comparing the inhibition of expression in each of the cell, tissue or animal for each of the one or more concentrations of the agent to determine whether the agent selectively inhibits expression of the first allelic variant of the gene relative to the second allelic variant of the gene.

In certain embodiments, the methods described herein further comprise the step of contacting a third cell, tissue or animal with the agent at one or more concentrations, wherein the third cell, tissue or animal is homozygous for the second nucleotide at the SNP position or is heterozygous for the first and the second nucleotides at the SNP position and has a different genotype than the first cell, tissue or animal and the second cell, tissue or animal.

In certain embodiments, the cell, tissue or animal is a a YAC18 cell, tissue or mouse, a BACHD cell, tissue or mouse, a cell line derived from a human Huntington's Disease patient or any combination thereof.

In certain embodiments, the first allelic variant is a mutant allele and the second allelic variant is a wild-type allele. In certain embodiments, the first allelic variant is a wild-type allele and the second allelic variant is a mutant allele.

In certain embodiments, the mutant allelic variant is associated with disease. In certain embodiments, the disease is the result of a gain of toxic function. In certain embodiments, the disease is Huntington's Disease.

In certain embodiments, the first nucleotide is in linkage disequilibrium with a disease associated mutation. In certain embodiments, the second nucleotide is in linkage disequilibrium with a disease associated mutation. In certain embodiments, the disease associated mutation is a tri-nucleotide repeat expansion. In certain embodiments, the tri-nucleotide repeat expansion is a CAG expansion. In certain embodiments, the CAG expansion is in a HTT gene.

In certain embodiments, the first nucleotide is A and the second nucleotide is any of C, G, or T. In certain embodiments, the first nucleotide is C and the second nucleotide is any of A, G, or T. In certain embodiments, the first nucleotide is G and the second nucleotide is any of A, C, or T. In certain embodiments, the first nucleotide is T and the second nucleotide is any of A, C, or G. In certain embodiments, the first nucleotide is in linkage disequilibrium with a disease associated mutation. In certain embodiments, the disease associated mutation is a CAG expansion. In certain embodiments, the CAG expansion is in a HTT gene.

In certain embodiments, the agent is an antisense compound. In certain embodiments, the antisense compound is an antisense oligonucleotide. In certain embodiments, the antisense oligonucleotide is a gapmer. In certain embodiments, the gapmer has a wing-gap-wing motif. In certain embodiments, the wing-gap-wing motif is any one of the group consisting of 5-10-5, 2-9-6, 3-9-3, 3-9-4, 3-9-5, 4-7-4, 4-9-3, 4-9-4, 4-9-5, 4-10-5, 4-11-4, 4-11-5, 5-7-5, 5-8-6, 5-9-3, 5-9-5, 5-10-4, 5-10-5, 6-7-6, 6-8-5, and 6-9-2. In certain embodiments, at least one internucleoside linkage is a modified internucleoside linkage. In certain embodiments, each internucleoside linkage is a phosphorothioate internucleoside linkage. In certain embodiments, at least one nucleoside comprises a modified nucleobase. In certain embodiments, the modified nucleobase is a 5′-methylcytosine. In certain embodiments, at least one nucleoside of at least one of the wing regions comprises a modified sugar or sugar surrogate. In certain embodiments, each of the nucleosides of each wing region comprises a modified sugar or sugar surrogate. In certain embodiments, the sugar or sugar surrogate is a 2′-O-methoxyethyl modified sugar. In certain embodiments, at least one of the wing regions comprises a 4′ to 2′ bicyclic nucleoside and at least one of the remaining wing nucleosides is a non-bicyclic 2′-modified nucleoside. In certain embodiments, the non-bicyclic 2′-modified nucleoside is a 2′-O-methoxyethyl nucleoside. In certain embodiments, the 4′ to 2′ bicyclic nucleoside is 4′-CH(CH3)—O-2′ bicyclic nucleoside.

In certain embodiments, the first cell line is any of the group consisting of fibroblast cell line, neuronal cell line, or lymphoblast cell line. In certain embodiments, the second cell line is any of the group consisting of fibroblast cell line, neuronal cell line, or lymphoblast cell line. In certain embodiments, the third cell line is any of the group consisting of fibroblast cell line, neuronal cell line, or lymphoblast cell line. In certain embodiments, the fibroblast cell line is any of the group consisting of GM04281, GM02171, and GM02173B. In certain embodiments, the neuronal cell line comprises a cell line derived from a YAC18 mouse, a cell line derived from a BACHD mouse, or a cell line derived from a human Huntington's Disease patient.

In certain embodiments, the SNP position is any of the group consisting of rs6446723, rs3856973, rs2285086, rs363092, rs916171, rs6844859, rs7691627, rs4690073, rs2024115, rs11731237, rs362296, rs10015979, rs7659144, rs363096, rs362273, rs16843804, rs362271, rs362275, rs3121419, rs362272, rs3775061, rs34315806, rs363099, rs2298967, rs363088, rs363064, rs363102, rs2798235, rs363080, rs363072, rs363125, rs362303, rs362310, rs10488840, rs362325, rs35892913, rs363102, rs363096, rs11731237, rs10015979, rs363080, rs2798235, rs1936032, rs2276881, rs363070, rs35892913, rs12502045, rs6446723, rs7685686, rs3733217, rs6844859, rs362331, rs1143646, rs2285086, rs2298969, rs4690072, rs916171, rs3025849, rs7691627, rs4690073, rs3856973, rs363092, rs362310, rs362325, rs363144, rs362303, rs34315806, rs363099, rs363081, rs3775061, rs2024115, rs10488840, rs363125, rs362296, rs2298967, rs363088, rs363064, rs362275, rs3121419, rs3025849, rs363070, rs362273, rs362272, rs362306, rs362271, rs363072, rs16843804, rs7659144, rs363120, and rs12502045.

In certain embodiments, the one or more concentration is any of the group consisting of two concentrations, three concentrations, four concentrations, and five concentrations.

In certain embodiments, selectivity is determined by increased inhibition of the allelic variant in one cell line as compared to another cell line.

In certain embodiments, described herein are methods for determining whether an agent selectively inhibits expression of a first allelic variant of a gene relative to a second allelic variant of the gene wherein the first allelic variant comprises a first nucleotide at a SNP position and the second allelic variant comprises a second nucleotide at the SNP position, comprising:

  • (i) contacting a first cell line with the agent at one or more concentrations, wherein the first cell line is homozygous for the first nucleotide at the SNP position;
  • (ii) contacting a second cell line with the agent at one or more concentrations, wherein the second cell line is homozygous for the second nucleotide at the SNP position or is heterozygous for the first and the second nucleotides at the SNP position;
  • (iii) measuring inhibition of expression of the allelic variant in each of the cell lines for each of the one or more concentrations of the agent; and
  • (iv) comparing the inhibition of expression in each of the cell lines for each of the one or more concentrations of the agent to determine whether the agent selectively inhibits expression of the first allelic variant of the gene relative to the second allelic variant of the gene.
  • (v) calculating IC50 values for the agent in each of the first cell line, the second cell line, and the third cell line; and
  • (vi) comparing the IC50 values to determine whether the agent selectively inhibits expression of the first allelic variant or mutant allele.

In certain embodiments, the huntingtinSNP targeting oligonucleotides provided herein are selective for the mutant allele and demonstrate a potency of within at least 10-fold, at least 9-fold, at least 8-fold, at least 7-fold, at least 6-fold, at least 5-fold, at least 4-fold, at least 3-fold or at least 2-fold of the potency of the benchmark PAN ASO, ISIS 387916. In certain embodiments, the SNP targeting oligonucleotides provided herein have an IC50 in a cell line homozygous for the mutant allele of no more than 6.5, 6, 5.5, 5, 4.5, 4, 3.5, 3, 2.5, 2, 1.5, 1 or 0.75. In certain embodiments, the SNP targeting oligonucleotides provided herein have an IC50 in a cell line that is homozygous for the wild-type allele that is at least 2 fold, 3 fold, 4 fold or 5 fold, 6 fold, 7 fold, 8 fold, 9 fold or 10 fold less than the IC50 in a cell line homozygous for the mutant allele. In certain embodiments, the SNP targeting oligonucleotides provided herein have an IC50 in a cell line that is heterozygous for the mutant and wild-type allele that is at least 1.4 fold, 1.7 fold, 2 fold, 2.2, 2.4, 2.5, 2.6, 2.8 or 3 fold less than the IC50 in a cell line homozygous for the mutant allele. In certain embodiments, the SNP targeting oligonucleotides include or are selected from ISIS Nos 435869, 460069, 460206, 460019, 460071, 460207, 460212, 460218, 460231, 474892, 460012, 460208, 435879, 460065, 460085, 460209, 474871, 474891, 474919, 474923, 476333, 476337, 435890, 460026, 460210, 463571, 476444.

Therapeutically Effective Dosages

In certain embodiments, administration of a therapeutically effective amount of an antisense compound targeted to the mutant huntingtin allele is accompanied by monitoring of expression of a gene product in an individual, to determine an individual's response to administration of the antisense compound. In certain embodiments, the gene product is huntingtin mRNA or protein. An individual's response to administration of the antisense compound is used by a physician to determine the amount and duration of therapeutic intervention.

In certain embodiments, administration of an antisense compound targeted to a mutant nucleic acid results in reduction of mRNA or protein expression by at least 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95 or 99%, or a range defined by any two of these values. In certain embodiments, the mutant nucleic acid is huntingtin nucleic acid, the mRNA is huntingtin mRNA, and the protein is huntingtin protein.

In certain embodiments, pharmaceutical compositions comprising an antisense compound targeted to a mutant allele are used for the preparation of a medicament for treating a patient suffering or susceptible to Huntington's Disease.

Certain Combination Therapies

In certain embodiments, one or more pharmaceutical compositions of the present invention are co-administered with one or more other pharmaceutical agents. In certain embodiments, such one or more other pharmaceutical agents are designed to treat the same disease, disorder, or condition as the one or more pharmaceutical compositions of the present invention. In certain embodiments, such one or more other pharmaceutical agents are designed to treat a different disease, disorder, or condition as the one or more pharmaceutical compositions of the present invention. In certain embodiments, such one or more other pharmaceutical agents are designed to treat an undesired side effect of one or more pharmaceutical compositions of the present invention. In certain embodiments, one or more pharmaceutical compositions of the present invention are co-administered with another pharmaceutical agent to treat an undesired effect of that other pharmaceutical agent. In certain embodiments, one or more pharmaceutical compositions of the present invention are co-administered with another pharmaceutical agent to produce a combinational effect. In certain embodiments, one or more pharmaceutical compositions of the present invention are co-administered with another pharmaceutical agent to produce a synergistic effect.

In certain embodiments, one or more pharmaceutical compositions of the present invention and one or more other pharmaceutical agents are administered at the same time. In certain embodiments, one or more pharmaceutical compositions of the present invention and one or more other pharmaceutical agents are administered at different times. In certain embodiments, one or more pharmaceutical compositions of the present invention and one or more other pharmaceutical agents are prepared together in a single formulation. In certain embodiments, one or more pharmaceutical compositions of the present invention and one or more other pharmaceutical agents are prepared separately.

EXAMPLES Non-Limiting Disclosure and Incorporation by Reference

While certain compounds, compositions and methods described herein have been described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds described herein and are not intended to limit the same. Each of the patents, applications, printed publications, and other published documents mentioned or referred to in this specification are herein incorporated by reference in their entirety.

Example 1: Single Nucleotide Polymorphisms (SNPs) in the Huntingtin (HTT) Gene Sequence

The HTT genomic sequence, designated herein as SEQ ID NO: 1 (NT_006081.18 truncated from nucleotides 1566000 to 1768000) was aligned with the HTT mRNA, designated herein as SEQ ID NO: 2 (NM_002111.6), using the EMBL-EBI sequence database (ClustalW2, http://www.ebi.ac.uk/Tools/clustalw2/index.html), and the output is presented in FIG. 1. SNP positions (identified by Hayden et al, WO/2009/135322) associated with the HTT gene were mapped to the two sequences and have been demarcated in FIG. 1 by their reference SNP ID number from the Entrez SNP database at the National Center for Biotechnology Information (NCBI, http://www.ncbi.nlm.nih.gov/sites/entrez?db=snp), incorporated herein by reference. Table 2 furnishes further details on each SNP. The ‘Reference SNP ID number’ or ‘RS number’ is the number designated to each SNP from the Entrez SNP database at NCBI, incorporated herein by reference. ‘SNP position’ refers to the nucleotide position of the SNP on SEQ ID NO: 1. ‘Polymorphism’ indicates the nucleotide variants at that SNP position. ‘Major allele’ indicates the nucleotide associated with the major allele, or the nucleotide present in a statistically significant proportion of individuals in the human population. ‘Minor allele’ indicates the nucleotide associated with the minor allele, or the nucleotide present in a relatively small proportion of individuals in the human population.

TABLE 2 Single Nuclear Polymorphisms (SNPs) and their positions on SEQ ID NO: 1 SNP Major Minor RS No. position Polymorphism allele allele rs2857936 1963 C/T C T rs12506200 3707 A/G G A rs762855 14449 A/G G A rs3856973 19826 G/A G A rs2285086 28912 G/A A G rs7659144 37974 C/G C G rs16843804 44043 C/T C T rs2024115 44221 G/A A G rs10015979 49095 A/G A G rs7691627 51063 A/G G A rs2798235 54485 G/A G A rs4690072 62160 G/T T G rs6446723 66466 C/T T C rs363081 73280 G/A G A rs363080 73564 T/C C T rs363075 77327 G/A G A rs363064 81063 T/C C T rs3025849 83420 A/G A G rs6855981 87929 A/G G A rs363102 88669 G/A A G rs11731237 91466 C/T C T rs4690073 99803 A/G G A rs363144 100948 T/G T G rs3025838 101099 C/T C T rs34315806 101687 A/G G A rs363099 101709 T/C C T rs363096 119674 T/C T C rs2298967 125400 C/T T C rs2298969 125897 A/G G A rs6844859 130139 C/T T C rs363092 135682 C/A C A rs7685686 146795 A/G A G rs363088 149983 A/T A T rs362331 155488 C/T T C rs916171 156468 G/C C G rs362322 161018 A/G A G rs362275 164255 T/C C T rs362273 167080 A/G A G rs2276881 171314 G/A G A rs3121419 171910 T/C C T rs362272 174633 G/A G A rs362271 175171 G/A G A rs3775061 178407 C/T C T rs362310 179429 A/G G A rs362307 181498 T/C C T rs362306 181753 G/A G A rs362303 181960 T/C C T rs362296 186660 C/A C A rs1006798 198026 A/G A G

Example 2: Design of Antisense Oligonucleotides Targeting Huntingtin Gene SNPs and Inhibition of HTT mRNA in Coriell Fibroblast Cell Lines (GM04281, GM02171, and GM02173B)

Antisense oligonucleotides targeting nucleotides overlapping SNP positions presented in Table 1 were designed and tested for potency in three huntingtin patient-derived Coriell fibroblast cell lines, GM04281, GM02171, and GM02173B (from the Coriell Institute for Medical Research). Cultured GM04281 cells or GM02171 cells or GM02173B cells at a density of 20,000 cells per well were transfected using electroporation with 10,000 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real time PCR using primer probe set RTS2617 (forward sequence CTCCGTCCGGTAGACATGCT, designated herein as SEQ ID NO: 3; reverse sequence GGAAATCAGAACCCTCAAAATGG, designated herein as SEQ ID NO: 4; probe sequence TGAGCACTGTTCAACTGTGGATATCGGGAX, designated herein as SEQ ID NO: 5). HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells.

ISIS 387916 (TCTCTATTGCACATTCCAAG, 5-10-5 MOE (SEQ ID NO: 6)) and ISIS 388816 (GCCGTAGCCTGGGACCCGCC, 5-10-5 MOE (SEQ ID NO: 7)) were included in each study as benchmark oligonucleotides against which the potency of the antisense oligonucleotides targeting nucleotides overlapping each SNP position could be compared.

The chimeric antisense oligonucleotides in Tables 3 and 4 were designed as 5-9-5 MOE gapmers. The gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both sides (in the 5′ and 3′ directions) by wings comprising five nucleotides each. Each nucleotide in the 5′ wing segment and each nucleotide in the 3′ wing segment has a 2′-MOE modification. The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine nucleobases throughout each gapmer are 5-methylcytosines.

The oligonucleotides are further described in Table 3. The percent inhibition of HTT mRNA by the antisense oligonucleotides in each cell line is shown in Table 4. ‘Target allele’ indicates whether the gapmer is targeted to the major or the minor allele at the SNP position. The number in parentheses indicates the nucleotide position in the gapmer opposite to the SNP position, starting from the 5′-terminus of the oligonucleotide. ‘Start site’ indicates the 5′-most nucleotide to which the gapmer is targeted. “Stop site” indicates the 3′-most nucleotide to which the gapmer is targeted. Each gapmer listed in Tables 3 and 4 is targeted to human HTT pre-mRNA, which is SEQ ID NO: 1.

TABLE 3 Chimeric oligonucleotides targeting SNP positions on the HTT gene SEQ ISIS SNP RS Target Start Stop ID No No. allele Sequence Site Site NO 387916 n/a n/a TCTCTATTGCACATTCCAAG 145466 145485 6 388816 n/a n/a GCCGTAGCCTGGGACCCGCC 16501 16520 7 435330 rs3856973 Major (8) TAACACTCGATTAACCCTG 19815 19833 8 435348 rs3856973 Minor (8) TAACACTTGATTAACCCTG 19815 19833 9 435294 rs3856973 Major (10) GTTAACACTCGATTAACCC 19817 19835 10 435312 rs3856973 Minor (10) GTTAACACTTGATTAACCC 19817 19835 11 435864 rs2285086 Major (10) GCTAGTTCATCCCAGTGAG 28903 28921 12 435889 rs2285086 Minor (10) GCTAGTTCACCCCAGTGAG 28903 28921 13 435878 rs7659144 Major (10) TGGAAATGGGTTTTTCCAC 37965 37983 14 435903 rs7659144 Minor (10) TGGAAATGGCTTTTTCCAC 37965 37983 15 435863 rs16843804 Major (10) TTTAACCGTGGCATGGGCA 44034 44052 16 435888 rs16843804 Minor (10) TTTAACCGTAGCATGGGCA 44034 44052 17 435331 rs2024115 Major (8) TTCAAGCTAGTAACGATGC 44210 44228 18 435349 rs2024115 Minor (8) TTCAAGCCAGTAACGATGC 44210 44228 19 435295 rs2024115 Major (10) ACTTCAAGCTAGTAACGAT 44212 44230 20 435313 rs2024115 Minor (10) ACTTCAAGCCAGTAACGAT 44212 44230 21 435862 rs10015979 Major (10) GCAGCTAGGTTAAAGAGTC 49086 49104 22 435887 rs10015979 Minor (10) GCAGCTAGGCTAAAGAGTC 49086 49104 23 435880 rs7691627 Major (10) AATAAGAAACACAATCAAA 51054 51072 24 435905 rs7691627 Minor (10) AATAAGAAATACAATCAAA 51054 51072 25 435885 rs2798235 Major (10) CAGAGGAGGCATACTGTAT 54476 54494 26 435910 rs2798235 Minor (10) CAGAGGAGGTATACTGTAT 54476 54494 27 435874 rs4690072 Major (10) CACAGTGCTACCCAACCTT 62151 62169 28 435899 rs4690072 Minor (10) CACAGTGCTCCCCAACCTT 62151 62169 29 435875 rs6446723 Major (10) TAATTTTCTAGACTTTATG 66457 66475 30 435900 rs6446723 Minor (10) TAATTTTCTGGACTTTATG 66457 66475 31 435332 rs363081 Major (8) GCTACAACGCAGGTCAAAT 73269 73287 32 435350 rs363081 Minor (8) GCTACAATGCAGGTCAAAT 73269 73287 33 435296 rs363081 Major (10) GAGCTACAACGCAGGTCAA 73271 73289 34 435314 rs363081 Minor (10) GAGCTACAATGCAGGTCAA 73271 73289 35 435886 rs363080 Major (10) AGAGAGAACGAGAAGGCTC 73555 73573 36 435911 rs363080 Minor (10) AGAGAGAACAAGAAGGCTC 73555 73573 37 435914 rs363075 Major (6) AGCCCCTCTGTGTAAGTTT 77314 77332 38 435926 rs363075 Minor (6) AGCCCTTCTGTGTAAGTTT 77314 77332 39 435916 rs363075 Major (7) GAGCCCCTCTGTGTAAGTT 77315 77333 40 435928 rs363075 Minor (7) GAGCCCTTCTGTGTAAGTT 77315 77333 41 435333 rs363075 Major (8) TGAGCCCCTCTGTGTAAGT 77316 77334 42 435351 rs363075 Minor (8) TGAGCCCTTCTGTGTAAGT 77316 77334 43 435918 rs363075 Major (9) ATGAGCCCCTCTGTGTAAG 77317 77335 44 435930 rs363075 Minor (9) ATGAGCCCTTCTGTGTAAG 77317 77335 45 435297 rs363075 Major (10) GATGAGCCCCTCTGTGTAA 77318 77336 46 435315 rs363075 Minor (10) GATGAGCCCTTCTGTGTAA 77318 77336 47 435920 rs363075 Major (11) TGATGAGCCCCTCTGTGTA 77319 77337 48 435932 rs363075 Minor (11) TGATGAGCCCTTCTGTGTA 77319 77337 49 435366 rs363075 Major (12) ATGATGAGCCCCTCTGTGT 77320 77338 50 435924 rs363075 Minor (12) ATGATGAGCCCTTCTGTGT 77320 77338 51 435922 rs363075 Major (14) TAATGATGAGCCCCTCTGT 77322 77340 52 435934 rs363075 Minor (14) TAATGATGAGCCCTTCTGT 77322 77340 53 435334 rs363064 Major (8) AGAATACGGGTAACATTTT 81052 81070 54 435352 rs363064 Minor (8) AGAATACAGGTAACATTTT 81052 81070 55 435298 rs363064 Major (10) GGAGAATACGGGTAACATT 81054 81072 56 435316 rs363064 Minor (10) GGAGAATACAGGTAACATT 81054 81072 57 435335 rs3025849 Major (8) TTAGTAATCAATTTTAATG 83409 83427 58 435353 rs3025849 Minor (8) TTAGTAACCAATTTTAATG 83409 83427 59 435299 rs3025849 Major (10) AGTTAGTAATCAATTTTAA 83411 83429 60 435317 rs3025849 Minor (10) AGTTAGTAACCAATTTTAA 83411 83429 61 435877 rs6855981 Major (10) GAAGGAATGCTTTTACTAG 87920 87938 62 435902 rs6855981 Minor (10) GAAGGAATGTTTTTACTAG 87920 87938 63 435336 rs363102 Major (8) CTAAAACTAACTTGAGAAT 88658 88676 64 435354 rs363102 Minor (8) CTAAAACCAACTTGAGAAT 88658 88676 65 435300 rs363102 Major (10) ATCTAAAACTAACTTGAGA 88660 88678 66 435318 rs363102 Minor (10) ATCTAAAACCAACTTGAGA 88660 88678 67 435884 rs11731237 Major (10) GGTGGGCAGGAAGGACTGA 91457 91475 68 435909 rs11731237 Minor (10) GGTGGGCAGAAAGGACTGA 91457 91475 69 435337 rs4690073 Major (8) CCTAAATCAATCTACAAGT 99792 99810 70 435355 rs4690073 Minor (8) CCTAAATTAATCTACAAGT 99792 99810 71 435301 rs4690073 Major (10) TCCCTAAATCAATCTACAA 99794 99812 72 435319 rs4690073 Minor (10) TCCCTAAATTAATCTACAA 99794 99812 73 435883 rs363144 Major (10) GAAAATGTGAGTGGATCTA 100939 100957 74 435908 rs363144 Minor (10) GAAAATGTGCGTGGATCTA 100939 100957 75 435338 rs3025838 Major (8) GTAAGGCGAGACTGACTAG 101088 101106 76 435356 rs3025838 Minor (8) GTAAGGCAAGACTGACTAG 101088 101106 77 435302 rs3025838 Major (10) AGGTAAGGCGAGACTGACT 101090 101108 78 435320 rs3025838 Minor (10) AGGTAAGGCAAGACTGACT 101090 101108 79 435339 rs363099 Major (8) CTGAGCGGAGAAACCCTCC 101698 101716 80 435357 rs363099 Minor (8) CTGAGCGAAGAAACCCTCC 101698 101716 81 435303 rs363099 Major (10) GGCTGAGCGGAGAAACCCT 101700 101718 82 435321 rs363099 Minor (10) GGCTGAGCGAAGAAACCCT 101700 101718 83 435367 rs363099 Major (12) AAGGCTGAGCGGAGAAACC 101702 101720 84 435340 rs363096 Major (8) TTCCCTAAAAACAAAAACA 119663 119681 85 435358 rs363096 Minor (8) TTCCCTAGAAACAAAAACA 119663 119681 86 435304 rs363096 Major (10) GATTCCCTAAAAACAAAAA 119665 119683 87 435322 rs363096 Minor (10) GATTCCCTAGAAACAAAAA 119665 119683 88 435341 rs2298967 Major (8) CTTTTCTATTGTCTGTCCC 125389 125407 89 435359 rs2298967 Minor (8) CTTTTCTGTTGTCTGTCCC 125389 125407 90 435305 rs2298967 Major (10) TGCTTTTCTATTGTCTGTC 125391 125409 91 435323 rs2298967 Minor (10) TGCTTTTCTGTTGTCTGTC 125391 125409 92 435865 rs2298969 Major (10) AAGGGATGCCGACTTGGGC 125888 125906 93 435890 rs2298969 Minor (10) AAGGGATGCTGACTTGGGC 125888 125906 94 435876 rs6844859 Major (10) ACCTTCCTCACTGAGGATG 130130 130148 95 435901 rs6844859 Minor (10) ACCTTCCTCGCTGAGGATG 130130 130148 96 435872 rs363092 Major (10) CAAACCACTGTGGGATGAA 135673 135691 97 435897 rs363092 Minor (10) CAAACCACTTTGGGATGAA 135673 135691 98 435879 rs7685686 Major (10) AATAAATTGTCATCACCAG 146786 146804 99 435904 rs7685686 Minor (10) AATAAATTGCCATCACCAG 146786 146804 100 435871 rs363088 Major (10) TCACAGCTATCTTCTCATC 149974 149992 101 435896 rs363088 Minor (10) TCACAGCTAACTTCTCATC 149974 149992 102 435870 rs362331 Major (10) GCACACAGTAGATGAGGGA 155479 155497 103 435895 rs362331 Minor (10) GCACACAGTGGATGAGGGA 155479 155497 104 435881 rs916171 Major (10) CAGAACAAAGAGAAGAATT 156459 156477 105 435906 rs916171 Minor (10) CAGAACAAACAGAAGAATT 156459 156477 106 435342 rs362322 Major (8) GCTTACATGCCTTCAGTGA 161007 161025 107 435360 rs362322 Minor (8) GCTTACACGCCTTCAGTGA 161007 161025 108 435306 rs362322 Major (10) CAGCTTACATGCCTTCAGT 161009 161027 109 435324 rs362322 Minor (10) CAGCTTACACGCCTTCAGT 161009 161027 110 435868 rs362275 Major (10) AAGAAGCCTGATAAAATCT 164246 164264 111 435893 rs362275 Minor (10) AAGAAGCCTAATAAAATCT 164246 164264 112 435343 rs2276881 Major (8) CATACATCAGCTCAAACTG 171303 171321 113 435361 rs2276881 Minor (8) CATACATTAGCTCAAACTG 171303 171321 114 435307 rs2276881 Major (10) CACATACATCAGCTCAAAC 171305 171323 115 435325 rs2276881 Minor (10) CACATACATTAGCTCAAAC 171305 171323 116 435368 rs2276881 Major (12) GTCACATACATCAGCTCAA 171307 171325 117 435866 rs3121419 Major (10) GAGACTATAGCACCCAGAT 171901 171919 118 435891 rs3121419 Minor (10) GAGACTATAACACCCAGAT 171901 171919 119 435344 rs362272 Major (8) TAGAGGACGCCGTGCAGGG 174622 174640 120 435362 rs362272 Minor (8) TAGAGGATGCCGTGCAGGG 174622 174640 121 435308 rs362272 Major (10) CATAGAGGACGCCGTGCAG 174624 174642 122 435326 rs362272 Minor (10) CATAGAGGATGCCGTGCAG 174624 174642 123 435369 rs362272 Major (12) CACATAGAGGACGCCGTGC 174626 174644 124 435867 rs362271 Major (10) ACGTGTGTACAGAACCTGC 175162 175180 125 435892 rs362271 Minor (10) ACGTGTGTATAGAACCTGC 175162 175180 126 435873 rs3775061 Major (10) TGTTCAGAATGCCTCATCT 178398 178416 127 435898 rs3775061 Minor (10) TGTTCAGAACGCCTCATCT 178398 178416 128 435345 rs362310 Major (8) AAACGGCGCAGCGGGAAGG 179418 179436 129 435363 rs362310 Minor (8) AAACGGCACAGCGGGAAGG 179418 179436 130 435309 rs362310 Major (10) AGAAACGGCGCAGCGGGAA 179420 179438 131 435327 rs362310 Minor (10) AGAAACGGCACAGCGGGAA 179420 179438 132 435915 rs362307 Major (6) AGGGCGCAGACTTCCAAAG 181485 181503 133 435927 rs362307 Minor (6) AGGGCACAGACTTCCAAAG 181485 181503 134 435917 rs362307 Major (7) AAGGGCGCAGACTTCCAAA 181486 181504 135 435929 rs362307 Minor (7) AAGGGCACAGACTTCCAAA 181486 181504 136 435346 rs362307 Major (8) CAAGGGCGCAGACTTCCAA 181487 181505 137 435364 rs362307 Minor (8) CAAGGGCACAGACTTCCAA 181487 181505 138 435919 rs362307 Major (9) ACAAGGGCGCAGACTTCCA 181488 181506 139 435931 rs362307 Minor (9) ACAAGGGCACAGACTTCCA 181488 181506 140 435310 rs362307 Major (10) CACAAGGGCGCAGACTTCC 181489 181507 141 435328 rs362307 Minor (10) CACAAGGGCACAGACTTCC 181489 181507 142 435921 rs362307 Major (11) GCACAAGGGCGCAGACTTC 181490 181508 143 435933 rs362307 Minor (11) GCACAAGGGCACAGACTTC 181490 181508 144 435370 rs362307 Major (12) GGCACAAGGGCGCAGACTT 181491 181509 145 435925 rs362307 Minor (12) GGCACAAGGGCACAGACTT 181491 181509 146 435923 rs362307 Major (14) AGGGCACAAGGGCGCAGAC 181493 181511 147 435935 rs362307 Minor (14) AGGGCACAAGGGCACAGAC 181493 181511 148 435869 rs362306 Major (10) GAGCAGCTGCAACCTGGCA 181744 181762 149 435894 rs362306 Minor (10) GAGCAGCTGTAACCTGGCA 181744 181762 150 435347 rs362303 Major (8) TGGTGCCGGGTGTCTAGCA 181949 181967 151 435365 rs362303 Minor (8) TGGTGCCAGGTGTCTAGCA 181949 181967 152 435311 rs362303 Major (10) AATGGTGCCGGGTGTCTAG 181951 181969 153 435329 rs362303 Minor (10) AATGGTGCCAGGTGTCTAG 181951 181969 154 435882 rs362296 Major (10) GGGGACAGGGTGTGCTCTC 186651 186669 155 435907 rs362296 Minor (10) GGGGACAGGTTGTGCTCTC 186651 186669 156

TABLE 4 Comparison of inhibition of HTT mRNA levels by ISIS 387916 and ISIS 388816 with that by chimeric oligonucleotides targeting SNP positions on the HTT gene (SEQ ID NO: 1) SEQ SNP RS Target % inhibition ID ISIS No No. allele GM04281 GM02171 GM02173B NO 387916 n/a n/a 96 96 98 6 388816 n/a n/a 76 88 85 7 435330 rs3856973 Major (8) 64 51 36 8 435348 rs3856973 Minor (8) 50 88 80 9 435294 rs3856973 Major (10) 54 46 54 10 435312 rs3856973 Minor (10) 20 82 58 11 435864 rs2285086 Major (10) 54 28 26 12 435889 rs2285086 Minor (10) 17 43 41 13 435878 rs7659144 Major (10) 43 32 39 14 435903 rs7659144 Minor (10) 16 37 29 15 435863 rs16843804 Major (10) 63 78 81 16 435888 rs16843804 Minor (10) 58 75 77 17 435331 rs2024115 Major (8) 56 27 56 18 435349 rs2024115 Minor (8) 26 91 66 19 435295 rs2024115 Major (10) 53 57 62 20 435313 rs2024115 Minor (10) 25 87 53 21 435862 rs10015979 Major (10) 8 51 40 22 435887 rs10015979 Minor (10) 40 22 28 23 435880 rs7691627 Major (10) 43 17 21 24 435905 rs7691627 Minor (10) 13 27 15 25 435885 rs2798235 Major (10) 38 39 30 26 435910 rs2798235 Minor (10) 17 30 16 27 435874 rs4690072 Major (10) 61 34 48 28 435899 rs4690072 Minor (10) 50 41 45 29 435875 rs6446723 Major (10) 28 13 35 30 435900 rs6446723 Minor (10) 24 56 37 31 435332 rs363081 Major (8) 76 95 88 32 435350 rs363081 Minor (8) 27 61 43 33 435296 rs363081 Major (10) 59 77 66 34 435314 rs363081 Minor (10) 38 66 40 35 435886 rs363080 Major (10) 74 72 79 36 435911 rs363080 Minor (10) 57 58 54 37 435914 rs363075 Major (6) 95 92 95 38 435926 rs363075 Minor (6) 88 81 79 39 435916 rs363075 Major (7) 90 92 94 40 435928 rs363075 Minor (7) 83 79 85 41 435333 rs363075 Major (8) 86 97 91 42 435351 rs363075 Minor (8) 59 80 58 43 435918 rs363075 Major (9) 83 90 91 44 435930 rs363075 Minor (9) 29 49 49 45 435297 rs363075 Major (10) 74 84 83 46 435315 rs363075 Minor (10) 47 63 45 47 435920 rs363075 Major (11) 78 66 83 48 435932 rs363075 Minor (11) 39 20 19 49 435366 rs363075 Major (12) 80 91 85 50 435924 rs363075 Minor (12) 37 49 58 51 435922 rs363075 Major (14) 80 90 91 52 435934 rs363075 Minor (14) 63 70 80 53 435334 rs363064 Major (8) 50 59 44 54 435352 rs363064 Minor (8) 12 37 48 55 435298 rs363064 Major (10) 81 92 87 56 435316 rs363064 Minor (10) 69 90 80 57 435335 rs3025849 Major (8) 0 40 37 58 435353 rs3025849 Minor (8) 0 29 18 59 435299 rs3025849 Major (10) 0 34 67 60 435317 rs3025849 Minor (10) 0 38 34 61 435877 rs6855981 Major (10) 31 59 58 62 435902 rs6855981 Minor (10) 0 43 27 63 435336 rs363102 Major (8) 0 21 19 64 435354 rs363102 Minor (8) 0 36 33 65 435300 rs363102 Major (10) 0 34 24 66 435318 rs363102 Minor (10) 0 30 20 67 435884 rs11731237 Major (10) 7 46 51 68 435909 rs11731237 Minor (10) 30 47 41 69 435337 rs4690073 Major (8) 12 0 12 70 435355 rs4690073 Minor (8) 0 26 33 71 435301 rs4690073 Major (10) 23 0 10 72 435319 rs4690073 Minor (10) 0 45 53 73 435883 rs363144 Major (10) 24 23 39 74 435908 rs363144 Minor (10) 27 20 22 75 435338 rs3025838 Major (8) 31 46 69 76 435356 rs3025838 Minor (8) 3 25 17 77 435302 rs3025838 Major (10) 39 73 67 78 435320 rs3025838 Minor (10) 21 49 32 79 435339 rs363099 Major (8) 84 87 76 80 435357 rs363099 Minor (8) 71 91 90 81 435303 rs363099 Major (10) 83 92 85 82 435321 rs363099 Minor (10) 84 95 89 83 435367 rs363099 Major (12) 76 82 72 84 435340 rs363096 Major (8) 0 47 52 85 435358 rs363096 Minor (8) 0 25 35 86 435304 rs363096 Major (10) 5 33 36 87 435322 rs363096 Minor (10) 2 30 32 88 435341 rs2298967 Major (8) 54 72 56 89 435359 rs2298967 Minor (8) 25 59 63 90 435305 rs2298967 Major (10) 66 80 78 91 435323 rs2298967 Minor (10) 36 79 66 92 435865 rs2298969 Major (10) 53 72 79 93 435890 rs2298969 Minor (10) 65 46 54 94 435876 rs6844859 Major (10) 70 67 77 95 435901 rs6844859 Minor (10) 39 83 80 96 435872 rs363092 Major (10) 46 41 54 97 435897 rs363092 Minor (10) 37 69 57 98 435879 rs7685686 Major (10) 83 31 70 99 435904 rs7685686 Minor (10) 30 92 72 100 435871 rs363088 Major (10) 70 55 70 101 435896 rs363088 Minor (10) 66 74 80 102 435870 rs362331 Major (10) 88 74 88 103 435895 rs362331 Minor (10) 78 92 86 104 435881 rs916171 Major (10) 0 57 51 105 435906 rs916171 Minor (10) 14 26 17 106 435342 rs362322 Major (8) 47 74 67 107 435360 rs362322 Minor (8) 17 58 52 108 435306 rs362322 Major (10) 50 77 65 109 435324 rs362322 Minor (10) 42 61 64 110 435868 rs362275 Major (10) 54 35 43 111 435893 rs362275 Minor (10) 3 27 33 112 435343 rs2276881 Major (8) 59 76 65 113 435361 rs2276881 Minor (8) 58 44 20 114 435307 rs2276881 Major (10) 69 82 81 115 435325 rs2276881 Minor (10) 17 47 43 116 435368 rs2276881 Major (12) 84 96 92 117 435866 rs3121419 Major (10) 67 61 64 118 435891 rs3121419 Minor (10) 53 76 73 119 435344 rs362272 Major (8) 35 46 36 120 435362 rs362272 Minor (8) 34 68 57 121 435308 rs362272 Major (10) 26 30 35 122 435326 rs362272 Minor (10) 29 50 39 123 435369 rs362272 Major (12) 66 74 65 124 435867 rs362271 Major (10) 73 74 75 125 435892 rs362271 Minor (10) 52 74 79 126 435873 rs3775061 Major (10) 40 32 47 127 435898 rs3775061 Minor (10) 13 20 24 128 435345 rs362310 Major (8) 38 55 52 129 435363 rs362310 Minor (8) 45 67 60 130 435309 rs362310 Major (10) 33 44 56 131 435327 rs362310 Minor (10) 33 71 61 132 435915 rs362307 Major (6) 61 54 58 133 435927 rs362307 Minor (6) 31 35 44 134 435917 rs362307 Major (7) 67 76 66 135 435929 rs362307 Minor (7) 33 34 55 136 435346 rs362307 Major (8) 67 89 66 137 435364 rs362307 Minor (8) 46 72 66 138 435919 rs362307 Major (9) 84 79 70 139 435931 rs362307 Minor (9) 74 74 86 140 435310 rs362307 Major (10) 74 81 71 141 435328 rs362307 Minor (10) 47 69 75 142 435921 rs362307 Major (11) 74 77 69 143 435933 rs362307 Minor (11) 38 47 74 144 435370 rs362307 Major (12) 64 74 38 145 435925 rs362307 Minor (12) 60 66 80 146 435923 rs362307 Major (14) 73 66 71 147 435935 rs362307 Minor (14) 68 75 87 148 435869 rs362306 Major (10) 82 77 81 149 435894 rs362306 Minor (10) 28 79 72 150 435347 rs362303 Major (8) 68 74 71 151 435365 rs362303 Minor (8) 69 83 76 152 435311 rs362303 Major (10) 46 56 72 153 435329 rs362303 Minor (10) 49 62 39 154 435882 rs362296 Major (10) 29 48 56 155 435907 rs362296 Minor (10) 42 56 52 156

Example 3: Dose-Dependent Antisense Inhibition of Human Huntingtin mRNA Levels in Coriell Fibroblast Cell Lines

Gapmers from the study described in Example 2 were selected and tested at various doses in GM04281, GM02171, and GM02173B cell lines. Each cell line was plated at a density of 25,000 cells per well and transfected using electroporation with 750 nM, 1,500 nM, 3,000 nM, 6,000 nM, and 12,000 nM concentrations of antisense oligonucleotide, as specified in Table 5, 6, and 7. After a treatment period of approximately 16 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. Human HTT primer probe set RTS2617 was used to measure mRNA levels. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells. IC50 values are also provided in Tables 5, 6, and 7.

TABLE 5 Dose-dependent antisense inhibition of human HTT in GM04281 cells ISIS 750 IC50 No. nM 1,500 nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 51 81 80 91 97 0.6 435330 24 49 50 73 85 2.5 435331 23 38 64 72 74 2.4 435868 3 17 7 29 63 6.7 435870 53 73 77 86 93 0.6 435871 28 51 52 78 89 1.7 435874 14 21 28 64 82 3.3 435879 42 57 57 81 91 1.1 435890 48 56 62 76 91 0.9 435929 10 0 5 12 48 13.8 435931 20 17 53 62 81 2.9 435933 0 7 24 43 49 10.7 435935 0 38 38 62 29 4.2

TABLE 6 Dose-dependent antisense inhibition of human HTT in GM02171 cells ISIS 750 IC50 No. nM 1,500 nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 57 73 81 93 98 0.4 435330 27 37 0 44 63 4.4 435331 35 34 19 41 63 3.5 435868 21 21 39 24 12 >12.0 435870 50 53 57 70 79 0.9 435871 32 46 45 58 62 3.9 435874 1 0 4 11 6 >12.0 435879 32 14 17 45 38 >12.0 435890 34 33 40 51 62 5.4 435929 25 22 31 5 29 >12.0 435931 15 28 27 60 79 3.7 435933 13 36 21 43 48 12.2 435935 25 42 27 61 68 3.2

TABLE 7 Dose-dependent antisense inhibition of human HTT in GM02173B cells ISIS 750 IC50 No. nM 1,500 nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 43 67 80 86 97 1.1 435330 22 21 0 52 62 5.3 435331 19 17 32 50 55 9.4 435868 17 25 41 13 26 >12.0 435870 24 57 70 78 75 1.8 435871 8 30 42 50 48 5.0 435874 31 35 28 35 42 >12.0 435879 39 44 42 60 64 2.5 435890 38 36 50 65 73 3.1 435929 19 17 19 42 35 7.7 435931 40 19 31 48 71 5.8 435933 35 24 47 52 59 4.4 435935 25 23 40 73 77 3.7

Example 4: Dose-Dependent Antisense Inhibition of Human Huntingtin mRNA Levels in Coriell Fibroblast Cell Lines

Gapmers from the study described in Example 2 were selected and tested at various doses in GM04281, GM02171, and GM02173B cell lines. Each cell line was plated at a density of 25,000 cells per well and transfected using electroporation with 750 nM, 1,500 nM, 3,000 nM, 6,000 nM, and 12,000 nM concentrations of antisense oligonucleotide, as specified in Table 8, 9, and 10. After a treatment period of approximately 16 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. Human HTT primer probe set RTS2617 was used to measure mRNA levels. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA relative to untreated control cells. IC50 values are also provided in Tables 8, 9, and 10.

TABLE 8 Dose-dependent antisense inhibition of human HTT in GM04281 cells 1,500 IC50 ISIS No. 750 nM nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 61 78 90 94 97 <0.8 435303 33 39 69 79 91 1.5 435328 0 12 16 51 75 5.3 435331 27 48 48 70 82 2.1 435339 46 37 61 73 89 2.3 435869 17 35 44 66 80 3.3 435870 44 60 64 84 84 1.1 435871 41 50 71 78 87 1.2 435874 24 36 35 65 73 3.1 435879 46 52 78 81 92 0.9 435890 41 53 63 80 86 1.3 435925 0 14 39 60 87 4.2 435926 20 28 67 81 89 2.0 435928 32 49 73 86 86 1.8 435931 22 24 40 59 90 3.8

TABLE 9 Dose-dependent antisense inhibition of human HTT in GM02171 cells 1,500 IC50 ISIS No. 750 nM nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 50 64 90 95 96 0.7 435303 14 32 68 79 85 2.8 435328 0 12 20 38 55 10.3 435331 0 13 5 30 36 >12.0 435339 30 40 58 63 49 2.5 435869 13 25 31 47 87 4.0 435870 18 31 44 66 74 3.5 435871 1 20 29 49 64 6.5 435874 3 6 12 17 31 >12.0 435879 0 2 12 35 44 >12.0 435890 15 16 30 48 72 5.8 435925 0 0 22 48 29 6.3 435926 25 28 58 74 85 2.3 435928 18 53 61 86 83 2.5 435931 0 4 25 46 68 6.7

TABLE 10 Dose-dependent antisense inhibition of human HTT in GM02173B cells 1,500 IC50 ISIS No. 750 nM nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 27 65 84 81 96 1.9 435303 23 48 52 76 76 2.9 435328 8 14 19 34 50 15.7 435331 10 17 16 27 32 >12.0 435339 28 26 38 67 82 3.8 435869 12 24 37 45 79 4.2 435870 20 26 58 53 78 2.7 435871 15 16 32 45 71 6.0 435874 13 8 28 36 31 >12.0 435879 22 20 36 53 60 6.0 435890 21 28 34 54 71 4.3 435925 2 10 28 43 78 5.9 435926 7 25 37 73 79 3.5 435928 15 39 60 73 87 2.5 435931 13 13 32 61 62 6.7

Example 5: Antisense Inhibition of Human HTT in GM04281 Cells

Additional antisense oligonucleotides were designed based on the gapmers selected from studies described in Example 4. These oligonucleotides were designed by creating gapmers shifted slightly upstream and downstream (i.e. “microwalk”) of the original gapmers from Tables 8, 9, and 10. Antisense oligonucleotides were also created with uniform MOE, as well as with various motifs, 2-9-6 MOE, 3-9-3 MOE, 3-9-4 MOE, 3-9-5 MOE, 4-10-5 MOE, 4-11-4 MOE, 4-7-4 MOE, 4-9-4 MOE, 4-9-5 MOE, 5-10-4 MOE, 5-7-5 MOE, 5-8-6 MOE, 5-9-3 MOE, 5-9-5 MOE, 6-7-6 MOE, 6-9-2 MOE, and 6-8-5 MOE.

In addition, antisense oligonucleotides were designed targeting SNP RS Nos. rs2857936, rs12506200, rs762855, and rs1006798 (refer to Table 2). The oligonucleotides were designed targeting either the major allele or the minor allele, and with the SNP position opposite either position 8 or position 10 of the gapmer.

These gapmers were tested in vitro. Cultured GM04281 cells at a density of 25,000 cells per well were transfected using electroporation with 10,000 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented in Tables 11-19 as percent inhibition of HTT mRNA, relative to untreated control cells.

The gapmers, ISIS 435869, ISIS 435870, ISIS 435874, ISIS 435879, and ISIS 435890, from which some of the newly designed gapmers were derived are marked with an asterisk (*) in the table. ISIS 387916 was included in the study as a benchmark oligonucleotide against which the potency of the antisense oligonucleotides targeting nucleotides overlapping each SNP position could be compared.

The uniform MOE oligonucleotides are 15 nucleotides in length.

The 2-9-6 gapmers are 17 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 2 nucleotides and on the 3′ direction by a wing comprising 6 nucleotides.

The 3-9-3 gapmers are 15 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 3 nucleotides each.

The 3-9-4 gapmers are 16 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 3 nucleotides and on the 3′ direction by a wing comprising 4 nucleotides.

The 3-9-5 gapmers are 17 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 3 nucleotides and on the 3′ direction by a wing comprising 5 nucleotides.

The 4-10-5 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of ten 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 4 nucleotides and on the 3′ direction by a wing comprising 5 nucleotides.

The 4-11-4 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of eleven 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 4 nucleotides each.

The 4-7-4 gapmers are 15 nucleotides in length, wherein the central gap segment is comprised of seven 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 4 nucleotides each.

The 4-9-4 gapmers are 17 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 4 nucleotides each.

The 4-9-5 gapmers are 18 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 4 nucleotides and on the 3′ direction by a wing comprising 5 nucleotides.

The 5-10-4 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of ten 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 5 nucleotides and on the 3′ direction by a wing comprising 4 nucleotides.

The 5-7-5 gapmers are 17 nucleotides in length, wherein the central gap segment is comprised of seven 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 5 nucleotides each.

The 5-8-6 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of eight 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 5 nucleotides and on the 3′ direction by a wing comprising 6 nucleotides.

The 5-9-3 gapmers are 17 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 5 nucleotides and on the 3′ direction by a wing comprising 3 nucleotides.

The 5-9-5 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 5 nucleotides each.

The 6-7-6 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of seven 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 6 nucleotides each.

The 6-9-2 gapmers are 17 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 6 nucleotides and on the 3′ direction by a wing comprising 2 nucleotides.

The 6-8-5 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of eight 2′-deoxynucleotides and is flanked on the 5′ direction by a wing comprising 6 nucleotides and on the 3′ direction by a wing comprising 5 nucleotides.

For each of the motifs, each nucleotide in the 5′ wing segment and each nucleotide in the 3′ wing segment has a 2′-MOE modification. The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine nucleobases throughout each gapmer are 5-methylcytosines.

The oligonucleotides are organized in tables according to the SNP they target. “Start site” indicates the 5′-most nucleotide to which the gapmer is targeted. “Stop site” indicates the 3′-most nucleotide to which the gapmer is targeted. ‘Target allele’ indicates whether the gapmer is targeted to the major or the minor allele. The number in parentheses indicates the position on the oligonucleotide opposite to the SNP position.

TABLE 11 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs2857936 (nucleobases 1952 to 1972 of SEQ ID NO: 1) SEQ Start Stop Target ISIS % ID Site Site allele No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 1952 1970 Minor (8) 459908 GCTTTTCATTGAAAAGAAA 5-9-5 26 157 1952 1970 Major (8) 459916 GCTTTTCGTTGAAAAGAAA 5-9-5 8 158 1954 1972 Minor (10) 459904 CTGCTTTTCATTGAAAAGA 5-9-5 23 159 1954 1972 Major (10) 459912 CTGCTTTTCGTTGAAAAGA 5-9-5 8 160

TABLE 12 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs12506200 (nucleobases 3695 to 3715 of SEQ ID NO: 1) SEQ Start Stop Target ISIS % ID Site Site allele No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 3695 3713 Major (8) 459909 ACTAGGCCGGGCATGCTGG 5-9-5 48 161 3695 3713 Minor (8) 459917 ACTAGGCTGGGCATGCTGG 5-9-5 35 162 3697 3715 Major (10) 459905 AGACTAGGCCGGGCATGCT 5-9-5 33 163 3697 3715 Minor (10) 459913 AGACTAGGCTGGGCATGCT 5-9-5 45 164

TABLE 13 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs762855 (nucleobases 14437 to 14457 of SEQ ID NO: 1) SEQ Start Stop Target ISIS % ID Site Site allele No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 14437 14455 Minor (8) 459910 AAACAGCTGTTAGTTCCCA 5-9-5 27 165 14437 14455 Major (8) 459918 AAACAGCCGTTAGTTCCCA 5-9-5 39 166 14439 14457 Minor (10) 459906 AGAAACAGCTGTTAGTTCC 5-9-5 24 167 14439 14457 Major (10) 459914 AGAAACAGCCGTTAGTTCC 5-9-5 28 168

TABLE 14 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs4690072 (nucleobases 62147 to 62173 of SEQ ID NO: 1) SEQ Start Stop % ID Site Site Target allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 62147 62165 Major (6) 460145 GTGCTACCCAACCTTTCTG 5-9-5 62 169 62148 62166 Major (7) 460144 AGTGCTACCCAACCTTTCT 5-9-5 61 170 62149 62167 Major (8) 460143 CAGTGCTACCCAACCTTTC 5-9-5 65 171 62150 62168 Major (9) 460142 ACAGTGCTACCCAACCTTT 5-9-5 83 172 62151 62169 Major (10) *435874 CACAGTGCTACCCAACCTT 5-9-5 76 28 62151 62169 Major (10) 460022 CACAGTGCTACCCAACCTT 4-10-5 75 28 62151 62169 Major (10) 460033 CACAGTGCTACCCAACCTT 4-11-4 89 28 62151 62168 Major (9) 460063 ACAGTGCTACCCAACCTT 4-9-5 77 173 62151 62169 Major (10) 460073 CACAGTGCTACCCAACCTT 5-10-4 86 28 62151 62169 Major (10) 460093 CACAGTGCTACCCAACCTT 5-8-6 61 28 62151 62169 Major (10) 460169 CACAGTGCTACCCAACCTT 6-7-6 16 28 62151 62169 Major (10) 460188 CACAGTGCTACCCAACCTT 6-8-5 53 28 62152 62168 Major (9) 459978 ACAGTGCTACCCAACCT 2-9-6 87 174 62152 62167 Major (8) 459999 CAGTGCTACCCAACCT 3-9-4 48 175 62152 62168 Major (9) 460012 ACAGTGCTACCCAACCT 3-9-5 84 174 62152 62168 Major (9) 460052 ACAGTGCTACCCAACCT 4-9-4 51 174 62152 62168 Major (9) 460083 ACAGTGCTACCCAACCT 5-7-5 37 174 62152 62168 Major (9) 460103 ACAGTGCTACCCAACCT 5-9-3 80 174 62152 62170 Major (11) 460137 TCACAGTGCTACCCAACCT 5-9-5 65 176 62152 62168 Major (9) 460179 ACAGTGCTACCCAACCT 6-9-2 67 174 62153 62167 Major (8) 459989 CAGTGCTACCCAACC 3-9-3 60 177 62153 62167 Major (8) 460043 CAGTGCTACCCAACC 4-7-4 24 177 62153 62171 Major (12) 460138 ATCACAGTGCTACCCAACC 5-9-5 76 178 62154 62172 Major (13) 460139 TATCACAGTGCTACCCAAC 5-9-5 68 179 62155 62173 Major (14) 460140 ATATCACAGTGCTACCCAA 5-9-5 79 180

TABLE 15 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs2298969 (nucleobases 125883 to 125911 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 125883 125901 Minor (5) 460166 ATGCTGACTTGGGCCATTC 5-9-5 83 181 125884 125902 Minor (6) 460165 GATGCTGACTTGGGCCATT 5-9-5 88 182 125885 125903 Minor (7) 460164 GGATGCTGACTTGGGCCAT 5-9-5 68 183 125886 125904 Minor (8) 460163 GGGATGCTGACTTGGGCCA 5-9-5 73 184 125887 125905 Minor (9) 460162 AGGGATGCTGACTTGGGCC 5-9-5 88 185 125888 125906 Minor (10) *435890 AAGGGATGCTGACTTGGGC 5-9-5 83 94 125888 125906 Minor (10) 460026 AAGGGATGCTGACTTGGGC 4-10-5 90 94 125888 125906 Minor (10) 460037 AAGGGATGCTGACTTGGGC 4-11-4 86 94 125888 125905 Minor (9) 460068 AGGGATGCTGACTTGGGC 4-9-5 90 186 125888 125906 Minor (10) 460076 AAGGGATGCTGACTTGGGC 5-10-4 90 94 125888 125906 Minor (10) 460096 AAGGGATGCTGACTTGGGC 5-8-6 88 94 125888 125906 Minor (10) 460171 AAGGGATGCTGACTTGGGC 6-7-6 87 94 125888 125906 Minor (10) 460190 AAGGGATGCTGACTTGGGC 6-8-5 69 94 125889 125905 Minor (9) 459983 AGGGATGCTGACTTGGG 2-9-6 80 187 125889 125904 Minor (8) 460005 GGGATGCTGACTTGGG 3-9-4 80 284 125889 125905 Minor (9) 460016 AGGGATGCTGACTTGGG 3-9-5 90 187 125889 125905 Minor (9) 460057 AGGGATGCTGACTTGGG 4-9-4 86 187 125889 125905 Minor (9) 460087 AGGGATGCTGACTTGGG 5-7-5 86 187 125889 125905 Minor (9) 460107 AGGGATGCTGACTTGGG 5-9-3 79 187 125889 125907 Major (11) 460157 CAAGGGATGCTGACTTGGG 5-9-5 88 188 125889 125905 Minor (9) 460181 AGGGATGCTGACTTGGG 6-9-2 62 187 125890 125904 Minor (8) 459972 GGGATGCTGACTTGG Uniform 18 189 125890 125904 Minor (8) 459992 GGGATGCTGACTTGG 3-9-3 90 189 125890 125904 Minor (8) 460046 GGGATGCTGACTTGG 4-7-4 59 189 125890 125908 Major (12) 460158 CCAAGGGATGCTGACTTGG 5-9-5 79 190 125891 125909 Major (13) 460159 GCCAAGGGATGCTGACTTG 5-9-5 82 191 125892 125910 Major (14) 460160 TGCCAAGGGATGCTGACTT 5-9-5 87 192 125893 125911 Major (15) 460161 CTGCCAAGGGATGCTGACT 5-9-5 78 193

TABLE 16 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs7685686 (nucleobases 146781 to 146809 of SEQ ID NO: 1) SEQ Start Stop Target ISIS % ID Site Site allele No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 146781 146799 Major (5) 460156 ATTGTCATCACCAGAAAAA 5-9-5 88 194 146782 146800 Major (6) 460155 AATTGTCATCACCAGAAAA 5-9-5 89 195 146783 146801 Major (7) 460154 AAATTGTCATCACCAGAAA 5-9-5 89 196 146784 146802 Major (8) 460153 TAAATTGTCATCACCAGAA 5-9-5 93 197 146785 146803 Major (9) 460152 ATAAATTGTCATCACCAGA 5-9-5 95 198 146786 146804 Major (10) *435879 AATAAATTGTCATCACCAG 5-9-5 94 99 146786 146804 Major (10) 460024 AATAAATTGTCATCACCAG 4-10-5 88 99 146786 146804 Major (10) 460035 AATAAATTGTCATCACCAG 4-11-4 91 99 146786 146803 Major (9) 460065 ATAAATTGTCATCACCAG 4-9-5 96 199 146786 146804 Major (10) 460074 AATAAATTGTCATCACCAG 5-10-4 94 99 146786 146804 Major (10) 460095 AATAAATTGTCATCACCAG 5-8-6 92 99 146786 146804 Major (10) 460170 AATAAATTGTCATCACCAG 6-7-6 91 99 146786 146804 Major (10) 460189 AATAAATTGTCATCACCAG 6-8-5 94 99 146787 146803 Major (9) 459981 ATAAATTGTCATCACCA 2-9-6 85 200 146787 146802 Major (8) 460002 TAAATTGTCATCACCA 3-9-4 86 201 146787 146803 Major (9) 460014 ATAAATTGTCATCACCA 3-9-5 91 200 146787 146803 Major (9) 460055 ATAAATTGTCATCACCA 4-9-4 90 200 146787 146803 Major (9) 460085 ATAAATTGTCATCACCA 5-7-5 94 200 146787 146803 Major (9) 460104 ATAAATTGTCATCACCA 5-9-3 93 200 146787 146805 Major (11) 460147 TAATAAATTGTCATCACCA 5-9-5 91 202 146787 146803 Major (9) 460180 ATAAATTGTCATCACCA 6-9-2 91 200 146788 146802 Major (8) 459970 TAAATTGTCATCACC Uniform 9 203 146788 146802 Major (8) 459990 TAAATTGTCATCACC 3-9-3 67 203 146788 146802 Major (8) 460045 TAAATTGTCATCACC 4-7-4 84 203 146788 146806 Major (12) 460148 TTAATAAATTGTCATCACC 5-9-5 88 204 146789 146807 Major (13) 460149 ATTAATAAATTGTCATCAC 5-9-5 32 205 146790 146808 Major (14) 460150 TATTAATAAATTGTCATCA 5-9-5 29 206 146791 146809 Major (15) 460151 CTATTAATAAATTGTCATC 5-9-5 33 207

TABLE 17 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362331 (nucleobases 155474 to 155502 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 155474 155492 Major (5) 460136 CAGTAGATGAGGGAGCAGG 5-9-5 81 208 155475 155493 Major (6) 460135 ACAGTAGATGAGGGAGCAG 5-9-5 84 209 155476 155494 Major (7) 460134 CACAGTAGATGAGGGAGCA 5-9-5 87 210 155477 155495 Major (8) 460133 ACACAGTAGATGAGGGAGC 5-9-5 85 211 155478 155496 Major (9) 460132 CACACAGTAGATGAGGGAG 5-9-5 86 212 155479 155497 Major (10) *435870 GCACACAGTAGATGAGGGA 5-9-5 91 103 155479 155497 Major (10) 460019 GCACACAGTAGATGAGGGA 4-10-5 92 103 155479 155497 Major (10) 460031 GCACACAGTAGATGAGGGA 4-11-4 95 103 155479 155496 Major (9) 460061 CACACAGTAGATGAGGGA 4-9-5 87 213 155479 155497 Major (10) 460071 GCACACAGTAGATGAGGGA 5-10-4 94 103 155479 155497 Major (10) 460090 GCACACAGTAGATGAGGGA 5-8-6 86 103 155479 155497 Major (10) 460168 GCACACAGTAGATGAGGGA 6-7-6 84 103 155479 155497 Major (10) 460187 GCACACAGTAGATGAGGGA 6-8-5 89 103 155480 155496 Major (9) 459977 CACACAGTAGATGAGGG 2-9-6 90 214 155480 155495 Major (8) 459996 ACACAGTAGATGAGGG 3-9-4 37 215 155480 155496 Major (9) 460009 CACACAGTAGATGAGGG 3-9-5 90 214 155480 155496 Major (9) 460051 CACACAGTAGATGAGGG 4-9-4 73 214 155480 155496 Major (9) 460081 CACACAGTAGATGAGGG 5-7-5 77 214 155480 155496 Major (9) 460101 CACACAGTAGATGAGGG 5-9-3 84 214 155480 155498 Major (11) 460127 TGCACACAGTAGATGAGGG 5-9-5 89 216 155480 155496 Major (9) 460178 CACACAGTAGATGAGGG 6-9-2 92 214 155481 155495 Major (8) 459967 ACACAGTAGATGAGG Uniform 81 217 155481 155495 Major (8) 459987 ACACAGTAGATGAGG 3-9-3 18 217 155481 155495 Major (8) 460041 ACACAGTAGATGAGG 4-7-4 54 217 155481 155499 Major (12) 460128 GTGCACACAGTAGATGAGG 5-9-5 73 218 155482 155500 Major (13) 460129 AGTGCACACAGTAGATGAG 5-9-5 86 219 155483 155501 Major (14) 460130 AAGTGCACACAGTAGATGA 5-9-5 60 220 155484 155502 Major (15) 460131 GAAGTGCACACAGTAGATG 5-9-5 73 221

TABLE 18 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362306 (nucleobases 181739 to 181767 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 181739 181757 Major (5) 460126 GCTGCAACCTGGCAACAAC 5-9-5 87 222 181740 181758 Major (6) 460125 AGCTGCAACCTGGCAACAA 5-9-5 70 223 181741 181759 Major (7) 460123 CAGCTGCAACCTGGCAACA 5-9-5 83 224 181742 181760 Major (8) 460121 GCAGCTGCAACCTGGCAAC 5-9-5 47 225 181743 181761 Major (9) 460118 AGCAGCTGCAACCTGGCAA 5-9-5 75 226 181744 181762 Major (10) *435869 GAGCAGCTGCAACCTGGCA 5-9-5 91 149 181744 181762 Major (10) 460018 GAGCAGCTGCAACCTGGCA 4-10-5 86 149 181744 181762 Major (10) 460028 GAGCAGCTGCAACCTGGCA 4-11-4 89 149 181744 181761 Major (9) 460058 AGCAGCTGCAACCTGGCA 4-9-5 85 227 181744 181762 Major (10) 460069 GAGCAGCTGCAACCTGGCA 5-10-4 91 149 181744 181762 Major (10) 460089 GAGCAGCTGCAACCTGGCA 5-8-6 54 149 181744 181762 Major (10) 460167 GAGCAGCTGCAACCTGGCA 6-7-6 85 149 181744 181762 Major (10) 460186 GAGCAGCTGCAACCTGGCA 6-8-5 84 149 181745 181761 Major (9) 459975 AGCAGCTGCAACCTGGC 2-9-6 86 228 181745 181760 Major (8) 459995 GCAGCTGCAACCTGGC 3-9-4 87 229 181745 181761 Major (9) 460008 AGCAGCTGCAACCTGGC 3-9-5 83 228 181745 181761 Major (9) 460049 AGCAGCTGCAACCTGGC 4-9-4 88 228 181745 181761 Major (9) 460079 AGCAGCTGCAACCTGGC 5-7-5 46 228 181745 181761 Major (9) 460099 AGCAGCTGCAACCTGGC 5-9-3 44 228 181745 181763 Major (11) 460108 AGAGCAGCTGCAACCTGGC 5-9-5 50 230 181745 181761 Major (9) 460177 AGCAGCTGCAACCTGGC 6-9-2 67 228 181746 181760 Major (8) 459966 GCAGCTGCAACCTGG Uniform 26 231 181746 181760 Major (8) 459985 GCAGCTGCAACCTGG 3-9-3 69 231 181746 181760 Major (8) 460039 GCAGCTGCAACCTGG 4-7-4 56 231 181746 181764 Major (12) 460110 AAGAGCAGCTGCAACCTGG 5-9-5 75 232 181747 181765 Major (13) 460113 CAAGAGCAGCTGCAACCTG 5-9-5 36 233 181748 181766 Major (14) 460115 GCAAGAGCAGCTGCAACCT 5-9-5 78 234 181749 181767 Major (15) 460117 TGCAAGAGCAGCTGCAACC 5-9-5 73 235

TABLE 19 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs1006798 (nucleobases 198015 to 198035 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 98 6 198015 198033 Minor (8) 459911 ACCATGATATCTCCAGCAC 5-9-5 33 236 198015 198033 Minor (8) 459919 ACCATGACATCTCCAGCAC 5-9-5 26 237 198017 198035 Major (10) 459907 CCACCATGATATCTCCAGC 5-9-5 32 238 198017 198035 Minor (10) 459915 CCACCATGACATCTCCAGC 5-9-5 51 239

Example 6: Dose-Dependent Antisense Inhibition of Human Huntingtin mRNA Levels in Coriell Fibroblast Cell Lines

Gapmers from the studies described in Example 5 were selected and tested at various doses in GM04281, GM02171, and GM02173B cell lines. Each cell line was plated at a density of 25,000 cells per well and transfected using electroporation with 750 nM, 1,500 nM, 3,000 nM, 6,000 nM, and 12,000 nM concentrations of antisense oligonucleotide, as specified in Tables 20, 21, and 22. After a treatment period of approximately 16 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. Human HTT primer probe set RTS2617 was used to measure mRNA levels. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells. IC50 values are also provided in Tables 20, 21, and 22.

TABLE 20 Dose-dependent antisense inhibition of human HTT in GM04281 cells ISIS 750 IC50 No. nM 1,500 nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 56 81 89 96 98 0.6 435869 38 49 66 86 91 1.4 435874 33 27 37 49 62 8.4 435879 42 55 73 86 96 1.1 435890 39 51 74 83 89 1.3 459978 29 33 51 69 86 2.5 459992 14 27 51 54 84 3.2 460012 15 24 54 70 81 3.1 460016 3 36 48 71 77 3.3 460019 54 59 74 87 94 0.7 460026 48 47 71 79 88 0.8 460028 39 38 73 77 87 1.4 460031 44 62 72 87 92 0.9 460033 11 38 52 64 87 3.0 460065 43 54 74 89 96 1.1 460068 47 28 63 76 90 2.6 460069 38 50 65 77 91 1.4 460071 53 61 80 89 93 0.6 460073 16 39 42 58 75 4.0 460076 26 47 54 70 86 2.1 460085 48 60 79 89 94 0.8 460140 6 24 44 44 64 6.6 460142 2 38 46 46 68 4.8 460152 35 61 76 92 94 1.2 460157 51 36 53 74 89 2.6 460162 64 41 71 76 85 2.1 460165 41 50 56 76 84 1.5

TABLE 21 Dose-dependent antisense inhibition of human HTT in GM02171 cells ISIS 750 IC50 No. nM 1,500 nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 53 66 88 96 98 0.7 435869 4 20 36 63 86 3.9 435870 25 39 48 62 83 2.8 435874 12 20 18 27 37 >12.0 435879 10 7 11 42 51 10.6 435890 10 23 29 29 55 9.2 459978 15 7 6 29 52 12.7 459992 11 19 26 39 62 8.7 460012 3 3 10 19 41 >12.0 460016 0 14 12 22 48 >12.0 460019 27 21 41 60 73 4.4 460026 9 25 30 46 58 7.8 460028 24 8 32 54 77 5.3 460031 8 25 42 60 83 3.8 460033 11 25 30 40 75 4.1 460065 11 16 11 31 53 10.3 460068 15 13 39 44 53 8.8 460069 17 28 37 60 79 3.9 460071 16 36 58 70 88 2.6 460073 5 19 24 33 56 8.7 460076 19 29 44 54 83 3.3 460085 10 15 17 28 31 >12.0 460140 8 22 22 28 47 >12.0 460142 11 24 28 36 38 >12.0 460152 14 21 8 25 44 22 460157 22 21 29 44 66 6.7 460162 24 55 52 62 82 2.8 460165 14 34 50 69 81 3.1

TABLE 22 Dose-dependent antisense inhibition of human HTT in GM02173B cells ISIS 750 IC50 No. nM 1,500 nM 3,000 nM 6,000 nM 12,000 nM (μM) 387916 37 63 86 88 98 1.0 435869 10 20 43 70 85 3.5 435870 24 24 56 72 87 2.3 435874 0 11 12 30 44 >12.0 435879 4 17 43 64 74 4.3 435890 31 29 54 57 69 4.4 459978 7 13 17 35 64 8.4 459992 18 15 30 51 71 5.7 460012 0 10 24 37 72 7.1 460016 15 5 30 38 59 9.5 460019 10 32 51 65 87 3.1 460026 0 34 21 55 65 6.4 460028 0 14 31 51 77 5.2 460031 0 31 53 71 88 3.2 460033 11 8 6 52 84 5.0 460065 19 37 53 58 74 3.6 460068 17 11 31 59 69 5.5 460069 11 21 37 55 75 4.6 460071 6 42 61 83 88 2.6 460073 7 13 19 49 66 6.3 460076 27 31 49 43 81 2.9 460085 17 34 51 54 68 4.4 460140 0 2 28 18 46 >12.0 460142 2 32 37 42 59 7.6 460152 17 32 35 51 66 5.5 460157 9 34 38 52 74 4.5 460162 22 45 57 65 79 2.5 460165 5 45 52 72 84 3.2

Example 7: Antisense Inhibition of Human HTT in GM04281 Cells and GM02171 Cells

Additional antisense oligonucleotides were designed based on the gapmers selected from studies described in Example 2. These oligonucleotides were designed by creating gapmers shifted slightly upstream and downstream (i.e. “microwalk”) of the original gapmers from Table 4.

The gapmers were tested in the GM04281 and the GM02171 cell lines. Cultured GM04281 or GM02171 cells at a density of 25,000 cells per well were transfected using electroporation with 10,000 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR using primer probe set RTS2617. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells.

The gapmers, from which the newly designed oligonucleotides were derived, were also included in the assay. These parent gapmers, ISIS 435294, ISIS 435295, ISIS 435301, ISIS 435303, ISIS 435304, ISIS 435305, ISIS 435308, ISIS 435330, ISIS 435331, ISIS 435337, ISIS 435339, ISIS 435340, ISIS 435341, ISIS 435344, ISIS 435862, ISIS 435863, ISIS 435864, ISIS 435866, ISIS 435867, ISIS 435868, ISIS 435871, ISIS 435873, ISIS 435875, ISIS 435876, ISIS 435878, ISIS 435880, ISIS 435881, ISIS 435882, ISIS 435884, ISIS 435890, and ISIS 435897 are marked with an asterisk (*) in the table. ISIS 387916 was included in the study as a benchmark oligonucleotide against which the potency of the antisense oligonucleotides targeting nucleotides overlapping each SNP position could be compared.

The chimeric antisense oligonucleotides in Tables 23-48 were designed as 5-9-5 MOE gapmers. The 5-9-5 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 5 nucleotides each. Each nucleotide in the 5′ wing segment and each nucleotide in the 3′ wing segment has a 2′-MOE modification. The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine nucleobases throughout each gapmer are 5-methylcytosines.

The gapmers are organized in Tables 23-48, according to the SNP site they target. “Start site” indicates the 5′-most nucleotide to which the gapmer is targeted. “Stop site” indicates the 3′-most nucleotide to which the gapmer is targeted. ‘Target allele’ indicates whether the gapmer is targeted to the major or the minor allele. The number in parentheses indicates the position on the oligonucleotide opposite to the SNP position.

TABLE 23 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs3856973 (nucleobases 19815 to 19835 of SEQ ID NO: 1) % % inhibition inhibition Start Stop Target in in SEQ Site Site ISIS No allele Sequence GM04281 GM02171 ID NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 19815 19833 *435330 Major (8) TAACACTCGATTAACCCTG 88 31 8 19816 19834 476441 Major (9) TTAACACTCGATTAACCCT 88 0 240 19817 19835 *435294 Major (10) GTTAACACTCGATTAACCC 72 30 10

TABLE 24 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs2285086 (nucleobases 28901 to 28921 of SEQ ID NO: 1) % % inhibition inhibition Start Stop Target in in SEQ Site Site ISIS No allele Sequence GM04281 GM02171 ID NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 28901 28919 463570 Major (8) TAGTTCATCCCAGTGAGAA 66 12 241 28902 28920 463573 Major (9) CTAGTTCATCCCAGTGAGA 66 36 242 28903 28921 *435864 Major (10) GCTAGTTCATCCCAGTGAG 40 18 12

TABLE 25 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs7659144 (nucleobases 37963 to 37983 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 37963 37981 476462 Major (8) GAAATGGGTTTTTCCACAT 38 0 243 37964 37982 476439 Major (9) GGAAATGGGTTTTTCCACA 80 45 244 37965 37983 *435878 Major (10) TGGAAATGGGTTTTTCCAC 76 3 14

TABLE 26 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs16843804 (nucleobases 44032 to 44052 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 44032 44050 476471 Major (8) TAACCGTGGCATGGGCAGT 82 53 245 44033 44051 476452 Major (9) TTAACCGTGGCATGGGCAG 84 44 246 44034 44052 *435863 Major (10) TTTAACCGTGGCATGGGCA 89 89 16

TABLE 27 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs2024115 (nucleobases 44210 to 44230 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 44210 44228 *435331 Major (8) TTCAAGCTAGTAACGATGC 84 20 18 44211 44229 476447 Major (9) CTTCAAGCTAGTAACGATG 87 57 247 44212 44230 *435295 Major (10) ACTTCAAGCTAGTAACGAT 85 67 20

TABLE 28 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs10015979 (nucleobases 49084 to 49104 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 49084 49102 476470 Major (8) AGCTAGGTTAAAGAGTCAC 55 74 248 49085 49103 476450 Major (9) CAGCTAGGTTAAAGAGTCA 44 5 249 49086 49104 *435862 Major (10) GCAGCTAGGTTAAAGAGTC 56 49 22

TABLE 29 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs7691627 (nucleobases 51052 to 51072 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 51052 51070 476467 Major (8) TAAGAAACACAATCAAAGA 45 21 250 51053 51071 476445 Major (9) ATAAGAAACACAATCAAAG 34 1 251 51054 51072 *435880 Major (10) AATAAGAAACACAATCAAA 68 7 24

TABLE 30 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs6446723 (nucleobases 66455 to 66475 of SEQ ID NO: 1) % % inhibition inhibition Start Stop Target in in SEQ Site Site ISIS No allele Sequence GM04281 GM02171 ID NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 66455 66473 476463 Major (8) ATTTTCTAGACTTTATGAT 37 7 252 66456 66474 476440 Major (9) AATTTTCTAGACTTTATGA 57 0 253 66457 66475 *435875 Major (10) TAATTTTCTAGACTTTATG 42 0 30

TABLE 31 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and a chimeric antisense oligonucleotide targeted to SNP rs363064 (nucleobases 81053 to 81071 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 81053 81071 476461 Major (9) GAGAATACGGGTAACATTT 87 62 254

TABLE 32 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs11731237 (nucleobases 91455 to 91475 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 91455 91473 476468 Major (8) TGGGCAGGAAGGACTGAAC 58 56 255 91456 91474 476448 Major (9) GTGGGCAGGAAGGACTGAA 61 69 256 91457 91475 *435884 Major (10) GGTGGGCAGGAAGGACTGA 59 49 68

TABLE 33 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs4690073 (nucleobases 99792 to 99812 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 99792 99810 *435337 Major (8) CCTAAATCAATCTACAAGT 69 7 70 99793 99811 476446 Major (9) CCCTAAATCAATCTACAAG 61 0 257 99794 99812 *435301 Major (10) TCCCTAAATCAATCTACAA 63 1 72

TABLE 34 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs34315806 (nucleobases 101676 to 101696 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 101676 101694 463569 Major (8) CTTTTCCGTGCTGTTCTGA 96 95 258 101677 101695 463572 Major (9) ACTTTTCCGTGCTGTTCTG 93 91 259 101678 101696 463567 Major (10) AACTTTTCCGTGCTGTTCT 98 97 260

TABLE 35 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs363099 (nucleobases 101698 to 101718 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 101698 101716 *435339 Major (8) CTGAGCGGAGAAACCCTCC 94 85 80 101699 101717 476458 Major (9) GCTGAGCGGAGAAACCCTC 92 79 261 101700 101718 *435303 Major (10) GGCTGAGCGGAGAAACCCT 96 93 82

TABLE 36 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs363096 (nucleobases 119663 to 119683 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 119663 119681 *435340 Major (8) TTCCCTAAAAACAAAAACA 42 21 85 119664 119682 476451 Major (9) ATTCCCTAAAAACAAAAAC 0 0 262 119665 119683 *435304 Major (10) GATTCCCTAAAAACAAAAA 41 27 87

TABLE 37 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs2298967 (nucleobases 125389 to 125409 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 125389 125407 *435341 Major (8) CTTTTCTATTGTCTGTCCC 83 65 89 125390 125408 476459 Major (9) GCTTTTCTATTGTCTGTCC 89 82 263 125391 125409 *435305 Major (10) TGCTTTTCTATTGTCTGTC 92 85 91

TABLE 38 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and a chimeric antisense oligonucleotide targeted to SNP rs2298969 (nucleobases 125888 to 125906 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 125888 125906 *435890 Minor (10) AAGGGATGCTGACTTGGGC 91 64 94

TABLE 39 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs6844859 (nucleobases 130128 to 130148 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 130128 130146 476466 Major (8) CTTCCTCACTGAGGATGAA 87 64 264 130129 130147 476444 Major (9) CCTTCCTCACTGAGGATGA 92 77 265 130130 130148 *435876 Major (10) ACCTTCCTCACTGAGGATG 94 87 95

TABLE 40 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs363092 (nucleobases 135671 to 135691 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 135671 135689 476464 Major (8) AACCACTTTGGGATGAATA 51 71 266 135672 135690 476442 Major (9) AAACCACTTTGGGATGAAT 58 59 267 135673 135691 *435897 Minor (10) CAAACCACTTTGGGATGAA 48 78 98

TABLE 41 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs363088 (nucleobases 149972 to 149992 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 149972 149990 476476 Major (8) ACAGCTATCTTCTCATCAA 90 65 268 149973 149991 476460 Major (9) CACAGCTATCTTCTCATCA 86 39 269 149974 149992 *435871 Major (10) TCACAGCTATCTTCTCATC 91 54 101

TABLE 42 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs916171 (nucleobases 156457 to 156477 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop in in ID Site Site ISIS No Target allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 156457 156475 476465 Major (8) GAACAAAGAGAAGAATTTC 38 0 270 156458 156476 476443 Major (9) AGAACAAAGAGAAGAATTT 58 0 271 156459 156477 *435881 Major (10) CAGAACAAAGAGAAGAATT 59 16 105

TABLE 43 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362275 (nucleobases 164244 to 164264 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop in in ID Site Site ISIS No Target allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 164244 164262 476473 Major (8) GAAGCCTGATAAAATCTCT 83 51 272 164245 164263 476454 Major (9) AGAAGCCTGATAAAATCTC 79 61 273 164246 164264 *435868 Major (10) AAGAAGCCTGATAAAATCT 69 56 111

TABLE 44 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362273 (nucleobases 167061 to 167081 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 167061 167079 463568 Major (8) TGATCTGTAGCAGCAGCTT 96 78 274 167062 167080 463571 Major (9) TTGATCTGTAGCAGCAGCT 95 86 275 167063 167081 463566 Major (10) GTTGATCTGTAGCAGCAGC 94 78 276

TABLE 45 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362272 (nucleobases 174622 to 174642 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 174622 174640 *435344 Major (8) TAGAGGACGCCGTGCAGGG 78 63 120 174623 174641 476456 Major (9) ATAGAGGACGCCGTGCAGG 87 60 277 174624 174642 *435308 Major (10) CATAGAGGACGCCGTGCAG 76 48 122

TABLE 46 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362271 (nucleobases 175160 to 175180 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop in in ID Site Site ISIS No Target allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 175160 175178 476472 Major (8) GTGTGTACAGAACCTGCCG 85 52 278 175161 175179 476453 Major (9) CGTGTGTACAGAACCTGCC 88 69 279 175162 175180 *435867 Major (10) ACGTGTGTACAGAACCTGC 91 80 125

TABLE 47 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs3775061 (nucleobases 178396 to 178416 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 178396 178414 476475 Major (8) TTCAGAATGCCTCATCTGG 61 1 280 178397 178415 476457 Major (9) GTTCAGAATGCCTCATCTG 80 50 281 178398 178416 *435873 Major (10) TGTTCAGAATGCCTCATCT 80 43 127

TABLE 48 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362296 (nucleobases 186649 to 1786669 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Target in in ID Site Site ISIS No allele Sequence GM04281 GM02171 NO 145466 145485 387916 n/a TCTCTATTGCACATTCCAAG 100 99 6 186649 186667 476469 Major (8) GGACAGGGTGTGCTCTCCG 80 58 282 186650 186668 476449 Major (9) GGGACAGGGTGTGCTCTCC 80 64 283 186651 186669 *435882 Major (10) GGGGACAGGGTGTGCTCTC 61 61 155

Example 8: Dose-Dependent Antisense Inhibition of Human Huntingtin mRNA Levels in Coriell Fibroblast Cell Lines

Gapmers from the studies described in Example 7 were selected and tested at various doses in GM04281, GM02171, and GM02173B cell lines. Each cell line was plated at a density of 25,000 cells per well and transfected using electroporation with 750 nM, 1,500 nM, 3,000 nM, 6,000 nM, and 12,000 nM concentrations of antisense oligonucleotide, as specified in Tables 49, 50, and 51. After a treatment period of approximately 16 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. Human HTT primer probe set RTS2617 was used to measure mRNA levels. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells. IC50 values are also provided in Tables 49, 50, and 51.

TABLE 49 Dose-dependent antisense inhibition of human HTT in GM04281 cells ISIS IC50 No. 750 nM 1500 nM 3000 nM 6000 nM 12000 nM (μM) 387916 67 88 95 97 99 <0.8 463566 25 65 79 88 95 1.5 463567 34 73 90 93 98 1.1 463568 33 56 75 87 92 1.3 463571 32 21 70 90 93 1.4 476441 11 27 50 70 87 3.1 476444 20 31 68 49 93 2.3 476449 4 28 34 47 77 4.9 476453 21 21 48 73 85 2.7 476455 5 19 34 56 80 4.6 476458 36 72 83 93 96 1.1 476459 23 59 75 85 91 1.5 476469 17 27 47 47 67 5.5 476473 0 6 32 50 68 6.2 476476 3 7 32 53 86 4.9

TABLE 50 Dose-dependent antisense inhibition of human HTT in GM02171 cells ISIS IC50 No. 750 nM 1500 nM 3000 nM 6000 nM 12000 nM (μM) 387916 59 79 93 98 98 <0.8 463566 4 33 42 62 79 3.8 463567 38 41 69 85 94 1.5 463568 21 26 41 58 64 4.8 463571 8 23 56 63 75 3.7 476441 0 13 7 0 12 >12.0 476444 11 0 0 67 59 8.8 476449 4 27 37 51 63 5.8 476453 6 40 40 51 73 4.9 476455 32 15 18 47 61 7.8 476458 42 54 71 86 84 1.2 476459 22 38 70 44 73 4.3 476469 7 24 30 56 58 7.8 476473 4 10 15 33 43 >12.0 476476 5 16 18 23 41 >12.0

TABLE 51 Dose-dependent antisense inhibition of human HTT in GM02171 cells ISIS IC50 No. 750 nM 1500 nM 3000 nM 6000 nM 12000 nM (μM) 387916 66 89 95 97 99 <0.8 463566 32 55 76 77 93 1.3 463567 51 61 87 94 97 0.7 463568 26 23 72 87 94 1.6 463571 32 34 60 86 94 1.9 476441 18 18 27 47 44 >12.0 476444 15 0 31 51 58 7.1 476449 27 33 56 80 81 2.6 476453 24 28 55 75 83 2.7 476455 24 26 52 55 73 3.7 476458 63 77 87 89 94 0.2 476459 37 55 56 62 86 1.5 476469 22 41 40 63 76 2.9 476473 7 28 33 51 73 5.0 476476 11 29 26 55 69 4.6

Example 9: Antisense Inhibition of Human HTT in GM04281 Cells by Oligonucleotides Designed by Microwalk

Additional gapmers were designed based on the gapmers selected from studies described in Example 4. These gapmers were designed by creating gapmers shifted slightly upstream and downstream (i.e. “microwalk”) of the original gapmers from Tables 8, 9, and 10. Gapmers were also created with 3-9-3 or 5-9-5 motifs, and with constrained 6(S)—CH3-bicyclic nucleic acid (BNA) molecules at various nucleoside positions.

These gapmers were tested in vitro. Cultured GM04281 cells at a density of 25,000 cells per well were transfected using electroporation with 5,000 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells.

The chimeric antisense oligonucleotides in Tables 52-56 were designed as 3-9-3 or 5-9-5 gapmers. The parent gapmers, ISIS 435869, ISIS 435870, ISIS 435874, ISIS 435879, and ISIS 435890, from which the newly designed gapmers were derived are marked with an asterisk (*) in the table. ISIS 387916 was included in the study as a benchmark oligonucleotide against which the potency of the antisense oligonucleotides targeting nucleotides overlapping each SNP position could be compared.

The 3-9-3 gapmers are 15 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleosides and is flanked on both 5′ and 3′ directions by wings comprising 3 sugar modified nucleosides each.

The 5-9-5 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleosides and is flanked on both 5′ and 3′ directions by wings comprising 5 sugar modified nucleosides each.

The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine nucleobases throughout each gapmer are 5-methylcytosines. Bolded and underlined nucleotides in Tables 52-56 indicate the positions of the 6(S)—CH3-BNA molecules (e.g. cEt molecules) in each gapmer. Italicized nucleotides are MOE subunits.

“Start site” indicates the 5′-most nucleotide to which the gapmer is targeted. “Stop site” indicates the 3′-most nucleotide to which the gapmer is targeted. ‘Target allele’ indicates whether the gapmer is targeted to the major or the minor allele. The number in parentheses indicates the position on the oligonucleotide opposite to the SNP position.

TABLE 52 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs4690072 (nucleobases 62147 to 62173 of SEQ ID NO: 1) Start Stop Target % SEQ Site Site allele ISIS No. Sequence Motif inhibition ID NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 97 6 62147 62165 Major (6) 460266 GTGCTACCCAACCTTTCTG 5-9-5 63 169 62151 62169 Major (10) *435874 CACAGTGCTACCCAACCTT 5-9-5 50 28 62151 62169 Major (10) 460213 CACAGTGCTACCCAACCTT 5-9-5 22 28 62151 62169 Major (10) 460220 CACAGTGCTACCCAACCTT 5-9-5 24 28 62151 62169 Major (10) 460221 CACAGTGCTACCCAACCTT 5-9-5 28 28 62153 62167 Major (8) 460208 CAGTGCTACCCAACC 3-9-3 81 177 62155 62173 Major (14) 460267 ATATCACAGTGCTACCCAA 5-9-5 37 180

TABLE 53 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs2298969 (nucleobases 125884 to 125910 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 97 6 125884 125902 Minor (6) 460233 GATGCTGACTTGGGCCATT 5-9-5 76 182 125888 125906 Minor (10) *435890 AAGGGATGCTGACTTGGGC 5-9-5 75 94 125888 125906 Minor (10) 460215 AAGGGATGCTGACTTGGGC 5-9-5 26 94 125888 125906 Minor (10) 460224 AAGGGATGCTGACTTGGGC 5-9-5 38 94 125888 125906 Minor (10) 460225 AAGGGATGCTGACTTGGGC 5-9-5 49 94 125890 125904 Minor (8) 460210 GGGATGCTGACTTGG 3-9-3 97 189 125892 125910 Minor (14) 460229 TGCCAAGGGATGCTGACTT 5-9-5 60 192

TABLE 54 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs7685686 (nucleobases 146782 to 146808 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 97 6 146782 146800 Major (6) 460232 AATTGTCATCACCAGAAAA 5-9-5 82 195 146786 146804 Major (10) *435879 AATAAATTGTCATCACCAG 5-9-5 84 99 146786 146804 Major (10) 460214 AATAAATTGTCATCACCAG 5-9-5 33 99 146786 146804 Major (10) 460222 AATAAATTGTCATCACCAG 5-9-5 87 99 146786 146804 Major (10) 460223 AATAAATTGTCATCACCAG 5-9-5 75 99 146788 146802 Major (8) 460209 TAAATTGTCATCACC 3-9-3 96 203 146790 146808 Major (14) 460228 TATTAATAAATTGTCATCA 5-9-5 0 206

TABLE 55 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362331 (nucleobases 155475 to 155501 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 97 6 155475 155493 Major (6) 460231 ACAGTAGATGAGGGAGCAG 5-9-5 88 209 155479 155497 Major (10) *435870 GCACACAGTAGATGAGGGA 5-9-5 86 103 155479 155497 Major (10) 460212 GCACACAGTAGATGAGGGA 5-9-5 89 103 155479 155497 Major (10) 460218 GCACACAGTAGATGAGGGA 5-9-5 90 103 155479 155497 Major (10) 460219 GCACACAGTAGATGAGGGA 5-9-5 88 103 155481 155495 Major (8) 460207 ACACAGTAGATGAGG 3-9-3 89 217 155483 155501 Major (14) 460227 AAGTGCACACAGTAGATGA 5-9-5 45 220

TABLE 56 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362306 (nucleobases 181740 to 181766 of SEQ ID NO: 1) SEQ Start Stop Target % ID Site Site allele ISIS No. Sequence Motif inhibition NO 145466 145485 n/a 387916 TCTCTATTGCACATTCCAAG 5-10-5 97 6 181740 181758 Major (6) 460230 AGCTGCAACCTGGCAACAA 5-9-5 66 223 181744 181762 Major (10) *435869 GAGCAGCTGCAACCTGGCA 5-9-5 69 149 181744 181762 Major (10) 460211 GAGCAGCTGCAACCTGGCA 5-9-5 22 149 181744 181762 Major (10) 460216 GAGCAGCTGCAACCTGGCA 5-9-5 18 149 181744 181762 Major (10) 460217 GAGCAGCTGCAACCTGGCA 5-9-5 56 149 181746 181760 Major (8) 460206 GCAGCTGCAACCTGG 3-9-3 83 231 181748 181766 Major (14) 460226 GCAAGAGCAGCTGCAACCT 5-9-5 51 234

Example 10: Dose-Dependent Antisense Inhibition of Human Huntingtin mRNA Levels in Coriell Fibroblast Cell Lines

Gapmers from studies described in Example 9 were selected and tested at various doses in GM04281, GM02171 and GM02173B cell lines. Each cell line was plated at a density of 25,000 cells per well and transfected using electroporation with 312.5 nM, 625 nM, 1,250 nM, 2,500 nM, 5,000 nM and 10,000 nM concentrations of antisense oligonucleotide, as specified in Tables 75, 58, and 59. After a treatment period of approximately 16 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. Human HTT primer probe set RTS2617 was used to measure mRNA levels. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells. IC50 values are also provided in Tables 57, 58, and 59.

TABLE 57 Dose-dependent antisense inhibition of human HTT in GM04281 cells ISIS 312.5 1,250 2,500 IC50 No. nM 625 nM nM nM 5,000 nM 10,000 nM (μM) 387916 26 49 68 86 94 97 0.7 435869 0 0 23 48 62 82 3.2 435870 15 38 50 65 85 88 1.3 435874 14 22 32 49 65 73 2.7 435879 0 17 40 61 83 94 1.8 435890 5 13 37 56 70 82 2.3 460206 10 18 37 52 66 85 2.3 460207 20 27 50 65 80 91 1.4 460208 21 34 51 63 70 79 1.5 460209 52 74 89 94 94 95 0.2 460210 34 61 84 91 97 98 0.5 460212 13 31 50 62 75 82 1.6 460218 14 27 50 63 78 86 1.8 460219 9 32 42 64 77 87 1.6 460222 19 21 42 57 73 78 1.7 460231 12 24 41 57 71 84 1.9 460233 16 28 59 66 72 74 1.8 460266 4 17 32 48 60 75 2.9

TABLE 58 Dose-dependent antisense inhibition of human HTT in GM02171 cells 312.5 625 1,250 2,500 IC50 ISIS No. nM nM nM nM 5,000 nM 10,000 nM (μM) 387916 32 56 77 89 95 97 0.7 435869 0 6 22 40 69 84 2.9 435870 15 19 32 51 68 77 2.4 435874 0 5 1 17 17 30 >10.0 435879 0 8 0 16 36 47 15.3 435890 14 16 19 19 39 57 9.3 460206 5 13 33 41 68 80 2.7 460207 13 10 22 22 33 39 45.6 460208 13 15 11 11 15 53 10.8 460209 8 27 46 70 80 86 1.6 460210 19 37 55 75 88 96 1.1 460212 8 23 30 43 57 74 2.2 460218 15 26 27 36 52 78 3.2 460219 16 17 32 44 69 76 2.5 460222 14 3 0 0 13 0 >10.0 460231 6 8 13 16 33 56 10.4 460233 27 30 39 46 61 73 2.4 460266 0 15 20 15 18 34 >10.0

TABLE 59 Dose-dependent antisense inhibition of human HTT in GM02173B cells ISIS 312.5 1,250 2,500 IC50 No. nM 625 nM nM nM 5,000 nM 10,000 nM (μM) 387916 22 47 76 88 96 98 0.7 435869 10 0 16 38 59 76 3.9 435870 22 36 44 58 69 81 2.0 435874 11 6 25 23 32 42 >10.0 435879 0 9 21 30 52 68 4.8 435890 12 16 30 31 48 66 4.5 460206 11 13 18 35 59 74 3.5 460207 15 25 30 37 42 66 4.3 460208 5 14 27 32 52 51 9.0 460209 27 49 61 79 81 74 0.8 460210 19 40 61 77 89 95 1.0 460212 0 19 32 32 61 78 2.9 460218 4 17 26 38 64 82 3.0 460219 5 6 26 47 68 84 2.9 460222 13 19 23 30 35 50 16.1 460231 7 33 25 35 54 77 3.7 460233 11 20 37 52 68 69 2.3 460266 12 6 10 21 25 47 >10.0

Example 11: Dose-Dependent Antisense Inhibition of Human HTT in GM04281 and GM02171 Cells by Oligonucleotides Designed by Microwalk

Additional gapmers were designed based on the gapmers selected from studies described in Example 10. These gapmers were designed by creating gapmers shifted slightly upstream and downstream (i.e. “microwalk”) of the original gapmers from Tables 57, 58, and 59. Gapmers were also created with 4-9-4 MOE or 5-9-5 MOE motifs, and with constrained 6(S)—CH3-bicyclic nucleic acid (BNA) molecules at various nucleotide positions.

These gapmers were tested in the GM04281 and GM02171 cell lines. Cultured GM04281 or GM02171 cells at a density of 25,000 cells per well were transfected using electroporation with 2,500 nM or 5,000 nM antisense oligonucleotide. After a treatment period of approximately 24 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells.

The chimeric antisense oligonucleotides in Tables 60, 61, and 62 were designed as 3-9-3, 4-9-4, or 5-9-5 MOE gapmers. The parent gapmers, ISIS 435890, ISIS 460210, ISIS 435879, ISIS 460209, ISIS 435870, and ISIS 460207, from which the newly designed gapmers were derived are marked with an asterisk (*) in the table. ISIS 387916 was included in the study as a benchmark oligonucleotide against which the potency of the antisense oligonucleotides targeting nucleotides overlapping each SNP position could be compared.

The 3-9-3 gapmers are 15 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 3 nucleotides each.

The 4-9-4 gapmers are 17 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 4 nucleotides each.

The 5-9-5 gapmers are 19 nucleotides in length, wherein the central gap segment is comprised of nine 2′-deoxynucleotides and is flanked on both 5′ and 3′ directions by wings comprising 5 nucleotides each.

The internucleoside linkages throughout each gapmer are phosphorothioate (P═S) linkages. All cytosine nucleobases throughout each gapmer are 5-methylcytosines. Bolded and underlined nucleotides in Tables 60, 61, and 62 indicate the positions of the 6(S)—CH3-BNA (e.g. cEt molecules)molecules in each gapmer. Italicized nucleotides are MOE subunits.

The gapmers are organized in Tables 60, 61, and 62, according to the SNP site they target. “Start site” indicates the 5′-most nucleotide to which the gapmer is targeted. “Stop site” indicates the 3′-most nucleotide to which the gapmer is targeted. ‘Target allele’ indicates whether the gapmer is targeted to the major or the minor allele. The number in parentheses indicates the position on the oligonucleotide opposite to the SNP position.

TABLE 60 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs2298969 (nucleobases 125888 to 125907 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Concentration in in ID position position ISIS No. Sequence Motif (nM) GM04281 GM02171 NO 145466 145485 387916 TCTCTATTGCA 5-10-5 5000 57 24 6 CATTCCAAG 125888 125907 *435890 AAGGGATGCTG 5-9-5 2500 22 0 94 ACTTGGGC 5000 41 23 125890 125904 *460210 GGGATGCTGAC 3-9-3 2500 59 24 189 TTGG 5000 81 33 125889 125905 474870 AGGGATGCTG 4-9-4 2500 23 3 187 ACTTGGG 5000 44 34 125889 125905 474890 AGGGATGCTG 4-9-4 2500 38 6 187 ACTTGGG 5000 49 25 125889 125905 474910 AGGGATGCTGA 4-9-4 2500 34 8 187 CTTGGG 5000 49 41 125889 125905 474914 AGGGATGCTGA 4-9-4 2500 44 14 187 CTTGGG 5000 44 21 125888 125907 474918 AAGGGATGCT 5-9-5 2500 31 0 94 GACTTGGGC 5000 26 25 125888 125907 474922 AAGGGATGCT 5-9-5 2500 33 14 94 GACTTGGGC 5000 65 24 125889 125905 476332 AGGGATGCTG 4-9-4 2500 23 13 187 ACTTGGG 5000 51 42 125888 125907 476336 AAGGGATGCTG 5-9-5 2500 5 0 94 ACTTGGGC 5000 43 9

TABLE 61 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs7685686 (nucleobases 146786 to 146805 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Concentration in in ID position position ISIS No. Sequence Motif (nM) GM04281 GM02171 NO 145466 145485 387916 TCTCTATTGCA 5-10-5 5000 57 24 6 CATTCCAAG 146786 146805 *435879 AATAAATTGTC 5-9-5 2500 39 0 99 ATCACCAG 5000 59 19 146788 146802 *460209 TAAATTGTCAT 3-9-3 2500 3 0 203 CACC 5000 13 5 146787 146803 474871 ATAAATTGTCA 4-9-4 2500 82 32 200 TCACCA 5000 83 58 146787 146803 474891 ATAAATTGTCA 4-9-4 2500 84 29 200 TCACCA 5000 89 56 146787 146803 474911 ATAAATTGTCA 4-9-4 2500 70 18 200 TCACCA 5000 83 40 146787 146803 474915 ATAAATTGTCA 4-9-4 2500 38 9 200 TCACCA 5000 74 14 146786 146805 474919 AATAAATTGTC 5-9-5 2500 80 7 99 ATCACCAG 5000 84 37 146786 146805 474923 AATAAATTGTC 5-9-5 2500 74 32 99 ATCACCAG 5000 83 51 146787 146803 476333 ATAAATTGTCA 4-9-4 2500 75 28 200 TCACCA 5000 86 21 146786 146805 476337 AATAAATTGTC 5-9-5 2500 71 6 99 ATCACCAG 5000 83 31

TABLE 62 Comparison of inhibition of human HTT mRNA levels by ISIS 387916 and chimeric antisense oligonucleotides targeted to SNP rs362331 (nucleobases 155478 to 155498 of SEQ ID NO: 1) % % inhibition inhibition SEQ Start Stop Concentration in in ID position position ISIS No. Sequence Motif (nM) GM04281 GM02171 NO 145466 145485 387916 TCTCTATTGCAC 5-10-5 5000 57 24 6 ATTCCAAG 155479 155498 *435870 GCACACAGTAG 5-9-5 2500 19 1 103 ATGAGGGA 5000 49 34 155481 155495 *460207 ACACAGTAGAT 3-9-3 2500 0 0 217 GAGG 5000 7 8 155480 155496 474872 CACACAGTAGA 4-9-4 2500 35 9 214 TGAGGG 5000 63 37 155480 155496 474892 CACACAGTAGA 4-9-4 2500 43 16 214 TGAGGG 5000 69 31 155480 155496 474912 CACACAGTAGA 4-9-4 2500 16 9 214 TGAGGG 5000 36 6 155480 155496 474916 CACACAGTAGA 4-9-4 2500 22 5 214 TGAGGG 5000 47 7 155479 155498 474920 GCACACAGTAG 5-9-5 2500 19 0 103 ATGAGGGA 5000 43 23 155479 155498 474924 GCACACAGTAG 5-9-5 2500 29 8 103 ATGAGGGA 5000 48 22 155480 155496 476334 CACACAGTAGA 4-9-4 2500 35 7 214 TGAGGG 5000 62 32 155479 155498 476338 GCACACAGTAG 5-9-5 2500 26 9 103 ATGAGGGA 5000 40 4 155479 155495 474873 ACACAGTAGAT 4-9-4 2500 53 9 285 GAGGGA 5000 61 29 155479 155495 474893 ACACAGTAGAT 4-9-4 2500 47 5 285 GAGGGA 5000 59 30 155479 155495 474913 ACACAGTAGAT 4-9-4 2500 30 16 285 GAGGGA 5000 29 17 155479 155495 474917 ACACAGTAGAT 4-9-4 2500 23 12 285 GAGGGA 5000 40 5 155478 155497 474921 CACACAGTAGA 5-9-5 2500 28 0 212 TGAGGGAG 5000 43 23 155478 155497 474925 CACACAGTAGA 5-9-5 2500 30 9 212 TGAGGGAG 5000 61 34 155479 155495 476335 ACACAGTAGAT 4-9-4 2500 35 2 285 GAGGGA 5000 53 31 155478 155497 476339 CACACAGTAGA 5-9-5 2500 15 0 212 TGAGGGAG 5000 34 13

Example 12: Dose-Dependent Antisense Inhibition of Human Huntingtin mRNA Levels in Coriell Fibroblast Cell Lines

Gapmers from the studies described in Example 11 were selected and tested at various doses in GM04281, GM02171 and GM02173B cell lines. Each cell line was plated at a density of 25,000 cells per well and transfected using electroporation with 625 nM, 1,250 nM, 2,500 nM, 5,000 nM and 10,000 nM concentrations of antisense oligonucleotide, as specified in Tables 63, 64, and 65. After a treatment period of approximately 16 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. Human HTT primer probe set RTS2617 was used to measure mRNA levels. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells. IC50 values are also provided in Tables 63, 64, and 65.

TABLE 63 Dose-dependent antisense inhibition of human HTT in GM04281 cells IC50 ISIS No 625 nM 1250 nM 2500 nM 5000 nM 10000 nM (μM) 387916 70 83 94 96 98 <0.6 460207 51 63 83 91 93 0.5 460209 83 93 96 97 97 <0.6 460210 70 89 94 97 98 0.6 474871 94 97 96 96 95 <0.6 474873 51 73 89 94 95 0.5 474891 93 95 97 96 95 <0.6 474892 48 72 89 93 95 0.6 474911 85 92 96 95 94 <0.6 474919 89 94 95 94 96 <0.6 474922 21 47 73 86 96 1.5 474923 86 94 96 95 94 <0.6 476333 92 94 95 95 96 <0.6 476334 45 70 87 92 95 0.6 476337 83 92 95 96 96 <0.6

TABLE 64 Dose-dependent antisense inhibition of human HTT in GM02171 cells IC50 ISIS No 625 nM 1250 nM 2500 nM 5000 nM 10000 nM (μM) 387916 28 38 63 82 99 1.6 460207 16 0 20 22 55 10.0 460209 27 50 61 87 94 9.9 460210 34 60 80 86 97 0.9 474871 62 74 84 87 90 0.1 474873 13 29 61 77 89 2.2 474891 57 72 80 83 88 0.2 474892 23 26 51 68 81 2.5 474911 47 58 68 72 82 0.7 474919 44 48 65 71 83 1.1 474922 15 27 49 74 79 2.6 474923 27 53 74 79 84 1.5 476333 42 53 75 76 84 1.0 476334 20 23 58 71 87 2.3 476337 23 34 60 62 75 2.7

TABLE 65 Dose-dependent antisense inhibition of human HTT in GM02173B cells IC50 ISIS No 625 nM 1250 nM 2500 nM 5000 nM 10000 nM (μM) 387916 38 75 89 95 99 0.9 460207 13 27 52 46 63 6.5 460209 79 68 84 90 92 <0.6 460210 37 62 79 92 97 0.9 474871 74 83 87 92 89 <0.6 474873 22 32 67 72 92 1.9 474891 69 78 84 89 89 <0.6 474892 26 50 75 83 91 1.3 474911 50 66 76 86 86 0.6 474919 57 67 74 87 82 <0.6 474922 15 32 61 71 90 2.2 474923 49 67 78 83 85 0.5 476333 58 71 78 87 89 <0.6 476334 20 42 63 76 91 1.8 476337 48 63 71 79 80 0.6

Example 13: Strategy for Selection of Antisense Oligonucleotides Based on Potency and Selectivity

Gapmers from each of the studies described above were selected for further analysis based on potency and selectivity.

Potency was based on the percent inhibition of HTT mRNA achieved by the antisense oligonucleotides targeting a SNP compared to the percent inhibition of HTT mRNA achieved by the benchmark oligonucleotide, ISIS 387916.

Selectivity was based on the ability of the antisense oligonucleotides targeting a SNP to inhibit expression of the major allele and not of the minor allele. The usage of the three cell lines with different genotypes at each SNP position facilitated this process.

ISIS 460065 (5′-ATAAATTGTCATCACCAG-3′ (SEQ ID NO: 199)) is a 4-9-5 MOE gapmer targeted to SNP rs7685686 (major allele A, minor allele G) at position 9 of the oligonucleotide. The GM04281 cell line is homozygous AA at SNP position rs7685686. The GM02173B cell line is heterozygous AG at SNP position rs7685686. The GM02171 cell line is homozygous GG at SNP position rs7685686. Therefore, selectivity is shown if ISIS 460065 causes potent inhibition of HTT mRNA in GM04281, less potent inhibition of HTT mRNA in GM02173, and little to no significant inhibition of HTT mRNA in GM02171. IC50 values taken from Table 20, 21, and 22, and presented below in Table 66, confirm varying degrees of inhibition in the three cell lines, wherein expression was most reduced in the homozygous AA cell line, moderately reduced in the heterozygous AG cell line, and less reduced in the homozygous GG cell line. IC50 is the concentration of antisense oligonucleotide required for 50 percent inhibition HTT mRNA. IC50 values are in μM.

TABLE 66 Genotype of the Coriell cell lines for SNP rs7685686 and comparison of inhibition of HTT mRNA by ISIS 460065 in each cell line GM04281 GM02173B GM02171 Genotype AA AG GG IC50 with ISIS 1.1 3.6 10.3 460065

ISIS 459978 (5′-ACAGTGCTACCCAACCT-3′ (SEQ ID NO: 174)) is a 2-9-6 MOE gapmer targeted to SNP rs4690072 (major allele T, minor allele G) at position 9 of the oligonucleotide. The GM04281 cell line is homozygous TT at SNP position rs4690072. The GM02173B cell line is heterozygous TG at SNP position rs4690072. The GM02171 cell line is homozygous GG at SNP position rs4690072. Therefore, selectivity is shown if ISIS 459978 causes potent inhibition of HTT mRNA in GM04281, less potent inhibition of HTT mRNA in GM02173, and little to no significant inhibition of HTT mRNA in GM02171. IC50 values taken from Table 20, 21, and 22, and presented below in Table 67, confirm varying degrees of inhibition in the three cell lines, wherein expression was most reduced in the homozygous TT cell line, moderately reduced in the heterozygous TG cell line, and less reduced in the homozygous GG cell line. IC50 is the concentration of antisense oligonucleotide required for 50 percent inhibition HTT mRNA. IC50 values are in μM.

TABLE 67 Genotype of the Coriell cell lines for SNP rs4690072 and comparison of inhibition of HTT mRNA by ISIS 459978 in each cell line GM04281 GM02173B GM02171 Genotype TT TG GG IC50 with ISIS 2.5 8.4 12.7 459978

ISIS 460028 (5′-GAGCAGCTGCAACCTGGCA-3′ (SEQ ID NO: 149)) is a 4-11-4 MOE gapmer targeted to SNP rs362306 (major allele G, minor allele A) at position 10 of the oligonucleotide. The GM04281 cell line is homozygous GG at SNP position rs362306. The GM02173B and GM02171 cell lines are heterozygous GA at SNP position rs362306. Therefore, selectivity is shown if ISIS 460028 causes potent inhibition of HTT mRNA in GM04281 and less potent inhibition of HTT mRNA in GM02173 and GM02171. IC50 values taken from Table 20, 21, and 22, and presented below in Table 68, confirm varying degrees of inhibition between the GM04281 cell line and the GM02173B and GM02171 cell lines, wherein expression was most reduced in the homozygous GG cell line and less reduced in the heterozygous AG cell line. IC50 is the concentration of antisense oligonucleotide required for 50 percent inhibition HTT mRNA. IC50 values are in μM.

TABLE 68 Genotype of the Coriell cell lines for SNP rs362306 and comparison of inhibition of HTT mRNA by ISIS 460028 in each cell line GM04281 GM02173B GM02171 Genotype GG AG AG IC50 with ISIS 1.4 5.2 5.3 460028

Example 14: Strategy for Selection of Antisense Oligonucleotides with cEt Motifs Based on Potency and Selectivity

Gapmers from each of the studies described above were selected for further analysis based on potency and selectivity.

Potency was based on the percent inhibition of HTT mRNA achieved by the antisense oligonucleotides targeting a SNP compared to the percent inhibition of HTT mRNA achieved by the benchmark oligonucleotide, ISIS 387916.

Selectivity was based on the ability of the antisense oligonucleotides targeting a SNP to inhibit expression of the major allele and not of the minor allele. The usage of the three cell lines with different genotypes at each SNP position facilitated this process.

ISIS 460209 (5′-TAAATTGTCATCACC-3′ (SEQ ID NO: 203)) is a 3-9-3 gapmer with cEt subunits at positions 2, 3, 13, and 14, targeted to SNP rs7685686 (major allele A, minor allele G) at position 8 of the oligonucleotide. The GM04281 cell line is homozygous AA at SNP position rs7685686. The GM02173B cell line is heterozygous AG at SNP position rs7685686. The GM02171 cell line is homozygous GG at SNP position rs7685686. Therefore, selectivity is shown if ISIS 460209 causes potent inhibition of HTT mRNA in GM04281, less potent inhibition of HTT mRNA in GM02173, and little to no significant inhibition of HTT mRNA in GM02171. IC50 values taken from Table 57, 58, and 59, and presented below in Table 69, confirm varying degrees of inhibition in the three cell lines, wherein expression was most reduced in the homozygous AA cell line, moderately reduced in the heterozygous AG cell line, and less reduced in the homozygous GG cell line. IC50 is the concentration of antisense oligonucleotide required for 50 percent inhibition HTT mRNA. IC50 values are in μM.

TABLE 69 Genotype of the Coriell cell lines for SNP rs7685686 and comparison of inhibition of HTT mRNA by ISIS 460209 in each cell line GM04281 GM02173B GM02171 Genotype AA AG GG IC50 with ISIS 0.2 0.8 1.6 460209

ISIS 460208 (5′-CAGTGCTACCCAACC-3′ (SEQ ID NO: 177)) is a 3-9-3 gapmer with cEt subunits at positions 2, 3, 13, and 14, targeted to SNP rs4690072 (major allele T, minor allele G) at position 8 of the oligonucleotide. The GM04281 cell line is homozygous TT at SNP position rs4690072. The GM02173B cell line is heterozygous TG at SNP position rs4690072. The GM02171 cell line is homozygous GG at SNP position rs4690072. Therefore, selectivity is shown if ISIS 460208 causes potent inhibition of HTT mRNA in GM04281, less potent inhibition of HTT mRNA in GM02173, and little to no significant inhibition of HTT mRNA in GM02171. IC50 values taken from Table 57, 58, and 59, and presented below in Table 70, confirm varying degrees of inhibition in the three cell lines, wherein expression was most reduced in the homozygous TT cell line, moderately reduced in the heterozygous TG cell line, and less reduced in the homozygous GG cell line. IC50 is the concentration of antisense oligonucleotide required for 50 percent inhibition HTT mRNA. IC50 values are in μM.

TABLE 70 Genotype of the Coriell cell lines for SNP rs4690072 and comparison of inhibition of HTT mRNA by ISIS 460208 in each cell line GM04281 GM02173B GM02171 Genotype TT TG GG IC50 with ISIS 1.5 9.0 10.8 460208

ISIS 460206 (5′-GCAGCTGCAACCTGG-3′ (SEQ ID NO: 231)) is a 3-9-3 gapmer with cEt subunits at positions 2, 3, 13, and 14, targeted to SNP rs362306 (major allele G, minor allele A) at position 8 of the oligonucleotide. The GM04281 cell line is homozygous GG at SNP position rs362306. The GM02173B and GM02171 cell lines are heterozygous GA at SNP position rs362306. Therefore, selectivity is shown if ISIS 460206 causes potent inhibition of HTT mRNA in GM04281 and less potent inhibition of HTT mRNA in GM02173 and GM02171. IC50 values taken from Table 57, 58, and 59, and presented below in Table 71, confirm varying degrees of inhibition between the GM04281 cell line and the GM02173B and GM02171 cell lines, wherein expression was most reduced in the homozygous GG cell line and less reduced in the heterozygous AG cell line. IC50 is the concentration of antisense oligonucleotide required for 50 percent inhibition HTT mRNA. IC50 values are in μM.

TABLE 71 Genotype of the Coriell cell lines for SNP rs362306 and comparison of inhibition of HTT mRNA by ISIS 460206 in each cell line GM04281 GM02173B GM02171 Genotype GG AG AG IC50 with ISIS 2.3 2.7 2.7 460206

Example 15: Comparison of SNPs in Various Cell Lines and Mouse Models Associated with Huntington's Disease

The genotype at various SNP positions associated with Huntington's disease was compared amongst the three Corriell cell lines, used in the above Examples, as well as with the GM04022 fibroblast, the BACHD mouse model and the YAC18 mouse model.

The donor patient of the GM04022 fibroblast cell line was heterozygous at SNP position rs363125 (NCBI Entrez SNP database), harboring an A allele (adenine) and a C allele (cytosine) at nucleotide 5310 of SEQ ID NO: 2 (van Bilsen, P. H. J. et al., Human Gene Therapy. 19: 710-718, 2008). YAC18 mice were developed with a YAC transgene containing human huntingtin gene (Hodgson, et al. Hum. Mol. Genet. 5: 1875-85, 1996). BACHD mice were developed expressing a full-length mutant huntingtin gene with 97 glutamine repeats under the control of a bacterial artificial chromosome (Gray, M. et al., J. Neurosc. 28: 6182-95, 2008). The comparative genotype at the indicated SNP positions in all four cell lines and mouse models is presented in Table 72.

TABLE 72 Genotypes of the Coriell cell lines and Huntington mouse models SNP GM02171 GM02173 GM04281 GM04022 BACHD YAC18 rs3856973 AA AG GG AG GG AA rs2285086 GG AG AA AG AA GG rs7659144 CG CG CC CG CC GG rs16843804 TC TC CC CC CC TT rs2024115 GG AG AA AG AA GG rs3733217 CC CC CC CC CC CC rs10015979 AA AG GG AA AA AA rs7691627 AA AG GG AG GG AA rs2798235 GG GG GG AG GG GG rs4690072 GG TG TT TG TT GG rs6446723 CC TC TT TC TT CC rs363081 GG GG GG GG GG GG rs363080 CC CC CC TC CC CC rs363075 GG GG GG GG GG GG rs363064 TC TC CC CC CC TT rs3025849 AA AA AA AA AA AA rs363102 AA AA AA AG AA AA rs11731237 CC TC TT CC CC CC rs4690073 AA AG GG AG GG AA rs363144 TT TT TT TT TT TT rs3025838 CC CC CC CC CC CC rs34315806 TC TC CC CC CC TT rs363099 TC TC CC CC CC TT rs363096 CC TC TT CC TT CC rs2298967 TC TC TT TT TT CC rs2298969 GG AG AA AG AA GG rs6844859 CC TC TT TC TT CC rs363092 AA AC CC AC AA AA rs7685686 GG AG AA AG AA GG rs363088 TA TA AA AA AA TT rs362331 CC TC TT TC TT CC rs916171 GG GC CC GC CC GG rs362322 AA AA AA AA AA AA rs362275 TC TC CC CC CC TT rs362273 AG AG AA AA AA GG rs2276881 GG GG GG GG GG GG rs3121419 TC TC CC CC CC TT rs362272 AG GG GG GG AA rs362271 AG AG GG GG GG AA rs3775061 AG AG AA AA AA GG rs362310 TC CC CC TC CC CC rs362307 CC TC CC CC CC CC rs362306 AG AG GG GG GG AA rs362303 TC CC CC TC CC CC rs362296 AC AC AC CC CC AA

Example 16: Allele-Specific Inhibition Measured in BacHD Cortical Neurons

Antisense oligonucleotides, ISIS 460209 (5′-TAAATTGTCATCACC-3′ (SEQ ID NO: 203)), targeting SNP rs7685686 of human HTT, and ISIS 387916 (TCTCTATTGCACATTCCAAG (SEQ ID NO: 6)), and with no human or murine SNP target site, were tested for their effect on Htt protein levels in vitro. ISIS 387916 is cross-reactive with murine Htt mRNA (GENBANK Accession No. NM_010414.1, designated herein as SEQ ID NO: 286) at target start site 5763 with one mismatch. ISIS 460209 is cross-reactive with murine Htt mRNA at target start site 6866 with three mismatches.

Primary BacHD cortical neurons, which express human Htt and murine Htt, were isolated in the following way: Embryos were dissected from E15.5-E17.5 pregnant females. Cortices were dissected into ice-cold divalent-free Hank's Balanced Salt Solution (Invitrogen, 14025-134). The cortices were chopped into pieces and digested with 0.05% Trypsin-EDTA (Invitrogen, 25300-120) at 37° C. for 8 minutes. The digestion was halted by addition of complete neurobasal media (Invitrogen, 10888-022). Cells were resuspended in media and treated with DNAse I (Invitrogen, 18047-019). After titration through a 100 ul pipette tip, cells are resuspended in neurobasal media with B27 supplement (Invitrogen, 17504-044), and counted. 1.7×105 cells/well were plated in 24-well plates precoated with poly-D-lysine (BD Biosciences, 354210). Neurons were fed with 200 μl neurobasal media with B27 on the second day in vitro.

ISIS 460209 or ISIS 387916 was added to the supplementary media fed to neurons on division 2 at 0.7 μM, 1.4 μM or 1.5 μM final concentrations. Cells were harvested after 8 days with into 1 mL of media using a cell scraper. Cells were centrifuged at 2,500 rpm for 5 min at 4° C. and the pellets were resuspended in a buffer of 50 mM Tris, pH=8.0, 150 mM NaCl, 1% Igepal, 40 mM β-glycerophosphate, 10 mM NaF, 1× Roche complete protease inhibitor, 1 mM Sodium Orthovanadate and 800 μM PMSF. The lysates were centrifuged after 15 min incubation and protein concentration was measured with the DC assay (BioRad).

Protein lysates were run on low-bis gels to separate huntingtin alleles (resolving gel—2001:Acrylamide:BIS (10% acrylamide, 0.5% BIS, 375 mM Tris pH 8.8; stacking gel—4% Acrylamide-BIS(29:1), 156 mM Tris pH6.8; Running buffer—25 mM Tris, 190 mM Glycine, 0.1% SDS+10 μM beta-mercaptoethanol added fresh). After electrophoresis, proteins in the gel were transferred to a nitrocellulose membrane (Hybond-C Extra; GE Healthcare Bio-Sciences) at 90V for 40′ to allow samples to penetrate the stacking gel and then at 190V for 2.5 h to resolve proteins.

Primary antibodies specific for human Htt and murine calnexin protein were used at 1:10,000 dilutions. HRP-conjugated anti-mouse secondary antibody (1:10,000, Jackson ImmunoResearch Laboratories) was used for visualizing proteins using SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific). Protein bands were quantified using ImageJ software and normalized to calnexin levels. Protein bands were quantified using ImageJ software. Table 73 provides an estimate of the percentage inhibition relative to the negative control sample. The comparative percent inhibitions of the human Htt protein and the murine Htt protein are presented.

TABLE 73 Effect of antisense inhibition on mutant human and wild-type murine Htt protein (percent inhibition normalized to PBS control) Dose (μM) Human Murine ISIS 387916 0.7 54 38 1.4 75 58 1.5 92 88 ISIS 460209 0.2 71 35 0.4 82 41 1.5 94 56

Example 17: Dose-Dependent Antisense Inhibition of Human Huntingtin mRNA Levels in Coriell Fibroblast Cell Lines

Gapmers from the studies described in Examples, 3, 4, 10, and 12 were selected and tested at various doses in GM04281, GM02171 and GM02173B cell lines. Each cell line was plated at a density of 25,000 cells per well and transfected using electroporation with 0.4747 nM, 1.5011 nM, 4.7463 nM, 15.0079 nM 45.455 nM, 150.0527 nM, 474.4673 nM, 1,500.27 nM, 4,743.833 nM, and 15,000 nM concentrations of antisense oligonucleotide, as specified in Tables 72, 73, and 74. After a treatment period of approximately 16 hours, RNA was isolated from the cells and HTT mRNA levels were measured by quantitative real-time PCR. Human HTT primer probe set RTS2617 was used to measure mRNA levels. HTT mRNA levels were adjusted according to total RNA content, as measured by RIBOGREEN. Results are presented as percent inhibition of HTT mRNA, relative to untreated control cells. IC50 values are also provided in Tables 72, 73, and 74.

TABLE 74 Dose-dependent antisense inhibition of human HTT in GM04281 cells ISIS 0.4747 1.5011 4.7463 15.0079 47.455 150.0527 474.4673 IC50 No nM nM nM nM nM nM nM 1500.27 nM 4743.833 nM 15000.0 nM (μM) 387916 15 12 4 5 7 26 70 89 98 99 0.33 435879 0 8 19 13 24 23 45 53 84 93 0.25 435890 16 1 8 12 25 23 32 52 61 91 0.82 460209 2 9 21 17 36 46 80 89 94 93 0.09 460210 4 7 5 19 20 35 69 85 98 98 0.21 476333 7 10 8 11 42 65 86 93 93 95 0.05

TABLE 75 Dose-dependent antisense inhibition of human HTT in GM02171 cells 0.4747 1.5011 4.7463 15.0079 47.455 150.0527 IC50 ISIS No nM nM nM nM nM nM 474.4673 nM 1500.27 nM 4743.833 nM 15000.0 nM (μM) 387916 22 8 0 9 0 32 60 90 96 97 0.27 435879 0 1 6 2 0 0 8 9 46 57 7.62 435890 0 0 0 6 0 0 0 31 27 71 4.37 460209 11 5 15 0 0 7 30 69 82 88 0.96 460210 0 0 0 2 17 18 38 70 93 95 0.56 476333 0 0 0 0 13 18 44 69 72 91 0.75

TABLE 76 Dose-dependent antisense inhibition of human HTT in GM02173B cells ISIS 0.4747 1.5011 4.7463 15.0079 47.455 150.0527 IC50 No nM nM nM nM nM nM 474.4673 nM 1500.27 nM 4743.833 nM 15000.0 nM (μM) 387916 3 17 7 25 27 33 65 88 98 99 0.19 435879 0 6 0 8 3 10 16 24 50 68 3.72 435890 0 13 0 1 2 12 16 23 49 82 4.60 460209 0 7 29 2 9 32 52 71 82 86 0.27 460210 0 13 0 5 16 18 49 74 93 97 0.27 476333 11 13 20 7 23 36 63 75 83 90 0.13

Example 18: Validation of the Specificity of ISIS Oligonucleotides Targeting SNPs of Human Huntingtin by the Molecular Beacon Assay

Some of the gapmers from the study described in Example 17 were tested in GM04022 fibroblasts (from the Coriell Institute for Medical Research).

To verify allele-specific suppression of HTT mRNA in GM04022 fibroblasts by ISIS 435879, ISIS 460209, and ISIS 476333, the Molecular Beacon assay, as described in the van Bilsen at el publication (van Bilsen, P. H. J. et al., Human Gene Therapy. 19: 710-718, 2008), was conducted using ‘molecular beacon’ synthetic oligonucleotides linked with a fluorophore and quencher. GM04022 fibroblasts were transfected by electroporation with ISIS 435879, ISIS 460209, or ISIS 476333 at 0.06 μM, 0.19 μM, 0.56 μM, 1.67 μM, 5 μM and 15 μM concentrations of antisense oligonucleotide, as specified in Tables 75-77. ISIS 387916 was included in the assay as a benchmark oligonucleotide. The qRT-PCR assay for molecular beacon for the A allele was conducted with the annealing temperature at 56.5° C. The qRT-PCR assay for molecular beacon for the C allele was conducted with the annealing temperature at 62.0° C. Primer probe set RTS2617 was used to measure the total HTT mRNA reduction. The results of the assay are presented in Tables 77-79 as percent inhibition over the PBS control. The results demonstrate that the SNP-specific ISIS oligonucleotides specifically target the C allele of rs7685686 compared to the A allele (Table 80).

TABLE 77 Dose-dependent antisense inhibition of the A allele of rs7685686 in GM04022 fibroblasts 0.19 0.56 1.67 IC50 ISIS No 0.06 μM μM μM μM 5.00 μM 15.00 μM (μM) 387916 33 40 53 90 99 98 0.56 435879 0 0 50 29 38 47 10.8 460209 14 4 54 73 81 95 0.53 476333 2 44 41 77 91 86 0.64

TABLE 78 Dose-dependent antisense inhibition of the C allele of rs7685686 in GM04022 fibroblasts 0.06 0.19 0.56 IC50 ISIS No μM μM μM 1.67 μM 5.00 μM 15.00 μM (μM) 387916 41 42 46 86 95 92 0.54 435879 0 0 75 60 68 81 2.9 460209 35 48 76 84 88 92 0.19 476333 22 60 75 84 90 93 0.15

TABLE 79 Dose-dependent antisense inhibition of total HTT mRNA in GM04022 fibroblasts 0.06 0.19 ISIS No μM μM 0.56 μM 1.67 μM 5.00 μM 15.00 μM 387916 32 59 49 89 98 99 435879 0 0 42 25 41 62 460209 26 27 54 75 84 96 476333 25 51 58 82 92 90

TABLE 80 IC50 ratio (A/C) in GM04022 fibroblasts ISIS No Ratio 387916 1.0 435879 4.2 460209 2.8 476333 4.3

Example 19: Allele-Specific Inhibition Measured in Cortical Neurons from BACHD and YAC18 Mice

In order to identify potential SNPs for screening of human allele-specific ISIS oligonucleotides, the HTT mRNA of YAC18 and BACHD mice were sequenced by the Goldengate 96SNP assay. It was determined that the BAC and YAC mice carried different alleles at several key SNP positions (Table 72) and could therefore be used as a screening tool for allele-specific knockdown. Each of the SNP positions chosen for targeting in the mouse strains were also compared to human HD chromosomes. For each target, approximately 50% of the human HD population is heterozygous for the target expressed in the BACHD mice, but not the YAC18 mice.

In order to verify the allele-specificity of the ISIS oligonucleotides (described in Examples 2, 9, 17 and 18), the antisense oligonucleotides, ISIS 460207, targeting SNP rs362331; ISIS 460209, targeting SNP rs7685686; ISIS 435879, targeting SNP rs7685686; ISIS 476333, targeting SNP rs7685686; ISIS 460210, targeting SNP rs2298969; ISIS 435874, targeting SNP rs4690072; ISIS 460208, targeting SNP rs4690072; ISIS 435331, targeting SNP rs2024115; and ISIS 435871, targeting SNP rs363088, were tested for their effect on HTT protein levels in BACHD and YAC18 cortical neurons. ISIS 387916, which has no human or murine SNP target site, was used as the benchmark. ISIS 387916 is cross-reactive with murine HTT mRNA (GENBANK Accession No. NM_010414.1, designated herein as SEQ ID NO: 286) at target start site 5763 with one mismatch. It was expected that treatment with the allele-specific antisense oligonucleotides would cause significant inhibition of HTT mRNA in the BACHD neurons and not in the YAC18 neurons. It was also expected that treatment with ISIS 387916 would cause inhibition of HTT mRNA in both sets of neurons.

YAC18 cultures were prepared from E16.5 pregnant female YAC18 (line 60, +/+) mice who had been bred with YAC18 (line 60, +/+) males. All progeny are thus homozygous YAC18 (line 60), facilitating pooled cortical cultures. BACHD E16.5 embryos were isolated from pregnant BACHD (+/−) mice who had been bred with pregnant BACHD (+/−) male mice, necessitating single pup cultures and genotyping. Single cortices were isolated, using caution to prevent cross-contamination of samples. Each dissociated cortex was used to seed 5 wells of a 6-well plate. After genotyping, only BACHD (+/−) cultures were used for ASO treatment. The antisense oligonucleotides were added to the supplementary media fed to the neurons on division 2. Cells were harvested after 8 days with into 1 mL of media using a cell scraper. Cells were centrifuged at 2,500 rpm for 5 min at 4° C. and the pellets were resuspended in a buffer of 50 mM Tris, pH=8.0, 150 mM NaCl, 1% Igepal, 40 mM β-glycerophosphate, 10 mM NaF, 1× Roche complete protease inhibitor, 1 mM Sodium Orthovanadate and 800 μM PMSF. The lysates were centrifuged after 15 min incubation and protein concentration was measured with the DC assay (BioRad).

Protein lysates were run on low-bis gels to separate huntingtin alleles (resolving gel—2001:Acrylamide:BIS (10% acrylamide, 0.5% BIS, 375 mM Tris pH 8.8; stacking gel—4% Acrylamide-BIS(29:1), 156 mM Tris pH6.8; Running buffer—25 mM Tris, 190 mM Glycine, 0.1% SDS+10 μM beta-mercaptoethanol added fresh). After electrophoresis, proteins in the gel were transferred to a nitrocellulose membrane (Hybond-C Extra; GE Healthcare Bio-Sciences) at 90V for 40′ to allow samples to penetrate the stacking gel and then at 190V for 2.5 h to resolve proteins.

Primary antibodies specific for human HTT and murine calnexin protein were used at 1:10,000 dilutions. HRP-conjugated anti-mouse secondary antibody (1:10,000, Jackson ImmunoResearch Laboratories) was used for visualizing proteins using SuperSignal West Pico Chemiluminescent Substrate (Thermo Scientific). Protein bands were quantified using ImageJ software and normalized to calnexin levels. Tables 81-91 provide the percentage inhibition relative to the untreated control sample. The percentage inhibition of human HTT protein levels in BACHD and YAC18 neurons are presented.

TABLE 81 HTT SNPs in BACHD and YAC18 mice and correlation with human HTT SNPs Allele present in % of human human Allele Allele patients patients present present with heterozgous in in high at the YAC18 BACHD CAG SNP SNP Mice Mice repeats position rs2024115 G A A 48 rs2298969 G A A 52 rs362331 C T T 49 rs363088 G T T 38 rs4690072 T A A 49 rs7685686 G A A 49

TABLE 82 Effect of antisense inhibition by ISIS 387916 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 69 81 BACHD 84 90

TABLE 83 Effect of antisense inhibition by ISIS 435331, targeting rs2024115 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 0 0 BACHD 39 43

TABLE 84 Effect of antisense inhibition by ISIS 460210, targeting rs2298969 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 31 51 BACHD 79 89

TABLE 85 Effect of antisense inhibition by ISIS 460207, targeting rs362331 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 0 0 BACHD 29 44

TABLE 86 Effect of antisense inhibition by ISIS 435871, targeting rs363088 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 0 0 BACHD 51 68

TABLE 87 Effect of antisense inhibition by ISIS 435874, targeting rs4690072 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 9 5 BACHD 30 44

TABLE 88 Effect of antisense inhibition by ISIS 460208, targeting rs4690072 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 1 8 BACHD 54 68

TABLE 89 Effect of antisense inhibition by ISIS 460209, targeting rs7685686 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 12 32 BACHD 72 83

TABLE 90 Effect of antisense inhibition by ISIS 435879, targeting rs7685686 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 0 7 BACHD 36 58

TABLE 91 Effect of antisense inhibition by ISIS 476333, targeting rs7685686 in BACHD and YAC18 neurons 500 nM 1500 nM YAC18 46 61 BACHD 89 91

Claims

1.-97. (canceled)

98. A method of selectively reducing the expression of a mutant huntingtin allele in a cell, tissue, or animal comprising administering to the cell, tissue, or animal a compound comprising a modified, single-stranded antisense oligonucleotide complementary to the mutant huntingtin allele at a position comprising a single nucleotide polymorphism, wherein the expression of the mutant huntingtin allele is selectively reduced, and wherein the single nucleotide polymorphism is of rs6446723, rs3856973, rs2285086, rs363092, rs916171, rs6844859, rs7691627, rs4690073, rs11731237, rs362296, rs10015979, rs7659144, rs363096, rs362273, rs16843804, rs362271, rs362275, rs3121419, rs362272, rs3775061, rs34315806, rs363099, rs2298967, rs363064, rs363102, rs2798235, rs363080, rs363072, rs363125, rs362303, rs362310, rs10488840, rs362325, rs35892913, rs1936032, rs2276881, rs363070, rs12502045, rs3733217, rs2857936, rs12506200, rs762855, rs363081, rs363075, rs3025849, rs6855981, rs363144, rs3025838, rs362322, rs362307, and rs1006798.

99. The method of claim 98, wherein the modified oligonucleotide consists of 15 to 20 linked nucleosides and position 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 of the modified oligonucleotide, as counted from the 5′ terminus of the modified oligonucleotide, aligns with the single nucleotide polymorphism.

100. The method of claim 99, wherein at least one nucleoside comprises a modified sugar.

101. The method of claim 98, wherein the modified oligonucleotide consists of 15 to 20 linked nucleosides and position 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, or 15 of the modified oligonucleotide, as counted from the 5′ terminus of the modified oligonucleotide, aligns with the single nucleotide polymorphism.

102. The method of claim 99, wherein at least one nucleoside comprises a modified sugar.

103. The method of claim 100, wherein the modified sugar is a high-affinity sugar modification.

104. The method of claim 101, wherein the high-affinity sugar is a bicyclic sugar.

105. The method of claim 102, wherein each bicyclic sugar comprises a 4′-CH(CH3)—O-2′ bridge.

106. The method of claim 101, wherein the at least one modified sugar comprises a 2′-O-methoxyethyl.

107. The method of claim 99, wherein the modified oligonucleotide comprises a wing-gap-wing motif.

108. The method of claim 105, wherein the wing-gap-wing motif is any of the group consisting of 2-9-6, 3-9-3, 3-9-4, 3-9-5, 4-7-4, 4-9-4, 4-9-5, 4-10-5, 4-11-4, 4-11-5, 5-7-5, 5-8-6, 5-9-3, 5-9-5, 5-10-4, 5-10-5, 6-7-6, 6-8-5, and 6-9-2.

109. The method of claim 106, wherein each nucleoside positioned in the wing segment of the modified oligonucleotide comprises a 2′-O-methoxyethyl modification.

110. The method of claim 106, wherein at least one of the wing regions comprises a 4′ to 2′ bicyclic nucleoside and at least one of the remaining wing nucleosides is a non-bicyclic 2′-modified nucleoside.

111. The method of claim 108, wherein the non-bicyclic 2′-modified nucleoside is a 2′-O-methoxyethyl nucleoside.

112. The method of claim 108, wherein the 4′ to 2′ bicyclic nucleoside is a 4′-CH(CH3)—O-2′ bicyclic nucleoside.

113. The method of claim 105, wherein at least one nucleoside comprises a modified nucleobase.

114. The method of claim 111, wherein the at least one modified nucleobase is a 5′-methylcytosine.

115. The method of claim 99, wherein the modified oligonucleotide is 95% complementary to the mutant huntingtin allele.

116. The method of claim 99, wherein the modified oligonucleotide is 100% complementary to the mutant huntingtin allele.

117. The method of claim 106, wherein at least one internucleoside linkage is a modified internucleoside linkage.

118. The method of claim 115, wherein each internucleoside linkage is a phosphorothioate internucleoside linkage.

119. A method of treating, ameliorating, or slowing the onset or progression of Huntington's Disease, comprising administering to an animal a compound comprising a modified, single-stranded antisense oligonucleotide complementary to a mutant huntingtin allele at a position on the allele comprising a single nucleotide polymorphism site, wherein the compound administered to the animal treats, ameliorates or slows the onset or progression Huntington's Disease by selectively reducing the mutant huntingtin allele, and wherein the single nucleotide polymorphism site is rs6446723, rs3856973, rs2285086, rs363092, rs916171, rs6844859, rs7691627, rs4690073, rs11731237, rs362296, rs10015979, rs7659144, rs363096, rs362273, rs16843804, rs362271, rs362275, rs3121419, rs362272, rs3775061, rs34315806, rs363099, rs2298967, rs363064, rs363102, rs2798235, rs363080, rs363072, rs363125, rs362303, rs362310, rs10488840, rs362325, rs35892913, rs1936032, rs2276881, rs363070, rs12502045, rs3733217, rs2857936, rs12506200, rs762855, rs363081, rs363075, rs3025849, rs6855981, rs363144, rs3025838, rs362322, rs362307, and rs1006798.

Patent History
Publication number: 20180291453
Type: Application
Filed: Dec 18, 2017
Publication Date: Oct 11, 2018
Applicants: Ionis Pharmaceuticals, Inc. (Carlsbad, CA), The University of British Columbia (Vancouver)
Inventors: C. Frank Bennett (Carlsbad, CA), Michael Hayden (Vancouver), Susan M. Freier (San Diego, CA), Sarah Greenlee (San Diego, CA), Jeffrey Carroll (Vancouver), Simon Warby (Victoria), Eric E. Swayze (Encinitas, CA)
Application Number: 15/846,039
Classifications
International Classification: C12Q 1/6883 (20060101); C12N 15/113 (20060101); C12Q 1/6897 (20060101);