TARGETING OLIGONUCLEOTIDES AND USES THEREOF TO MODULATE GENE EXPRESSION

Abstract
Aspects of the invention provide single stranded oligonucleotides for modulating expression of genes based on targeting of low abundance non-coding RNA transcripts. Further aspects provide compositions and kits comprising single stranded oligonucleotides for modulating expression of genes. Methods for modulating expression of genes using the single stranded oligonucleotides are also provided. Further aspects of the invention provide methods for selecting a candidate oligonucleotide for modulating expression of genes.
Description
FIELD OF THE INVENTION

The invention relates to oligonucleotide based compositions, as well as methods of using oligonucleotide based compositions for treating disease.


BACKGROUND OF THE INVENTION

Modulation of gene expression is an important tool for basic research and for treating diseases caused by defective expression (either upregulation or downregulation) of one or more genes. Obtaining specificity with respect to modulation of a target gene as well as achieving sufficient modulation (e.g., sufficient upregulation or downregulation) to obtain a desired result, e.g., treatment of disease, remains a challenge. Additionally, limited approaches are available for increasing the expression of genes.


SUMMARY OF THE INVENTION

Aspects of the invention disclosed herein provide methods and compositions that are useful for modulating (e.g., upregulating) expression of a target gene in cells. In some embodiments, single stranded oligonucleotides are provided that target a low-abundance non-coding RNA (lancRNA) of a target gene, e.g., encoding a protein of interest. In some embodiments, single stranded oligonucleotides are provided that target a lancRNA of a target gene (e.g., a human gene) and thereby cause modulation (e.g., upregulation) of the gene. In some embodiments, the target gene is a gene listed in Table 1. In some embodiments, these single stranded oligonucleotides modulate (e.g., activate or enhance) expression of a target gene by degrading the lancRNA or blocking the activity of the lancRNA. In some embodiments, these single stranded oligonucleotides modulate (e.g., activate or enhance) expression of a target gene to treat a disease or condition associated with reduced expression of the target gene. In some embodiments, the disease or condition associated with reduced expression of the target gene is listed is Table 2.


Further aspects of the invention provide methods for selecting oligonucleotides for modulating (e.g., activating or enhancing) expression of a target gene. In some embodiments, the target gene may be a target gene listed in Table 1, such as ABCA1, APOA1, ATP2A2, BDNF, FXN, HBA2, HBB, HBD, HBE1, HBG1, HBG2, SMN, UTRN, PTEN, MECP2, FOXP3, NFE2L2 (NRF2), THRB, NR1H4 (FXR), HAMP, ADIPOQ, PRKAA1, PRKAA2, PRKAB1, PRKAB2, PRKAG1, PRKAG2, or PRKAG3. In some embodiments, methods are provided for selecting a set of oligonucleotides that is enriched in candidates (e.g., compared with a random selection of oligonucleotides) for modulating (e.g., activating or enhancing) expression of a target gene. Accordingly, the methods may be used to establish sets of clinical candidates that are enriched in oligonucleotides that modulate (e.g., activate or enhance) expression of a target. Such libraries may be utilized, for example, to identify lead oligonucleotides for developing therapeutics to treat a disease or condition associated with reduced or enhanced expression of the target gene. In some embodiments, the disease or condition associated with reduced expression of the target gene is listed is Table 2 or otherwise disclosed herein. Furthermore, in some embodiments, oligonucleotide chemistries are provided that are useful for controlling the pharmacokinetics, biodistribution, bioavailability and/or efficacy of the single stranded oligonucleotides for modulating (e.g., activating) expression of a target gene.


In some aspects, a method of modulating expression of a target gene in cells is provided, the method comprising: delivering to the cells a single-stranded oligonucleotide of 8 to 50 nucleotides in length that comprises a region of complementarity that is complementary with at least 5 contiguous nucleotides of a low-abundance non-coding RNA (lancRNA) that modulates expression of a target gene in the cells, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 5 kb of a transcriptional boundary of the target gene.


In some embodiments, the lancRNA is represented at a level of less than 0.01 fragments per kilobase per million mapped reads (FPKM) based sequencing of RNA of the cells. In some embodiments, the lancRNA is represented at an average copy number of less than 10 (e.g., less than 0.1 or less than 0.0001) transcripts per cell. In some embodiments, the average copy number of the lancRNA is less than 1% of the average copy number of transcripts expressed from the target gene in the cells.


In some embodiments, the lancRNA is transcribed from the same strand of the chromosomal region as the target gene. In some embodiments, the lancRNA is transcribed from the opposite strand of the chromosomal region as the target gene.


In some embodiments, the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 5 kb (e.g., within 2 kb, within 1 kb, within 500 kb or within 250 bp) of a transcriptional boundary of the target gene.


In some embodiments, the transcriptional boundary is a transcriptional start site. In some embodiments, the transcriptional boundary is a transcriptional end site.


In some embodiments, the lancRNA is no more than 200 nucleotides in length.


In some embodiments, the target gene is ABCA1, APOA1, ATP2A2, BDNF, FXN, HBA2, HBB, HBD, HBE1, HBG1, HBG2, SMN, UTRN, PTEN, MECP2, FOXP3, NFE2L2 (NRF2), THRB, NR1H4 (FXR), HAMP, ADIPOQ, PRKAA1, PRKAA2, PRKAB1, PRKAB2, PRKAG1, PRKAG2, or PRKAG3. In some embodiments, the target gene is FXN.


In some embodiments, the oligonucleotide does not comprise three or more consecutive guanosine nucleotides. In some embodiments, the oligonucleotide does not comprise four or more consecutive guanosine nucleotides.


In some embodiments, the oligonucleotide is 8 to 30 nucleotides in length. In some embodiments, the oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the lancRNA are cytosine or guanosine nucleotides.


In some embodiments, at least one nucleotide of the oligonucleotide is a nucleotide analogue. In some embodiments, the at least one nucleotide analogue results in an increase in Tm of the oligonucleotide in a range of 1 to 5° C. compared with an oligonucleotide that does not have the at least one nucleotide analogue.


In some embodiments, at least one nucleotide of the oligonucleotide comprises a 2′ O-methyl. In some embodiments, each nucleotide of the oligonucleotide comprises a 2′ O-methyl.


In some embodiments, wherein the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide. In some embodiments, the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide. In some embodiments, each nucleotide of the oligonucleotide is a LNA nucleotide.


In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-fluoro-deoxyribonucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-O-methyl nucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and ENA nucleotide analogues. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and LNA nucleotides. In some embodiments, the 5′ nucleotide of the oligonucleotide is a deoxyribonucleotide.


In some embodiments, the nucleotides of the oligonucleotide comprise alternating LNA nucleotides and 2′-O-methyl nucleotides. In some embodiments, the 5′ nucleotide of the oligonucleotide is a LNA nucleotide.


In some embodiments, the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one LNA nucleotide on each of the 5′ and 3′ ends of the deoxyribonucleotides.


In some embodiments, the oligonucleotide further comprises phosphorothioate internucleotide linkages between at least two nucleotides. In some embodiments, the oligonucleotide further comprises phosphorothioate internucleotide linkages between all nucleotides.


In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group. In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ thiophosphate.


In some embodiments, the oligonucleotide further comprises a biotin moiety conjugated to the 5′ nucleotide.


In some embodiments, the oligonucleotide comprises a nucleotide sequence as set for in Table 3.


Other aspects provide a single stranded oligonucleotide having a nucleotide sequence as set forth in Table 3.


In some embodiments, at least one nucleotide of the oligonucleotide comprises a 2′ O-methyl. In some embodiments, each nucleotide of the oligonucleotide comprises a 2′ O-methyl.


In some embodiments, the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide. In some embodiments, the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide. In some embodiments, each nucleotide of the oligonucleotide is a LNA nucleotide.


In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-fluoro-deoxyribonucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-O-methyl nucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and ENA nucleotide analogues. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and LNA nucleotides. In some embodiments, the 5′ nucleotide of the oligonucleotide is a deoxyribonucleotide.


In some embodiments, the nucleotides of the oligonucleotide comprise alternating LNA nucleotides and 2′-O-methyl nucleotides. In some embodiments, the 5′ nucleotide of the oligonucleotide is a LNA nucleotide.


In some embodiments, the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one LNA nucleotide on each of the 5′ and 3′ ends of the deoxyribonucleotides.


In some embodiments, the oligonucleotide further comprises phosphorothioate internucleotide linkages between at least two nucleotides.


In some embodiments, the oligonucleotide further comprises phosphorothioate internucleotide linkages between all nucleotides.


In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group.


In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ thiophosphate.


In some embodiments, the oligonucleotide further comprises a biotin moiety conjugated to the 5′ nucleotide.


In some embodiments, the modification pattern for the oligonucleotide is the modification pattern provided in Table 3.


Other aspects relate to a composition comprising a single stranded oligonucleotide as described herein, such as in any embodiment described above, and a carrier. In some embodiments, the carrier is a peptide. In some embodiments, the carrier is a steroid. In some embodiments, the oligonucleotide is conjugated to the carrier.


Yet other aspects relate to a composition comprising a single stranded oligonucleotide as described herein, such as in any embodiment described above, in a buffered solution.


In another aspect a pharmaceutical composition is provided comprising a composition as described herein, such as in any embodiment described above, and a pharmaceutically acceptable carrier.


In yet another aspect a kit is provided comprising a container housing a composition as described herein, such as in any embodiment described above.


In other aspects, a method of modulating expression of a target gene in cells is provided, the method comprising:


i) determining presence of a low-abundance non-coding RNA (lancRNA) in cells; and


ii) based on the determination made in i), delivering to the cells a single-stranded oligonucleotide of 8 to 50 nucleotides in length that comprises a region of complementarity that is complementary with at least 5 contiguous nucleotides of a lancRNA that modulates expression of a target gene in the cells, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 5 kb of a transcriptional boundary of the target gene.


In some embodiments, in step i) the lancRNA is determined to be present at a level of less than 0.01 fragments per kilobase per million mapped reads (FPKM) based sequencing of RNA of the cells. In some embodiments, in step i) the lancRNA is determined to be present at an average copy number of less than 10 (e.g., less than 0.1 or less than 0.0001) transcripts per cell. In some embodiments, in step i) the lancRNA is determined to be present at less than 1% of the average copy number of transcripts expressed from the target gene in the cells.


In another aspect, a method of modulating expression of a target gene in cells is provided, the method comprising: delivering to the cells a single-stranded oligonucleotide of 8 to 50 nucleotides in length that comprises a region of complementarity that is complementary with at least 5 contiguous nucleotides of a chromosomal region that corresponds to a 3′ UTR of the target gene, wherein the at least 5 contiguous nucleotides are antisense to the target gene. In some embodiments, the method comprises delivering to the cells a single-stranded oligonucleotide of 8 to 50 nucleotides in length that comprises a region of complementarity that is complementary with at least 5 contiguous nucleotides of a chromosomal region that encodes a 3′ UTR of the target gene, wherein the at least 5 contiguous nucleotides are on the opposite strand of the chromosomal region as the target gene.


In some embodiments, the target gene is ABCA1, APOA1, ATP2A2, BDNF, FXN, HBA2, HBB, HBD, HBE1, HBG1, HBG2, SMN, UTRN, PTEN, MECP2, FOXP3, NFE2L2 (NRF2), THRB, NR1H4 (FXR), HAMP, ADIPOQ, PRKAA1, PRKAA2, PRKAB1, PRKAB2, PRKAG1, PRKAG2, or PRKAG3. In some embodiments, the target gene is FXN.


In some embodiments, the oligonucleotide does not comprise three or more consecutive guanosine nucleotides. In some embodiments, the oligonucleotide does not comprise four or more consecutive guanosine nucleotides.


In some embodiments, the oligonucleotide is 8 to 30 nucleotides in length. In some embodiments, the oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the lancRNA are cytosine or guanosine nucleotides.


In some embodiments, at least one nucleotide of the oligonucleotide is a nucleotide analogue. In some embodiments, the at least one nucleotide analogue results in an increase in Tm of the oligonucleotide in a range of 1 to 5° C. compared with an oligonucleotide that does not have the at least one nucleotide analogue. In some embodiments, at least one nucleotide of the oligonucleotide comprises a 2′ O-methyl. In some embodiments, the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide. In some embodiments, the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide.


In some embodiments, the oligonucleotide further comprises phosphorothioate internucleotide linkages between at least two nucleotides. In some embodiments, the oligonucleotide further comprises phosphorothioate internucleotide linkages between all nucleotides.


In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group. In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ thiophosphate.


In some embodiments, a single stranded oligonucleotide provided herein comprises a fragment of at least 8 nucleotides of a nucleotide sequence as set forth in Table 3. In some embodiments, the single stranded oligonucleotide comprises or consists of a nucleotide sequence as set forth in Table 3. In some embodiments, the single stranded oligonucleotide comprises or consists of a modification pattern as set forth in Table 3. In some embodiments, one or more sequences in Table 3 are excluded, e.g., FXN-375, FXN-390, FXN-577, and FXN-578 in Table 3 are excluded.


In some embodiments, the single stranded oligonucleotide does not comprise three or more consecutive guanosine nucleotides. In some embodiments, the single stranded oligonucleotide does not comprise four or more consecutive guanosine nucleotides.


In some embodiments, the single stranded oligonucleotide is 8 to 30 nucleotides in length. In some embodiments, the single stranded oligonucleotide is up to 50 nucleotides in length. In some embodiments, the single stranded oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the lancRNA are cytosine or guanosine nucleotides.


In some embodiments, the single stranded oligonucleotide is complementary with at least 8 consecutive nucleotides of a lancRNA of a target gene, in which the nucleotide sequence of the single stranded oligonucleotide comprises one or more of a nucleotide sequence selected from the group consisting of


(a) (X)Xxxxxx, (X)xXxxxx, (X)xxXxxx, (X)xxxXxx, (X)xxxxXx and (X)xxxxxX,


(b) (X)XXxxxx, (X)XxXxxx, (X)XxxXxx, (X)XxxxXx, (X)XxxxxX, (X)xXXxxx, (X)xXxXxx, (X)xXxxXx, (X)xXxxxX, (X)xxXXxx, (X)xxXxXx, (X)xxXxxX, (X)xxxXXx, (X)xxxXxX and (X)xxxxXX,


(c) (X)XXXxxx, (X)xXXXxx, (X)xxXXXx, (X)xxxXXX, (X)XXxXxx, (X)XXxxXx, (X)XXxxxX, (X)xXXxXx, (X)xXXxxX, (X)xxXXxX, (X)XxXXxx, (X)XxxXXx (X)XxxxXX, (X)xXxXXx, (X)xXxxXX, (X)xxXxXX, (X)xXxXxX and (X)XxXxXx,


(d) (X)xxXXX, (X)xXxXXX, (X)xXXxXX, (X)xXXXxX, (X)xXXXXx, (X)XxxXXXX, (X)XxXxXX, (X)XxXXxX, (X)XxXXx, (X)XXxxXX, (X)XXxXxX, (X)XXxXXx, (X)XXXxxX, (X)XXXxXx, and (X)XXXXxx,


(e) (X)xXXXXX, (X)XxXXXX, (X)XXxXXX, (X)XXXxXX, (X)XXXXxX and (X)XXXXXx, and


(f) XXXXXX, XxXXXXX, XXxXXXX, XXXxXXX, XXXXxXX, XXXXXxX and XXXXXXx, wherein “X” denotes a nucleotide analogue, (X) denotes an optional nucleotide analogue, and “x” denotes a DNA or RNA nucleotide unit.


In some embodiments, at least one nucleotide of the oligonucleotide is a nucleotide analogue. In some embodiments, the at least one nucleotide analogue results in an increase in Tm of the oligonucleotide in a range of 1 to 5° C. compared with an oligonucleotide that does not have the at least one nucleotide analogue.


In some embodiments, at least one nucleotide of the oligonucleotide comprises a 2′ O-methyl. In some embodiments, each nucleotide of the oligonucleotide comprises a 2′ O-methyl. In some embodiments, the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide. In some embodiments, the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide. In some embodiments, each nucleotide of the oligonucleotide is a LNA nucleotide.


In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-fluoro-deoxyribonucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-O-methyl nucleotides. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and ENA nucleotide analogues. In some embodiments, the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and LNA nucleotides. In some embodiments, the 5′ nucleotide of the oligonucleotide is a deoxyribonucleotide. In some embodiments, the nucleotides of the oligonucleotide comprise alternating LNA nucleotides and 2′-O-methyl nucleotides. In some embodiments, the 5′ nucleotide of the oligonucleotide is a LNA nucleotide. In some embodiments, the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one LNA nucleotide on each of the 5′ and 3′ ends of the deoxyribonucleotides.


In some embodiments, the single stranded oligonucleotide comprises modified internucleotide linkages (e.g., phosphorothioate internucleotide linkages or other linkages) between at least two, at least three, at least four, at least five or more nucleotides. In some embodiments, the single stranded oligonucleotide comprises modified internucleotide linkages (e.g., phosphorothioate internucleotide linkages or other linkages) between all nucleotides.


In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group. In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ thiophosphate. In some embodiments, the single stranded oligonucleotide has a biotin moiety or other moiety conjugated to its 5′ or 3′ nucleotide. In some embodiments, the single stranded oligonucleotide has cholesterol, Vitamin A, folate, sigma receptor ligands, aptamers, peptides, such as CPP, hydrophobic molecules, such as lipids, ASGPR or dynamic polyconjugates and variants thereof at its 5′ or 3′ end.


According to some aspects of the invention compositions are provided that comprise any of the oligonucleotides disclosed herein, and a carrier. In some embodiments, compositions are provided that comprise any of the oligonucleotides in a buffered solution. In some embodiments, the oligonucleotide is conjugated to the carrier. In some embodiments, the carrier is a peptide. In some embodiments, the carrier is a steroid. According to some aspects of the invention pharmaceutical compositions are provided that comprise any of the oligonucleotides disclosed herein, and a pharmaceutically acceptable carrier.


According to other aspects of the invention, kits are provided that comprise a container housing any of the compositions disclosed herein.


According to some aspects of the invention, methods of modulatin (e.g., increasing) expression of a target gene in a cell are provided. In some embodiments, the methods involve delivering any one or more of the single stranded oligonucleotides disclosed herein into the cell. In some embodiments, delivery of the single stranded oligonucleotide into the cell results in a level of expression of the target gene that is greater (e.g., at least 50% greater) than a level of expression of the target gene in a control cell that does not comprise the single stranded oligonucleotide.


According to some aspects of the invention, methods of increasing levels of a target gene in a subject are provided. According to some aspects of the invention, methods of treating a disease or condition (e.g., a disease or condition provided in Table 2) associated with decreased levels of a target gene in a subject are provided. In some embodiments, the methods involve administering any one or more of the single stranded oligonucleotides disclosed herein to the subject.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1 is a diagram showing the APOA1 gene locus with oligos targeting 5′/3′ antisense regions encoding lancRNAs.



FIG. 2A is a diagram FXN gene locus with oligos targeting 3′ antisense regions encoding lancRNAs.



FIG. 2B is a diagram FXN gene locus with oligos targeting 5′ antisense regions encoding lancRNAs. The sequences correspond to SEQ ID NO: 296.



FIG. 3A is a graph showing APOA1 mRNA levels in cells treated with APOA1 oligos (oligos are those shown in Table 3, “26”=Apoa1_mus-26, “27”=Apoa1_mus-27, etc. in Table 3).



FIG. 3B is a graph showing APOA1 mRNA levels in cells treated with APOA1 oligos (oligos are those shown in Table 3, “33”=Apoa1_mus-33, “34”=Apoa1_mus-34, etc. in Table 3).



FIG. 3C is a graph showing APOA1 mRNA levels in cells treated with APOA1 oligos (oligos are those shown in Table 3, “40”=Apoa1_mus-40, “41”=Apoa1_mus-41, etc. in Table 3).



FIG. 4A is a photograph of a Western blot showing APOA1 protein levels in cells treated with APOA1 oligos (oligos are those shown in Table 3, “26”=Apoa1_mus-26, “27”=Apoa1_mus-27, etc. in Table 3).



FIG. 4B is a photograph of a Western blot showing APOA1 protein levels in cells treated with APOA1 oligos (oligos are those shown in Table 3, “33”=Apoa1_mus-33, “34”=Apoa1_mus-34, etc. in Table 3).



FIG. 4C is a photograph of a Western blot showing APOA1 protein levels in cells treated with APOA1 oligos (oligos are those shown in Table 3, “40”=Apoa1_mus-40, “41”=Apoa1_mus-41, etc. in Table 3).



FIG. 5 is a graph showing FXN mRNA levels is Sarsero fibroblasts treated with FXN oligos (oligos are those shown in Table 3, “606”=FXN-606, “607”=FXN-607, etc. in Table 3). The oligo names on the X-axis are, from left to right, 606-653 in numerical order.



FIG. 6 is a graph showing FXN mRNA levels is GM03816 cells treated with FXN oligos (oligos are those shown in Table 3, “606”=FXN-606, “607”=FXN-607, etc. in Table 3). The oligo names on the X-axis are, from left to right, 606-653 in numerical order.



FIG. 7 is a graph showing FXN mRNA levels is GM03816 cells treated with FXN oligos (oligos are those shown in Table 3, “800”=FXN-800, “801”=FXN-801, etc. in Table 3). For each oligo on the X-axis, the concentrations are, from left to right, 50 nM, 25 nM, 12.5 nM, 6.25 nM, 3.125 nM, or water. The oligo names on the X-axis are, from left to right, 800-804 in numerical order, 800-812 in numerical order, 588, 594, 40, 823-827 in numerical order, and 816-822 in numerical order.



FIG. 8 is a photograph of a Western blot showing FXN protein levels in Sarsero cells treated with FXN oligos (oligos are those shown in Table 3, “606”=FXN-606, “607”=FXN-607, etc. in Table 3).



FIG. 9 is a photograph of a Western blot showing FXN protein levels in GM03816 cells treated with FXN oligos (oligos are those shown in Table 3, “606”=FXN-606, “607”=FXN-607, etc. in Table 3).



FIG. 10A is a graph showing FXN mRNA levels is cardiomyocytes treated with FXN oligos (oligos are those shown in Table 3, “603”=FXN-603, “62”=FXN-62, etc. in Table 3).



FIG. 10B is a graph showing FXN mRNA levels is cardiomyocytes treated with FXN oligos (oligos are those shown in Table 3, “634”=FXN-634, “643”=FXN-643, etc. in Table 3).



FIG. 10C is a photograph of a Western blot showing FXN protein levels is cardiomyocytes treated with FXN oligos (oligos are those shown in Table 3, “603”=FXN-603, “607”=FXN-607, etc. in Table 3).



FIG. 11A is a graph showing FXN mRNA levels in a liver from a mouse treated with FXN oligos (oligos are those shown in Table 3, “603”=FXN-603, “607”=FXN-607, etc. in Table 3).



FIG. 11B is a graph showing FXN mRNA levels in a liver from a mouse treated with FXN oligos (oligos are those shown in Table 3, “643”=FXN-643, etc. in Table 3).



FIG. 11C is a graph showing FXN total mRNA levels a mouse treated with FXN oligos (oligos are those shown in Table 3, “603”=FXN-603, “607”=FXN-607, etc. in Table 3).



FIG. 11D is a graph showing FXN total mRNA levels a mouse treated with FXN oligos (oligos are those shown in Table 3, “643”=FXN-643, etc. in Table 3).





DETAILED DESCRIPTION OF CERTAIN EMBODIMENTS OF THE INVENTION

Aspects of the invention relate to modulation of gene expression. A considerable portion of human diseases can be treated by selectively altering protein and/or RNA levels of disease-associated transcription units (noncoding short and long RNAs, protein-coding RNAs or other regulatory coding or noncoding genomic regions).


Genomic regions encoding main RNA transcript units (e.g., genes) also produce RNA species such as PARs (promoter-associated RNAs) and TARs (termini-associated RNAs), which are a class of short (e.g., <200 nucleotides) or long noncoding RNAs expressed at low abundance at or near the 5′ and 3′ end of genes. The gene may be a protein coding gene or a gene that encodes a noncoding RNA. The low abundance noncoding RNAs (lancRNAs) from these regions can be both in sense or antisense orientation to the main transcript being produced. As described herein, single stranded oligonucleotides were designed to be complementary to chromosomal regions encoding lancRNAs, thereby targeting the lancRNAs. It was found that gene expression was modulated after administration of these oligonucleotides to cells, resulting in many instances in upregulation of genes tested (e.g., APOA1, FXN). Without wishing to be bound by theory, it is thought that targeting these lancRNAs resulted in modulation of gene expression. Again, without wishing to be bound by theory, the regulation of parent RNA behavior through these lancRNAs can be through various mechanisms, including, but not limited to, transcriptional mechanisms, splicing mechanisms, posttranscriptional mechanisms and mechanisms affecting translation efficiency and levels. The chromosomal regions containing these lancRNAs can be +/−200 nucleotides, +/−500 nucleotides, +/−1000 nucleotides, +/−5000 nucleotides, or more, of transcriptional boundaries (e.g., 5′ and 3′ ends) of genes. As used herein, the term “low abundance noncoding RNA (lancRNA)” has its common meaning in the art and generally refers to a non-coding RNA that is present at low levels in cells, for example, at levels of less than 50 transcripts per cell. In some embodiments, a low abundance noncoding RNA has a copy number (e.g., average copy number) in a population of appropriate cells of less than 50, less than 40, less than 30, less than 20, less than 10, less than 5, less than 3, less than 1, less than 0.1, less than 0.01, less than 0.001 or less than 0.0001 transcripts per cell in the population. In some embodiments, a low abundance noncoding RNA is at a level of less than 100, less than 50, less than 40, less than 30, less than 20, less than 10, less than 1, less than 0.1, less than 0.01, less than 0.001, less than 0.0001 fragments per kilobase per million mapped reads (FPKM) based sequencing of RNA obtained from cells of an appropriate cell population. In some embodiments, a low abundance noncoding RNA is at a level of less than 100, less than 50, less than 40, less than 30, less than 20, less than 10, less than 1, less than 0.1, less than 0.01, less than 0.001, less than 0.0001 reads per kilobase per million mapped reads (RPKM) based sequencing of RNA obtained from cells of an appropriate cell population. In some embodiments, a low abundance noncoding RNA has a copy number (e.g., an average copy number) in a population of appropriate cells of less than 50%, less than 40%, less than 30%, less than 20%, less than 10%, less than 5%, less than 1%, less than 0.5%, less than 0.05%, less than 0.01% or less than 0.001%, of the average copy number of transcripts expressed from a target gene of the low abundance noncoding RNA in cells of the population. Methods for calculating FPKM, RPKM, and copy number are well known in the art (see, e.g., Hart et al. Finding the active genes in deep RNA-seq gene expression studies. BMC Genomics. 2013 Nov. 11; 14:778; and Trapnell et al. Transcript assembly and quantification by RNA-Seq reveals unannotated transcripts and isoform switching during cell differentiation. Nat Biotechnol. 2010 May; 28(5):511-5).


In some embodiments, the lancRNA has a length of no more than 1000, 500, 400, 300, or 200 nucleotides. In some embodiments, the lancRNA has a length of between 10 and 1000 nucleotides, 10 and 500 nucleotides, 10 and 400 nucleotides, 10 and 300 nucleotides, 10 and 200 nucleotides, 50 and 1000 nucleotides, 50 and 500 nucleotides, 50 and 400 nucleotides, 50 and 300 nucleotides, 50 and 200 nucleotides, 100 and 1000 nucleotides, 100 and 500 nucleotides, 100 and 400 nucleotides, 100 and 300 nucleotides, or 100 and 200 nucleotides.


In some embodiments, single stranded oligonucleotides are provided that specifically bind to, or are complementary to, a lancRNA transcribed from a genomic region that is within, spans or is in proximity to a target gene. In some embodiments, single stranded oligonucleotides are provided that specifically bind to, or are complementary to, a lancRNA that is transcribed from a chromosomal region that encompasses +/−100 nucleotides, +/−200 nucleotides, +/−300 nucleotides, +/−400 nucleotides, +/−500 nucleotides, +/−600 nucleotides, +/−700 nucleotides, +/−800 nucleotides, +/−900 nucleotides, +/−1000 nucleotides, +/−2000 nucleotides, +/−3000 nucleotides, +/−4000 nucleotides, +/−5000 nucleotides, or more, of a 5′ or 3′ end of a target gene. The lancRNA may be transcribed from the strand that is antisense to the target gene or sense to the target gene.


Accordingly, in some aspects the invention contemplates single stranded oligonucleotides that specifically bind to, or are complementary to, a sense strand or antisense strand of a chromosomal region that encompasses +/−100 nucleotides, +/−200 nucleotides, +/−300 nucleotides, +/−400 nucleotides, +/−500 nucleotides, +/−600 nucleotides, +/−700 nucleotides, +/−800 nucleotides, +/−900 nucleotides, +/−1000 nucleotides, +/−2000 nucleotides, +/−3000 nucleotides, +/−4000 nucleotides, +/−5000 nucleotides, or more, of a transcriptional boundary (e.g., a 5′ or 3′ end) of a target gene. In some embodiments, the single stranded oligonucleotide specifically binds to, or is complementary to, a region of an antisense strand (relative to the target gene) within a chromosomal region that encodes a 3′UTR of the target gene. In some embodiments, the single stranded oligonucleotide specifically binds to, or is complementary to, a region of a sense strand (relative to the target gene) within a chromosomal region that encodes a 3′UTR of the target gene. In some embodiments, the single stranded oligonucleotide specifically binds to, or is complementary to, a region of an antisense strand (relative to the target gene) within a chromosomal region that encodes a 5′UTR of the target gene. In some embodiments, the single stranded oligonucleotide specifically binds to, or is complementary to, a region of a sense strand (relative to the target gene) within a chromosomal region that encodes a 5′UTR of the target gene.


Methods for identifying transcript ends (e.g., transcriptional start sites and polyadenylation junctions) are known in the art and may be used in selecting oligonucleotides that specifically bind to lancRNAs transcribed from chromosomal regions encompassing these ends. In some embodiments, 3′ end oligonucleotides may be designed by identifying RNA 3′ ends (also referred to herein as transcription end sites) using quantitative end analysis of poly-A tails, designating a window (e.g., 200 nucleotides, 500 nucleotides, 1000 nucleotides, 2000 nucleotides, 5000 nucleotides, or more) that encompasses the 3′ end, and designing oligonucleotides that are complementary to either the sense or antisense strand relative to the target gene within the designated window. In some embodiments, 5′ end oligonucleotides may be designed by identifying 5′ start sites (also referred to herein as transcriptional start sites) using Cap analysis gene expression (CAGE), designating a window (e.g., 200 nucleotides, 500 nucleotides, 1000 nucleotides, 2000 nucleotides, 5000 nucleotides, or more) that encompasses the 5′ start site, and designing oligonucleotides that are complementary to either the sense or antisense strand relative to the target gene within the designated window. Appropriate methods are disclosed, for example, in Ozsolak et al. Comprehensive Polyadenylation Site Maps in Yeast and Human Reveal Pervasive Alternative Polyadenylation. Cell. Volume 143, Issue 6, 2010, Pages 1018-1029; Shiraki, T, et al., Cap analysis gene expression for high-throughput analysis of transcriptional starting point and identification of promoter usage. Proc Natl Acad Sci USA. 100 (26): 15776-81. 2003 Dec. 23; and Zhao, X, et al., (2011). Systematic Clustering of Transcription Start Site Landscapes. PLoS ONE (Public Library of Science) 6 (8): e23409, the contents of each of which are incorporated herein by reference. Other appropriate methods for identifying transcript start sites and polyadenylation junctions may also be used, including, for example, RNA-Paired-end tags (PET) (See, e.g., Ruan X, Ruan Y. Methods Mol Biol. 2012; 809:535-62); use of standard EST databases; RACE combined with microarray or sequencing, PAS-Seq (See, e.g., Peter J. Shepard, et al., RNA. 2011 April; 17(4): 761-772); and 3P-Seq (See, e.g., Calvin H. Jan, Nature. 2011 Jan. 6; 469(7328): 97-101; and others.


In some embodiments, the target gene is a gene provided in Table 1. In some embodiments, the transcriptional boundaries of the target gene refer to the 5′ and 3′ end of the exemplary mRNA provided in Table 1 for the target gene.









TABLE 1







Non-limiting examples of RNA transcripts for certain genes










GENE





SYMBOL
MRNA
SPECIES
GENE NAME





ABCA1
NM_013454

Mus

ATP-binding cassette, sub-family A (ABC1),





musculus

member 1


ABCA1
NM_005502

Homo

ATP-binding cassette, sub-family A (ABC1),





sapiens

member 1


ABCA4
NM_007378

Mus

ATP-binding cassette, sub-family A (ABC1),





musculus

member 4


ABCA4
NM_000350

Homo

ATP-binding cassette, sub-family A (ABC1),





sapiens

member 4


ABCB11
NM_003742

Homo

ATP-binding cassette, sub-family B





sapiens

(MDR/TAP), member 11


ABCB11
NM_021022

Mus

ATP-binding cassette, sub-family B





musculus

(MDR/TAP), member 11


ABCB4
NM_018850

Homo

ATP-binding cassette, sub-family B





sapiens

(MDR/TAP), member 4


ABCB4
NM_000443

Homo

ATP-binding cassette, sub-family B





sapiens

(MDR/TAP), member 4


ABCB4
NM_018849

Homo

ATP-binding cassette, sub-family B





sapiens

(MDR/TAP), member 4


ABCB4
NM_008830

Mus

ATP-binding cassette, sub-family B





musculus

(MDR/TAP), member 4


ABCG5
NM_022436

Homo

ATP-binding cassette, sub-family G (WHITE),





sapiens

member 5


ABCG5
NM_031884

Mus

ATP-binding cassette, sub-family G (WHITE),





musculus

member 5


ABCG8
NM_026180

Mus

ATP-binding cassette, sub-family G (WHITE),





musculus

member 8


ABCG8
NM_022437

Homo

ATP-binding cassette, sub-family G (WHITE),





sapiens

member 8


ADIPOQ
NM_009605

Mus

adiponectin, C1Q and collagen domain





musculus

containing


ADIPOQ
NM_004797

Homo

adiponectin, C1Q and collagen domain





sapiens

containing


ALB
NM_000477

Homo

albumin





sapiens




ALB
NM_009654

Mus

albumin





musculus




APOA1
NM_000039

Homo

apolipoprotein A-I





sapiens




APOA1
NM_009692

Mus

apolipoprotein A-l





musculus




APOE
NM_009696

Mus

apolipoprotein E





musculus




APOE
XM_001724655

Homo

hypothetical LOC100129500;





sapiens

apolipoprotein E


APOE
XM_001722911

Homo

hypothetical LOC100129500;





sapiens

apolipoprotein E


APOE
XM_001724653

Homo

hypothetical LOC100129500;





sapiens

apolipoprotein E


APOE
NM_000041

Homo

hypothetical LOC100129500;





sapiens

apolipoprotein E


APOE
XM_001722946

Homo

hypothetical LOC100129500;





sapiens

apolipoprotein E


ATP2A2
NM_009722

Mus

ATPase, Ca++ transporting, cardiac muscle,





musculus

slow twitch 2


ATP2A2
NM_001110140

Mus

ATPase, Ca++ transporting, cardiac muscle,





musculus

slow twitch 2


ATP2A2
NM_001135765

Homo

ATPase, Ca++ transporting, cardiac muscle,





sapiens

slow twitch 2


ATP2A2
NM_170665

Homo

ATPase, Ca++ transporting, cardiac muscle,





sapiens

slow twitch 2


ATP2A2
NM_001681

Homo

ATPase, Ca++ transporting, cardiac muscle,





sapiens

slow twitch 2


BCL2L11
NM_006538

Homo

BCL2-like 11 (apoptosis facilitator)





sapiens




BCL2L11
NM_207002

Homo

BCL2-like 11 (apoptosis facilitator)





sapiens




BCL2L11
NM_138621

Homo

BCL2-like 11 (apoptosis facilitator)





sapiens




BCL2L11
NM_207680

Mus

BCL2-like 11 (apoptosis facilitator)





musculus




BCL2L11
NM_207681

Mus

BCL2-like 11 (apoptosis facilitator)





musculus




BCL2L11
NM_009754

Mus

BCL2-like 11 (apoptosis facilitator)





musculus




BDNF
NM_001143816

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143815

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143814

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143813

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143812

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143806

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143811

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143805

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143810

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001709

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_170735

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_170734

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_170733

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_170732

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_170731

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143809

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143807

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_001143808

Homo

brain-derived neurotrophic factor





sapiens




BDNF
NM_007540

Mus

brain derived neurotrophic factor





musculus




BDNF
NM_001048141

Mus

brain derived neurotrophic factor





musculus




BDNF
NM_001048142

Mus

brain derived neurotrophic factor





musculus




BDNF
NM_001048139

Mus

brain derived neurotrophic factor





musculus




BRCA1
NM_009764

Mus

breast cancer 1





musculus




BRCA1
NM_007296

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007300

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007297

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007303

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007298

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007302

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007299

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007304

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007294

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007305

Homo

breast cancer 1, early onset





sapiens




BRCA1
NM_007295

Homo

breast cancer 1, early onset





sapiens




CD274
NM_014143

Homo

CD274 molecule





sapiens




CD274
NM_021893

Mus

CD274 antigen





musculus




CEP290
NM_025114

Homo

centrosomal protein 290 kDa





sapiens




CEP290
NM_146009

Mus

centrosomal protein 290





musculus




CFTR
NM_000492

Homo

cystic fibrosis transmembrane conductance





sapiens

regulator (ATP-binding cassette sub-family C,





member 7)


CFTR
NM_021050

Mus

cystic fibrosis transmembrane conductance





musculus

regulator homolog


EPO
NM_000799

Homo

erythropoietin





sapiens




EPO
NM_007942

Mus

erythropoietin





musculus




F7
NM_000131

Homo

coagulation factor VII (serum prothrombin





sapiens

conversion accelerator)


F7
NM_019616

Homo

coagulation factor VII (serum prothrombin





sapiens

conversion accelerator)


F7
NM_010172

Mus

coagulation factor VII





musculus




F8
NM_019863

Homo

coagulation factor VIII, procoagulant





sapiens

component


F8
NM_000132

Homo

coagulation factor VIII, procoagulant





sapiens

component


F8
NM_001161373

Mus

coagulation factor VIII





musculus




F8
NM_001161374

Mus

coagulation factor VIII





musculus




F8
NM_007977

Mus

coagulation factor VIII





musculus




FLI1
NM_002017

Homo

Friend leukemia virus integration 1





sapiens




FLI1
NM_001167681

Homo

Friend leukemia virus integration 1





sapiens




FLI1
NM_008026

Mus

Friend leukemia integration 1





musculus




FMR1
NM_008031

Mus

fragile X mental retardation syndrome 1





musculus

homolog


FMR1
NM_002024

Homo

fragile X mental retardation 1





sapiens




FNDC5
NM_001171941

Homo

fibronectin type III domain containing 5





sapiens




FNDC5
NM_153756

Homo

fibronectin type III domain containing 5





sapiens




FNDC5
NM_001171940

Homo

fibronectin type III domain containing 5





sapiens




FNDC5
NM_027402

Mus

fibronectin type III domain containing 5





musculus




FOXP3
NM_054039

Mus

forkhead box P3





musculus




FOXP3
NM_001114377

Homo

forkhead box P3





sapiens




FOXP3
NM_014009

Homo

forkhead box P3





sapiens




FXN
NM_001161706

Homo

frataxin





sapiens




FXN
NM_181425

Homo

frataxin





sapiens




FXN
NM_000144

Homo

frataxin





sapiens




FXN
NM_008044

Mus

frataxin





musculus




GCH1
NM_008102

Mus

GTP cyclohydrolase 1





musculus




GCH1
NM_000161

Homo

GTP cyclohydrolase 1





sapiens




GCH1
NM_001024070

Homo

GTP cyclohydrolase 1





sapiens




GCH1
NM_001024071

Homo

GTP cyclohydrolase 1





sapiens




GCH1
NM_001024024

Homo

GTP cyclohydrolase 1





sapiens




GCK
NM_010292

Mus

glucokinase





musculus




GCK
NM_000162

Homo

glucokinase (hexokinase 4)





sapiens




GCK
NM_033508

Homo

glucokinase (hexokinase 4)





sapiens




GCK
NM_033507

Homo

glucokinase (hexokinase 4)





sapiens




GLP1R
NM_021332

Mus

glucagon-like peptide 1 receptor; similar to





musculus

glucagon-like peptide-1 receptor


GLP1R
XM_001471951

Mus

glucagon-like peptide 1 receptor; similar to





musculus

glucagon-like peptide-1 receptor


GLP1R
NM_002062

Homo

glucagon-like peptide 1 receptor





sapiens




GRN
NM_002087

Homo

granulin





sapiens




GRN
NM_008175

Mus

granulin





musculus




HAMP
NM_021175

Homo

hepcidin antimicrobial peptide





sapiens




HAMP
NM_032541

Mus

hepcidin antimicrobial peptide





musculus




HBA2
NM_000517

Homo

hemoglobin, alpha 2; hemoglobin, alpha 1





sapiens




HBA2
NM_000558

Homo

hemoglobin, alpha 2; hemoglobin, alpha 1





sapiens




HBB
NM_000518

Homo

hemoglobin, beta





sapiens




HBB
XM_921413

Mus

hemoglobin beta chain complex





musculus




HBB
XM_903245

Mus

hemoglobin beta chain complex





musculus




HBB
XM_921395

Mus

hemoglobin beta chain complex





musculus




HBB
XM_903244

Mus

hemoglobin beta chain complex





musculus




HBB
XM_903246

Mus

hemoglobin beta chain complex





musculus




HBB
XM_909723

Mus

hemoglobin beta chain complex





musculus




HBB
XM_921422

Mus

hemoglobin beta chain complex





musculus




HBB
XM_489729

Mus

hemoglobin beta chain complex





musculus




HBB
XM_903242

Mus

hemoglobin beta chain complex





musculus




HBB
XM_903243

Mus

hemoglobin beta chain complex





musculus




HBB
XM_921400

Mus

hemoglobin beta chain complex





musculus




HBD
NM_000519

Homo

hemoglobin, delta





sapiens




HBE1
NM_005330

Homo

hemoglobin, epsilon 1





sapiens




HBG1
NM_000559

Homo

hemoglobin, gamma A





sapiens




HBG2
NM_000184

Homo

hemoglobin, gamma G





sapiens




HPRT1
NM_000194

Homo

hypoxanthine phosphoribosyltransferase 1





sapiens




IDO1
NM_008324

Mus

indoleamine 2,3-dioxygenase 1





musculus




IDO1
NM_002164

Homo

indoleamine 2,3-dioxygenase 1





sapiens




IGF1
NM_001111284

Homo

insulin-like growth factor 1 (somatomedin C)





sapiens




IGF1
NM_001111285

Homo

insulin-like growth factor 1 (somatomedin C)





sapiens




IGF1
NM_001111283

Homo

insulin-like growth factor 1 (somatomedin C)





sapiens




IGF1
NM_000618

Homo

insulin-like growth factor 1 (somatomedin C)





sapiens




IGF1
NM_001111274

Mus

insulin-like growth factor 1





musculus




IGF1
NM_010512

Mus

insulin-like growth factor 1





musculus




IGF1
NM_184052

Mus

insulin-like growth factor 1





musculus




IGF1
NM_001111276

Mus

insulin-like growth factor 1





musculus




IGF1
NM_001111275

Mus

insulin-like growth factor 1





musculus




IL10
NM_000572

Mus

interleukin 10





musculus




IL10
NM_010548

Mus

interleukin 10





musculus




IL6
NM_031168

Mus

interleukin 6





musculus




IL6
NM_000600

Homo

interleukin 6 (interferon, beta 2)





sapiens




KCNMA1
NM_002247

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, alpha





member 1


KCNMA1
NM_001161352

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, alpha





member 1


KCNMA1
NM_001014797

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, alpha





member 1


KCNMA1
NM_001161353

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, alpha





member 1


KCNMA1
NM_010610

Mus

potassium large conductance calcium-





musculus

activated channel, subfamily M, alpha





member 1


KCNMB1
NM_031169

Mus

potassium large conductance calcium-





musculus

activated channel, subfamily M, beta





member 1


KCNMB1
NM_004137

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, beta





member 1


KCNMB2
NM_028231

Mus

potassium large conductance calcium-





musculus

activated channel, subfamily M, beta





member 2


KCNMB2
NM_005832

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, beta





member 2


KCNMB2
NM_181361

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, beta





member 2


KCNMB3
NM_171829

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M beta





member 3


KCNMB3
NM_171828

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M beta





member 3


KCNMB3
NM_001163677

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M beta





member 3


KCNMB3
NM_014407

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M beta





member 3


KCNMB3
NM_171830

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M beta





member 3


KCNMB3
XM_001475546

Mus

potassium large conductance calcium-





musculus

activated channel, subfamily M, beta





member 3


KCNMB3
XM_912348

Mus

potassium large conductance calcium-





musculus

activated channel, subfamily M, beta





member 3


KCNMB4
NM_021452

Mus

potassium large conductance calcium-





musculus

activated channel, subfamily M, beta





member 4


KCNMB4
NM_014505

Homo

potassium large conductance calcium-





sapiens

activated channel, subfamily M, beta





member 4


KLF1
NM_010635

Mus

Kruppel-like factor 1 (erythroid)





musculus




KLF1
NM_006563

Homo

Kruppel-like factor 1 (erythroid)





sapiens




KLF4
NM_010637

Mus

Kruppel-Iike factor 4 (gut)





musculus




KLF4
NM_004235

Homo

Kruppel-like factor 4 (gut)





sapiens




LAMA1
NM_005559.3

Homo

laminin, alpha 1





sapiens




LAMA1
NM_008480.2

Mus

laminin, alpha 1





musculus




LDLR
NM_000527

Homo

low density lipoprotein receptor





sapiens




LDLR
NM_010700

Mus

low density lipoprotein receptor





musculus




MBNL1
NM_021038.3,

Homo

muscleblind-like splicing regulator 1



NM_020007.3,

sapiens





NM_207293.1,





NM_207294.1,





NM_207295.1,





NM_207296.1,





NM_207297.1




MBNL1
NM_001253708.1,

Mus

muscleblind-like 1 (Drosophila)



NM_001253709.1,

musculus





NM_001253710.1,





NM_001253711.1,





NM_001253713.1,





NM_020007.3




MECP2
NM_010788

Mus

methyl CpG binding protein 2





musculus




MECP2
NM_001081979

Mus

methyl CpG binding protein 2





musculus




MECP2
NM_001110792

Homo

methyl CpG binding protein 2





sapiens

(Rett syndrome)


MECP2
NM_004992

Homo

methyl CpG binding protein 2





sapiens

(Rett syndrome)


MERTK
NM_006343.2

Homo

MER proto-oncogene, tyrosine kinase





sapiens




MERTK
NM_008587.1

Mus

c-mer proto-oncogene tyrosine kinase





musculus




MSX2
NM_013601

Mus

similar to homeobox protein; homeobox,





musculus

msh-like 2


MSX2
XM_001475886

Mus

similar to homeobox protein; homeobox,





musculus

msh-like 2


MSX2
NM_002449

Homo

msh homeobox 2





sapiens




MYBPC3
NM_008653

Mus

myosin binding protein C, cardiac





musculus




MYBPC3
NM_000256

Homo

myosin binding protein C, cardiac





sapiens




NANOG
NM_024865

Homo

Nanog homeobox pseudogene 8; Nanog





sapiens

homeobox


NANOG
XM_001471588

Mus

similar to Nanog homeobox; Nanog





musculus

homeobox


NANOG
NM_028016

Mus

similar to Nanog homeobox; Nanog





musculus

homeobox


NANOG
NM_001080945

Mus

similar to Nanog homeobox; Nanog





musculus

homeobox


NF1
NM_000267

Homo

neurofibromin 1





sapiens




NF1
NM_001042492

Homo

neurofibromin 1





sapiens




NF1
NM_001128147

Homo

neurofibromin 1





sapiens




NF1
NM_010897

Mus

neurofibromatosis 1





musculus




NKX2-1
NM_001079668

Homo

NK2 homeobox 1





sapiens




NKX2-1
NM_003317

Homo

NK2 homeobox 1





sapiens




NKX2-1
XM_002344771

Homo

KK2 homeobox 1





sapiens




NKX2-1
NM_009385

Mus

KK2 homeobox 1





musculus




NKX2-1
NM_001146198

Mus

NK2 homeobox 1





musculus




PAH
NM_008777

Mus

phenylalanine hydroxylase





musculus




PAH
NM_000277

Homo

phenylalanine hydroxylase





sapiens




PTEN
NM_000314

Homo

phosphatase and tensin homolog;





sapiens

phosphatase and tensin homolog





pseudogene 1


PTEN
NM_177096

Mus

phosphatase and tensin homolog





musculus




PTEN
NM_008960

Mus

phosphatase and tensin homolog





musculus




PTGS2
NM_011198

Mus

prostaglandin-endoperoxide synthase 2





musculus




PTGS2
NM_000963

Homo

prostaglandin-endoperoxide synthase 2





sapiens

(prostaglandin G/H synthase and





cyclooxygenase)


RB1
NM_009029

Mus

retinoblastoma 1





musculus




RB1
NM_000321

Homo

retinoblastoma 1





sapiens




RPS14
NM_020600

Mus

predicted gene 6204; ribosomal protein S14





musculus




RPS14
NM_001025071

Homo

ribosomal protein S14





sapiens




RPS14
NM_005617

Homo

ribosomal protein S14





sapiens




RPS14
NM_001025070

Homo

ribosomal protein S14





sapiens




RPS19
XM_204069

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_991053

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_905004

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_001005575

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
NM_023133

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_994263

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_001481027

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_913504

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_001479631

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_902221

Mus

predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein S19;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
XM_893968
Mus
predicted gene 4327; predicted gene 8683;





musculus

similar to 40S ribosomal protein S19;





predicted gene 4510; predicted gene 13143;





predicted gene 9646; ribosomal protein 519;





predicted gene 9091; predicted gene 6636;





predicted gene 14072


RPS19
NM_001022

Homo

ribosomal protein S19 pseudogene 3;





sapiens

ribosomal protein S19


SCARB1
NM_016741

Mus

scavenger receptor class B, member 1





musculus




SCARB1
NM_001082959

Homo

scavenger receptor class B, member 1





sapiens




SCARB1
NM_005505

Homo

scavenger receptor class B, member 1





sapiens




SERPINF1
NM_011340

Mus

serine (or cysteine) peptidase inhibitor,





musculus

clade F, member 1


SERPINF1
NM_002615

Homo

serpin peptidase inhibitor, clade F (alpha-2





sapiens

antiplasmin, pigment epithelium derived





factor), member 1


SIRT1
NM_001159590

Mus

sirtuin 1 (silent mating type information





musculus

regulation 2, homolog) 1 (S. cerevisiae)


SIRT1
NM_019812

Mus

sirtuin 1 (silent mating type information





musculus

regulation 2, homolog) 1 (S. cerevisiae)


SIRT1
NM_001159589

Mus

sirtuin 1 (silent mating type information





musculus

regulation 2, homolog) 1 (S. cerevisiae)


SIRT1
NM_012238

Homo

sirtuin (silent mating type information





sapiens

regulation 2 homolog) 1 (S. cerevisiae)


SIRT1
NM_001142498

Homo

sirtuin (silent mating type information





sapiens

regulation 2 homolog) 1 (S. cerevisiae)


SIRT6
NM_016539

Homo

sirtuin (silent mating type information





sapiens

regulation 2 homolog) 6 (S. cerevisiae)


SIRT6
NM_001163430

Mus

sirtuin 6 (silent mating type information





musculus

regulation 2, homolog) 6 (S. cerevisiae)


SIRT6
NM_181586

Mus

sirtuin 6 (silent mating type information





musculus

regulation 2, homolog) 6 (S. cerevisiae)


SMAD7
NM_005904

Homo

SMAD family member 7





sapiens




SMAD7
NM_001042660

Mus

MAD homolog 7 (Drosophila)





musculus




SMN1
NM_000344.3

Homo

Survival Motor Neuron 1





sapiens




SMN1
NM_022874.2

Homo

Survival Motor Neuron 1





sapiens




SMN2
NM_017411.3

Homo

Survival Motor Neuron 2



NM_022875.2

sapiens





NM_022876.2





NM_022877.2




SSPN
NM_001135823.1,

Homo

sarcospan



NM_005086.4

sapiens




SSPN
NM_010656.2

Homo

sarcospan





sapiens




ST7
NM_021908

Homo

suppression of tumorigenicity 7





sapiens




ST7
NM_018412

Homo

suppression of tumorigenicity 7





sapiens




STAT3
NM_213660

Mus

similar to Stat3B; signal transducer and





musculus

activator of transcription 3


STAT3
XM_001474017

Mus

similar to Stat3B; signal transducer and





musculus

activator of transcription 3


STAT3
NM_213659

Mus

similar to Stat3B; signal transducer and





musculus

activator of transcription 3


STAT3
NM_011486

Mus

similar to Stat3B; signal transducer and





musculus

activator of transcription 3


STAT3
NM_213662

Homo

signal transducer and activator of





sapiens

transcription 3 (acute-phase response factor)


STAT3
NM_003150

Homo

signal transducer and activator of





sapiens

transcription 3 (acute-phase response factor)


STAT3
NM_139276

Homo

signal transducer and activator of





sapiens

transcription 3 (acute-phase response factor)


UTRN
NM_007124

Homo

utrophin





sapiens




UTRN
NM_011682

Mus

utrophin





musculus




NFE2L2
NM_001145412.2,

Homo

nuclear factor, erythroid 2-like 2



NM_001145413.2,

sapiens





NM_006164.4




NFE2L2
NM_010902.3

Mus

nuclear factor, erythroid 2-like 2





musculus




ACTB
NM_001101.3

Homo

actin, beta





sapiens




ACTB
NM_007393.3

Mus

actin, beta





musculus




ANRIL
NR_003529.3,

Homo

CDKN2B antisense RNA 1



NR_047532.1,

sapiens

(also called CDKN2B)



NR_047533.1,





NR_047534.1,





NR_047535.1,





NR_047536.1,





NR_047538.1,





NR_047539.1,





NR_047540.1,





NR_047541.1,





NR_047542.1,





NR 047543.1




HOTAIR
NR_003716.3,

Homo

HOX transcript antisense RNA



NR_047517.1,

sapiens





NR_047518.1




HOT AIR
NR_047528.1

Mus

HOX transcript antisense RNA





musculus




DINO
JX993265

Homo

Damage Induced NOncoding





sapiens




DINO
JX993266

Mus

Damage Induced NOncoding





musculus




HOTTIP
NR_037843.3

Homo

HOXA distal transcript antisense RNA





sapiens




HOTTIP
NR_110441.1,

Mus

Hoxa distal transcript antisense RNA



NR 110442.1

musculus




NEST
NR_104124.1

Homo


Homo
sapiens IFNG antisense RNA 1






sapiens

(IFNG-AS1), transcript variant 1, long





non-coding RNA.


NEST
NR_104123.1

Mus

Theiler's murine encephalomyelitis virus





musculus

persistence candidate gene 1


THRB
NM_000461.4

Homo

thyroid hormone receptor, beta





sapiens




THRB
NM_001128176.2

Homo

thyroid hormone receptor, beta





sapiens




THRB
NM_001128177.1

Homo

thyroid hormone receptor, beta





sapiens




THRB
NM_001252634.1

Homo

thyroid hormone receptor, beta





sapiens




THRB
NM_00H13417.1

Mus

thyroid hormone receptor, beta





musculus




THRB
NM_009380.3

Mus

thyroid hormone receptor, beta





musculus




NR1H4
NM_001206977.1

Homo

nuclear receptor subfamily 1, group H,





sapiens

member 4


NR1H4
NM_001206978.1

Homo

nuclear receptor subfamily 1, group H,





sapiens

member 4


NR1H4
NM_001206979.1

Homo

nuclear receptor subfamily 1, group H,





sapiens

member 4


NR1H4
NM_001206992.1

Homo

nuclear receptor subfamily 1, group H,





sapiens

member 4


NR1H4
NM_001206993.1

Homo

nuclear receptor subfamily 1, group H,





sapiens

member 4


NR1H4
NM_005123.3

Homo

nuclear receptor subfamily 1, group H,





sapiens

member 4


NR1H4
NM_001163504.1

Mus

nuclear receptor subfamily 1, group H,





musculus

member 4


NR1H4
NM_001163700.1

Mus

nuclear receptor subfamily 1, group H,





musculus

member 4


NR1H4
NM_009108.2

Mus

nuclear receptor subfamily 1, group H,





musculus

member 4


PRKAA1
NM_006251.5

Homo

protein kinase, AMP-activated, alpha 1





sapiens

catalytic subunit


PRKAA1
NM_206907.3

Homo

protein kinase, AMP-activated, alpha 1





sapiens

catalytic subunit


PRKAA1
NM_001013367.3

Mus

protein kinase, AMP-activated, alpha 1





musculus

catalytic subunit


PRKAA2
NM_006252.3

Homo

protein kinase, AMP-activated, alpha 2





sapiens

catalytic subunit


PRKAA2
NM_178143.2

Mus

protein kinase, AMP-activated, alpha 2





musculus

catalytic subunit


PRKAB1
NM_006253.4

Homo

protein kinase, AMP-activated, beta 1





sapiens

non-catalytic subunit


PRKAB1
NM_031869.2

Mus

protein kinase, AMP-activated, beta 1





musculus

non-catalytic subunit


PRKAB2
NM_005399.4

Homo

protein kinase, AMP-activated, beta 2





sapiens

non-catalytic subunit


PRKAB2
NM_182997.2

Mus

protein kinase, AMP-activated, beta 2





musculus

non-catalytic subunit


PRKAG1
NM_001206709.1

Homo

protein kinase, AMP-activated, gamma 1





sapiens

non-catalytic subunit


PRKAG1
NM_001206710.1

Homo

protein kinase, AMP-activated, gamma 1





sapiens

non-catalytic subunit


PRKAG1
NM_002733.4

Homo

protein kinase, AMP-activated, gamma 1





sapiens

non-catalytic subunit


PRKAG1
NM_016781.2

Mus

protein kinase, AMP-activated, gamma 1





musculus

non-catalytic subunit


PRKAG2
NM_001040633.1

Homo

protein kinase, AMP-activated, gamma 2





sapiens

non-catalytic subunit


PRKAG2
NM_001304527.1

Homo

protein kinase, AMP-activated, gamma 2





sapiens

non-catalytic subunit


PRKAG2
NM_001304531.1

Homo

protein kinase, AMP-activated, gamma 2





sapiens

non-catalytic subunit


PRKAG2
NM_016203.3

Homo

protein kinase, AMP-activated, gamma 2





sapiens

non-catalytic subunit


PRKAG2
NM_024429.1

Homo

protein kinase, AMP-activated, gamma 2





sapiens

non-catalytic subunit


PRKAG2
NM_001170555.1

Mus

protein kinase, AMP-activated, gamma 2





musculus

non-catalytic subunit


PRKAG2
NM_001170556.1

Mus

protein kinase, AMP-activated, gamma 2





musculus

non-catalytic subunit


PRKAG2
NM_145401.2

Mus

protein kinase, AMP-activated, gamma 2





musculus

non-catalytic subunit


PRKAG3
NM_017431.2

Homo

protein kinase, AMP-activated, gamma 3





sapiens

non-catalytic subunit


PRKAG3
NM_153744.3

Mus

protein kinase, AMP-activated, gamma 3





musculus

non-catalytic subunit









Methods of modulating (e.g., upregulating or downregulating) gene expression are provided, in some embodiments, that may be carried out in vitro, ex vivo, or in vivo. It is understood that any reference to uses of compounds throughout the description contemplates use of the compound in preparation of a pharmaceutical composition or medicament for use in the treatment of a condition associated with increased or decreased levels or activity of a target gene. Thus, as one nonlimiting example, this aspect of the invention includes use of such single stranded oligonucleotides in the preparation of a medicament for use in the treatment of disease, wherein the treatment involves upregulating or downregulating expression of a target gene.


In further aspects of the invention, methods are provided for selecting a candidate oligonucleotide for modulating (e.g., upregulating or downregulating) expression of a target gene. The methods generally involve selecting as a candidate oligonucleotide, a single stranded oligonucleotide comprising a nucleotide sequence that is complementary to a lancRNA or to a chromosomal region that encodes a lancRNA, e.g., a region within 5 kb of a transcriptional boundary of a target gene. In some embodiments, sets of oligonucleotides may be selected that are enriched (e.g., compared with a random selection of oligonucleotides) in oligonucleotides that modulate (e.g., upregulate or downregulate) expression of a target gene.


Single Stranded Oligonucleotides for Modulating Expression of a Target Gene

In one aspect of the invention, single stranded oligonucleotides complementary to a lancRNA or to a chromosomal region that encodes a lancRNA, e.g., a region within 5 kb of a transcriptional boundary of a target gene, are provided for modulating expression of the target gene in a cell. In some embodiments, expression of the target gene is upregulated or increased. The oligonucleotide may be selected using any of the methods disclosed herein for selecting a candidate oligonucleotide for modulating expression of a target gene.


The single stranded oligonucleotide may comprise a region of complementarity that is complementary with a lancRNA or with a chromosomal region that encodes a lancRNA, e.g., a region within 5 kb of a transcriptional boundary of a target gene. The region of complementarity of the single stranded oligonucleotide may be complementary with at least 5, e.g., at least 6, at least 7, at least 8, at least 9, at least 10, at least 15 or more consecutive nucleotides of the lancRNA or chromosomal region that encodes the lancRNA, e.g., a region within 5 kb of a transcriptional boundary of a target gene.


The chromosomal region encoding the lancRNA may map to a position in a chromosome between 10 kilobases (e.g., 5 kb, 4, kb, 2 kb, 1 kb, 500 bp, 400 bp, 300 bp, 200 bp, 100 bp) upstream and 10 kilobases (e.g., 5 kb, 4, kb, 2 kb, 1 kb, 500 bp, 400 bp, 300 bp, 200 bp, 100 bp) downstream of a transcriptional start site of the target gene or 10 kilobases (e.g., 5 kb, 4, kb, 2 kb, 1 kb, 500 bp, 400 bp, 300 bp, 200 bp, 100 bp) upstream and 10 kilobases (e.g., 5 kb, 4, kb, 2 kb, 1 kb, 500 bp, 400 bp, 300 bp, 200 bp, 100 bp) downstream of a transcriptional end site of the target gene.


The single stranded oligonucleotide may have a sequence that does not contain guanosine nucleotide stretches (e.g., 3 or more, 4 or more, 5 or more, 6 or more consecutive guanosine nucleotides). In some embodiments, oligonucleotides having guanosine nucleotide stretches have increased non-specific binding and/or off-target effects, compared with oligonucleotides that do not have guanosine nucleotide stretches.


The single stranded oligonucleotide may have a sequence that has less than a threshold level of sequence identity with every sequence of nucleotides, of equivalent length, that map to a genomic position encompassing or in proximity to an off-target gene. For example, an oligonucleotide may be designed to ensure that it does not have a sequence that maps to genomic positions encompassing or in proximity with all known genes (e.g., all known protein coding genes) other than the target gene. In a similar embodiment, an oligonucleotide may be designed to ensure that it does not have a sequence that maps to any other known lancRNAs. In either case, the oligonucleotide is expected to have a reduced likelihood of having off-target effects. The threshold level of sequence identity may be 50%, 60%, 70%, 80%, 85%, 90%, 95%, 99% or 100% sequence identity.


The single stranded oligonucleotide may have a sequence that is has greater than 30% G-C content, greater than 40% G-C content, greater than 50% G-C content, greater than 60% G-C content, greater than 70% G-C content, or greater than 80% G-C content. The single stranded oligonucleotide may have a sequence that has up to 100% G-C content, up to 95% G-C content, up to 90% G-C content, or up to 80% G-C content. In some embodiments in which the oligonucleotide is 8 to 10 nucleotides in length, all but 1, 2, 3, 4, or 5 of the nucleotides of the complementary sequence of the lancRNA are cytosine or guanosine nucleotides. In some embodiments, the sequence of the lancRNA to which the single stranded oligonucleotide is complementary comprises no more than 3 nucleotides selected from adenine and uracil.


The single stranded oligonucleotide may be complementary to a chromosome of a different species (e.g., a mouse, rat, rabbit, goat, monkey, etc.) at a position that encompasses or that is in proximity to that species' homolog of a target gene. The single stranded oligonucleotide may be complementary to a human genomic region encompassing or in proximity to the target gene and also be complementary to a mouse genomic region encompassing or in proximity to the mouse homolog of the target gene. Oligonucleotides having these characteristics may be tested in vivo or in vitro for efficacy in multiple species (e.g., human and mouse). This approach also facilitates development of clinical candidates for treating human disease by selecting a species in which an appropriate animal exists for the disease.


According to some aspects, single stranded oligonucleotides are provided that have a region of complementarity that is complementarty with (e.g., at least 5 consecutive nucleotides of) a lancRNA of a target gene. In some embodiments, the oligonucleotide has at least one of the following features: a) a sequence that is 5′X-Y-Z, in which X is any nucleotide and in which X is at the 5′ end of the oligonucleotide, Y is a nucleotide sequence of 6 nucleotides in length that is not a human seed sequence of a microRNA, and Z is a nucleotide sequence of 1 to 23 nucleotides in length; b) a sequence that does not comprise three or more consecutive guanosine nucleotides; c) a sequence that has less than a threshold level of sequence identity with every sequence of nucleotides, of equivalent length to the second nucleotide sequence, that are between 50 kilobases upstream of a 5′-end of an off-target gene and 50 kilobases downstream of a 3′-end of the off-target gene; and d) a sequence that has greater than 60% G-C content. In some embodiments, the single stranded oligonucleotide has at least two of features a), b), c), and d), each independently selected. In some embodiments, the single stranded oligonucleotide has at least three of features a), b), c), and d), each independently selected. In some embodiments, the single stranded oligonucleotide has at least four of features a), b), c), and d), each independently selected. In some embodiments, the single stranded oligonucleotide has each of features a), b), c), and d). In certain embodiments, the oligonucleotide has the sequence 5′X-Y-Z, in which the oligonucleotide is 8-50 nucleotides in length.


In some embodiments, the region of complementarity of the single stranded oligonucleotide is complementary with 5 to 15, 6 to 15, 7 to 15, 8 to 15, 5 to 30, 6 to 30, 7 to 30, 8 to 30, 8 to 40, or 10 to 50, or 5 to 50, or 5 to 40 bases, e.g., 5, 6, 7, 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, or 50 consecutive nucleotides of a lancRNA. In some embodiments, the region of complementarity is complementary with at least 8 consecutive nucleotides of a lancRNA.


Complementary, as the term is used in the art, refers to the capacity for precise pairing between two nucleotides. For example, if a nucleotide at a certain position of an oligonucleotide is capable of hydrogen bonding with a nucleotide at the same position of lancRNA, then the single stranded nucleotide and lancRNA are considered to be complementary to each other at that position. The single stranded nucleotide and lancRNA are complementary to each other when a sufficient number of corresponding positions in each molecule are occupied by nucleotides that can hydrogen bond with each other through their bases. Thus, “complementary” is a term which is used to indicate a sufficient degree of complementarity or precise pairing such that stable and specific binding occurs between the single stranded nucleotide and lancRNA. For example, if a base at one position of a single stranded nucleotide is capable of hydrogen bonding with a base at the corresponding position of a lancRNA, then the bases are considered to be complementary to each other at that position. 100% complementarity is not required.


The single stranded oligonucleotide may be at least 80% complementary to (optionally one of at least 85%, 90%, 91%, 92%, 93%, 94%, 95%, 96%, 97%, 98%, 99% or 100% complementary to) the consecutive nucleotides of a lancRNA. In some embodiments the single stranded oligonucleotide may contain 1, 2 or 3 base mismatches compared to the portion of the consecutive nucleotides of a lancRNA. In some embodiments the single stranded oligonucleotide may have up to 3 mismatches over 15 bases, or up to 2 mismatches over 10 bases.


It is understood in the art that a complementary nucleotide sequence need not be 100% complementary to that of its target to be specifically hybridizable. In some embodiments, a complementary nucleic acid sequence for purposes of the present disclosure is specifically hybridizable when binding of the sequence to the target molecule (e.g., lancRNA) interferes with the normal function of the target (e.g., lancRNA) to cause a loss of activity and there is a sufficient degree of complementarity to avoid non-specific binding of the sequence to non-target sequences under conditions in which avoidance of non-specific binding is desired, e.g., under physiological conditions in the case of in vivo assays or therapeutic treatment, and in the case of in vitro assays, under conditions in which the assays are performed under suitable conditions of stringency.


In some embodiments, the single stranded oligonucleotide is 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 35, 40, 45, 50 or more nucleotides in length. In some embodiments, the oligonucleotide is 8 to 30 nucleotides in length.


In some embodiments, the chromosomal region encoding the lancRNA occurs on the same DNA strand as a gene sequence (sense). In some embodiments, the chromosomal region encoding the lancRNA occurs on the opposite DNA strand as a gene sequence (anti-sense). Oligonucleotides complementary to a lancRNA or the chromosomal region encoding the lancRNA can bind either sense or anti-sense sequences. Base pairings may include both canonical Watson-Crick base pairing and non-Watson-Crick base pairing (e.g., Wobble base pairing and Hoogsteen base pairing). It is understood that for complementary base pairings, adenosine-type bases (A) are complementary to thymidine-type bases (T) or uracil-type bases (U), that cytosine-type bases (C) are complementary to guanosine-type bases (G), and that universal bases such as 3-nitropyrrole or 5-nitroindole can hybridize to and are considered complementary to any A, C, U, or T. Inosine (I) has also been considered in the art to be a universal base and is considered complementary to any A, C, U or T.


In some embodiments, any one or more thymidine (T) nucleotides (or modified nucleotide thereof) or uridine (U) nucleotides (or a modified nucleotide thereof) in a sequence provided herein, including a sequence provided in the sequence listing, may be replaced with any other nucleotide suitable for base pairing (e.g., via a Watson-Crick base pair) with an adenosine nucleotide. In some embodiments, any one or more thymidine (T) nucleotides (or modified nucleotide thereof) or uridine (U) nucleotides (or a modified nucleotide thereof) in a sequence provided herein, including a sequence provided in the sequence listing, may be suitably replaced with a different pyrimidine nucleotide or vice versa. In some embodiments, any one or more thymidine (T) nucleotides (or modified nucleotide thereof) in a sequence provided herein, including a sequence provided in the sequence listing, may be suitably replaced with a uridine (U) nucleotide (or a modified nucleotide thereof) or vice versa.


In some embodiments, GC content of the single stranded oligonucleotide is preferably between about 30-60%. Contiguous runs of three or more Gs or Cs may not be preferable in some embodiments. Accordingly, in some embodiments, the oligonucleotide does not comprise a stretch of three or more guanosine nucleotides.


In some embodiments, it has been found that single stranded oligonucleotides disclosed herein may increase expression of mRNA corresponding to the gene by at least about 50% (i.e. 150% of normal or 1.5 fold), or by about 2 fold to about 5 fold. In some embodiments, expression may be increased by at least about 15 fold, 20 fold, 30 fold, 40 fold, 50 fold or 100 fold, or any range between any of the foregoing numbers. It has also been found that increased mRNA expression has been shown to correlate to increased protein expression.


In some or any of the embodiments of oligonucleotides described herein, or processes for designing or synthesizing them, the oligonucleotides will upregulate gene expression and may specifically bind or specifically hybridize or be complementary to a lancRNA that is transcribed from the same strand (the sense strand) of a protein coding reference gene. In some or any of the embodiments of oligonucleotides described herein, or processes for designing or synthesizing them, the oligonucleotides will upregulate gene expression and may specifically bind or specifically hybridize or be complementary to a lancRNA that is transcribed from the opposite strand (the antisense strand) of a protein coding reference gene. The oligonucleotide may bind to a region of the lancRNA that is transcribed from a region within or overlaps with an 5′ UTR, 3′ UTR, a translation initiation region, or a translation termination region of a target gene. The oligonucleotide may bind to a region of the lancRNA that is transcribed from a region upstream of an 5′ UTR or a translation initiation region or from a region downstream of a 3′ UTR or a translation termination region of a target gene.


The oligonucleotides described herein may be modified, e.g., comprise a modified sugar moiety, a modified internucleoside linkage, a modified nucleotide and/or combinations thereof.


Any of the oligonucleotides disclosed herein may be linked to one or more other oligonucleotides disclosed herein by a linker, e.g., a cleavable linker.


Method for Selecting Candidate Oligonucleotides for Modulating Expression of a Target Gene

Methods are provided herein for selecting a candidate oligonucleotide for modulating (e.g., activating or enhancing) expression of a target gene. The target selection methods may generally involve steps for selecting single stranded oligonucleotides having any of the structural and functional characteristics disclosed herein. Typically, the methods involve one or more steps aimed at identifying oligonucleotides that target a lancRNA that is functionally related to a target gene, for example a lancRNA that regulates expression of a target gene (e.g., in a cis-regulatory manner). In some embodiments, “cis-regulatory manner” means that the lancRNA regulates expression of genes in the locus from which the lancRNA is expressed.


Methods of selecting a candidate oligonucleotide may involve selecting a region that encodes a lancRNA that maps to a chromosomal position encompassing or in proximity to a transcriptional boundary of the target gene. The region encoding the lancRNA may map to the strand of the chromosome comprising the sense strand of the target gene, in which case the candidate oligonucleotide is complementary to the sense strand of the target gene (i.e., the oligonucleotide is antisense to the target gene). Alternatively, the region encoding the lancRNA may map to the strand of the chromosome comprising the antisense strand of the target gene, in which case the oligonucleotide is complementary to the antisense strand (the template strand) of the target gene (i.e., the oligonucleotide is sense to the target gene).


Methods for selecting a set of candidate oligonucleotides that is enriched in oligonucleotides that modulate (e.g., activate) expression of a target gene may involve selecting one or more regions that encode lancRNAs that map to a chromosomal position that encompasses or that is in proximity to a transcriptional boundary of the target gene and selecting a set of oligonucleotides, in which each oligonucleotide in the set comprises a nucleotide sequence that is complementary with the one or more regions. As used herein, the phrase, “a set of oligonucleotides that is enriched in oligonucleotides that modulate (e.g., activate) expression of” refers to a set of oligonucleotides that has a greater number of oligonucleotides that modulate (e.g., activate) expression of a target gene compared with a random selection of oligonucleotides of the same physicochemical properties (e.g., the same GC content, Tm, length etc.) as the enriched set.


Where the design and/or synthesis of a single stranded oligonucleotide involves design and/or synthesis of a sequence that is complementary to a nucleic acid or lancRNA described by such sequence information, the skilled person is readily able to determine the complementary sequence, e.g., through understanding of Watson Crick base pairing rules which form part of the common general knowledge in the field.


In some embodiments design and/or synthesis of a single stranded oligonucleotide involves manufacture of an oligonucleotide from starting materials by techniques known to those of skill in the art, where the synthesis may be based on a sequence of a lancRNA, a region encoding a lancRNA, or portion thereof.


Methods of design and/or synthesis of a single stranded oligonucleotide may involve one or more of the steps of:


Identifying and/or selecting a chromosomal region within 5 kb of a transcriptional boundary;


Designing a nucleic acid sequence having a desired degree of sequence identity or complementarity to the region or a portion thereof;


Synthesizing a single stranded oligonucleotide to the designed sequence;


Purifying the synthesized single stranded oligonucleotide; and


Optionally mixing the synthesized single stranded oligonucleotide with at least one pharmaceutically acceptable diluent, carrier or excipient to form a pharmaceutical composition or medicament.


Single stranded oligonucleotides so designed and/or synthesized may be useful in method of modulating gene expression as described herein.


Preferably, single stranded oligonucleotides of the invention are synthesized chemically. Oligonucleotides used to practice this invention can be synthesized in vitro by well-known chemical synthesis techniques.


Oligonucleotides of the invention can be stabilized against nucleolytic degradation such as by the incorporation of a modification, e.g., a nucleotide modification. For example, nucleic acid sequences of the invention include a phosphorothioate at least the first, second, or third internucleotide linkage at the 5′ or 3′ end of the nucleotide sequence. As another example, the nucleic acid sequence can include a 2′-modified nucleotide, e.g., a 2′-deoxy, 2′-deoxy-2′-fluoro, 2′-O-methyl, 2′-O-methoxyethyl (2′-O-MOE), 2′-O-aminopropyl (2′-O-AP), 2′-O-dimethylaminoethyl (2′-O-DMAOE), 2′-O-dimethylaminopropyl (2′-O-DMAP), 2′-O-dimethylaminoethyloxyethyl (2′-O-DMAEOE), or 2′-O—N-methylacetamido (2′-O-NMA). As another example, the nucleic acid sequence can include at least one 2′-O-methyl-modified nucleotide, and in some embodiments, all of the nucleotides include a 2′-O-methyl modification. In some embodiments, the nucleic acids are “locked,” i.e., comprise nucleic acid analogues in which the ribose ring is “locked” by a methylene bridge connecting the 2′-O atom and the 4′-C atom.


It is understood that any of the modified chemistries or formats of single stranded oligonucleotides described herein can be combined with each other, and that one, two, three, four, five, or more different types of modifications can be included within the same molecule.


In some embodiments, the method may further comprise the steps of amplifying the synthesized single stranded oligonucleotide, and/or purifying the single stranded oligonucleotide (or amplified single stranded oligonucleotide), and/or sequencing the single stranded oligonucleotide so obtained.


As such, the process of preparing a single stranded oligonucleotide may be a process that is for use in the manufacture of a pharmaceutical composition or medicament for use in the treatment of disease, optionally wherein the treatment involves modulating expression of a target gene.


In the methods described above a lancRNA may be, or have been, identified, or obtained, by a method that involves a detection of the lancRNA. Exemplary methods include RNase protection assays, FISH (fluorescence in situ hybridization), single molecule imaging, deep and/or targeted next generation sequencing. and Northern blots, which are known in the art.


Where the single stranded oligonucleotide is based on a lancRNA sequence, or a portion of such a sequence, it may be based on information about that sequence, e.g., sequence information available in written or electronic form, which may include sequence information contained in publicly available scientific publications or sequence databases.


Nucleotide Analogues

In some embodiments, the oligonucleotide may comprise at least one ribonucleotide, at least one deoxyribonucleotide, and/or at least one bridged nucleotide. In some embodiments, the oligonucleotide may comprise a bridged nucleotide, such as a locked nucleic acid (LNA) nucleotide, a constrained ethyl (cEt) nucleotide, or an ethylene bridged nucleic acid (ENA) nucleotide. Examples of such nucleotides are disclosed herein and known in the art. In some embodiments, the oligonucleotide comprises a nucleotide analog disclosed in one of the following United States patent or patent application Publications: U.S. Pat. No. 7,399,845, U.S. Pat. No. 7,741,457, U.S. Pat. No. 8,022,193, U.S. Pat. No. 7,569,686, U.S. Pat. No. 7,335,765, U.S. Pat. No. 7,314,923, U.S. Pat. No. 7,335,765, and U.S. Pat. No. 7,816,333, US 20110009471, the entire contents of each of which are incorporated herein by reference for all purposes. The oligonucleotide may have one or more 2′ O-methyl nucleotides. The oligonucleotide may consist entirely of 2′ O-methyl nucleotides.


Often the single stranded oligonucleotide has one or more nucleotide analogues. For example, the single stranded oligonucleotide may have at least one nucleotide analogue that results in an increase in Tm of the oligonucleotide in a range of 1° C., 2° C., 3° C., 4° C., or 5° C. compared with an oligonucleotide that does not have the at least one nucleotide analogue. The single stranded oligonucleotide may have a plurality of nucleotide analogues that results in a total increase in Tm of the oligonucleotide in a range of 2° C., 3° C., 4° C., 5° C., 6° C., 7° C., 8° C., 9° C., 10° C., 15° C., 20° C., 25° C., 30° C., 35° C., 40° C., 45° C. or more compared with an oligonucleotide that does not have the nucleotide analogue.


The oligonucleotide may be of up to 50 nucleotides in length in which 2 to 10, 2 to 15, 2 to 16, 2 to 17, 2 to 18, 2 to 19, 2 to 20, 2 to 25, 2 to 30, 2 to 40, 2 to 45, or more nucleotides of the oligonucleotide are nucleotide analogues. The oligonucleotide may be of 8 to 30 nucleotides in length in which 2 to 10, 2 to 15, 2 to 16, 2 to 17, 2 to 18, 2 to 19, 2 to 20, 2 to 25, 2 to 30 nucleotides of the oligonucleotide are nucleotide analogues.


The oligonucleotide may be of 8 to 15 nucleotides in length in which 2 to 4, 2 to 5, 2 to 6, 2 to 7, 2 to 8, 2 to 9, 2 to 10, 2 to 11, 2 to 12, 2 to 13, 2 to 14 nucleotides of the oligonucleotide are nucleotide analogues. Optionally, the oligonucleotides may have every nucleotide except 1, 2, 3, 4, 5, 6, 7, 8, 9, or 10 nucleotides modified.


The oligonucleotide may consist entirely of bridged nucleotides (e.g., LNA nucleotides, cEt nucleotides, ENA nucleotides). The oligonucleotide may comprise alternating deoxyribonucleotides and 2′-fluoro-deoxyribonucleotides. The oligonucleotide may comprise alternating deoxyribonucleotides and 2′-O-methyl nucleotides. The oligonucleotide may comprise alternating deoxyribonucleotides and ENA nucleotide analogues. The oligonucleotide may comprise alternating deoxyribonucleotides and LNA nucleotides. The oligonucleotide may comprise alternating LNA nucleotides and 2′-O-methyl nucleotides. The oligonucleotide may have a 5′ nucleotide that is a bridged nucleotide (e.g., a LNA nucleotide, cEt nucleotide, ENA nucleotide). The oligonucleotide may have a 5′ nucleotide that is a deoxyribonucleotide.


The oligonucleotide may comprise deoxyribonucleotides flanked by at least one bridged nucleotide (e.g., a LNA nucleotide, cEt nucleotide, ENA nucleotide) on each of the 5′ and 3′ ends of the deoxyribonucleotides. The oligonucleotide may comprise deoxyribonucleotides flanked by 1, 2, 3, 4, 5, 6, 7, 8 or more bridged nucleotides (e.g., LNA nucleotides, cEt nucleotides, ENA nucleotides) on each of the 5′ and 3′ ends of the deoxyribonucleotides. The 3′ position of the oligonucleotide may have a 3′ hydroxyl group. The 3′ position of the oligonucleotide may have a 3′ thiophosphate.


The oligonucleotide may be conjugated with a label. For example, the oligonucleotide may be conjugated with a biotin moiety, cholesterol, Vitamin A, folate, sigma receptor ligands, aptamers, peptides, such as CPP, hydrophobic molecules, such as lipids, ASGPR or dynamic polyconjugates and variants thereof at its 5′ or 3′ end.


Preferably the single stranded oligonucleotide comprises one or more modifications comprising: a modified sugar moiety, and/or a modified internucleoside linkage, and/or a modified nucleotide and/or combinations thereof. It is not necessary for all positions in a given oligonucleotide to be uniformly modified, and in fact more than one of the modifications described herein may be incorporated in a single oligonucleotide or even at within a single nucleoside within an oligonucleotide.


In some embodiments, the single stranded oligonucleotides are chimeric oligonucleotides that contain two or more chemically distinct regions, each made up of at least one nucleotide. These oligonucleotides typically contain at least one region of modified nucleotides that confers one or more beneficial properties (such as, for example, increased nuclease resistance, increased uptake into cells, increased binding affinity for the target) and a region that is a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. Chimeric single stranded oligonucleotides of the invention may be formed as composite structures of two or more oligonucleotides, modified oligonucleotides, oligonucleosides and/or oligonucleotide mimetics as described above. Such compounds have also been referred to in the art as hybrids or gapmers. Representative United States patents that teach the preparation of such hybrid structures comprise, but are not limited to, U.S. Pat. Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355; 5,652,356; and 5,700,922, each of which is herein incorporated by reference.


In some embodiments, the single stranded oligonucleotide comprises at least one nucleotide modified at the 2′ position of the sugar, most preferably a 2′-O-alkyl, 2′-O-alkyl-O-alkyl or 2′-fluoro-modified nucleotide. In other preferred embodiments, RNA modifications include 2′-fluoro, 2′-amino and 2′ O-methyl modifications on the ribose of pyrimidines, abasic residues or an inverted base at the 3′ end of the RNA. Such modifications are routinely incorporated into oligonucleotides and these oligonucleotides have been shown to have a higher Tm (i.e., higher target binding affinity) than 2′-deoxyoligonucleotides against a given target.


A number of nucleotide and nucleoside modifications have been shown to make the oligonucleotide into which they are incorporated more resistant to nuclease digestion than the native oligodeoxynucleotide; these modified oligos survive intact for a longer time than unmodified oligonucleotides. Specific examples of modified oligonucleotides include those comprising modified backbones, for example, phosphorothioates, phosphotriesters, methyl phosphonates, short chain alkyl or cycloalkyl intersugar linkages or short chain heteroatomic or heterocyclic intersugar linkages. Most preferred are oligonucleotides with phosphorothioate backbones and those with heteroatom backbones, particularly CH2—NH—O—CH2, CH, ˜N(CH3)˜O˜CH2 (known as a methylene(methylimino) or MMI backbone, CH2—O—N(CH3)—CH2, CH2—N(CH3)—N(CH3)—CH2 and O—N(CH3)—CH2—CH2 backbones, wherein the native phosphodiester backbone is represented as O—P—O—CH); amide backbones (see De Mesmaeker et al. Ace. Chem. Res. 1995, 28:366-374); morpholino backbone structures (see Summerton and Weller, U.S. Pat. No. 5,034,506); peptide nucleic acid (PNA) backbone (wherein the phosphodiester backbone of the oligonucleotide is replaced with a polyamide backbone, the nucleotides being bound directly or indirectly to the aza nitrogen atoms of the polyamide backbone, see Nielsen et al., Science 1991, 254, 1497). Phosphorus-containing linkages include, but are not limited to, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates comprising 3′alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates comprising 3′-amino phosphoramidate and aminoalkylphosphoramidates, thionophosphoramidates, thionoalkylphosphonates, thionoalkylphosphotriesters, and boranophosphates having normal 3′-5′ linkages, 2′-5′ linked analogs of these, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3′-5′ to 5′-3′ or 2′-5′ to 5′-2′; see U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,196; 5,188,897; 5,264,423; 5,276,019; 5,278,302; 5,286,717; 5,321,131; 5,399,676; 5,405,939; 5,453,496; 5,455, 233; 5,466,677; 5,476,925; 5,519,126; 5,536,821; 5,541,306; 5,550,111; 5,563, 253; 5,571,799; 5,587,361; and 5,625,050.


Morpholino-based oligomeric compounds are described in Dwaine A. Braasch and David R. Corey, Biochemistry, 2002, 41(14), 4503-4510); Genesis, volume 30, issue 3, 2001; Heasman, J., Dev. Biol., 2002, 243, 209-214; Nasevicius et al., Nat. Genet., 2000, 26, 216-220; Lacerra et al., Proc. Natl. Acad. Sci., 2000, 97, 9591-9596; and U.S. Pat. No. 5,034,506, issued Jul. 23, 1991. In some embodiments, the morpholino-based oligomeric compound is a phosphorodiamidate morpholino oligomer (PMO) (e.g., as described in Iverson, Curr. Opin. Mol. Ther., 3:235-238, 2001; and Wang et al., J. Gene Med., 12:354-364, 2010; the disclosures of which are incorporated herein by reference in their entireties).


Cyclohexenyl nucleic acid oligonucleotide mimetics are described in Wang et al., J. Am. Chem. Soc., 2000, 122, 8595-8602.


Modified oligonucleotide backbones that do not include a phosphorus atom therein have backbones that are formed by short chain alkyl or cycloalkyl internucleoside linkages, mixed heteroatom and alkyl or cycloalkyl internucleoside linkages, or one or more short chain heteroatomic or heterocyclic internucleoside linkages. These comprise those having morpholino linkages (formed in part from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide and sulfone backbones; formacetyl and thioformacetyl backbones; methylene formacetyl and thioformacetyl backbones; alkene containing backbones; sulfamate backbones; methyleneimino and methylenehydrazino backbones; sulfonate and sulfonamide backbones; amide backbones; and others having mixed N, O, S and CH2 component parts; see U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,264, 562; 5, 264,564; 5,405,938; 5,434,257; 5,466,677; 5,470,967; 5,489,677; 5,541,307; 5,561,225; 5,596,086; 5,602,240; 5,610,289; 5,602,240; 5,608,046; 5,610,289; 5,618,704; 5,623,070; 5,663,312; 5,633,360; 5,677,437; and 5,677,439, each of which is herein incorporated by reference.


Modified oligonucleotides are also known that include oligonucleotides that are based on or constructed from arabinonucleotide or modified arabinonucleotide residues. Arabinonucleosides are stereoisomers of ribonucleosides, differing only in the configuration at the 2′-position of the sugar ring. In some embodiments, a 2′-arabino modification is 2′-F arabino. In some embodiments, the modified oligonucleotide is 2′-fluoro-D-arabinonucleic acid (FANA) (as described in, for example, Lon et al., Biochem., 41:3457-3467, 2002 and Min et al., Bioorg. Med. Chem. Lett., 12:2651-2654, 2002; the disclosures of which are incorporated herein by reference in their entireties). Similar modifications can also be made at other positions on the sugar, particularly the 3′ position of the sugar on a 3′ terminal nucleoside or in 2′-5′ linked oligonucleotides and the 5′ position of 5′ terminal nucleotide.


PCT Publication No. WO 99/67378 discloses arabinonucleic acids (ANA) oligomers and their analogues for improved sequence specific inhibition of gene expression via association to complementary messenger RNA.


Other preferred modifications include ethylene-bridged nucleic acids (ENAs) (e.g., International Patent Publication No. WO 2005/042777, Morita et al., Nucleic Acid Res., Suppl 1:241-242, 2001; Surono et al., Hum. Gene Ther., 15:749-757, 2004; Koizumi, Curr. Opin. Mol. Ther., 8:144-149, 2006 and Horie et al., Nucleic Acids Symp. Ser (Oxf), 49:171-172, 2005; the disclosures of which are incorporated herein by reference in their entireties). Preferred ENAs include, but are not limited to, 2′-0,4′-C-ethylene-bridged nucleic acids.


Examples of LNAs are described in WO/2008/043753 and include compounds of the following general formula.




embedded image


where X and Y are independently selected among the groups —O—,


—S—, —N(H)—, N(R)—, —CH2— or —CH— (if part of a double bond),


—CH2—O—, —CH2—S—, —CH2—N(H)—, —CH2—N(R)—, —CH2—CH2— or —CH2—CH— (if part of a double bond),


—CH═CH—, where R is selected from hydrogen and C1-4-alkyl; Z and Z* are independently selected among an internucleoside linkage, a terminal group or a protecting group; B constitutes a natural or non-natural nucleotide base moiety; and the asymmetric groups may be found in either orientation.


Preferably, the LNA used in the oligonucleotides described herein comprises at least one LNA unit according any of the formulas




embedded image


wherein Y is —O—, —S—, —NH—, or N(RH); Z and Z* are independently selected among an internucleoside linkage, a terminal group or a protecting group; B constitutes a natural or non-natural nucleotide base moiety, and RH is selected from hydrogen and C1-4-alkyl.


In some embodiments, the Locked Nucleic Acid (LNA) used in the oligonucleotides described herein comprises at least one Locked Nucleic Acid (LNA) unit according any of the formulas shown in Scheme 2 of PCT/DK2006/000512.


In some embodiments, the LNA used in the oligomer of the invention comprises internucleoside linkages selected from —O—P(O)2—O—, —O—P(O,S)—O—, —O—P(S)2—O—, —S—P(O)2—O—, —S—P(O,S)—O—, —S—P(S)2—O—, —O—P(O)2—S—, —O—P(O,S)—S—, —S—P(O)2—S—, —O—PO(RH)—O—, O—PO(OCH3)—O—, —O—PO(NRH)—O—, —O—PO(OCH2CH2S—R)—O—, —O—PO(BH3)—O—, —O—PO(NHRH)—O—, —O—P(O)2—NRH—, —NRH—P(O)2—O—, —NRH—CO—O—, where RH is selected from hydrogen and C1-4-alkyl.


Specifically preferred LNA units are shown in scheme 2:




embedded image


The term “thio-LNA” comprises a locked nucleotide in which at least one of X or Y in the general formula above is selected from S or —CH2—S—. Thio-LNA can be in both beta-D and alpha-L-configuration.


The term “amino-LNA” comprises a locked nucleotide in which at least one of X or Y in the general formula above is selected from —N(H)—, N(R)—, CH2—N(H)—, and —CH2—N(R)— where R is selected from hydrogen and C1-4-alkyl. Amino-LNA can be in both beta-D and alpha-L-configuration.


The term “oxy-LNA” comprises a locked nucleotide in which at least one of X or Y in the general formula above represents —O— or —CH2—O—. Oxy-LNA can be in both beta-D and alpha-L-configuration.


The term “ena-LNA” comprises a locked nucleotide in which Y in the general formula above is —CH2—O— (where the oxygen atom of —CH2—O— is attached to the 2′-position relative to the base B).


LNAs are described in additional detail herein.


One or more substituted sugar moieties can also be included, e.g., one of the following at the 2′ position: OH, SH, SCH3, F, OCN, OCH3OCH3, OCH3O(CH2)n CH3, O(CH2)n NH2 or O(CH2)n CH3 where n is from 1 to about 10; C1 to C10 lower alkyl, alkoxyalkoxy, substituted lower alkyl, alkaryl or aralkyl; Cl; Br; CN; CF3; OCF3; O-, S-, or N-alkyl; O-, S-, or N-alkenyl; 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 the pharmacokinetic properties of an oligonucleotide; or a group for improving the pharmacodynamic properties of an oligonucleotide and other substituents having similar properties. A preferred modification includes 2′-methoxyethoxy [2′-O—CH2CH2OCH3, also known as 2′-O-(2-methoxyethyl)] (Martin et al, HeIv. Chim. Acta, 1995, 78, 486). Other preferred modifications include 2′-methoxy (2′-O—CH3), 2′-propoxy (2′-OCH2CH2CH3) and 2′-fluoro (2′-F). Similar modifications may also be made at other positions on the oligonucleotide, particularly the 3′ position of the sugar on the 3′ terminal nucleotide and the 5′ position of 5′ terminal nucleotide. Oligonucleotides may also have sugar mimetics such as cyclobutyls in place of the pentofuranosyl group.


Single stranded oligonucleotides can also include, additionally or alternatively, nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases include adenine (A), guanine (G), thymine (T), cytosine (C) and uracil (U). Modified nucleobases include nucleobases found only infrequently or transiently in natural nucleic acids, e.g., hypoxanthine, 6-methyladenine, 5-Me pyrimidines, particularly 5-methylcytosine (also referred to as 5-methyl-2′ deoxycytosine and often referred to in the art as 5-Me-C), 5-hydroxymethylcytosine (HMC), glycosyl HMC and gentobiosyl HMC, isocytosine, pseudoisocytosine, as well as synthetic nucleobases, e.g., 2-aminoadenine, 2-(methylamino)adenine, 2-(imidazolylalkyl)adenine, 2-(aminoalklyamino)adenine or other heterosubstituted alkyladenines, 2-thiouracil, 2-thiothymine, 5-bromouracil, 5-hydroxymethyluracil, 5-propynyluracil, 8-azaguanine, 7-deazaguanine, N6 (6-aminohexyl)adenine, 6-aminopurine, 2-aminopurine, 2-chloro-6-aminopurine and 2,6-diaminopurine or other diaminopurines. See, e.g., Kornberg, “DNA Replication,” W. H. Freeman & Co., San Francisco, 1980, pp 75-′7′7; and Gebeyehu, G., et al. Nucl. Acids Res., 15:4513 (1987)). A “universal” base known in the art, e.g., inosine, can also be included. 5-Me-C substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, in Crooke, and Lebleu, eds., Antisense Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and may be used as base substitutions.


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


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


Single stranded oligonucleotides can also include one or more nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases comprise the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases comprise other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudo-uracil), 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-methylquanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-deazaadenine and 3-deazaguanine and 3-deazaadenine. In some embodiments, a cytosine is substituted with a 5-methylcytosine. In some embodiments, an oligonucleotide has 2, 3, 4, 5, 6, 7, or more cytosines substituted with 5-methylcytosines. In some embodiments, an oligonucleotide does not have 2, 3, 4, 5, 6, 7, or more consecutive 5-methylcytosines. In some embodiments, an LNA cytosine nucleotide is replaced with an LNA 5-methylcytosine nucleotide.


Further, nucleobases comprise those disclosed in U.S. Pat. No. 3,687,808, those disclosed in “The Concise Encyclopedia of Polymer Science And Engineering”, pages 858-859, Kroschwitz, ed. John Wiley & Sons, 1990; those disclosed by Englisch et al., Angewandle Chemie, International Edition, 1991, 30, page 613, and those disclosed by Sanghvi, Chapter 15, Antisense Research and Applications,” pages 289-302, Crooke, and Lebleu, eds., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds of the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, comprising 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2<0>C (Sanghvi, et al., eds, “Antisense Research and Applications,” CRC Press, Boca Raton, 1993, pp. 276-278) and are presently preferred base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications. Modified nucleobases are described in U.S. Pat. No. 3,687,808, as well as U.S. Pat. Nos. 4,845,205; 5,130,302; 5,134,066; 5,175, 273; 5, 367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,596,091; 5,614,617; 5,750,692, and 5,681,941, each of which is herein incorporated by reference.


In some embodiments, the single stranded oligonucleotides are chemically linked to one or more moieties or conjugates that enhance the activity, cellular distribution, or cellular uptake of the oligonucleotide. For example, one or more single stranded oligonucleotides, of the same or different types, can be conjugated to each other; or single stranded oligonucleotides can be conjugated to targeting moieties with enhanced specificity for a cell type or tissue type. Such moieties include, but are not limited to, lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86, 6553-6556), cholic acid (Manoharan et al., Bioorg. Med. Chem. Let., 1994, 4, 1053-1060), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al, Ann. N.Y. Acad. Sci., 1992, 660, 306-309; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3, 2765-2770), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20, 533-538), an aliphatic chain, e.g., dodecandiol or undecyl residues (Kabanov et al., FEBS Lett., 1990, 259, 327-330; Svinarchuk et al., Biochimie, 1993, 75, 49-54), a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium 1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654; Shea et al., Nucl. Acids Res., 1990, 18, 3777-3783), a polyamine or a polyethylene glycol chain (Mancharan et al., Nucleosides & Nucleotides, 1995, 14, 969-973), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36, 3651-3654), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264, 229-237), or an octadecylamine or hexylamino-carbonyl-t oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277, 923-937). See also U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552, 538; 5,578,717, 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486, 603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762, 779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082, 830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5, 245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391, 723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5, 565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599, 928 and 5,688,941, each of which is herein incorporated by reference.


These moieties or conjugates can include conjugate groups covalently bound to functional groups such as primary or secondary hydroxyl groups. Conjugate groups of the invention include intercalators, reporter molecules, polyamines, polyamides, polyethylene glycols, polyethers, groups that enhance the pharmacodynamic properties of oligomers, and groups that enhance the pharmacokinetic properties of oligomers. Typical conjugate groups include cholesterols, lipids, phospholipids, biotin, phenazine, folate, phenanthridine, anthraquinone, acridine, fluoresceins, rhodamines, coumarins, and dyes. Groups that enhance the pharmacodynamic properties, in the context of this invention, include groups that improve uptake, enhance resistance to degradation, and/or strengthen sequence-specific hybridization with the target nucleic acid. Groups that enhance the pharmacokinetic properties, in the context of this invention, include groups that improve uptake, distribution, metabolism or excretion of the compounds of the present invention. Representative conjugate groups are disclosed in International Patent Application No. PCT/US92/09196, filed Oct. 23, 1992, and U.S. Pat. No. 6,287,860, which are incorporated herein by reference. Conjugate moieties include, but are not limited to, lipid moieties such as a cholesterol moiety, cholic acid, a thioether, e.g., hexyl-5-tritylthiol, a thiocholesterol, an aliphatic chain, e.g., dodecandiol or undecyl residues, a phospholipid, e.g., di-hexadecyl-rac-glycerol or triethylammonium1,2-di-O-hexadecyl-rac-glycero-3-H-phosphonate, a polyamine or a polyethylene glycol chain, or adamantane acetic acid, a palmityl moiety, or an octadecylamine or hexylamino-carbonyl-oxy cholesterol moiety. See, e.g., U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,580,731; 5,591,584; 5,109,124; 5,118,802; 5,138,045; 5,414,077; 5,486,603; 5,512,439; 5,578,718; 5,608,046; 4,587,044; 4,605,735; 4,667,025; 4,762,779; 4,789,737; 4,824,941; 4,835,263; 4,876,335; 4,904,582; 4,958,013; 5,082,830; 5,112,963; 5,214,136; 5,082,830; 5,112,963; 5,214,136; 5,245,022; 5,254,469; 5,258,506; 5,262,536; 5,272,250; 5,292,873; 5,317,098; 5,371,241, 5,391,723; 5,416,203, 5,451,463; 5,510,475; 5,512,667; 5,514,785; 5,565,552; 5,567,810; 5,574,142; 5,585,481; 5,587,371; 5,595,726; 5,597,696; 5,599,923; 5,599,928 and 5,688,941.


In some embodiments, single stranded oligonucleotide modification include modification of the 5′ or 3′ end of the oligonucleotide. In some embodiments, the 3′ end of the oligonucleotide comprises a hydroxyl group or a thiophosphate. It should be appreciated that additional molecules (e.g. a biotin moiety or a fluorophor) can be conjugated to the 5′ or 3′ end of the single stranded oligonucleotide. In some embodiments, the single stranded oligonucleotide comprises a biotin moiety conjugated to the 5′ nucleotide.


In some embodiments, the single stranded oligonucleotide comprises locked nucleic acids (LNA), ENA modified nucleotides, 2′-O-methyl nucleotides, or 2′-fluoro-deoxyribonucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and 2′-fluoro-deoxyribonucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and 2′-O-methyl nucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and ENA modified nucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating deoxyribonucleotides and locked nucleic acid nucleotides. In some embodiments, the single stranded oligonucleotide comprises alternating locked nucleic acid nucleotides and 2′-O-methyl nucleotides.


In some embodiments, the 5′ nucleotide of the oligonucleotide is a deoxyribonucleotide. In some embodiments, the 5′ nucleotide of the oligonucleotide is a locked nucleic acid nucleotide. In some embodiments, the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one locked nucleic acid nucleotide on each of the 5′ and 3′ ends of the deoxyribonucleotides. In some embodiments, the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group or a 3′ thiophosphate.


In some embodiments, the single stranded oligonucleotide comprises phosphorothioate internucleotide linkages. In some embodiments, the single stranded oligonucleotide comprises phosphorothioate internucleotide linkages between at least two nucleotides. In some embodiments, the single stranded oligonucleotide comprises phosphorothioate internucleotide linkages between all nucleotides.


It should be appreciated that the single stranded oligonucleotide can have any combination of modifications as described herein.


In some embodiments, an oligonucleotide described herein may be a mixmer or comprise a mixmer sequence pattern. The term ‘mixmer’ refers to oligonucleotides which comprise both naturally and non-naturally occurring nucleotides or comprise two different types of non-naturally occurring nucleotides. Mixmers are generally known in the art to have a higher binding affinity than unmodified oligonucleotides and may be used to specifically bind a target molecule, e.g., to block a binding site on the target molecule. Generally, mixmers do not recruit an RNAse to the target molecule and thus do not promote cleavage of the target molecule. Accordingly, in some embodiments, an oligonucleotide provided herein may be cleavage promoting (e.g., an siRNA or gapmer) or not cleavage promoting (e.g., a mixmer, siRNA, single stranded RNA or double stranded RNA).


In some embodiments, the mixmer comprises or consists of a repeating pattern of nucleotide analogues and naturally occurring nucleotides, or one type of nucleotide analogue and a second type of nucleotide analogue. However, it is to be understood that the mixmer need not comprise a repeating pattern and may instead comprise any arrangement of nucleotide analogues and naturally occurring nucleotides or any arrangement of one type of nucleotide analogue and a second type of nucleotide analogue. The repeating pattern, may, for instance be every second or every third nucleotide is a nucleotide analogue, such as LNA, and the remaining nucleotides are naturally occurring nucleotides, such as DNA, or are a 2′ substituted nucleotide analogue such as 2′MOE or 2′ fluoro analogues, or any other nucleotide analogues described herein. It is recognised that the repeating pattern of nucleotide analogues, such as LNA units, may be combined with nucleotide analogues at fixed positions—e.g. at the 5′ or 3′ termini.


In some embodiments, the mixmer does not comprise a region of more than 5, more than 4, more than 3, or more than 2 consecutive naturally occurring nucleotides, such as DNA nucleotides. In some embodiments, the mixmer comprises at least a region consisting of at least two consecutive nucleotide analogues, such as at least two consecutive LNAs. In some embodiments, the mixmer comprises at least a region consisting of at least three consecutive nucleotide analogue units, such as at least three consecutive LNAs.


In some embodiments, the mixmer does not comprise a region of more than 7, more than 6, more than 5, more than 4, more than 3, or more than 2 consecutive nucleotide analogues, such as LNAs. It is to be understood that the LNA units may be replaced with other nucleotide analogues, such as those referred to herein.


In some embodiments, the mixmer comprises at least one nucleotide analogue in one or more of six consecutive nucleotides. The substitution pattern for the nucleotides may be selected from the group consisting of Xxxxxx, xXxxxx, xxXxxx, xxxXxx, xxxxXx and xxxxxX, wherein “X” denotes a nucleotide analogue, such as an LNA, and “x” denotes a naturally occurring nucleotide, such as DNA or RNA.


In some embodiments, the mixmer comprises at least two nucleotide analogues in one or more of six consecutive nucleotides. The substitution pattern for the nucleotides may be selected from the group consisting of XXxxxx, XxXxxx, XxxXxx, XxxxXx, XxxxxX, xXXxxx, xXxXxx, xXxxXx, xXxxxX, xxXXxx, xxXxXx, xxXxxX, xxxXXx, xxxXxX and xxxxXX, wherein “X” denotes a nucleotide analogue, such as an LNA, and “x” denotes a naturally occurring nucleotide, such as DNA or RNA. In some embodiments, the substitution pattern for the nucleotides may be selected from the group consisting of XxXxxx, XxxXxx, XxxxXx, XxxxxX, xXxXxx, xXxxXx, xXxxxX, xxXxXx, xxXxxX and xxxXxX. In some embodiments, the substitution pattern is selected from the group consisting of xXxXxx, xXxxXx, xXxxxX, xxXxXx, xxXxxX and xxxXxX. In some embodiments, the substitution pattern is selected from the group consisting of xXxXxx, xXxxXx and xxXxXx. In some embodiments, the substitution pattern for the nucleotides is xXxXxx.


In some embodiments, the mixmer comprises at least three nucleotide analogues in one or more of six consecutive nucleotides. The substitution pattern for the nucleotides may be selected from the group consisting of XXXxxx, xXXXxx, xxXXXx, xxxXXX, XXxXxx, XXxxXx, XXxxxX, xXXxXx, xXXxxX, xxXXxX, XxXXxx, XxxXXx, XxxxXX, xXxXXx, xXxxXX, xxXxXX, xXxXxX and XxXxXx, wherein “X” denotes a nucleotide analogue, such as an LNA, and “x” denotes a naturally occuring nucleotide, such as DNA or RNA. In some embodiments, the substitution pattern for the nucleotides is selected from the group consisting of XXxXxx, XXxxXx, XXxxxX, xXXxXx, xXXxxX, xxXXxX, XxXXxx, XxxXXx, XxxxXX, xXxXXx, xXxxXX, xxXxXX, xXxXxX and XxXxXx. In some embodiments, the substitution pattern for the nucleotides is selected from the group consisting of xXXxXx, xXXxxX, xxXXxX, xXxXXx, xXxxXX, xxXxXX and xXxXxX. n some embodiments, the substitution pattern for the nucleotides is xXxXxX or XxXxXx. In some embodiments, the substitution pattern for the nucleotides is xXxXxX.


In some embodiments, the mixmer comprises at least four nucleotide analogues in one or more of six consecutive nucleotides. The substitution pattern for the nucleotides may be selected from the group consisting of xXXXX, xXxXXX, xXXxXX, xXXXxX, xXXXXx, XxxXXX, XxXxXX, XxXXxX, XxXXXx, XXxxXX, XXxXxX, XXxXXx, XXXxxX, XXXxXx and XXXXxx, wherein “X” denotes a nucleotide analogue, such as an LNA, and “x” denotes a naturally occurring nucleotide, such as DNA or RNA.


In some embodiments, the mixmer comprises at least five nucleotide analogues in one or more of six consecutive nucleotides. The substitution pattern for the nucleotides may be selected from the group consisting of xXXXXX, XxXXXX, XXxXXX, XXXxXX, XXXXxX and XXXXXx, wherein “X” denotes a nucleotide analogue, such as an LNA, and “x” denotes a naturally occurring nucleotide, such as DNA or RNA.


The oligonucleotide may comprise a nucleotide sequence having one or more of the following modification patterns.


(a) (X)Xxxxxx, (X)xXxxxx, (X)xxXxxx, (X)xxxXxx, (X)xxxxXx and (X)xxxxxX,


(b) (X)XXxxxx, (X)XxXxxx, (X)XxxXxx, (X)XxxxXx, (X)XxxxxX, (X)xXXxxx, (X)xXxXxx, (X)xXxxXx, (X)xXxxxX, (X)xxXXxx, (X)xxXxXx, (X)xxXxxX, (X)xxxXXx, (X)xxxXxX and (X)xxxxXX,


(c) (X)XXXxxx, (X)xXXXxx, (X)xxXXXx, (X)xxxXXX, (X)XXxXxx, (X)XXxxXx, (X)XXxxxX, (X)xXXxXx, (X)xXXxxX, (X)xxXXxX, (X)XxXXxx, (X)XxxXXx (X)XxxxXX, (X)xXxXXx, (X)xXxxXX, (X)xxXxXX, (X)xXxXxX and (X)XxXxXx,


(d) (X)xxXXX, (X)xXxXXX, (X)xXXxXX, (X)xXXXxX, (X)xXXXXx, (X)XxxXXXX, (X)XxXxXX, (X)XxXXxX, (X)XxXXx, (X)XXxxXX, (X)XXxXxX, (X)XXxXXx, (X)XXXxxX, (X)XXXxXx, and (X)XXXXxx,


(e) (X)xXXXXX, (X)XxXXXX, (X)XXxXXX, (X)XXXxXX, (X)XXXXxX and (X)XXXXXx, and


(f) XXXXXX, XxXXXXX, XXxXXXX, XXXxXXX, XXXXxXX, XXXXXxX and XXXXXXx, in which “X” denotes a nucleotide analogue, (X) denotes an optional nucleotide analogue, and “x” denotes a DNA or RNA nucleotide unit. Each of the above listed patterns may appear one or more times within an oligonucleotide, alone or in combination with any of the other disclosed modification patterns.


In some embodiments, the mixmer contains a modified nucleotide, e.g., an LNA, at the 5′ end. In some embodiments, the mixmer contains a modified nucleotide, e.g., an LNA, at the first two positions, counting from the 5′ end.


In some embodiments, the mixmer is incapable of recruiting RNAseH. Oligonucleotides that are incapable of recruiting RNAseH are well known in the literature, in example see WO2007/112754, WO2007/112753, or PCT/DK2008/000344. Mixmers may be designed to comprise a mixture of affinity enhancing nucleotide analogues, such as in non-limiting example LNA nucleotides and 2′-O-methyl nucleotides. In some embodiments, the mixmer comprises modified internucleoside linkages (e.g., phosphorothioate internucleoside linkages or other linkages) between at least two, at least three, at least four, at least five or more nucleotides.


A mixmer may be produced using any method known in the art or described herein. Representative U.S. patents, U.S. patent publications, and PCT publications that teach the preparation of mixmers include U.S. patent publication Nos. US20060128646, US20090209748, US20090298916, US20110077288, and US20120322851, and U.S. Pat. No. 7,687,617.


In some embodiments, the oligonucleotide is a gapmer. A gapmer oligonucleotide generally has the formula 5′-X-Y-Z-3′, with X and Z as flanking regions around a gap region Y. In some embodiments, the Y region is a contiguous stretch of nucleotides, e.g., a region of at least 6 DNA nucleotides, which are capable of recruiting an RNAse, such as RNAseH. Without wishing to be bound by theory, it is thought that the gapmer binds to the target nucleic acid, at which point an RNAse is recruited and can then cleave the target nucleic acid. In some embodiments, the Y region is flanked both 5′ and 3′ by regions X and Z comprising high-affinity modified nucleotides, e.g., 1-6 modified nucleotides. Exemplary modified oligonucleotides include, but are not limited to, 2′ MOE or 2′OMe or Locked Nucleic Acid bases (LNA). The flanks X and Z may be have a of length 1-20 nucleotides, preferably 1-8 nucleotides and even more preferred 1-5 nucleotides. The flanks X and Z may be of similar length or of dissimilar lengths. The gap-segment Y may be a nucleotide sequence of length 5-20 nucleotides, preferably 6-12 nucleotides and even more preferred 6-10 nucleotides. In some aspects, the gap region of the gapmer oligonucleotides of the invention may contain modified nucleotides known to be acceptable for efficient RNase H action in addition to DNA nucleotides, such as C4′-substituted nucleotides, acyclic nucleotides, and arabino-configured nucleotides. In some embodiments, the gap region comprises one or more unmodified internucleosides. In some embodiments, one or both flanking regions each independently comprise one or more phosphorothioate internucleoside linkages (e.g., phosphorothioate internucleoside linkages or other linkages) between at least two, at least three, at least four, at least five or more nucleotides. In some embodiments, the gap region and two flanking regions each independently comprise modified internucleoside linkages (e.g., phosphorothioate internucleoside linkages or other linkages) between at least two, at least three, at least four, at least five or more nucleotides.


A gapmer may be produced using any method known in the art or described herein. Representative U.S. patents, U.S. patent publications, and PCT publications that teach the preparation of gapmers include, but are not limited to, U.S. Pat. Nos. 5,013,830; 5,149,797; 5,220,007; 5,256,775; 5,366,878; 5,403,711; 5,491,133; 5,565,350; 5,623,065; 5,652,355; 5,652,356; 5,700,922; 5,898,031; 7,432,250; and 7,683,036; U.S. patent publication Nos. US20090286969, US20100197762, and US20110112170; and PCT publication Nos. WO2008049085 and WO2009090182, each of which is herein incorporated by reference in its entirety.


In some embodiments, an oligonucleotide described herein comprises a synthetic cap, e.g., to increase efficiency of translation, RNA half-life and/or function within cells. Synthetic caps are known in the art. Exemplary synthetic caps include, but are not limited to, N7-Methyl-Guanosine-5′-Triphosphate-5′-Guanosine, Guanosine-5′-Triphosphate-5*-Guanosine, N7-Methyl-3′-O-Methyl-Guanosine-5′-Triphosphate-5′-Guanosine (see, e.g., products available from TrilinkBiotech), and N7-benzylated dinucleoside tetraphosphate analogs (see, e.g., Grudzien et al. Novel cap analogs for in vitro synthesis of mRNAs with high translational efficiency. RNA. 2004 September; 10(9): 1479-1487).


Methods for Modulating Gene Expression

In one aspect, the invention relates to methods for modulating (e.g., upregulating or downregulating) gene expression in a cell (e.g., a cell for which levels of the target gene are reduced or enhanced) for research purposes (e.g., to study the function of the gene in the cell). In another aspect, the invention relates to methods for modulating gene expression in a cell (e.g., a cell for which levels of the target gene are reduced or enhanced) for gene or epigenetic therapy. The cells can be in vitro, ex vivo, or in vivo (e.g., in a subject who has a disease resulting from reduced expression or activity of a target gene. In some embodiments, methods for modulating gene expression in a cell comprise delivering a single stranded oligonucleotide as described herein. In some embodiments, delivery of the single stranded oligonucleotide to the cell results in a level of expression of gene that is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200% or more greater than a level of expression of gene in a control cell to which the single stranded oligonucleotide has not been delivered. In certain embodiments, delivery of the single stranded oligonucleotide to the cell results in a level of expression of gene that is at least 50% greater than a level of expression of gene in a control cell to which the single stranded oligonucleotide has not been delivered. In some embodiments, delivery of the single stranded oligonucleotide to the cell results in a level of expression of gene that is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200% or more less than a level of expression of gene in a control cell to which the single stranded oligonucleotide has not been delivered.


In another aspect of the invention, methods comprise administering to a subject (e.g. a human) a composition comprising a single stranded oligonucleotide as described herein to increase protein levels in the subject. In some embodiments, the increase in protein levels is at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, or more, higher than the amount of a protein in the subject before administering.


In another aspect of the invention, methods comprise administering to a subject (e.g. a human) a composition comprising a single stranded oligonucleotide as described herein to decrease protein levels in the subject. In some embodiments, the decrease in protein levels is a decrease of at least 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 200%, or more, compared to the amount of a protein in the subject before administering.


As another example, to increase or decrease expression of a target gene in a cell, the methods include introducing into the cell a single stranded oligonucleotide that is sufficiently complementary to a lancRNA that maps to a genomic position encompassing or in proximity to a transcriptional boundary of the target gene.


In another aspect of the invention provides methods of treating a disease or condition associated with decreased levels of expression of a target gene in a subject, the method comprising administering a single stranded oligonucleotide as described herein. Exemplary diseases and condition associated with certain genes are provided in Table 2.









TABLE 2







Examples of diseases or conditions treatable with oligonucleotides


targeting lancRNAs associated with genes.








Gene
Disease of conditions





FXN
Friedreich's Ataxia


SMN
Spinal muscular atrophy (SMA) types I-IV


UTRN
Muscular dystrophy (MD) (e.g., Duchenne's muscular dystrophy,



Becker's muscular dystrophy, myotonic dystrophy)


HEMOGLOBIN
Anemia, microcytic anemia, sickle cell anemia and/or thalassemia (e.g.,



alpha-thalassemia, beta-thalaseemia, delta-thalessemia), beta-thalaseemia



(e.g., thalassemia minor/intermedia/major)


ATP2A2
Cardiac conditions (e.g., congenital heart disease, aortic aneurysms,



aortic dissections, arrhythmia, cardiomyopathy, and congestive heart



failure), Darier-White disease and Acrokeratosis verruciformi


APOA1/
Dyslipidemia (e.g. Hyperlipidemia) and atherosclerosis (e.g. coronary


ABCA1
artery disease (CAD) and myocardial infarction (MI))


PTEN
Cancer, such as, leukemias, lymphomas, myelomas, carcinomas,



metastatic carcinomas, sarcomas, adenomas, nervous system cancers and



genito-urinary cancers. In some embodiments, the cancer is adult and



pediatric acute lymphoblastic leukemia, acute myeloid leukemia,



adrenocortical carcinoma, AIDS-related cancers, anal cancer, cancer of



the appendix, astrocytoma, basal cell carcinoma, bile duct cancer,



bladder cancer, bone cancer, osteosarcoma, fibrous histiocytoma, brain



cancer, brain stem glioma, cerebellar astrocytoma, malignant glioma,



ependymoma, medulloblastoma, supratentorial primitive



neuroectodermal tumors, hypothalamic glioma, breast cancer, male



breast cancer, bronchial adenomas, Burkitt lymphoma, carcinoid tumor,



carcinoma of unknown origin, central nervous system lymphoma,



cerebellar astrocytoma, malignant glioma, cervical cancer, childhood



cancers, chronic lymphocytic leukemia, chronic myelogenous leukemia,



chronic myeloproliferative disorders, colorectal cancer, cutaneous T-cell



lymphoma, endometrial cancer, ependymoma, esophageal cancer, Ewing



family tumors, extracranial germ cell tumor, extragonadal germ cell



tumor, extrahepatic bile duct cancer, intraocular melanoma,



retinoblastoma, gallbladder cancer, gastric cancer, gastrointestinal



stromal tumor, extracranial germ cell tumor, extragonadal germ cell



tumor, ovarian germ cell tumor, gestational trophoblastic tumor, glioma,



hairy cell leukemia, head and neck cancer, hepatocellular cancer,



Hodgkin lymphoma, non-Hodgkin lymphoma, hypopharyngeal cancer,



hypothalamic and visual pathway glioma, intraocular melanoma, islet



cell tumors, Kaposi sarcoma, kidney cancer, renal cell cancer, laryngeal



cancer, lip and oral cavity cancer, small cell lung cancer, non-small cell



lung cancer, primary central nervous system lymphoma, Waldenstrom



macroglobulinema, malignant fibrous histiocytoma, medulloblastoma,



melanoma, Merkel cell carcinoma, malignant mesothelioma, squamous



neck cancer, multiple endocrine neoplasia syndrome, multiple myeloma,



mycosis fungoides, myelodysplastic syndromes, myeloproliferative



disorders, chronic myeloproliferative disorders, nasal cavity and



paranasal sinus cancer, nasopharyngeal cancer, neuroblastoma,



oropharyngeal cancer, ovarian cancer, pancreatic cancer, parathyroid



cancer, penile cancer, pharyngeal cancer, pheochromocytoma,



pineoblastoma and supratentorial primitive neuroectodermal tumors,



pituitary cancer, plasma cell neoplasms, pleuropulmonary blastoma,



prostate cancer, rectal cancer, rhabdomyosarcoma, salivary gland cancer,



soft tissue sarcoma, uterine sarcoma, Sezary syndrome, non-melanoma



skin cancer, small intestine cancer, squamous cell carcinoma, squamous



neck cancer, supratentorial primitive neuroectodermal tumors, testicular



cancer, throat cancer, thymoma and thymic carcinoma, thyroid cancer,



transitional cell cancer, trophoblastic tumors, urethral cancer, uterine



cancer, uterine sarcoma, vaginal cancer, vulvar cancer, or Wilms tumor


BDNF
Amyotrophic lateral sclerosis (ALS, also known as Lou Gehrig's



disease), Alzheimer's Disease (AD), and Parkinson's Disease (PD),



Neurodegeneration


MECP2
Rett Syndrome, MECP2-related severe neonatal encephalopathy,



Angelman syndrome, or PPM-X syndrome


FOXP3
Diseases or disorders associated with aberrant immune cell (e.g., T cell)



activation, e.g., autoimmune or inflammatory diseases or disorders.



Examples of autoimmune diseases and disorders that may be treated



according to the methods disclosed herein include, but are not limited to,



Acute Disseminated Encephalomyelitis (ADEM), Acute necrotizing



hemorrhagic leukoencephalitis, Addison's disease,



Agammaglobulinemia, Alopecia areata, Amyloidosis, Ankylosing



spondylitis, Anti-GBM/Anti-TBM nephritis, Antiphospholipid syndrome



(APS), Autoimmune angioedema, Autoimmune aplastic anemia,



Autoimmune dysautonomia, Autoimmune hepatitis, Autoimmune



hyperlipidemia, Autoimmune immunodeficiency, Autoimmune inner ear



disease (AIED), Autoimmune myocarditis, Autoimmune oophoritis,



Autoimmune pancreatitis, Autoimmune retinopathy, Autoimmune



thrombocytopenic purpura (ATP), Autoimmune thyroid disease,



Autoimmune urticaria, Axonal & neuronal neuropathies, Balo disease,



Behcet's disease, Bullous pemphigoid, Cardiomyopathy, Castleman



disease, Celiac disease, Chagas disease, Chronic inflammatory



demyelinating polyneuropathy (CIDP), Chronic recurrent multifocal



ostomyelitis (CRMO), Churg-Strauss syndrome, Cicatricial



pemphigoid/benign mucosal pemphigoid, inflammatory bowel disease



(e.g., Crohn's disease or Ulcerative colitis), Cogans syndrome, Cold



agglutinin disease, Congenital heart block, Coxsackie myocarditis,



CREST disease, Essential mixed cryoglobulinemia, Demyelinating



neuropathies, Dermatitis herpetiformis, Dermatomyositis, Devic's



disease (neuromyelitis optica), Discoid lupus, Dressler's syndrome,



Endometriosis, Eosinophilic esophagitis, Eosinophilic fasciitis, Erythema



nodosum, Experimental allergic encephalomyelitis, Evans syndrome,



Fibrosing alveolitis, Giant cell arteritis (temporal arteritis), Giant cell



myocarditis, Glomerulonephritis, Goodpasture's syndrome,



Granulomatosis with Polyangiitis (GPA) (formerly called Wegener's



Granulomatosis), Graves' disease, Guillain-Barre syndrome, Hashimoto's



encephalitis, Hashimoto's thyroiditis, Hemolytic anemia, Henoch-



Schonlein purpura, Herpes gestationis, Hypogammaglobulinemia,



Idiopathic thrombocytopenic purpura (ITP), IgA nephropathy, IgG4-



related sclerosing disease, Immunoregulatory lipoproteins, Inclusion



body myositis, Interstitial cystitis, IPEX (Immunodysregulation,



Polyendocrinopathy, and Enteropathy, X-linked) syndrome, Juvenile



arthritis, Juvenile diabetes (Type 1 diabetes), Juvenile myositis,



Kawasaki syndrome, Lambert-Eaton syndrome, Leukocytoclastic



vasculitis, Lichen planus, Lichen sclerosus, Ligneous conjunctivitis,



Linear IgA disease (LAD), systemic lupus erythematosus (SLE), chronic



Lyme disease, Meniere's disease, Microscopic polyangiitis, Mixed



connective tissue disease (MCTD), Mooren's ulcer, Mucha-Habermann



disease, Multiple sclerosis, Myasthenia gravis, Myositis, Narcolepsy,



Neuromyelitis optica (Devic's), Neutropenia, Ocular cicatricial



pemphigoid, Optic neuritis, Palindromic rheumatism, PANDAS



(Pediatric Autoimmune Neuropsychiatric Disorders Associated with



Streptococcus), Paraneoplastic cerebellar degeneration, Paroxysmal



nocturnal hemoglobinuria (PNH), Parry Romberg syndrome,



Parsonnage-Turner syndrome, Pars planitis (peripheral uveitis),



Pemphigus, Peripheral neuropathy, Perivenous encephalomyelitis,



Pernicious anemia, POEMS syndrome, Polyarteritis nodosa, Type I, II,



& III autoimmune polyglandular syndromes, Polymyalgia rheumatica,



Polymyositis, Postmyocardial infarction syndrome, Postpericardiotomy



syndrome, Progesterone dermatitis, Primary biliary cirrhosis, Primary



sclerosing cholangitis, Psoriasis, Psoriatic arthritis, Idiopathic pulmonary



fibrosis, Pyoderma gangrenosum, Pure red cell aplasia, Raynauds



phenomenon, Reactive Arthritis, Reflex sympathetic dystrophy, Reiter's



syndrome, Relapsing polychondritis, Restless legs syndrome,



Retroperitoneal fibrosis, Rheumatic fever, Rheumatoid arthritis,



Sarcoidosis, Schmidt syndrome, Scleritis, Scleroderma, Sjogren's



syndrome, Sperm & testicular autoimmunity, Stiff person syndrome,



Subacute bacterial endocarditis (SBE), Susac's syndrome, Sympathetic



ophthalmia, Takayasu's arteritis, Temporal arteritis/Giant cell arteritis,



Thrombocytopenic purpura (TTP), Tolosa-Hunt syndrome, Transverse



myelitis, Type 1 diabetes, Undifferentiated connective tissue disease



(UCTD), Uveitis, Vasculitis, Vesiculobullous dermatosis, Vitiligo, and



Wegener's granulomatosis (also called Granulomatosis with Polyangiitis



(GPA)). Further examples of autoimmune disease or disorder include



inflammatory bowel disease (e.g., Crohn's disease or Ulcerative colitis),



IPEX syndrome, Multiple sclerosis, Psoriasis, Rheumatoid arthritis, SLE



or Type 1 diabetes. Examples of inflammatory diseases or disorders that



may be treated according to the methods disclosed herein include, but are



not limited to, Acne Vulgaris, Appendicitis, Arthritis, Asthma,



Atherosclerosis, Allergies (Type 1 Hypersensitivity), Bursitis, Colitis,



Chronic Prostatitis, Cystitis, Dermatitis, Glomerulonephritis,



Inflammatory Bowel Disease, Inflammatory Myopathy (e.g.,



Polymyositis, Dermatomyositis, or Inclusion-body Myositis),



Inflammatory Lung Disease, Interstitial Cystitis, Meningitis, Pelvic



Inflammatory Disease, Phlebitis, Psoriasis, Reperfusion Injury,



Rheumatoid Arthritis, Sarcoidosis, Tendonitis, Tonsilitis, Transplant



Rejection, and Vasculitis. In some embodiments, the inflammatory



disease or disorder is asthma.


THRB
Thyroid hormone resistance, mixed dyslipidemia, dyslipidemia,



hypercholesterolemia


NR1H4
Byler disease, cholestasis, cholestasis intrahepatic, dyslipidemia, biliary



cirrhosis primary, fragile x syndrome, hypercholesterolemia,



atherosclerosis, biliary atresia


HAMP
Hemochromatosis (juvenile), hemochromatosis , iron overload,



hereditary hemochromatosis, anemia, inflammation, thalassemia









A subject can include a non-human mammal, e.g. mouse, rat, guinea pig, rabbit, cat, dog, goat, cow, or horse. In preferred embodiments, a subject is a human. Single stranded oligonucleotides have been employed as therapeutic moieties in the treatment of disease states in animals, including humans. Single stranded oligonucleotides can be useful therapeutic modalities that can be configured to be useful in treatment regimens for the treatment of cells, tissues and animals, especially humans.


For therapeutics, an animal, preferably a human, suspected of having a disease or condition is treated by administering single stranded oligonucleotide in accordance with this invention. For example, in one non-limiting embodiment, the methods comprise the step of administering to the animal in need of treatment, a therapeutically effective amount of a single stranded oligonucleotide as described herein.


Formulation, Delivery, and Dosing

The oligonucleotides described herein can be formulated for administration to a subject for treating a condition or disease associated with increased or decreased levels of a target gene. It should be understood that the formulations, compositions and methods can be practiced with any of the oligonucleotides disclosed herein.


The formulations may conveniently be presented in unit dosage form and may be prepared by any methods well known in the art of pharmacy. The amount of active ingredient (e.g., an oligonucleotide or compound of the invention) which can be combined with a carrier material to produce a single dosage form will vary depending upon the host being treated, the particular mode of administration, e.g., intradermal or inhalation. The amount of active ingredient which can be combined with a carrier material to produce a single dosage form will generally be that amount of the compound which produces a therapeutic effect, e.g. tumor regression.


Pharmaceutical formulations of this invention can be prepared according to any method known to the art for the manufacture of pharmaceuticals. Such formulations can contain sweetening agents, flavoring agents, coloring agents and preserving agents. A formulation can be admixtured with nontoxic pharmaceutically acceptable excipients which are suitable for manufacture. Formulations may comprise one or more diluents, emulsifiers, preservatives, buffers, excipients, etc. and may be provided in such forms as liquids, powders, emulsions, lyophilized powders, sprays, creams, lotions, controlled release formulations, tablets, pills, gels, on patches, in implants, etc.


A formulated single stranded oligonucleotide composition can assume a variety of states. In some examples, the composition is at least partially crystalline, uniformly crystalline, and/or anhydrous (e.g., less than 80, 50, 30, 20, or 10% water). In another example, the single stranded oligonucleotide is in an aqueous phase, e.g., in a solution that includes water. The aqueous phase or the crystalline compositions can, e.g., be incorporated into a delivery vehicle, e.g., a liposome (particularly for the aqueous phase) or a particle (e.g., a microparticle as can be appropriate for a crystalline composition). Generally, the single stranded oligonucleotide composition is formulated in a manner that is compatible with the intended method of administration.


In some embodiments, the composition is prepared by at least one of the following methods: spray drying, lyophilization, vacuum drying, evaporation, fluid bed drying, or a combination of these techniques; or sonication with a lipid, freeze-drying, condensation and other self-assembly.


A single stranded oligonucleotide preparation can be formulated or administered (together or separately) in combination with another agent, e.g., another therapeutic agent or an agent that stabilizes a single stranded oligonucleotide, e.g., a protein that complexes with single stranded oligonucleotide. Still other agents include chelators, e.g., EDTA (e.g., to remove divalent cations such as Mg2+), salts, RNAse inhibitors (e.g., a broad specificity RNAse inhibitor such as RNAsin) and so forth.


In one embodiment, the single stranded oligonucleotide preparation includes another single stranded oligonucleotide, e.g., a second single stranded oligonucleotide that modulates expression of a second gene or a second single stranded oligonucleotide that modulates expression of the first gene. Still other preparation can include at least 3, 5, ten, twenty, fifty, or a hundred or more different single stranded oligonucleotide species. Such single stranded oligonucleotides can mediated gene expression with respect to a similar number of different genes. In one embodiment, the single stranded oligonucleotide preparation includes at least a second therapeutic agent (e.g., an agent other than an oligonucleotide).


Route of Delivery

A composition that includes a single stranded oligonucleotide can be delivered to a subject by a variety of routes. Exemplary routes include: intravenous, intradermal, topical, rectal, parenteral, anal, intravaginal, intranasal, pulmonary, ocular, and oral. The term “therapeutically effective amount” is the amount of oligonucleotide present in the composition that is needed to provide the desired level of target gene expression in the subject to be treated to give the anticipated physiological response. The term “physiologically effective amount” is that amount delivered to a subject to give the desired palliative or curative effect. The term “pharmaceutically acceptable carrier” means that the carrier can be administered to a subject with no significant adverse toxicological effects to the subject.


The single stranded oligonucleotide molecules of the invention can be incorporated into pharmaceutical compositions suitable for administration. Such compositions typically include one or more species of single stranded oligonucleotide and a pharmaceutically acceptable carrier. As used herein the language “pharmaceutically acceptable carrier” is intended to include any and all solvents, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents, and the like, compatible with pharmaceutical administration. The use of such media and agents for pharmaceutically active substances is well known in the art. Except insofar as any conventional media or agent is incompatible with the active compound, use thereof in the compositions is contemplated. Supplementary active compounds can also be incorporated into the compositions.


The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (including ophthalmic, vaginal, rectal, intranasal, transdermal), oral or parenteral. Parenteral administration includes intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, or intrathecal or intraventricular administration.


The route and site of administration may be chosen to enhance targeting. For example, to target muscle cells, intramuscular injection into the muscles of interest would be a logical choice. Lung cells might be targeted by administering the single stranded oligonucleotide in aerosol form. The vascular endothelial cells could be targeted by coating a balloon catheter with the single stranded oligonucleotide and mechanically introducing the oligonucleotide.


Topical administration refers to the delivery to a subject by contacting the formulation directly to a surface of the subject. The most common form of topical delivery is to the skin, but a composition disclosed herein can also be directly applied to other surfaces of the body, e.g., to the eye, a mucous membrane, to surfaces of a body cavity or to an internal surface. As mentioned above, the most common topical delivery is to the skin. The term encompasses several routes of administration including, but not limited to, topical and transdermal. These modes of administration typically include penetration of the skin's permeability barrier and efficient delivery to the target tissue or stratum. Topical administration can be used as a means to penetrate the epidermis and dermis and ultimately achieve systemic delivery of the composition. Topical administration can also be used as a means to selectively deliver oligonucleotides to the epidermis or dermis of a subject, or to specific strata thereof, or to an underlying tissue.


Formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful.


Transdermal delivery is a valuable route for the administration of lipid soluble therapeutics. The dermis is more permeable than the epidermis and therefore absorption is much more rapid through abraded, burned or denuded skin. Inflammation and other physiologic conditions that increase blood flow to the skin also enhance transdermal adsorption. Absorption via this route may be enhanced by the use of an oily vehicle (inunction) or through the use of one or more penetration enhancers. Other effective ways to deliver a composition disclosed herein via the transdermal route include hydration of the skin and the use of controlled release topical patches. The transdermal route provides a potentially effective means to deliver a composition disclosed herein for systemic and/or local therapy. In addition, iontophoresis (transfer of ionic solutes through biological membranes under the influence of an electric field), phonophoresis or sonophoresis (use of ultrasound to enhance the absorption of various therapeutic agents across biological membranes, notably the skin and the cornea), and optimization of vehicle characteristics relative to dose position and retention at the site of administration may be useful methods for enhancing the transport of topically applied compositions across skin and mucosal sites.


Both the oral and nasal membranes offer advantages over other routes of administration. For example, oligonucleotides administered through these membranes may have a rapid onset of action, provide therapeutic plasma levels, avoid first pass effect of hepatic metabolism, and avoid exposure of the oligonucleotides to the hostile gastrointestinal (GI) environment. Additional advantages include easy access to the membrane sites so that the oligonucleotide can be applied, localized and removed easily.


In oral delivery, compositions can be targeted to a surface of the oral cavity, e.g., to sublingual mucosa which includes the membrane of ventral surface of the tongue and the floor of the mouth or the buccal mucosa which constitutes the lining of the cheek. The sublingual mucosa is relatively permeable thus giving rapid absorption and acceptable bioavailability of many agents. Further, the sublingual mucosa is convenient, acceptable and easily accessible.


A pharmaceutical composition of single stranded oligonucleotide may also be administered to the buccal cavity of a human being by spraying into the cavity, without inhalation, from a metered dose spray dispenser, a mixed micellar pharmaceutical formulation as described above and a propellant. In one embodiment, the dispenser is first shaken prior to spraying the pharmaceutical formulation and propellant into the buccal cavity.


Compositions for oral administration include powders or granules, suspensions or solutions in water, syrups, slurries, emulsions, elixirs or non-aqueous media, tablets, capsules, lozenges, or troches. In the case of tablets, carriers that can be used include lactose, sodium citrate and salts of phosphoric acid. Various disintegrants such as starch, and lubricating agents such as magnesium stearate, sodium lauryl sulfate and talc, are commonly used in tablets. For oral administration in capsule form, useful diluents are lactose and high molecular weight polyethylene glycols. When aqueous suspensions are required for oral use, the nucleic acid compositions can be combined with emulsifying and suspending agents. If desired, certain sweetening and/or flavoring agents can be added.


Parenteral administration includes intravenous drip, subcutaneous, intraperitoneal or intramuscular injection, intrathecal or intraventricular administration. In some embodiments, parental administration involves administration directly to the site of disease (e.g. injection into a tumor).


Formulations for parenteral administration may include sterile aqueous solutions which may also contain buffers, diluents and other suitable additives. Intraventricular injection may be facilitated by an intraventricular catheter, for example, attached to a reservoir. For intravenous use, the total concentration of solutes should be controlled to render the preparation isotonic.


Any of the single stranded oligonucleotides described herein can be administered to ocular tissue. For example, the compositions can be applied to the surface of the eye or nearby tissue, e.g., the inside of the eyelid. For ocular administration, ointments or droppable liquids may be delivered by ocular delivery systems known to the art such as applicators or eye droppers. Such compositions can include mucomimetics such as hyaluronic acid, chondroitin sulfate, hydroxypropyl methylcellulose or poly(vinyl alcohol), preservatives such as sorbic acid, EDTA or benzylchronium chloride, and the usual quantities of diluents and/or carriers. The single stranded oligonucleotide can also be administered to the interior of the eye, and can be introduced by a needle or other delivery device which can introduce it to a selected area or structure.


Pulmonary delivery compositions can be delivered by inhalation by the patient of a dispersion so that the composition, preferably single stranded oligonucleotides, within the dispersion can reach the lung where it can be readily absorbed through the alveolar region directly into blood circulation. Pulmonary delivery can be effective both for systemic delivery and for localized delivery to treat diseases of the lungs.


Pulmonary delivery can be achieved by different approaches, including the use of nebulized, aerosolized, micellular and dry powder-based formulations. Delivery can be achieved with liquid nebulizers, aerosol-based inhalers, and dry powder dispersion devices. Metered-dose devices are preferred. One of the benefits of using an atomizer or inhaler is that the potential for contamination is minimized because the devices are self-contained. Dry powder dispersion devices, for example, deliver agents that may be readily formulated as dry powders. A single stranded oligonucleotide composition may be stably stored as lyophilized or spray-dried powders by itself or in combination with suitable powder carriers. The delivery of a composition for inhalation can be mediated by a dosing timing element which can include a timer, a dose counter, time measuring device, or a time indicator which when incorporated into the device enables dose tracking, compliance monitoring, and/or dose triggering to a patient during administration of the aerosol medicament.


The term “powder” means a composition that consists of finely dispersed solid particles that are free flowing and capable of being readily dispersed in an inhalation device and subsequently inhaled by a subject so that the particles reach the lungs to permit penetration into the alveoli. Thus, the powder is said to be “respirable.” Preferably the average particle size is less than about 10 μm in diameter preferably with a relatively uniform spheroidal shape distribution. More preferably the diameter is less than about 7.5 μm and most preferably less than about 5.0 μm. Usually the particle size distribution is between about 0.1 μm and about 5 μm in diameter, particularly about 0.3 μm to about 5 μm.


The term “dry” means that the composition has a moisture content below about 10% by weight (% w) water, usually below about 5% w and preferably less it than about 3% w. A dry composition can be such that the particles are readily dispersible in an inhalation device to form an aerosol.


The types of pharmaceutical excipients that are useful as carrier include stabilizers such as human serum albumin (HSA), bulking agents such as carbohydrates, amino acids and polypeptides; pH adjusters or buffers; salts such as sodium chloride; and the like. These carriers may be in a crystalline or amorphous form or may be a mixture of the two.


Suitable pH adjusters or buffers include organic salts prepared from organic acids and bases, such as sodium citrate, sodium ascorbate, and the like; sodium citrate is preferred. Pulmonary administration of a micellar single stranded oligonucleotide formulation may be achieved through metered dose spray devices with propellants such as tetrafluoroethane, heptafluoroethane, dimethylfluoropropane, tetrafluoropropane, butane, isobutane, dimethyl ether and other non-CFC and CFC propellants.


Exemplary devices include devices which are introduced into the vasculature, e.g., devices inserted into the lumen of a vascular tissue, or which devices themselves form a part of the vasculature, including stents, catheters, heart valves, and other vascular devices. These devices, e.g., catheters or stents, can be placed in the vasculature of the lung, heart, or leg.


Other devices include non-vascular devices, e.g., devices implanted in the peritoneum, or in organ or glandular tissue, e.g., artificial organs. The device can release a therapeutic substance in addition to a single stranded oligonucleotide, e.g., a device can release insulin.


In one embodiment, unit doses or measured doses of a composition that includes single stranded oligonucleotide are dispensed by an implanted device. The device can include a sensor that monitors a parameter within a subject. For example, the device can include pump, e.g., and, optionally, associated electronics.


Tissue, e.g., cells or organs can be treated with a single stranded oligonucleotide, ex vivo and then administered or implanted in a subject. The tissue can be autologous, allogeneic, or xenogeneic tissue. E.g., tissue can be treated to reduce graft v. host disease. In other embodiments, the tissue is allogeneic and the tissue is treated to treat a disorder characterized by unwanted gene expression in that tissue. E.g., tissue, e.g., hematopoietic cells, e.g., bone marrow hematopoietic cells, can be treated to inhibit unwanted cell proliferation. Introduction of treated tissue, whether autologous or transplant, can be combined with other therapies. In some implementations, the single stranded oligonucleotide treated cells are insulated from other cells, e.g., by a semi-permeable porous barrier that prevents the cells from leaving the implant, but enables molecules from the body to reach the cells and molecules produced by the cells to enter the body. In one embodiment, the porous barrier is formed from alginate.


In one embodiment, a contraceptive device is coated with or contains a single stranded oligonucleotide. Exemplary devices include condoms, diaphragms, IUD (implantable uterine devices, sponges, vaginal sheaths, and birth control devices.


Dosage

In one aspect, the invention features a method of administering a single stranded oligonucleotide (e.g., as a compound or as a component of a composition) to a subject (e.g., a human subject). In one embodiment, the unit dose is between about 10 mg and 25 mg per kg of bodyweight. In one embodiment, the unit dose is between about 1 mg and 100 mg per kg of bodyweight. In one embodiment, the unit dose is between about 0.1 mg and 500 mg per kg of bodyweight. In some embodiments, the unit dose is more than 0.001, 0.005, 0.01, 0.05, 0.1, 0.5, 1, 2, 5, 10, 25, 50 or 100 mg per kg of bodyweight.


The defined amount can be an amount effective to treat or prevent a disease or condition, e.g., a disease or condition associated with the target gene. The unit dose, for example, can be administered by injection (e.g., intravenous or intramuscular), an inhaled dose, or a topical application.


In some embodiments, the unit dose is administered daily. In some embodiments, less frequently than once a day, e.g., less than every 2, 4, 8 or 30 days. In another embodiment, the unit dose is not administered with a frequency (e.g., not a regular frequency). For example, the unit dose may be administered a single time. In some embodiments, the unit dose is administered more than once a day, e.g., once an hour, two hours, four hours, eight hours, twelve hours, etc.


In one embodiment, a subject is administered an initial dose and one or more maintenance doses of a single stranded oligonucleotide. The maintenance dose or doses are generally lower than the initial dose, e.g., one-half less of the initial dose. A maintenance regimen can include treating the subject with a dose or doses ranging from 0.0001 to 100 mg/kg of body weight per day, e.g., 100, 10, 1, 0.1, 0.01, 0.001, or 0.0001 mg per kg of bodyweight per day. The maintenance doses may be administered no more than once every 1, 5, 10, or 30 days. Further, the treatment regimen may last for a period of time which will vary depending upon the nature of the particular disease, its severity and the overall condition of the patient. In some embodiments the dosage may be delivered no more than once per day, e.g., no more than once per 24, 36, 48, or more hours, e.g., no more than once for every 5 or 8 days. Following treatment, the patient can be monitored for changes in his condition and for alleviation of the symptoms of the disease state. The dosage of the oligonucleotide may either be increased in the event the patient does not respond significantly to current dosage levels, or the dose may be decreased if an alleviation of the symptoms of the disease state is observed, if the disease state has been ablated, or if undesired side-effects are observed.


The effective dose can be administered in a single dose or in two or more doses, as desired or considered appropriate under the specific circumstances. If desired to facilitate repeated or frequent infusions, implantation of a delivery device, e.g., a pump, semi-permanent stent (e.g., intravenous, intraperitoneal, intracisternal or intracapsular), or reservoir may be advisable.


In some embodiments, the oligonucleotide pharmaceutical composition includes a plurality of single stranded oligonucleotide species. In another embodiment, the single stranded oligonucleotide species has sequences that are non-overlapping and non-adjacent to another species with respect to a naturally occurring target sequence (e.g., a lancRNA). In another embodiment, the plurality of single stranded oligonucleotide species is specific for different lancRNAs. In another embodiment, the single stranded oligonucleotide is allele specific. In some cases, a patient is treated with a single stranded oligonucleotide in conjunction with other therapeutic modalities.


Following successful treatment, it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the compound of the invention is administered in maintenance doses, ranging from 0.0001 mg to 100 mg per kg of body weight.


The concentration of the single stranded oligonucleotide composition is an amount sufficient to be effective in treating or preventing a disorder or to regulate a physiological condition in humans. The concentration or amount of single stranded oligonucleotide administered will depend on the parameters determined for the agent and the method of administration, e.g. nasal, buccal, pulmonary. For example, nasal formulations may tend to require much lower concentrations of some ingredients in order to avoid irritation or burning of the nasal passages. It is sometimes desirable to dilute an oral formulation up to 10-100 times in order to provide a suitable nasal formulation.


Certain factors may influence the dosage required to effectively treat a subject, including but not limited to the severity of the disease or disorder, previous treatments, the general health and/or age of the subject, and other diseases present. Moreover, treatment of a subject with a therapeutically effective amount of a single stranded oligonucleotide can include a single treatment or, preferably, can include a series of treatments. It will also be appreciated that the effective dosage of a single stranded oligonucleotide used for treatment may increase or decrease over the course of a particular treatment. For example, the subject can be monitored after administering a single stranded oligonucleotide composition. Based on information from the monitoring, an additional amount of the single stranded oligonucleotide composition can be administered.


Dosing is dependent on severity and responsiveness of the disease or condition to be treated, with the course of treatment lasting from several days to several months, or until a cure is effected or a diminution of disease state is achieved. Optimal dosing schedules can be calculated from measurements of target gene expression levels in the body of the patient. Persons of ordinary skill can easily determine optimum dosages, dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative potency of individual compounds, and can generally be estimated based on EC50s found to be effective in in vitro and in vivo animal models. In some embodiments, the animal models include transgenic animals that express a human target gene. In another embodiment, the composition for testing includes a single stranded oligonucleotide that is complementary, at least in an internal region, to a sequence that is conserved between the target gene in the animal model and the target gene in a human.


In one embodiment, the administration of the single stranded oligonucleotide composition is parenteral, e.g. intravenous (e.g., as a bolus or as a diffusible infusion), intradermal, intraperitoneal, intramuscular, intrathecal, intraventricular, intracranial, subcutaneous, transmucosal, buccal, sublingual, endoscopic, rectal, oral, vaginal, topical, pulmonary, intranasal, urethral or ocular. Administration can be provided by the subject or by another person, e.g., a health care provider. The composition can be provided in measured doses or in a dispenser which delivers a metered dose. Selected modes of delivery are discussed in more detail below.


Kits

In certain aspects of the invention, kits are provided, comprising a container housing a composition comprising a single stranded oligonucleotide. In some embodiments, the composition is a pharmaceutical composition comprising a single stranded oligonucleotide and a pharmaceutically acceptable carrier. In some embodiments, the individual components of the pharmaceutical composition may be provided in one container. Alternatively, it may be desirable to provide the components of the pharmaceutical composition separately in two or more containers, e.g., one container for single stranded oligonucleotides, and at least another for a carrier compound. The kit may be packaged in a number of different configurations such as one or more containers in a single box. The different components can be combined, e.g., according to instructions provided with the kit. The components can be combined according to a method described herein, e.g., to prepare and administer a pharmaceutical composition. The kit can also include a delivery device.


The present invention is further illustrated by the following Examples, which in no way should be construed as further limiting. The entire contents of all of the references (including literature references, issued patents, published patent applications, and co-pending patent applications) cited throughout this application are hereby expressly incorporated by reference.


EXAMPLES

The invention is further described in the following examples, which do not limit the scope of the invention described in the claims.


Example 1. APOA1 and FXN lancRNA-Targeting Oligonucleotides
Oligo Design

Oligonucleotides were designed to target sense and antisense regions located within a 500 nucleotide window of the transcription start and end sites of APOA1 and FXN. The oligonucleotide sequence and modification (“formatted”) patterns are provided in Table 3 below. Table 4 provides a description of the nucleotide analogs, modifications and intranucleotide linkages used for certain oligonucleotides tested and described in Table 3. A map of each gene showing where each oligonucleotide binds is provided in FIGS. 1 and 2.









TABLE 3







Oligonucleotides test












Oligo

Gene
SEQ




Name
Organism
Name
ID NO
Base Sequence
Formatted Sequence















FXN-
human
FXN
1
CGCAGTAGCCGGCCT
InaCs;omeGs;InaCs;omeAs;InaGs;omeU


62




s;InaAs;omeGs;InaCs;omeCs;InaGs;ome


m01




Gs;InaCs;omeCs;InaT-Sup





FXN-
human
FXN
2
CCTGGCGTCACCCAG
InaCs;omeCs;InaTs;omeGs;InaGs;omeC


601




s;InaGs;omeUs;InaCs;omeAs;InaCs;ome


m01




Cs;InaCs;omeAs;InaG-Sup





FXN-
human
FXN
3
CCCAGCCCAGGCCCA
InaCs;omeCs;InaCs;omeAs;InaGs;omeC


602




s;InaCs;omeCs;InaAs;omeGs;InaGs;ome


m01




Cs;InaCs;omeCs;InaA-Sup





FXN-
human
FXN
4
CCCAGACCCTCACCC
InaCs;omeCs;InaCs;omeAs;InaGs;omeA


603




s;InaCs;omeCs;InaCs;omeUs;InaCs;ome


m01




As;InaCs;omeCs;InaC-Sup





FXN-
human
FXN
5
TCCCGCGGCCGGCAG
InaTs;omeCs;InaCs;omeCs;InaGs;omeCs


604




;InaGs;omeGs;InaCs;omeCs;InaGs;ome


m01




Gs;InaCs;omeAs;InaG-Sup





FXN-
human
FXN
6
AGAGTTGGCCCCACT
InaAs;omeGs;InaAs;omeGs;InaTs;omeU


605




s;InaGs;omeGs;InaCs;omeCs;InaCs;ome


m01




Cs;InaAs;omeCs;InaT-Sup





FXN-
human
FXN
7
AGCAGCTGGTCAACC
dAs;InaGs;dCs;InaAs;dGs;InaCs;dTs;Ina


606




Gs;dGs;InaTs;dCs;InaAs;dAs;InaCs;dC-


m02




Sup





FXN-
human
FXN
8
TGCTCACTGTTCTAT
dTs;InaGs;dCs;InaTs;dCs;InaAs;dCs;InaT


607




s;dGs;InaTs;dTs;InaCs;dTs;InaAs;dT-Sup


m02










FXN-
human
FXN
9
CTCCAAATGAGACAC
dCs;InaTs;dCs;InaCs;dAs;InaAs;dAs;InaT


608




s;dGs;InaAs;dGs;InaAs;dCs;InaAs;dC-


m02




Sup





FXN-
human
FXN
10
ATTAAAGGGTAGCCT
dAs;InaTs;dTs;InaAs;dAs;InaAs;dGs;Ina


609




Gs;dGs;InaTs;dAs;InaGs;dCs;InaCs;dT-


m02




Sup





FXN-
human
FXN
11
ACAAATGTTTTCAGG
dAs;InaCs;dAs;InaAs;dAs;InaTs;dGs;InaT


610




s;dTs;InaTs;dTs;InaCs;dAs;InaGs;dG-Sup


m02










FXN-
human
FXN
12
CTTCTTTCAAAGTGT
dCs;InaTs;dTs;InaCs;dTs;InaTs;dTs;InaCs


611




;dAs;InaAs;dAs;InaGs;dTs;InaGs;dT-Sup


m02










FXN-
human
FXN
13
TAATGTCTTATGCCT
dTs;InaAs;dAs;InaTs;dGs;InaTs;dCs;InaT


612




s;dTs;InaAs;dTs;InaGs;dCs;InaCs;dT-Sup


m02










FXN-
human
FXN
14
ATACCTGAATATAAC
dAs;InaTs;dAs;InaCs;dCs;InaTs;dGs;InaA


613




s;dAs;InaTs;dAs;InaTs;dAs;InaAs;dC-Sup


m02










FXN-
human
FXN
15
AACCTTTAAAAAAGC
dAs;InaAs;dCs;InaCs;dTs;InaTs;dTs;InaA


614




s;dAs;InaAs;dAs;InaAs;dAs;InaGs;dC-


m02




Sup





FXN-
human
FXN
16
AAAATAATAAGAAGG
dAs;InaAs;dAs;InaAs;dTs;InaAs;dAs;InaT


615




s;dAs;InaAs;dGs;InaAs;dAs;InaGs;dG-


m02




Sup





FXN-
human
FXN
17
AAAAATTCCAGGAGG
dAs;InaAs;dAs;InaAs;dAs;InaTs;dTs;InaC


616




s;dCs;InaAs;dGs;InaGs;dAs;InaGs;dG-


m02




Sup





FXN-
human
FXN
18
GAAAATGAATTGTCT
dGs;InaAs;dAs;InaAs;dAs;InaTs;dGs;Ina


617




As;dAs;InaTs;dTs;InaGs;dTs;InaCs;dT-


m02




Sup





FXN-
human
FXN
19
TCACTCTTCATTCTT
dTs;InaCs;dAs;InaCs;dTs;InaCs;dTs;InaT


618




s;dCs;InaAs;dTs;InaTs;dCs;InaTs;dT-Sup


m02










FXN-
human
FXN
20
TGAAGGATTTACTGC
dTs;InaGs;dAs;InaAs;dGs;InaGs;dAs;Ina


619




Ts;dTs;InaTs;dAs;InaCs;dTs;InaGs;dC-


m02




Sup





FXN-
human
FXN
21
TTGGATTGTCGGAT
dTs;InaTs;dGs;InaGs;dAs;InaTs;dTs;InaG


620




s;dTs;InaCs;dGs;InaGs;dAs;InaT-Sup


m02










FXN-
human
FXN
22
TTCCTCCCTCACATG
dTs;InaTs;dCs;InaCs;dTs;InaCs;dCs;InaC


621




s;dTs;InaCs;dAs;InaCs;dAs;InaTs;dG-Sup


m02










FXN-
human
FXN
23
ATACCCCTTATCTTT
dAs;InaTs;dAs;InaCs;dCs;InaCs;dCs;InaT


622




s;dTs;InaAs;dTs;InaCs;dTs;InaTs;dT-Sup


m02










FXN-
human
FXN
24
TATAATGTCTTATGC
dTs;InaAs;dTs;InaAs;dAs;InaTs;dGs;InaT


623




s;dCs;InaTs;dTs;InaAs;dTs;InaGs;dC-Sup


m02










FXN-
human
FXN
25
TGAGGACAGTTGGGC
dTs;InaGs;dAs;InaGs;dGs;InaAs;dCs;Ina


624




As;dGs;InaTs;dTs;InaGs;dGs;InaGs;dC-


m02




Sup





FXN-
human
FXN
26
TATGTGTCACAGCTC
dTs;InaAs;dTs;InaGs;dTs;InaGs;dTs;InaC


625




s;dAs;InaCs;dAs;InaGs;dCs;InaTs;dC-Sup


m02










FXN-
human
FXN
27
TGTAGAAAGAATGTG
dTs;InaGs;dTs;InaAs;dGs;InaAs;dAs;Ina


626




As;dGs;InaAs;dAs;InaTs;dGs;InaTs;dG-


m02




Sup





FXN-
human
FXN
28
TTGCCTCCTACCTTG
dTs;InaTs;dGs;InaCs;dCs;InaTs;dCs;InaC


627




s;dTs;InaAs;dCs;InaCs;dTs;InaTs;dG-Sup


m02










FXN-
human
FXN
29
CCCCCAAGTTCTGAT
dCs;InaCs;dCs;InaCs;dCs;InaAs;dAs;InaG


628




s;dTs;InaTs;dCs;InaTs;dGs;InaAs;dT-Sup


m02










FXN-
human
FXN
30
ATGCTTGATGCCCAG
dAs;InaTs;dGs;InaCs;dTs;InaTs;dGs;InaA


629




s;dTs;InaGs;dCs;InaCs;dCs;InaAs;dG-


m02




Sup





FXN-
human
FXN
31
CCCCGTTTTAAGGAC
dCs;InaCs;dCs;InaCs;dGs;InaTs;dTs;InaT


630




s;dTs;InaAs;dAs;InaGs;dGs;InaAs;dC-


m02




Sup





FXN-
human
FXN
32
ATTAAAAGCTATCAG
dAs;InaTs;dTs;InaAs;dAs;InaAs;dAs;InaG


631




s;dCs;InaTs;dAs;InaTs;dCs;InaAs;dG-Sup


m02










FXN-
human
FXN
33
GCCAAGACCCCAGCT
dGs;InaCs;dCs;InaAs;dAs;InaGs;dAs;Ina


632




Cs;dCs;InaCs;dCs;InaAs;dGs;InaCs;dT-


m02




Sup





FXN-
human
FXN
34
TCATTATGCAGCTGA
dTs;InaCs;dAs;InaTs;dTs;InaAs;dTs;InaG


633




s;dCs;InaAs;dGs;InaCs;dTs;InaGs;dA-


m02




Sup





FXN-
human
FXN
35
GGTCTGTTTTTTGTT
dGs;InaGs;dTs;InaCs;dTs;InaGs;dTs;InaT


634




s;dTs;InaTs;dTs;InaTs;dGs;InaTs;dT-Sup


m02










FXN-
human
FXN
36
GTTGTTGTTGTTTAT
dGs;InaTs;dTs;InaGs;dTs;InaTs;dGs;InaT


635




s;dTs;InaGs;dTs;InaTs;dTs;InaAs;dT-Sup


m02










FXN-
human
FXN
37
GCAGAGCTCACTAAA
dGs;InaCs;dAs;InaGs;dAs;InaGs;dCs;Ina


636




Ts;dCs;InaAs;dCs;InaTs;dAs;InaAs;dA-


m02




Sup





FXN-
human
FXN
38
GCCTTAAAAACCAAA
dGs;InaCs;dCs;InaTs;dTs;InaAs;dAs;InaA


637




s;dAs;InaAs;dCs;InaCs;dAs;InaAs;dA-


m02




Sup





FXN-
human
FXN
39
CTGGACTTGTCTTCC
dCs;InaTs;dGs;InaGs;dAs;InaCs;dTs;InaT


638




s;dGs;InaTs;dCs;InaTs;dTs;InaCs;dC-Sup


m02










FXN-
human
FXN
40
CAGGACATTAAAATT
dCs;InaAs;dGs;InaGs;dAs;InaCs;dAs;Ina


640




Ts;dTs;InaAs;dAs;InaAs;dAs;InaTs;dT-


m02




Sup





FXN-
human
FXN
41
CATGCAAAGTTATGC
dCs;InaAs;dTs;InaGs;dCs;InaAs;dAs;InaA


641




s;dGs;InaTs;dTs;InaAs;dTs;InaGs;dC-Sup


m02










FXN-
human
FXN
42
AAGTGCAGTAGGCCA
dAs;InaAs;dGs;InaTs;dGs;InaCs;dAs;Ina


642




Gs;dTs;InaAs;dGs;InaGs;dCs;InaCs;dA-


m02




Sup





FXN-
human
FXN
43
GTGCCAGTGAGAAAA
dGs;InaTs;dGs;InaCs;dCs;InaAs;dGs;Ina


643




Ts;dGs;InaAs;dGs;InaAs;dAs;InaAs;dA-


m02




Sup





FXN-
human
FXN
44
TAAATAACATCATAC
dTs;InaAs;dAs;InaAs;dTs;InaAs;dAs;InaC


644




s;dAs;InaTs;dCs;InaAs;dTs;InaAs;dC-Sup


m02










FXN-
human
FXN
45
ATGTTTGTATGTGTT
dAs;InaTs;dGs;InaTs;dTs;InaTs;dGs;InaT


645




s;dAs;InaTs;dGs;InaTs;dGs;InaTs;dT-Sup


m02










FXN-
human
FXN
46
AATCTATAAAATGGA
dAs;InaAs;dTs;InaCs;dTs;InaAs;dTs;InaA


646




s;dAs;InaAs;dAs;InaTs;dGs;InaGs;dA-


m02




Sup





FXN-
human
FXN
47
CAGTTTGCATTAAAT
dCs;InaAs;dGs;InaTs;dTs;InaTs;dGs;InaC


647




s;dAs;InaTs;dTs;InaAs;dAs;InaAs;dT-Sup


m02










FXN-
human
FXN
48
TATAGGTTTACAATA
dTs;InaAs;dTs;InaAs;dGs;InaGs;dTs;InaT


648




s;dTs;InaAs;dCs;InaAs;dAs;InaTs;dA-Sup


m02










FXN-
human
FXN
49
GTTATAATTATATGT
dGs;InaTs;dTs;InaAs;dTs;InaAs;dAs;InaT


649




s;dTs;InaAs;dTs;InaAs;dTs;InaGs;dT-Sup


m02










FXN-
human
FXN
50
TTAAGATAGTTGTTC
dTs;InaTs;dAs;InaAs;dGs;InaAs;dTs;InaA


650




s;dGs;InaTs;dTs;InaGs;dTs;InaTs;dC-Sup


m02










FXN-
human
FXN
51
AATAAACTCTAAATA
dAs;InaAs;dTs;InaAs;dAs;InaAs;dCs;InaT


651




s;dCs;InaTs;dAs;InaAs;dAs;InaTs;dA-Sup


m02










FXN-
human
FXN
52
ACCCCAACTCCAAGA
dAs;InaCs;dCs;InaCs;dCs;InaAs;dAs;InaC


652




s;dTs;InaCs;dCs;InaAs;dAs;InaGs;dA-Sup


m02










FXN-
human
FXN
53
GTGTTAGCAAGAAAT
dGs;InaTs;dGs;InaTs;dTs;InaAs;dGs;InaC


653




s;dAs;InaAs;dGs;InaAs;dAs;InaAs;dT-


m02




Sup





Apoa
mouse
Apoa
54
TCCAAAATGGAATA
InaTs;omeCs;InaCs;omeAs;InaAs;om


1_mu

1

G
eAs;InaAs;omeTs;InaGs;omeGs;InaA


s-26




s;omeAs;InaTs;omeAs;InaG-Sup


m01










Apoa
mouse
Apoa
55
TTTCCAAAATGGAA
InaTs;omeTs;InaTs;omeCs;InaCs;om


1_mu

1

T
eAs;InaAs;omeAs;InaAs;omeTs;InaG


s-27




s;omeGs;InaAs;omeAs;InaT-Sup


m01










Apoa
mouse
Apoa
56
ACCTTTCCAAAATG
InaAs;omeCs;InaCs;omeTs;InaTs;om


1_mu

1

G
eTs;InaCs;omeCs;InaAs;omeAs;InaA


s-28




s;omeAs;InaTs;omeGs;InaG-Sup


m01










Apoa
mouse
Apoa
57
AACCTTTCCAAAAT
InaAs;omeAs;InaCs;omeCs;InaTs;om


1_mu

1

G
eTs;InaTs;omeCs;InaCs;omeAs;InaA


s-29




s;omeAs;InaAs;omeTs;InaG-Sup


m01










Apoa
mouse
Apoa
58
ACAATAAACCTTTC
InaAs;omeCs;InaAs;omeAs;InaTs;o


1_mu

1

C
meAs;InaAs;omeAs;InaCs;omeCs;In


s-30




aTs;omeTs;InaTs;omeCs;InaC-Sup


m01










Apoa
mouse
Apoa
59
GGTGCCCGCTTCCA
InaGs;omeGs;InaTs;omeGs;InaCs;o


1_mu

1

C
meCs;InaCs;omeGs;InaCs;omeTs;Ina


s-31




Ts;omeCs;InaCs;omeAs;InaC-Sup


m01










Apoa
mouse
Apoa
60
TCCACTCCCCACCC
InaTs;omeCs;InaCs;omeAs;InaCs;om


1_mu

1

C
eTs;InaCs;omeCs;InaCs;omeCs;InaA


s-32




s;omeCs;InaCs;omeCs;InaC-Sup


m01










Apoa
mouse
Apoa
61
ACCCCCGCATTGGC
InaAs;omeCs;InaCs;omeCs;InaCs;om


1_mu

1

T
eCs;InaGs;omeCs;InaAs;omeTs;InaT


s-33




s;omeGs;InaGs;omeCs;InaT-Sup


m01










Apoa
mouse
Apoa
62
TGGCTTTCTTACAA
InaTs;omeGs;InaGs;omeCs;InaTs;o


1_mu

1

T
meTs;InaTs;omeCs;InaTs;omeTs;Ina


s-34




As;omeCs;InaAs;omeAs;InaT-Sup


m01










Apoa
mouse
Apoa
63
GGAATAGCTTCTTT
InaGs;omeGs;InaAs;omeAs;InaTs;o


1_mu

1

C
meAs;InaGs;omeCs;InaTs;omeTs;Ina


s-35




Cs;omeTs;InaTs;omeTs;InaC-Sup


m01










Apoa
mouse
Apoa
64
CTTTCTTTGGGGGA
InaCs;omeTs;InaTs;omeTs;InaCs;om


1_mu



C
eTs;InaTs;omeTs;InaGs;omeGs;InaG


s-36

1


s;omeGs;InaGs;omeAs;InaC-Sup


m01










Apoa
mouse
Apoa
65
CACCCAGACTGTCG
InaCs;omeAs;InaCs;omeCs;InaCs;om


1_mu

1

G
eAs;InaGs;omeAs;InaCs;omeTs;InaG


s-37




s;omeTs;InaCs;omeGs;InaG-Sup


m01










Apoa
mouse
Apoa
66
CAGGGCCAGGCTG
InaCs;omeAs;InaGs;omeGs;InaGs;o


1_mu

1

AG
meCs;InaCs;omeAs;InaGs;omeGs;In


s-38




aCs;omeTs;InaGs;omeAs;InaG-Sup


m01










Apoa
mouse
Apoa
67
GCTGATCCTTGAAC
InaGs;omeCs;InaTs;omeGs;InaAs;o


1_mu

1

T
meTs;InaCs;omeCs;InaTs;omeTs;Ina


s-39




Gs;omeAs;InaAs;omeCs;InaT-Sup


m01










Apoa
mouse
Apoa
68
AGACTGTCGGAGA
InaAs;omeGs;InaAs;omeCs;InaTs;o


1_mu

1

GC
meGs;InaTs;omeCs;InaGs;omeGs;In


s-40




aAs;omeGs;InaAs;omeGs;InaC-Sup


m01










Apoa
mouse
Apoa
69
TGTCGGAGAGCTC
InaTs;omeGs;InaTs;omeCs;InaGs;o


1_mu

1

CG
meGs;InaAs;omeGs;InaAs;omeGs;In


s-41




aCs;omeTs;InaCs;omeCs;InaG-Sup


m01










Apoa
mouse
Apoa
70
GCTGGACACCCAG
InaGs;omeCs;InaTs;omeGs;InaGs;o


1_mu

1

AC
meAs;InaCs;omeAs;InaCs;omeCs;In


s-42




aCs;omeAs;InaGs;omeAs;InaC-Sup


m01










Apoa
mouse
Apoa
71
GAAGAGCTGGACA
InaGs;omeAs;InaAs;omeGs;InaAs;o


1_mu

1

CC
meGs;InaCs;omeTs;InaGs;omeGs;In


s-43




aAs;omeCs;InaAs;omeCs;InaC-Sup


m01










Apoa
mouse
Apoa
72
GGGAAGAAGAGCT
InaGs;omeGs;InaGs;omeAs;InaAs;o


1_mu

1

GG
meGs;InaAs;omeAs;InaGs;omeAs;In


s-44




aGs;omeCs;InaTs;omeGs;InaG-Sup


m01










Apoa
mouse
Apoa
73
GACCAGGGAAGAA
InaGs;omeAs;InaCs;omeCs;InaAs;o


1_mu

1

GA
meGs;InaGs;omeGs;InaAs;omeAs;In


s-45




aGs;omeAs;InaAs;omeGs;InaA-Sup


m01










Apoa
mouse
Apoa
74
CACATATATAGACC
InaCs;omeAs;InaCs;omeAs;InaTs;om


1_mu

1

A
eAs;InaTs;omeAs;InaTs;omeAs;InaG


s-46




s;omeAs;InaCs;omeCs;InaA-Sup


m01










FXN-
human
FXN
75
GTCTCCCTTGGGTC
InaGs;omeUs;InaCs;omeUs;InaCs;o


800



A
meCs;InaCs;omeUs;InaTs;omeGs;In


m01




aGs;omeGs;InaTs;omeCs;InaA-Sup





FXN-
human
FXN
76
TGCGGCCAGTGGC
InaTs;omeGs;InaCs;omeGs;InaGs;o


801



CA
meCs;InaCs;omeAs;InaGs;omeUs;In


m01




aGs;omeGs;InaCs;omeCs;InaA-Sup





FXN-
human
FXN
77
CACCAGGGGTCGC
InaCs;omeAs;InaCs;omeCs;InaAs;o


802



CG
meGs;deazaGs;omeGs;InaGs;omeU


m01




s;InaCs;omeGs;InaCs;omeCs;InaG-







Sup





FXN-
human
FXN
78
CAGCGCTGGAGGG
InaCs;omeAs;InaGs;omeCs;InaGs;o


803



CG
meCs;InaTs;omeGs;InaGs;omeAs;In


m01




aGs;omeGs;deazaGs;omeCs;InaG-







Sup





FXN-
human
FXN
79
CTGGAGGGCGGAG
InaCs;omeUs;InaGs;omeGs;InaAs;o


804



CG
meGs;deazaGs;omeGs;InaCs;omeGs


m01




;InaGs;omeAs;InaGs;omeCs;InaG-







Sup





FXN-
human
FXN
80
GTCTCCCTTGGGTC
InaGs;InaTs;InaCs;dTs;dCs;dCs;dCs;d


800



A
Ts;dTs;dGs;dGs;dGs;InaTs;InaCs;Ina


m08




A-Sup





FXN-
human
FXN
81
TGCGGCCAGTGGC
InaTs;InaGs;InaCs;dGs;dGs;dCs;dCs;


801



CA
dAs;dGs;dTs;dGs;dGs;InaCs;InaCs;In


m08




aA-Sup





FXN-
human
FXN
82
CACCAGGGGTCGC
InaCs;InaAs;InaCs;dCs;dAs;dGs;dGs;


802



CG
dGs;dGs;dTs;dCs;dGs;InaCs;InaCs;In


m08




aG-Sup





FXN-
human
FXN
83
CAGCGCTGGAGGG
InaCs;InaAs;InaGs;dCs;dGs;dCs;dTs;


803



CG
dGs;dGs;dAs;dGs;dGs;InaGs;InaCs;1


m08




naG-Sup





FXN-
human
FXN
84
CTGGAGGGCGGAG
InaCs;InaTs;InaGs;dGs;dAs;dGs;dGs;


804



CG
dGs;dCs;dGs;dGs;dAs;InaGs;InaCs;In


m08




aG-Sup





FXN-
human
FXN
85
AACTGCTGTAAACC
InaAs;InaAs;InaCs;dTs;dGs;dCs;dTs;


805



C
dGs;dTs;dAs;dAs;dAs;InaCs;InaCs;In


m08




aC-Sup





FXN-
human
FXN
86
ATACCGGCGGCCA
InaAs;InaTs;InaAs;dCs;dCs;dGs;dGs;


806



AG
dCs;dGs;dGs;dCs;dCs;InaAs;InaAs;In


m08




aG-Sup





FXN-
human
FXN
87
CAGCCTCAATTTGT
InaCs;InaAs;InaGs;dCs;dCs;dTs;dCs;


807



G
dAs;dAs;dTs;dTs;dTs;InaGs;InaTs;Ina


m08




G-Sup





FXN-
human
FXN
88
CATGCACCCACTTC
InaCs;InaAs;InaTs;dGs;dCs;dAs;dCs;


808



C
dCs;dCs;dAs;dCs;dTs;InaTs;InaCs;Ina


m08




C-Sup





FXN-
human
FXN
89
CAGCAAGACAGCA
InaCs;InaAs;InaGs;dCs;dAs;dAs;dGs;


809



GC
dAs;dCs;dAs;dGs;dCs;InaAs;InaGs;In


m08




aC-Sup





FXN-
human
FXN
90
TCCCAAGTTCCTCC
InaTs;InaCs;InaCs;dCs;dAs;dAs;dGs;


810



T
dTs;dTs;dCs;dCs;dTs;InaCs;InaCs;Ina


m08




T-Sup





FXN-
human
FXN
91
GTTTAGAATTTTAG
InaGs;InaTs;InaTs;dTs;dAs;dGs;dAs;


811



A
dAs;dTs;dTs;dTs;dTs;InaAs;InaGs;Ina


m08




A-Sup





FXN-
human
FXN
92
GGCTGCAGTCTCCC
InaGs;InaGs;InaCs;dTs;dGs;dCs;dAs;


812



T
dGs;dTs;dCs;dTs;dCs;InaCs;InaCs;Ina


m08




T-Sup





FXN-
human
FXN
93
TCCTGGTTGCACTC
InaTs;InaCs;InaCs;dTs;dGs;dGs;dTs;


588



C
dTs;dGs;dCs;dAs;dCs;InaTs;InaCs;In


m08




aC-Sup





FXN-
human
FXN
94
AGTTCTTCCTGAGG
InaAs;InaGs;InaTs;dTs;dCs;dTs;dTs;d


593



T
Cs;dCs;dTs;dGs;dAs;InaGs;InaGs;Ina


m08




T-Sup





FXN-
human
FXN
95
CTAACCTCTAGCTG
InaCs;InaTs;InaAs;dAs;dCs;dCs;dTs;d


40



C
Cs;dTs;dAs;dGs;dCs;InaTs;InaGs;Ina


m08




C-Sup





FXN-
human
FXN
96
CACAGAAGAGTGC
InaCs;InaAs;InaCs;dAs;dGs;dAs;dAs;


816



CT
dGs;dAs;dGs;dTs;dGs;InaCs;InaCs;In


m08




aT-Sup





FXN-
human
FXN
97
GCCAGTGGCCACC
InaGs;InaCs;InaCs;dAs;dGs;dTs;dGs;


817



AG
dGs;dCs;dCs;dAs;dCs;InaCs;InaAs;In


m08




aG-Sup





FXN-
human
FXN
98
GCAGCACCCAGCG
InaGs;InaCs;InaAs;dGs;dCs;dAs;dCs;


818



CT
dCs;dCs;dAs;dGs;dCs;InaGs;InaCs;In


m08




aT-Sup





FXN-
human
FXN
99
GAGCAGCATGTGG
InaGs;InaAs;InaGs;dCs;dAs;dGs;dCs;


819



AC
dAs;dTs;dGs;dTs;dGs;InaGs;InaAs;In


m08




aC-Sup





FXN-
human
FXN
100
TCTCCCACTCAACA
InaTs;InaCs;InaTs;dCs;dCs;dCs;dAs;d


820



C
Cs;dTs;dCs;dAs;dAs;InaCs;InaAs;Ina


m08




C-Sup





FXN-
human
FXN
101
TCACACCTGTTAGT
InaTs;InaCs;InaAs;dCs;dAs;dCs;dCs;


821



T
dTs;dGs;dTs;dTs;dAs;InaGs;InaTs;In


m08




aT-Sup





FXN-
human
FXN
102
TTCCTCTTGACACTT
InaTs;InaTs;InaCs;dCs;dTs;dCs;dTs;d


822




Ts;dGs;dAs;dCs;dAs;InaCs;InaTs;Ina


m08




T-Sup





FXN-
human
FXN
103
GTCATTTAGCATCC
InaGs;InaTs;InaCs;dAs;dTs;dTs;dTs;d


823



T
As;dGs;dCs;dAs;dTs;InaCs;InaCs;Ina


m08




T-Sup





FXN-
human
FXN
104
AAGTATGTAAACAT
InaAs;InaAs;InaGs;dTs;dAs;dTs;dGs;


824



G
dTs;dAs;dAs;dAs;dCs;InaAs;InaTs;In


m08




aG-Sup





FXN-
human
FXN
105
CACGATTCACAAAG
InaCs;InaAs;InaCs;dGs;dAs;dTs;dTs;


825



T
dCs;dAs;dCs;dAs;dAs;InaAs;InaGs;In


m08




aT-Sup





FXN-
human
FXN
106
GGCTTTGGAAGAA
InaGs;InaGs;InaCs;dTs;dTs;dTs;dGs;


826



CT
dGs;dAs;dAs;dGs;dAs;InaAs;InaCs;In


m08




aT-Sup





FXN-
human
FXN
107
TTAGTACCTTCCCA
InaTs;InaTs;InaAs;dGs;dTs;dAs;dCs;


827



T
dCs;dTs;dTs;dCs;dCs;InaCs;InaAs;Ina


m08




T-Sup





FXN-
human
FXN
108
GTCTCCCTTGGGTC
InaGs;omeUs;InaCs;omeUs;InaCs;o


800



A
meCs;InaCs;omeUs;InaTs;omeGs;In


m01




aGs;omeGs;InaTs;omeCs;InaA-Sup





FXN-
human
FXN
109
CGCTCCGCCCTCCAG
dCs;InaGs;dCs;InaTs;dCs;InaCs;dGs;InaC


375




s;dCs;InaCs;dTs;InaCs;dCs;InaAs;dG-Sup


m02










FXN-
human
FXN
110
ATTATTTTGCTTTTT
dAs;InaTs;dTs;InaAs;dTs;InaTs;dTs;InaTs


390




;dGs;InaCs;dTs;InaTs;dTs;InaTs;dT-Sup


m02










FXN-
human
FXN
111
AGGCCACGGCGGCC
InaAs;omeGs;InaGs;omeCs;InaCs;omeA


577



GCA
s;InaCs;omeGs;InaGs;omeCs;InaGs;ome


m01




Gs;InaCs;omeCs;InaGs;omeCs;InaA-Sup





FXN-
human
FXN
112
CATCGATGTCGGTGC
InaCs;omeAs;InaTs;omeCs;InaGs;omeA


578



GC
s;InaTs;omeGs;InaTs;omeCs;InaGs;ome


m01




Gs;InaTs;omeGs;InaCs;omeGs;InaC-Sup





FXN-
human
FXN
113
ACACATAGCCCAACT
InaAs;InaCs;InaAs;dCs;dAs;dTs;dAs;dGs;


695




dCs;dCs;dCs;dAs;InaAs;InaCs;InaT-Sup


m08










HAMP-
human
HAMP
114
CTCAGACCACCGCC
InaCs;omeUs;InaCs;omeAs;InaGs;o


17



T
meAs;InaCs;omeCs;InaAs;omeCs;In


m01




aCs;omeGs;InaCs;omeCs;InaT-Sup





HAMP-
human
HAMP
115
CCACCGCCTCCCCT
InaCs;omeCs;InaAs;omeCs;InaCs;om


18



G
eGs;InaCs;omeCs;InaTs;omeCs;InaC


m01




s;omeCs;InaCs;omeUs;InaG-Sup





HAMP-
human
HAMP
116
CAGGCCCCATAAAA
InaCs;omeAs;InaGs;omeGs;InaCs;o


19



G
meCs;InaCs;omeCs;InaAs;omeUs;In


m01




aAs;omeAs;InaAs;omeAs;InaG-Sup





HAMP-
human
HAMP
117
CATAAAAGCGACT
InaCs;omeAs;InaTs;omeAs;InaAs;o


20



GT
meAs;InaAs;omeGs;InaCs;omeGs;In


m01




aAs;omeCs;InaTs;omeGs;InaT-Sup





HAMP-
human
HAMP
118
ACTGTCACTCGGTC
InaAs;omeCs;InaTs;omeGs;InaTs;om


21



C
eCs;InaAs;omeCs;InaTs;omeCs;InaG


m01




s;omeGs;InaTs;omeCs;InaC-Sup





HAMP-
human
HAMP
119
ACTCGGTCCCAGAC
InaAs;omeCs;InaTs;omeCs;InaGs;o


22



A
meGs;InaTs;omeCs;InaCs;omeCs;Ina


m01




As;omeGs;InaAs;omeCs;InaA-Sup





HAMP-
human
HAMP
120
AGACACCAGAGCA
InaAs;omeGs;InaAs;omeCs;InaAs;o


23



AG
meCs;InaCs;omeAs;InaGs;omeAs;In


m01




aGs;omeCs;InaAs;omeAs;InaG-Sup





HAMP-
human
HAMP
121
GAGCAAGCTCAAG
InaGs;omeAs;InaGs;omeCs;InaAs;o


24



AC
meAs;InaGs;omeCs;InaTs;omeCs;In


m01




aAs;omeAs;InaGs;omeAs;InaC-Sup





HAMP-
human
HAMP
122
GCAAGACGTAGAA
InaGs;omeCs;InaAs;omeAs;InaGs;o


25



CC
meAs;InaCs;omeGs;InaTs;omeAs;In


m01




aGs;omeAs;InaAs;omeCs;InaC-Sup





HAMP-
human
HAMP
123
AGACGTAGAACCT
InaAs;omeGs;InaAs;omeCs;InaGs;o


26



AC
meUs;InaAs;omeGs;InaAs;omeAs;In


m01




aCs;omeCs;InaTs;omeAs;InaC-Sup





HAMP-
human
HAMP
124
AGAACCTACCTGCC
InaAs;omeGs;InaAs;omeAs;InaCs;o


27



C
meCs;InaTs;omeAs;InaCs;omeCs;Ina


m01




Ts;omeGs;InaCs;omeCs;InaC-Sup





HAMP-
human
HAMP
125
ACATAGGTCTTGGA
InaAs;omeCs;InaAs;omeUs;InaAs;o


28



A
meGs;InaGs;omeUs;InaCs;omeUs;In


m01




aTs;omeGs;InaGs;omeAs;InaA-Sup





HAMP-
human
HAMP
126
AGGTCTTGGAATA
InaAs;omeGs;InaGs;omeUs;InaCs;o


29



AA
meUs;InaTs;omeGs;InaGs;omeAs;In


m01




aAs;omeUs;InaAs;omeAs;InaA-Sup





HAMP-
human
HAMP
127
TGGAATAAAATGG
InaTs;omeGs;InaGs;omeAs;InaAs;o


30



CT
meUs;InaAs;omeAs;InaAs;omeAs;In


m01




aTs;omeGs;InaGs;omeCs;InaT-Sup





HAMP-
human
HAMP
128
TGGCTGGTTCTTTT
InaTs;omeGs;InaGs;omeCs;InaTs;o


31



G
meGs;InaGs;omeUs;InaTs;omeCs;In


m01




aTs;omeUs;InaTs;omeUs;InaG-Sup





HAMP-
human
HAMP
129
GTTTTCCAAACCAG
InaGs;omeUs;InaTs;omeUs;InaTs;o


32



A
meCs;InaCs;omeAs;InaAs;omeAs;In


m01




aCs;omeCs;InaAs;omeGs;InaA-Sup





HAMP-
human
HAMP
130
ACCAGAGTGTCTGT
InaAs;omeCs;InaCs;omeAs;InaGs;o


33



T
meAs;InaGs;omeUs;InaGs;omeUs;In


m01




aCs;omeUs;InaGs;omeUs;InaT-Sup





HAMP-
human
HAMP
131
AGTGTCTGTTGTCC
InaAs;omeGs;InaTs;omeGs;InaTs;o


34



T
meCs;InaTs;omeGs;InaTs;omeUs;In


m01




aGs;omeUs;InaCs;omeCs;InaT-Sup





HAMP-
human
HAMP
132
GTTGTCCTTTCTCTC
InaGs;omeUs;InaTs;omeGs;InaTs;o


35




meCs;InaCs;omeUs;InaTs;omeUs;In


m01




aCs;omeUs;InaCs;omeUs;InaC-Sup





HAMP-
human
HAMP
133
CTTTCTCTCTGCCG
InaCs;omeUs;InaTs;omeUs;InaCs;o


36



A
meUs;InaCs;omeUs;InaCs;omeUs;In


m01




aGs;omeCs;InaCs;omeGs;InaA-Sup





HAMP-
human
HAMP
134
CTGCCGAGTGTCTG
InaCs;omeUs;InaGs;omeCs;InaCs;o


37



T
meGs;InaAs;omeGs;InaTs;omeGs;In


m01




aTs;omeCs;InaTs;omeGs;InaT-Sup





HAMP-
human
HAMP
135
CTCAGACCACCGCC
InaCs;InaTs;InaCs;dAs;dGs;dAs;dCs;


17



T
dCs;dAs;dCs;dCs;dGs;InaCs;InaCs;In


m08




aT-Sup





HAMP-
human
HAMP
136
CCACCGCCTCCCCT
InaCs;InaCs;InaAs;dCs;dCs;dGs;dCs;


18




dCs;dTs;dCs;dCs;dCs;InaCs;InaTs;Ina


m08




G-Sup





HAMP-
human
HAMP
137
CAGGCCCCATAAAA
InaCs;InaAs;InaGs;dGs;dCs;dCs;dCs;


19



G
dCs;dAs;dTs;dAs;dAs;InaAs;InaAs;In


m08




aG-Sup





HAMP-
human
HAMP
138
CATAAAAGCGACT
InaCs;InaAs;InaTs;dAs;dAs;dAs;dAs;


20



GT
dGs;dCs;dGs;dAs;dCs;InaTs;InaGs;In


m08




aT-Sup





HAMP-
human
HAMP
139
ACTGTCACTCGGTC
InaAs;InaCs;InaTs;dGs;dTs;dCs;dAs;


21



C
dCs;dTs;dCs;dGs;dGs;InaTs;InaCs;In


m08




aC-Sup





HAMP-
human
HAMP
140
ACTCGGTCCCAGAC
InaAs;InaCs;InaTs;dCs;dGs;dGs;dTs;


22



A
dCs;dCs;dCs;dAs;dGs;InaAs;InaCs;In


m08




aA-Sup





HAMP-
human
HAMP
141
AGACACCAGAGCA
InaAs;InaGs;InaAs;dCs;dAs;dCs;dCs;


23



AG
dAs;dGs;dAs;dGs;dCs;InaAs;InaAs;In


m08




aG-Sup





HAMP-
human
HAMP
142
GAGCAAGCTCAAG
InaGs;InaAs;InaGs;dCs;dAs;dAs;dGs;


24



AC
dCs;dTs;dCs;dAs;dAs;InaGs;InaAs;In


m08




aC-Sup





HAMP-
human
HAMP
143
GCAAGACGTAGAA
InaGs;InaCs;InaAs;dAs;dGs;dAs;dCs;


25



CC
dGs;dTs;dAs;dGs;dAs;InaAs;InaCs;In


m08




aC-Sup





HAMP-
human
HAMP
144
AGACGTAGAACCT
InaAs;InaGs;InaAs;dCs;dGs;dTs;dAs;


26



AC
dGs;dAs;dAs;dCs;dCs;InaTs;InaAs;In


m08




aC-Sup





HAMP-
human
HAMP
145
AGAACCTACCTGCC
InaAs;InaGs;InaAs;dAs;dCs;dCs;dTs;


27



C
dAs;dCs;dCs;dTs;dGs;InaCs;InaCs;In


m08




aC-Sup





HAMP
human-
HAMP
146
ACATAGGTCTTGGA
InaAs;InaCs;InaAs;dTs;dAs;dGs;dGs;


28



A
dTs;dCs;dTs;dTs;dGs;InaGs;InaAs;In


m08




aA-Sup





HAMP-
human
HAMP
147
AGGTCTTGGAATA
InaAs;InaGs;InaGs;dTs;dCs;dTs;dTs;


29



AA
dGs;dGs;dAs;dAs;dTs;InaAs;InaAs;In


m08




aA-Sup





HAMP-
human
HAMP
148
TGGAATAAAATGG
InaTs;InaGs;InaGs;dAs;dAs;dTs;dAs;


30



CT
dAs;dAs;dAs;dTs;dGs;InaGs;InaCs;In


m08




aT-Sup





HAMP-
human
HAMP
149
TGGCTGGTTCTTTT
InaTs;InaGs;InaGs;dCs;dTs;dGs;dGs;


31



G
dTs;dTs;dCs;dTs;dTs;InaTs;InaTs;Ina


m08




G-Sup





HAMP-
human
HAMP
150
GTTTTCCAAACCAG
InaGs;InaTs;InaTs;dTs;dTs;dCs;dCs;d


32



A
As;dAs;dAs;dCs;dCs;InaAs;InaGs;Ina


m08




A-Sup





HAMP-
human
HAMP
151
ACCAGAGTGTCTGT
InaAs;InaCs;InaCs;dAs;dGs;dAs;dGs;


33



T
dTs;dGs;dTs;dCs;dTs;InaGs;InaTs;Ina


m08




T-Sup





HAMP-
human
HAMP
152
AGTGTCTGTTGTCC
InaAs;InaGs;InaTs;dGs;dTs;dCs;dTs;


34



T
dGs;dTs;dTs;dGs;dTs;InaCs;InaCs;In


m08




aT-Sup





HAMP-
human
HAMP
153
GTTGTCCTTTCTCTC
InaGs;InaTs;InaTs;dGs;dTs;dCs;dCs;


35




dTs;dTs;dTs;dCs;dTs;InaCs;InaTs;Ina


m08




C-Sup





HAMP-
human
HAMP
154
CTTTCTCTCTGCCG
InaCs;InaTs;InaTs;dTs;dCs;dTs;dCs;d


36



A
Ts;dCs;dTs;dGs;dCs;InaCs;InaGs;Ina


m08




A-Sup





HAMP-
human
HAMP
155
CTGCCGAGTGTCTG
InaCs;InaTs;InaGs;dCs;dCs;dGs;dAs;


37



T
dGs;dTs;dGs;dTs;dCs;InaTs;InaGs;In


m08




aT-Sup





NR1H
human
NR1
156
CTCTCCCAAGGTT
InaCs;omeUs;InaCs;omeUs;InaCs;o


4-27

H4

C
CmeCs;InaCs;omeAs;InaAs;omeGs;In


m01




aGs;omeUs;InaTs;omeCs;InaC-Sup





NR1H
human
NR1
157
AGGTTCCTTTCTAT
InaAs;omeGs;InaGs;omeUs;InaTs;o


4-28

H4

G
meCs;InaCs;omeUs;InaTs;omeUs;In


m01




aCs;omeUs;InaAs;omeUs;InaG-Sup





NR1H
human
NR1
158
ATGTTTATATCATTT
InaAs;omeUs;InaGs;omeUs;InaTs;o


4-29

H4


meUs;InaAs;omeUs;InaAs;omeUs;In


m01




aCs;omeAs;InaTs;omeUs;InaT-Sup





NR1H
human
NR1
159
ATATCATTTAGCAG
InaAs;omeUs;InaAs;omeUs;InaCs;o


4-30

H4

G
meAs;InaTs;omeUs;InaTs;omeAs;In


m01




aGs;omeCs;InaAs;omeGs;InaG-Sup





NR1H
human
NR1
160
ATTGTTAATGACT
InaAs;omeUs;InaTs;omeGs;InaTs;o


4-31

H4

A
AmeUs;InaAs;omeAs;InaTs;omeGs;In


m01




aAs;omeCs;InaTs;omeAs;InaA-Sup





NR1H
human
NR1
161
AGCTTCTAGTTCAG
InaAs;omeGs;InaCs;omeUs;InaTs;o


4-32

H4

T
meCs;InaTs;omeAs;InaGs;omeUs;In


m01




aTs;omeCs;InaAs;omeGs;InaT-Sup





NR1H
human
NR1
162
AGTGATAGAGCTA
InaAs;omeGs;InaTs;omeGs;InaAs;o


4-33

H4

TT
meUs;InaAs;omeGs;InaAs;omeGs;In


m01




aCs;omeUs;InaAs;omeUs;InaT-Sup





NR1H
human
NR1
163
AGAGAGGGAAGAT
InaAs;omeGs;InaAs;omeGs;InaAs;o


4-34

H4

GA
meGs;InaGs;omeGs;InaAs;omeAs;In


m01




aGs;omeAs;InaTs;omeGs;InaA-Sup





NR1H
human
NR1
164
AGTTGATGTGTACA
InaAs;omeGs;InaTs;omeUs;InaGs;o


4-35

H4

G
meAs;InaTs;omeGs;InaTs;omeGs;In


m01




aTs;omeAs;InaCs;omeAs;InaG-Sup





NR1H
human
NR1
165
ACGGGTGCCCAGG
InaAs;omeCs;InaGs;omeGs;InaGs;o


4-36

H4

AG
meUs;InaGs;omeCs;InaCs;omeCs;In


m01




aAs;omeGs;InaGs;omeAs;InaG-Sup





NR1H
human
NR1
166
CACAAAACGGCCA
InaCs;omeAs;InaCs;omeAs;InaAs;o


4-37

H4

GA
meAs;InaAs;omeCs;InaGs;omeGs;In


m01




aCs;omeCs;InaAs;omeGs;InaA-Sup





NR1H
human
NR1
167
ATATTGCATATATT
InaAs;omeUs;InaAs;omeUs;InaTs;o


4-38

H4

T
meGs;InaCs;omeAs;InaTs;omeAs;In


m01




aTs;omeAs;InaTs;omeUs;InaT-Sup





NR1H
human
NR1
168
ATATTTTATTAAAG
InaAs;omeUs;InaAs;omeUs;InaTs;o


4-39

H4

A
meUs;InaTs;omeAs;InaTs;omeUs;In


m01




aAs;omeAs;InaAs;omeGs;InaA-Sup





NR1H
human
NR1
169
AGAGTTGTATTCAA
InaAs;omeGs;InaAs;omeGs;InaTs;o


4-40

H4

T
meUs;InaGs;omeUs;InaAs;omeUs;In


m01




aTs;omeCs;InaAs;omeAs;InaT-Sup





NR1H
human
NR1
170
TGTATTCAATCTTG
InaTs;omeGs;InaTs;omeAs;InaTs;om


4-41

H4

G
eUs;InaCs;omeAs;InaAs;omeUs;InaC


m01




s;omeUs;InaTs;omeGs;InaG-Sup





NR1H
human
NR1
171
CAATCTTGGCAATA
InaCs;omeAs;InaAs;omeUs;InaCs;o


4-42

H4

A
meUs;InaTs;omeGs;InaGs;omeCs;In


m01




aAs;omeAs;InaTs;omeAs;InaA-Sup





NR1H
human
NR1
172
AGCAAACATAATG
InaAs;omeGs;InaCs;omeAs;InaAs;o


4-43

H4

GC
meAs;InaCs;omeAs;InaTs;omeAs;In


m01




aAs;omeUs;InaGs;omeGs;InaC-Sup





NR1H
human
NR1
173
ATGGCAACAGGAT
InaAs;omeUs;InaGs;omeGs;InaCs;o


4-44

H4

TT
meAs;InaAs;omeCs;InaAs;omeGs;In


m01




aGs;omeAs;InaTs;omeUs;InaT-Sup





NR1H
human
NR1
174
TTTTCTTTGGGAAC
InaTs;omeUs;InaTs;omeUs;InaCs;o


4-45

H4

A
meUs;InaTs;omeUs;InaGs;omeGs;In


m01




aGs;omeAs;InaAs;omeCs;InaA-Sup





NR1H
human
NR1
175
ATTCTAATTGGCAA
InaAs;omeUs;InaTs;omeCs;InaTs;o


4-46

H4

G
meAs;InaAs;omeUs;InaTs;omeGs;In


m01




aGs;omeCs;InaAs;omeAs;InaG-Sup





NR1H
human
NR1
176
ATTGGCAAGCCCTG
InaAs;omeUs;InaTs;omeGs;InaGs;o


4-47

H4

T
meCs;InaAs;omeAs;InaGs;omeCs;In


m01




aCs;omeCs;InaTs;omeGs;InaT-Sup





NR1H
human
NR1
177
AGCCCTGTTTGCCT
InaAs;omeGs;InaCs;omeCs;InaCs;o


4-48

H4

A
meUs;InaGs;omeUs;InaTs;omeUs;In


m01




aGs;omeCs;InaCs;omeUs;InaA-Sup





NR1H
human
NR1
178
CTAATTAAATTGAT
InaCs;omeUs;InaAs;omeAs;InaTs;o


4-49

H4

T
meUs;InaAs;omeAs;InaAs;omeUs;In


m01




aTs;omeGs;InaAs;omeUs;InaT-Sup





NR1H
human
NR1
179
ATTGTTACTTCAAT
InaAs;omeUs;InaTs;omeGs;InaTs;o


4-50

H4

T
meUs;InaAs;omeCs;InaTs;omeUs;In


m01




aCs;omeAs;InaAs;omeUs;InaT-Sup





NR1H
human
NR1
180
TTCTATCTGTTGA
InaTs;omeUs;InaCs;omeUs;InaAs;o


4-51

H4

C
AmeUs;InaCs;omeUs;InaGs;omeUs;In


m01




aTs;omeGs;InaAs;omeAs;InaC-Sup





NR1H
human
NR1
181
CTCTCCCAAGGTTC
InaCs;InaTs;InaCs;dTs;dCs;dCs;dCs;d


4-27

H4

C
As;dAs;dGs;dGs;dTs;InaTs;InaCs;Ina


m08




C-Sup





NR1H
human
NR1
182
AGGTTCCTTTCTAT
InaAs;InaGs;InaGs;dTs;dTs;dCs;dCs;


4-28

H4

G
dTs;dTs;dTs;dCs;dTs;InaAs;InaTs;Ina


m08




G-Sup





NR1H
human
NR1
183
ATGTTTATATCATTT
InaAs;InaTs;InaGs;dTs;dTs;dTs;dAs;d


4-29

H4


Ts;dAs;dTs;dCs;dAs;InaTs;InaTs;InaT


m08




-Sup





NR1H
human
NR1
184
ATATCATTTAGCAG
InaAs;InaTs;InaAs;dTs;dCs;dAs;dTs;d


4-30

H4

G
Ts;dTs;dAs;dGs;dCs;InaAs;InaGs;Ina


m08




G-Sup





NR1H
human
NR1
185
ATTGTTAATGACTA
InaAs;InaTs;InaTs;dGs;dTs;dTs;dAs;d


4-31

H4

A
As;dTs;dGs;dAs;dCs;InaTs;InaAs;Ina


m08




A-Sup





NR1H
human
NR1
186
AGCTTCTAGTTCAG
InaAs;InaGs;InaCs;dTs;dTs;dCs;dTs;d


4-32

H4

T
As;dGs;dTs;dTs;dCs;InaAs;InaGs;Ina


m08




T-Sup





NR1H
human
NR1
187
AGTGATAGAGCTA
InaAs;InaGs;InaTs;dGs;dAs;dTs;dAs;


4-33

H4

TT
dGs;dAs;dGs;dCs;dTs;InaAs;InaTs;In


m08




aT-Sup





NR1H
human
NR1
188
AGAGAGGGAAGAT
InaAs;InaGs;InaAs;dGs;dAs;dGs;dGs;


4-34

H4

GA
dGs;dAs;dAs;dGs;dAs;InaTs;InaGs;In


m08




aA-Sup





NR1H
human
NR1
189
AGTTGATGTGTACA
InaAs;InaGs;InaTs;dTs;dGs;dAs;dTs;


4-35

H4

G
dGs;dTs;dGs;dTs;dAs;InaCs;InaAs;In


m08




aG-Sup





NR1H
human
NR1
190
ACGGGTGCCCAGG
InaAs;InaCs;InaGs;dGs;dGs;dTs;dGs;


4-36

H4

AG
dCs;dCs;dCs;dAs;dGs;InaGs;InaAs;In


m08




aG-Sup





NR1H
human
NR1
191
CACAAAACGGCCA
InaCs;InaAs;InaCs;dAs;dAs;dAs;dAs;


4-37

H4

GA
dCs;dGs;dGs;dCs;dCs;InaAs;InaGs;In


m08




aA-Sup





NR1H
human
NR1
192
ATATTGCATATATT
InaAs;InaTs;InaAs;dTs;dTs;dGs;dCs;


4-38

H4

T
dAs;dTs;dAs;dTs;dAs;InaTs;InaTs;Ina


m08




T-Sup





NR1H
human
NR1
193
ATATTTTATTAAAG
InaAs;InaTs;InaAs;dTs;dTs;dTs;dTs;d


4-39

H4

A
As;dTs;dTs;dAs;dAs;InaAs;InaGs;Ina


m08




A-Sup





NR1H
human
NR1
194
AGAGTTGTATTCAA
InaAs;InaGs;InaAs;dGs;dTs;dTs;dGs;


4-40

H4

T
dTs;dAs;dTs;dTs;dCs;InaAs;InaAs;Ina


m08




T-Sup





NR1H
human
NR1
195
TGTATTCAATCTTG
InaTs;InaGs;InaTs;dAs;dTs;dTs;dCs;d


4-41

H4

G
As;dAs;dTs;dCs;dTs;InaTs;InaGs;Ina


m08




G-Sup





NR1H
human
NR1
196
CAATCTTGGCAATA
InaCs;InaAs;InaAs;dTs;dCs;dTs;dTs;d


4-42

H4

A
Gs;dGs;dCs;dAs;dAs;InaTs;InaAs;Ina


m08




A-Sup





NR1H
human
NR1
197
AGCAAACATAATG
InaAs;InaGs;InaCs;dAs;dAs;dAs;dCs;


4-43

H4

GC
dAs;dTs;dAs;dAs;dTs;InaGs;InaGs;In


m08




aC-Sup





NR1H
human
NR1
198
ATGGCAACAGGA
InaAs;InaTs;InaGs;dGs;dCs;dAs;dAs;


4-44

H4

TT
TdCs;dAs;dGs;dGs;dAs;InaTs;InaTs;In


m08




aT-Sup





NR1H
human
NR1
199
TTTTCTTTGGGAAC
InaTs;InaTs;InaTs;dTs;dCs;dTs;dTs;d


4-45

H4

A
Ts;dGs;dGs;dGs;dAs;InaAs;InaCs;Ina


m08




A-Sup





NR1H
human
NR1
200
ATTCTAATTGGCAA
InaAs;InaTs;InaTs;dCs;dTs;dAs;dAs;d


4-46

H4

G
Ts;dTs;dGs;dGs;dCs;InaAs;InaAs;Ina


m08




G-Sup





NR1H
human
NR1
201
ATTGGCAAGCCCTG
InaAs;InaTs;InaTs;dGs;dGs;dCs;dAs;


4-47

H4

T
dAs;dGs;dCs;dCs;dCs;InaTs;InaGs;In


m08




aT-Sup





NR1H
human
NR1
202
AGCCCTGTTTGCCT
InaAs;InaGs;InaCs;dCs;dCs;dTs;dGs;


4-48

H4

A
dTs;dTs;dTs;dGs;dCs;InaCs;InaTs;Ina


m08




A-Sup





NR1H
human
NR1
203
CTAATTAAATTGATA
InaCs;InaTs;InaAs;dAs;dTs;dTs;dAs;d


4-49

H4

T
s;dAs;dTs;dTs;dGs;InaAs;InaTs;Ina


m08




T-Sup





NR1H
human
NR1
204
ATTGTTACTTCAATC
InaAs;InaTs;InaTs;dGs;dTs;dTs;dAs;d


4-50

H4

T
s;dTs;dTs;dCs;dAs;InaAs;InaTs;InaT


m08




-Sup





NR1H
human
NR1
205
TTCTATCTGTTGAAT
InaTs;InaTs;InaCs;dTs;dAs;dTs;dCs;d


4-51

H4

C
s;dGs;dTs;dTs;dGs;InaAs;InaAs;Ina


m08




C-Sup





THRB-
human
THRB
206
GAATATAGTGGGC
InaGs;omeAs;InaAs;omeUs;InaAs;o


91



GT
meUs;InaAs;omeGs;InaTs;omeGs;In


m01




aGs;omeGs;InaCs;omeGs;InaT-Sup





THRB-
human
THRB
207
AGTGGGCGTAGAT
InaAs;omeGs;InaTs;omeGs;InaGs;o


92



AA
meGs;InaCs;omeGs;InaTs;omeAs;In


m01




aGs;omeAs;InaTs;omeAs;InaA-Sup





THRB-
human
THRB
208
CGTAGATAAACTCA
InaCs;omeGs;InaTs;omeAs;InaGs;o


93



T
meAs;InaTs;omeAs;InaAs;omeAs;In


m01




aCs;omeUs;InaCs;omeAs;InaT-Sup





THRB-
human
THRB
209
TAAACTCATAAGCT
InaTs;omeAs;InaAs;omeAs;InaCs;o


94



T
meUs;InaCs;omeAs;InaTs;omeAs;In


m01




aAs;omeGs;InaCs;omeUs;InaT-Sup





THRB-
human
THRB
210
CATAAGCTTAAATT
InaCs;omeAs;InaTs;omeAs;InaAs;o


95



C
meGs;InaCs;omeUs;InaTs;omeAs;In


m01




aAs;omeAs;InaTs;omeUs;InaC-Sup





THRB-
human
THRB
211
AAGCTTATAACAGA
InaAs;omeAs;InaGs;omeCs;InaTs;o


96



T
meUs;InaAs;omeUs;InaAs;omeAs;In


m01




aCs;omeAs;InaGs;omeAs;InaT-Sup





THRB-
human
THRB
212
ATAACAGATATATT
InaAs;omeUs;InaAs;omeAs;InaCs;o


97



T
meAs;InaGs;omeAs;InaTs;omeAs;In


m01




aTs;omeAs;InaTs;omeUs;InaT-Sup





THRB-
human
THRB
213
GATATATTTTCCTG
InaGs;omeAs;InaTs;omeAs;InaTs;o


98



T
meAs;InaTs;omeUs;InaTs;omeUs;In


m01




aCs;omeCs;InaTs;omeGs;InaT-Sup





THRB-
human
THRB
214
TTTCCTGTCTCTTTC
InaTs;omeUs;InaTs;omeCs;InaCs;om


99




eUs;InaGs;omeUs;InaCs;omeUs;Ina


m01




Cs;omeUs;InaTs;omeUs;InaC-Sup





THRB-
human
THRB
215
ATGGATTTTTACAT
InaAs;omeUs;InaGs;omeGs;InaAs;o


100



A
meUs;InaTs;omeUs;InaTs;omeUs;In


m01




aAs;omeCs;InaAs;omeUs;InaA-Sup





THRB-
human
THRB
216
TGTATGCAGATATA
InaTs;omeGs;InaTs;omeAs;InaTs;om


101



A
eGs;InaCs;omeAs;InaGs;omeAs;InaT


m01




s;omeAs;InaTs;omeAs;InaA-Sup





THRB-
human
THRB
217
CTGTAATTATGAAT
InaCs;omeUs;InaGs;omeUs;InaAs;o


102



A
meAs;InaTs;omeUs;InaAs;omeUs;In


m01




aGs;omeAs;InaAs;omeUs;InaA-Sup





THRB-
human
THRB
218
TACATAGGCAAAG
InaTs;omeAs;InaCs;omeAs;InaTs;om


103



AG
eAs;InaGs;omeGs;InaCs;omeAs;InaA


m01




s;omeAs;InaGs;omeAs;InaG-Sup





THRB-
human
THRB
219
CAAAGAGTTGCCT
InaCs;omeAs;InaAs;omeAs;InaGs;o


104



GC
meAs;InaGs;omeUs;InaTs;omeGs;In


m01




aCs;omeCs;InaTs;omeGs;InaC-Sup





THRB-
human
THRB
220
CCAGCCGCTTCCTG
InaCs;omeCs;InaAs;omeGs;InaCs;o


105



C
meCs;InaGs;omeCs;InaTs;omeUs;In


m01




aCs;omeCs;InaTs;omeGs;InaC-Sup





THRB-
human
THRB
221
TAGACATGGATGA
InaTs;omeAs;InaGs;omeAs;InaCs;o


106



AA
meAs;InaTs;omeGs;InaGs;omeAs;In


m01




aTs;omeGs;InaAs;omeAs;InaA-Sup





THRB-
human
THRB
222
GATGAAATTGCCCC
InaGs;omeAs;InaTs;omeGs;InaAs;o


107



T
meAs;InaAs;omeUs;InaTs;omeGs;In


m01




aCs;omeCs;InaCs;omeCs;InaT-Sup





THRB-
human
THRB
223
TGCCCCTTGAATGC
InaTs;omeGs;InaCs;omeCs;InaCs;o


108



G
meCs;InaTs;omeUs;InaGs;omeAs;In


m01




aAs;omeUs;InaGs;omeCs;InaG-Sup





THRB-
human
THRB
224
TGAATGCGGGTAC
InaTs;omeGs;InaAs;omeAs;InaTs;o


109



TT
meGs;InaCs;omeGs;InaGs;omeGs;In


m01




aTs;omeAs;InaCs;omeUs;InaT-Sup





THRB-
human
THRB
225
GTACTTGAAACTAT
InaGs;omeUs;InaAs;omeCs;InaTs;o


110



T
meUs;InaGs;omeAs;InaAs;omeAs;In


m01




aCs;omeUs;InaAs;omeUs;InaT-Sup





THRB-
human
THRB
226
ACTATTGCATTTCG
InaAs;omeCs;InaTs;omeAs;InaTs;om


111



T
eUs;InaGs;omeCs;InaAs;omeUs;InaT


m01




s;omeUs;InaCs;omeGs;InaT-Sup





THRB-
human
THRB
227
CGTTCTCCGGTCCT
InaCs;omeGs;InaTs;omeUs;InaCs;o


112



G
meUs;InaCs;omeCs;InaGs;omeGs;In


m01




aTs;omeCs;InaCs;omeUs;InaG-Sup





THRB-
human
THRB
228
CTGTGATGTGAATG
InaCs;omeUs;InaGs;omeUs;InaGs;o


113



C
meAs;InaTs;omeGs;InaTs;omeGs;In


m01




aAs;omeAs;InaTs;omeGs;InaC-Sup





THRB-
human
THRB
229
GTTCGAGGATTAG
InaGs;omeUs;InaTs;omeCs;InaGs;o


114



AC
meAs;InaGs;omeGs;InaAs;omeUs;In


m01




aTs;omeAs;InaGs;omeAs;InaC-Sup





THRB-
human
THRB
230
ATTAGACTGACTGG
InaAs;omeUs;InaTs;omeAs;InaGs;o


115



A
meAs;InaCs;omeUs;InaGs;omeAs;In


m01




aCs;omeUs;InaGs;omeGs;InaA-Sup





THRB-
human
THRB
231
ACTGGATTCATTCT
InaAs;omeCs;InaTs;omeGs;InaGs;o


116



C
meAs;InaTs;omeUs;InaCs;omeAs;In


m01




aTs;omeUs;InaCs;omeUs;InaC-Sup





THRB-
human
THRB
232
ATTCTCATAATTCCT
InaAs;omeUs;InaTs;omeCs;InaTs;o


117




meCs;InaAs;omeUs;InaAs;omeAs;In


m01




aTs;omeUs;InaCs;omeCs;InaT-Sup





THRB-
human
THRB
233
ATTCCTACAGCACT
InaAs;omeUs;InaTs;omeCs;InaCs;o


118



A
meUs;InaAs;omeCs;InaAs;omeGs;In


m01




aCs;omeAs;InaCs;omeUs;InaA-Sup





THRB-
human
THRB
234
TCATTTCATTCCATT
InaTs;omeCs;InaAs;omeUs;InaTs;o


119




meUs;InaCs;omeAs;InaTs;omeUs;In


m01




aCs;omeCs;InaAs;omeUs;InaT-Sup





THRB-
human
THRB
235
TCCATTGCCTAGCT
InaTs;omeCs;InaCs;omeAs;InaTs;om


120



C
eUs;InaGs;omeCs;InaCs;omeUs;Ina


m01




As;omeGs;InaCs;omeUs;InaC-Sup





THRB-
human
THRB
236
ACCAGGTCACCGG
InaAs;omeCs;InaCs;omeAs;InaGs;o


121



TT
meGs;InaTs;omeCs;InaAs;omeCs;In


m01




aCs;omeGs;InaGs;omeUs;InaT-Sup





THRB-
human
THRB
237
CGCAGTAGCTTCCT
InaCs;omeGs;InaCs;omeAs;InaGs;o


122



A
meUs;InaAs;omeGs;InaCs;omeUs;In


m01




aTs;omeCs;InaCs;omeUs;InaA-Sup





THRB-
human
THRB
238
CAAGGAGTTGACA
InaCs;omeAs;InaAs;omeGs;InaGs;o


123



TT
meAs;InaGs;omeUs;InaTs;omeGs;In


m01




aAs;omeCs;InaAs;omeUs;InaT-Sup





THRB-

THRB
239
ACATTTTGCAGGAC
InaAs;omeCs;InaAs;omeUs;InaTs;o


124
human


T
meUs;InaTs;omeGs;InaCs;omeAs;In


m01




aGs;omeGs;InaAs;omeCs;InaT-Sup





THRB-
human
THRB
240
CAAGGAAGGCGCA
InaCs;omeAs;InaAs;omeGs;InaGs;o


125



CA
meAs;InaAs;omeGs;InaGs;omeCs;In


m01




aGs;omeCs;InaAs;omeCs;InaA-Sup





THRB-
human
THRB
241
ATTAACTTTGCATG
InaAs;omeUs;InaTs;omeAs;InaAs;o


126



A
meCs;InaTs;omeUs;InaTs;omeGs;In


m01




aCs;omeAs;InaTs;omeGs;InaA-Sup





THRB-
human
THRB
242
TGAATAATGTGAGT
InaTs;omeGs;InaAs;omeAs;InaTs;o


127



G
meAs;InaAs;omeUs;InaGs;omeUs;In


m01




aGs;omeAs;InaGs;omeUs;InaG-Sup





THRB-
human
THRB
243
GTAATTTGGCTAGA
InaGs;omeUs;InaAs;omeAs;InaTs;o


128



G
meUs;InaTs;omeGs;InaGs;omeCs;In


m01




aTs;omeAs;InaGs;omeAs;InaG-Sup





THRB-
human
THRB
244
ACAGTTCCAACTGT
InaAs;omeCs;InaAs;omeGs;InaTs;o


129



C
meUs;InaCs;omeCs;InaAs;omeAs;In


m01




aCs;omeUs;InaGs;omeUs;InaC-Sup





THRB-
human
THRB
245
ATCACTCTGAACAT
InaAs;InaTs;InaCs;dAs;dCs;dTs;dCs;d


130



T
Ts;dGs;dAs;dAs;dCs;InaAs;InaTs;Ina


m08




T-Sup





THRB-
human
THRB
246
GAGCCTATATTCAT
InaGs;InaAs;InaGs;dCs;dCs;dTs;dAs;


131



A
dTs;dAs;dTs;dTs;dCs;InaAs;InaTs;Ina


m08




A-Sup





THRB-
human
THRB
247
ATGCATTTAGGTCT
InaAs;InaTs;InaGs;dCs;dAs;dTs;dTs;


132



A
dTs;dAs;dGs;dGs;dTs;InaCs;InaTs;In


m08




aA-Sup





THRB-
human
THRB
248
ATGCTGTGATAGA
InaAs;InaTs;InaGs;dCs;dTs;dGs;dTs;


133



GT
dGs;dAs;dTs;dAs;dGs;InaAs;InaGs;In


m08




aT-Sup





THRB-
human
THRB
249
CATATTAATGCATT
InaCs;InaAs;InaTs;dAs;dTs;dTs;dAs;d


134



T
As;dTs;dGs;dCs;dAs;InaTs;InaTs;InaT


m08




-Sup





THRB-
human
THRB
250
TCATCAGCCTGATT
InaTs;InaCs;InaAs;dTs;dCs;dAs;dGs;


135



A
dCs;dCs;dTs;dGs;dAs;InaTs;InaTs;Ina


m08




A-Sup





THRB-
human
THRB
251
TACGGAGTGGACA
InaTs;InaAs;InaCs;dGs;dGs;dAs;dGs;


136



GT
dTs;dGs;dGs;dAs;dCs;InaAs;InaGs;In


m08




aT-Sup





THRB-
human
THRB
252
CAATCGCAGCGGC
InaCs;InaAs;InaAs;dTs;dCs;dGs;dCs;


137



TC
dAs;dGs;dCs;dGs;dGs;InaCs;InaTs;In


m08




aC-Sup





THRB-
human
THRB
253
CAGCTGTTGACATG
InaCs;InaAs;InaGs;dCs;dTs;dGs;dTs;


138



T
dTs;dGs;dAs;dCs;dAs;InaTs;InaGs;In


m08




aT-Sup





THRB-
human
THRB
254
ATGGAGTTTGGCAT
InaAs;InaTs;InaGs;dGs;dAs;dGs;dTs;


139



C
dTs;dTs;dGs;dGs;dCs;InaAs;InaTs;In


m08




aC-Sup





THRB-
human
THRB
255
CATGATGAGGAAG
InaCs;InaAs;InaTs;dGs;dAs;dTs;dGs;


140



TT
dAs;dGs;dGs;dAs;dAs;InaGs;InaTs;In


m08




aT-Sup





THRB-
human
THRB
256
CTCTGTTCCTCAAA
InaCs;InaTs;InaCs;dTs;dGs;dTs;dTs;d


141



C
Cs;dCs;dTs;dCs;dAs;InaAs;InaAs;Ina


m08




C-Sup





THRB-
human
THRB
257
GAATATAGTGGGC
InaGs;InaAs;InaAs;dTs;dAs;dTs;dAs;


91



GT
dGs;dTs;dGs;dGs;dGs;InaCs;InaGs;In


m08




aT-Sup





THRB-
human
THRB
258
AGTGGGCGTAGAT
InaAs;InaGs;InaTs;dGs;dGs;dGs;dCs;


92



AA
dGs;dTs;dAs;dGs;dAs;InaTs;InaAs;In


m08




aA-Sup





THRB-
human
THRB
259
CGTAGATAAACTCA
InaCs;InaGs;InaTs;dAs;dGs;dAs;dTs;


93



T
dAs;dAs;dAs;dCs;dTs;InaCs;InaAs;In


m08




aT-Sup





THRB-
human
THRB
260
TAAACTCATAAGCT
InaTs;InaAs;InaAs;dAs;dCs;dTs;dCs;


94



T
dAs;dTs;dAs;dAs;dGs;InaCs;InaTs;In


m08




aT-Sup





THRB-
human
THRB
261
CATAAGCTTAAATT
InaCs;InaAs;InaTs;dAs;dAs;dGs;dCs;


95



C
dTs;dTs;dAs;dAs;dAs;InaTs;InaTs;Ina


m08




C-Sup





THRB-
human
THRB
262
AAGCTTATAACAGA
InaAs;InaAs;InaGs;dCs;dTs;dTs;dAs;


96



T
dTs;dAs;dAs;dCs;dAs;InaGs;InaAs;In


m08




aT-Sup





THRB-
human
THRB
263
ATAACAGATATATT
InaAs;InaTs;InaAs;dAs;dCs;dAs;dGs;


97



T
dAs;dTs;dAs;dTs;dAs;InaTs;InaTs;Ina


m08




T-Sup





THRB-
human
THRB
264
GATATATTTTCCTG
InaGs;InaAs;InaTs;dAs;dTs;dAs;dTs;


98



T
dTs;dTs;dTs;dCs;dCs;InaTs;InaGs;Ina


m08




T-Sup





THRB-
human
THRB
265
TTTCCTGTCTCTTTC
InaTs;InaTs;InaTs;dCs;dCs;dTs;dGs;d


99




Ts;dCs;dTs;dCs;dTs;InaTs;InaTs;InaC


m08




-Sup





THRB-
human
THRB
266
ATGGATTTTTACAT
InaAs;InaTs;InaGs;dGs;dAs;dTs;dTs;


100



A
dTs;dTs;dTs;dAs;dCs;InaAs;InaTs;Ina


m08




A-Sup





THRB-
human
THRB
267
TGTATGCAGATATA
InaTs;InaGs;InaTs;dAs;dTs;dGs;dCs;


101



A
dAs;dGs;dAs;dTs;dAs;InaTs;InaAs;In


m08




aA-Sup





THRB-
human
THRB
268
CTGTAATTATGAAT
InaCs;InaTs;InaGs;dTs;dAs;dAs;dTs;


102



A
dTs;dAs;dTs;dGs;dAs;InaAs;InaTs;In


m08




aA-Sup





THRB-
human
THRB
269
TACATAGGCAAAG
InaTs;InaAs;InaCs;dAs;dTs;dAs;dGs;


103



AG
dGs;dCs;dAs;dAs;dAs;InaGs;InaAs;In


m08




aG-Sup





THRB-
human
THRB
270
CAAAGAGTTGCCT
InaCs;InaAs;InaAs;dAs;dGs;dAs;dGs;


104



GC
dTs;dTs;dGs;dCs;dCs;InaTs;InaGs;In


m08




aC-Sup





THRB-
human
THRB
271
CCAGCCGCTTCCTG
InaCs;InaCs;InaAs;dGs;dCs;dCs;dGs;


105



C
dCs;dTs;dTs;dCs;dCs;InaTs;InaGs;Ina


m08




C-Sup





THRB-
human
THRB
272
TAGACATGGATGA
InaTs;InaAs;InaGs;dAs;dCs;dAs;dTs;


106



AA
dGs;dGs;dAs;dTs;dGs;InaAs;InaAs;In


m08




aA-Sup





THRB-
human
THRB
273
GATGAAATTGCCCC
InaGs;InaAs;InaTs;dGs;dAs;dAs;dAs;


107



T
dTs;dTs;dGs;dCs;dCs;InaCs;InaCs;Ina


m08




T-Sup





THRB-
human
THRB
274
TGCCCCTTGAATGC
InaTs;InaGs;InaCs;dCs;dCs;dCs;dTs;d


108



G
Ts;dGs;dAs;dAs;dTs;InaGs;InaCs;Ina


m08




G-Sup





THRB-
human
THRB
275
TGAATGCGGGTAC
InaTs;InaGs;InaAs;dAs;dTs;dGs;dCs;


109



TT
dGs;dGs;dGs;dTs;dAs;InaCs;InaTs;In


m08




aT-Sup





THRB-
human
THRB
276
GTACTTGAAACTAT
InaGs;InaTs;InaAs;dCs;dTs;dTs;dGs;


110



T
dAs;dAs;dAs;dCs;dTs;InaAs;InaTs;In


m08




aT-Sup





THRB-
human
THRB
277
ACTATTGCATTTCG
InaAs;InaCs;InaTs;dAs;dTs;dTs;dGs;


111



T
dCs;dAs;dTs;dTs;dTs;InaCs;InaGs;Ina


m08




T-Sup





THRB-
human
THRB
278
CGTTCTCCGGTCCT
InaCs;InaGs;InaTs;dTs;dCs;dTs;dCs;d


112



G
Cs;dGs;dGs;dTs;dCs;InaCs;InaTs;Ina


m08




G-Sup





THRB-
human
THRB
279
CTGTGATGTGAATG
InaCs;InaTs;InaGs;dTs;dGs;dAs;dTs;


113



C
dGs;dTs;dGs;dAs;dAs;InaTs;InaGs;In


m08




aC-Sup





THRB-
human
THRB
280
GTTCGAGGATTAG
InaGs;InaTs;InaTs;dCs;dGs;dAs;dGs;


114



AC
dGs;dAs;dTs;dTs;dAs;InaGs;InaAs;In


m08




aC-Sup





THRB-
human
THRB

ATTAGACTGACTGG
InaAs;InaTs;InaTs;dAs;dGs;dAs;dCs;


115


281
A
dTs;dGs;dAs;dCs;dTs;InaGs;InaGs;In


m08




aA-Sup





THRB-
human
THRB

ACTGGATTCATTCT
InaAs;InaCs;InaTs;dGs;dGs;dAs;dTs;


116


282
C
dTs;dCs;dAs;dTs;dTs;InaCs;InaTs;Ina


m08




C-Sup





THRB-
human
THRB
283
ATTCTCATAATTCCT
InaAs;InaTs;InaTs;dCs;dTs;dCs;dAs;d


117




Ts;dAs;dAs;dTs;dTs;InaCs;InaCs;InaT


m08




-Sup





THRB-
human
THRB
284
ATTCCTACAGCACT
InaAs;InaTs;InaTs;dCs;dCs;dTs;dAs;d


118



A
Cs;dAs;dGs;dCs;dAs;InaCs;InaTs;Ina


m08




A-Sup





THRB-
human
THRB
285
TCATTTCATTCCATT
InaTs;InaCs;InaAs;dTs;dTs;dTs;dCs;d


119




As;dTs;dTs;dCs;dCs;InaAs;InaTs;InaT


m08




-Sup





THRB-
human
THRB
286
TCCATTGCCTAGCT
InaTs;InaCs;InaCs;dAs;dTs;dTs;dGs;d


120



C
Cs;dCs;dTs;dAs;dGs;InaCs;InaTs;Ina


m08




C-Sup





THRB-
human
THRB
287
ACCAGGTCACCGG
InaAs;InaCs;InaCs;dAs;dGs;dGs;dTs;


121



TT
dCs;dAs;dCs;dCs;dGs;InaGs;InaTs;In


m08




aT-Sup





THRB-
human
THRB
288
CGCAGTAGCTTCCT
InaCs;InaGs;InaCs;dAs;dGs;dTs;dAs;


122



A
dGs;dCs;dTs;dTs;dCs;InaCs;InaTs;Ina


m08




A-Sup





THRB-
human
THRB
289
CAAGGAGTTGACA
InaCs;InaAs;InaAs;dGs;dGs;dAs;dGs;


123



TT
dTs;dTs;dGs;dAs;dCs;InaAs;InaTs;In


m08




aT-Sup





THRB-
human
THRB
290
ACATTTTGCAGGAC
InaAs;InaCs;InaAs;dTs;dTs;dTs;dTs;d


124



T
Gs;dCs;dAs;dGs;dGs;InaAs;InaCs;Ina


m08




T-Sup





THRB-
human
THRB
291
CAAGGAAGGCGCA
InaCs;InaAs;InaAs;dGs;dGs;dAs;dAs;


125



CA
dGs;dGs;dCs;dGs;dCs;InaAs;InaCs;In


m08




aA-Sup





THRB-
human
THRB
292
ATTAACTTTGCATG
InaAs;InaTs;InaTs;dAs;dAs;dCs;dTs;d


126



A
Ts;dTs;dGs;dCs;dAs;InaTs;InaGs;Ina


m08




A-Sup





THRB-
human
THRB
293
TGAATAATGTGAGT
InaTs;InaGs;InaAs;dAs;dTs;dAs;dAs;


127



G
dTs;dGs;dTs;dGs;dAs;InaGs;InaTs;In


m08




aG-Sup





THRB-
human
THRB
294
GTAATTTGGCTAGA
InaGs;InaTs;InaAs;dAs;dTs;dTs;dTs;d


128



G
Gs;dGs;dCs;dTs;dAs;InaGs;InaAs;Ina


m08




G-Sup





THRB-
human
THRB
295
ACAGTTCCAACTGT
InaAs;InaCs;InaAs;dGs;dTs;dTs;dCs;


129



C
dCs;dAs;dAs;dCs;dTs;InaGs;InaTs;In


m08




aC-Sup
















TABLE 4







A listing of oligonucleotide modifications.










Symbol
Feature Description






bio
5′ biotin



dAs
DNA w/3′ thiophosphate



dCs
DNA w/3′ thiophosphate



dGs
DNA w/3′ thiophosphate



dTs
DNA w/3′ thiophosphate



dG
DNA



enaAs
ENA w/3′ thiophosphate



enaCs
ENA w/3′ thiophosphate



enaGs
ENA w/3′ thiophosphate



enaTs
ENA w/3′ thiophosphate



fluAs
2′-fluoro w/3′ thiophosphate



fluCs
2′-fluoro w/3′ thiophosphate



fluGs
2′-fluoro w/3′ thiophosphate



fluUs
2′-fluoro w/3′ thiophosphate



lnaAs
LNA w/3′ thiophosphate



lnaCs
LNA w/3′ thiophosphate



lnaGs
LNA w/3′ thiophosphate



lnaTs
LNA w/3′ thiophosphate



omeAs
2′-OMe w/3′ thiophosphate



omeCs
2′-OMe w/3′ thiophosphate



omeGs
2′-OMe w/3′ thiophosphate



omeTs
2′-OMe w/3′ thiophosphate



lnaAs-Sup
LNA w/3′ thiophosphate at 3′ terminus



lnaCs-Sup
LNA w/3′ thiophosphate at 3′ terminus



lnaGs-Sup
LNA w/3′ thiophosphate at 3′ terminus



lnaTs-Sup
LNA w/3′ thiophosphate at 3′ terminus



lnaA-Sup
LNA w/3′ OH at 3′ terminus



lnaC-Sup
LNA w/3′ OH at 3′ terminus



lnaG-Sup
LNA w/3′ OH at 3′ terminus



lnaT-Sup
LNA w/3′ OH at 3′ terminus



omeA-Sup
2′-OMe w/3′ OH at 3′ terminus



omeC-Sup
2′-OMe w/3′ OH at 3′ terminus



omeG-Sup
2′-OMe w/3′ OH at 3′ terminus



omeU-Sup
2′-OMe w/3′ OH at 3′ terminus



dAs-Sup
DNA w/3′ thiophosphate at 3′ terminus



dCs-Sup
DNA w/3′ thiophosphate at 3′ terminus



dGs-Sup
DNA w/3′ thiophosphate at 3′ terminus



dTs-Sup
DNA w/3′ thiophosphate at 3′ terminus



dA-Sup
DNA w/3′ OH at 3′ terminus



dC-Sup
DNA w/3′ OH at 3′ terminus



dG-Sup
DNA w/3′ OH at 3′ terminus



dT-Sup
DNA w/3′ OH at 3′ terminus



Symbol
Feature Description



bio
5′ biotin



dAs
DNA w/3′ thiophosphate



dCs
DNA w/3′ thiophosphate



dGs
DNA w/3′ thiophosphate



dTs
DNA w/3′ thiophosphate



dG
DNA



enaAs
ENA w/3′ thiophosphate










The suffix “Sup” in Table 4 indicates that a 3′ end nucleotide may, for synthesis purposes, be conjugated to a solid support. It should be appreciated that in general when conjugated to a solid support for synthesis, the synthesized oligonucleotide is released such that the solid support is not part of the final oligonucleotide product.


Mouse APOA1 5′ and 3′ termini lancRNA targeting oligos were screened in primary mouse hepatocytes gymnotically at 20 uM, 8 uM and 3.2 uM concentrations in duplicates. APOA1 mRNA was measured and normalized relative to the water control well and B2M housekeeper. As shown in FIGS. 3A-3C, some of the oligos tested (such as oligos Apoa1_mus-27, 34, 35, 36, 37, 38, and 44) resulted in upregulation of APOA1 mRNA levels.


Next, mouse APOA1 5′ and 3′ termini lancRNA targeting oligos were screened in primary mouse hepatocytes gymnotically. APOA1 protein levels were measured in culture media at day5 at 8 uM oligo treatment condition. Abcam ab20453 was used as APOA1 antibody. Treatment with several oligos including Apoa1_mus-27, 35-39, and 41-45 resulted in increased APOA1 protein secretion (FIGS. 4A-4C). These results show that oligos targeting regions that encode APOA1 lancRNAs were useful for upregulation of APOA1 levels.


Oligos targeting FXN 3′ termini regions in antisense orientation were screened in Sarsero mouse-model derived skin fibroblasts via gymnosis and human FXN mRNA levels were measured. Oligos were screened at 10 uM concentration. Oligo and media changes were performed at day1, day4, day8. Data collection was done at day11. As shown in FIG. 5, some of the oligos tested (such as oligos FXN-607, 608, 609, 629, and 634) resulted in upregulation of FXN mRNA levels.


Oligos targeting FXN 3′ termini regions in antisense orientation were also screened in GM03816 cells via transfection and human FXN mRNA levels were measured. Oligos were screened at 20 nM and 50 nM concentration. Data collection was done at day4. As shown in FIG. 6, some of the oligos tested (such as oligo FXN-650) resulted in upregulation of FXN mRNA levels.


Oligos targeting FXN 5′ promoter associated regions in antisense orientation were also screened in GM03816 cells via transfection and human FXN mRNA levels were measured. Except for the oligos FXN-816 to 822, which were screened at three doses, the other oligos were screened at 5 doses. Measurements were taken at day3. As shown in FIG. 7, some of the oligos tested (such as oligos FXN-803, 823, 824, 819, and 822) resulted in upregulation of FXN mRNA levels.


Oligos targeting FXN 3′ termini regions in antisense orientation were also screened in Sarsero mouse-model derived skin fibroblasts via gymnosis for human FXN protein levels. Measurements were taken at day10. As shown in FIG. 8, all of the oligos tested (such as oligos FXN-603, 607, 609, 634, 643) resulted in upregulation of FXN mRNA levels.


Oligos targeting FXN 3′ termini regions in antisense orientation were also screened in GM03816 cells via transfection for human FXN mRNA levels. Oligos were screened at 30 nM concentration. Data collection was done at day4. As shown in FIG. 9, all of the oligos tested (such as oligos FXN-600, 603, 607, 609, 634, 643) resulted in upregulation of FXN mRNA levels.


Next, screens were performed in human normal terminally differentiated cardiomyocytes. 10 uM of oligos were gymnotically delivered to human normal cardiomyocytes. Oligo treatment and media changes were done at day1, day4 and day7. Measurements for RNA and protein were taken at day10. The FXN mRNA data was normalized to GAPDH. FXN-607 showed slight FXN RNA and protein upregulation (FIGS. 10A-C). FXN-695 is a FXN gapmer and therefore downregulated FXN levels.


Lastly, FXN oligos were tested in vivo. Oligos were injected at 100 mg/kg at day1, day2, day3 subcutaneously to 12-16 week old Sarsero mice. Sarsero mice are an animal model of Fredreich's Ataxia. The tissue collections were done at day5. The human FXN mRNA levels were measured in liver. The data normalization was done based on GAPDH and total RNA levels. Among others, oligos 607, 634 and 643 showed human FXN upregulation in livers of Sarsero mice (FIGS. 11A-D). These results show that oligos targeting regions that encode FXN lancRNAs were useful for upregulation of FXN levels.


Together, these data show that oligos targeting regions encoding lancRNAs can be used to upregulate gene expression.


The foregoing written specification is considered to be sufficient to enable one skilled in the art to practice the invention. The present invention is not to be limited in scope by examples provided, since the examples are intended as a single illustration of one aspect of the invention and other functionally equivalent embodiments are within the scope of the invention. Various modifications of the invention in addition to those shown and described herein will become apparent to those skilled in the art from the foregoing description and fall within the scope of the appended claims. The advantages and objects of the invention are not necessarily encompassed by each embodiment of the invention.

Claims
  • 1. A method of modulating expression of a target gene in cells, the method comprising: delivering to the cells a single-stranded oligonucleotide of 8 to 50 nucleotides in length that comprises a region of complementarity that is complementary with at least 5 contiguous nucleotides of a low-abundance non-coding RNA (lancRNA) that modulates expression of a target gene in the cells, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 5 kb of a transcriptional boundary of the target gene.
  • 2. The method of claim 1, wherein the lancRNA is represented at a level of less than 0.01 fragments per kilobase per million mapped reads (FPKM) based sequencing of RNA of the cells.
  • 3. The method of claim 1, wherein the lancRNA is represented at an average copy number of less than 10 transcripts per cell.
  • 4. The method of claim 3, wherein the lancRNA is represented at an average copy number of less than 0.1 transcripts per cell.
  • 5. The method of claim 4, wherein the lancRNA is represented at an average copy number of less than 0.0001 transcripts per cell.
  • 6. The method of claim 1, wherein the average copy number of the lancRNA is less than 1% of the average copy number of transcripts expressed from the target gene in the cells.
  • 7. The method of any preceding claim, wherein the lancRNA is transcribed from the same strand of the chromosomal region as the target gene.
  • 8. The method of any preceding claim, wherein the lancRNA is transcribed from the opposite strand of the chromosomal region as the target gene.
  • 9. The method of any preceding claim, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 5 kb of a transcriptional boundary of the target gene.
  • 10. The method of any preceding claim, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 2 kb of a transcriptional boundary of the target gene.
  • 11. The method of any preceding claim, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 1 kb of a transcriptional boundary of the target gene.
  • 12. The method of any preceding claim, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 500 bp of a transcriptional boundary of the target gene.
  • 13. The method of any preceding claim, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 250 bp of a transcriptional boundary of the target gene.
  • 14. The method of any preceding claim, wherein the transcriptional boundary is a transcriptional start site.
  • 15. The method of any preceding claim, wherein the transcriptional boundary is a transcriptional end site.
  • 16. The method of any preceding claim, wherein the lancRNA is no more than 200 nucleotides in length.
  • 17. The method of any preceding claim, wherein the target gene is ABCA1, APOA1, ATP2A2, BDNF, FXN, HBA2, HBB, HBD, HBE1, HBG1, HBG2, SMN, UTRN, PTEN, MECP2, FOXP3, NFE2L2 (NRF2), THRB, NR1H4 (FXR), HAMP, ADIPOQ, PRKAA1, PRKAA2, PRKAB1, PRKAB2, PRKAG1, PRKAG2, or PRKAG3.
  • 18. The method of claim 17, wherein the target gene is FXN.
  • 19. The method of any preceding claim, wherein the oligonucleotide does not comprise three or more consecutive guanosine nucleotides.
  • 20. The method of any preceding claim, wherein the oligonucleotide does not comprise four or more consecutive guanosine nucleotides.
  • 21. The method of any preceding claim, wherein the oligonucleotide is 8 to 30 nucleotides in length.
  • 22. The method of any preceding claim, wherein the oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the lancRNA are cytosine or guanosine nucleotides.
  • 23. The method of any preceding claim, wherein at least one nucleotide of the oligonucleotide is a nucleotide analogue.
  • 24. The method of any preceding claim, wherein the at least one nucleotide analogue results in an increase in Tm of the oligonucleotide in a range of 1 to 5° C. compared with an oligonucleotide that does not have the at least one nucleotide analogue.
  • 25. The method of any preceding claim, wherein at least one nucleotide of the oligonucleotide comprises a 2′ O-methyl.
  • 26. The method of any preceding claim, wherein each nucleotide of the oligonucleotide comprises a 2′ O-methyl.
  • 27. The method of any one of claims 1 to 26, wherein the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide.
  • 28. The method of claim 27, wherein the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide.
  • 29. The method of any one of claims 1 to 26, wherein each nucleotide of the oligonucleotide is a LNA nucleotide.
  • 30. The method of any one of claims 1 to 26, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-fluoro-deoxyribonucleotides.
  • 31. The method of any one of claims 1 to 26, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-O-methyl nucleotides.
  • 32. The method of any one of claims 1 to 26, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and ENA nucleotide analogues.
  • 33. The method of any one of claims 1 to 26, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and LNA nucleotides.
  • 34. The single stranded oligonucleotide of any one of claims 31 to 33, wherein the 5′ nucleotide of the oligonucleotide is a deoxyribonucleotide.
  • 35. The method of any one of claims 1 to 26, wherein the nucleotides of the oligonucleotide comprise alternating LNA nucleotides and 2′-O-methyl nucleotides.
  • 36. The single stranded oligonucleotide of claim 35, wherein the 5′ nucleotide of the oligonucleotide is a LNA nucleotide.
  • 37. The method of any one of claims 1 to 26, wherein the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one LNA nucleotide on each of the 5′ and 3′ ends of the deoxyribonucleotides.
  • 38. The method of any preceding claim, further comprising phosphorothioate internucleotide linkages between at least two nucleotides.
  • 39. The method of claim 38, further comprising phosphorothioate internucleotide linkages between all nucleotides.
  • 40. The method of any preceding claim, wherein the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group.
  • 41. The method of any preceding claim, wherein the nucleotide at the 3′ position of the oligonucleotide has a 3′ thiophosphate.
  • 42. The method of any preceding claim, further comprising a biotin moiety conjugated to the 5′ nucleotide.
  • 43. The method of any preceding claim, wherein the single stranded oligonucleotide comprises a nucleotide sequence as set for in Table 3.
  • 44. A single stranded oligonucleotide having a nucleotide sequence as set forth in Table 3.
  • 45. The single stranded oligonucleotide of claim 44, wherein at least one nucleotide of the oligonucleotide comprises a 2′ O-methyl.
  • 46. The single stranded oligonucleotide of claim 44 or 45, wherein each nucleotide of the oligonucleotide comprises a 2′ O-methyl.
  • 47. The single stranded oligonucleotide of any one of claims 44 to 45, wherein the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide.
  • 48. The single strand oligonucleotide of claim 47, wherein the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide.
  • 49. The single stranded oligonucleotide of any one of claims 44 to 46, wherein each nucleotide of the oligonucleotide is a LNA nucleotide.
  • 50. The single stranded oligonucleotide of any one of claims 44 to 46, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-fluoro-deoxyribonucleotides.
  • 51. The single stranded oligonucleotide of any one of claims 44 to 46, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and 2′-O-methyl nucleotides.
  • 52. The single stranded oligonucleotide of any one of claims 44 to 46, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and ENA nucleotide analogues.
  • 53. The single stranded oligonucleotide of any one of claims 44 to 46, wherein the nucleotides of the oligonucleotide comprise alternating deoxyribonucleotides and LNA nucleotides.
  • 54. The single stranded oligonucleotide of any one of claims 51 to 53, wherein the 5′ nucleotide of the oligonucleotide is a deoxyribonucleotide.
  • 55. The single stranded oligonucleotide of any one of claims 44 to 46, wherein the nucleotides of the oligonucleotide comprise alternating LNA nucleotides and 2′-O-methyl nucleotides.
  • 56. The single stranded oligonucleotide of claim 55, wherein the 5′ nucleotide of the oligonucleotide is a LNA nucleotide.
  • 57. The single stranded oligonucleotide of any one of claims 44 to 46, wherein the nucleotides of the oligonucleotide comprise deoxyribonucleotides flanked by at least one LNA nucleotide on each of the 5′ and 3′ ends of the deoxyribonucleotides.
  • 58. The single stranded oligonucleotide of any one of claims 44 to 57, further comprising phosphorothioate internucleotide linkages between at least two nucleotides.
  • 59. The single stranded oligonucleotide of claim 58, further comprising phosphorothioate internucleotide linkages between all nucleotides.
  • 60. The single stranded oligonucleotide of any one of claims 44 to 59, wherein the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group.
  • 61. The single stranded oligonucleotide of any one of claims 44 to 60, wherein the nucleotide at the 3′ position of the oligonucleotide has a 3′ thiophosphate.
  • 62. The single stranded oligonucleotide of any one of claims 44 to 61, further comprising a biotin moiety conjugated to the 5′ nucleotide.
  • 63. The single stranded oligonucleotide of any one of claims 44 to 62, wherein the modification pattern for the oligonucleotide is the modification pattern provided in Table 3.
  • 64. A composition comprising a single stranded oligonucleotide of any one of claims 44 to 63 and a carrier.
  • 65. A composition comprising a single stranded oligonucleotide of any one of claims 44 to 63 in a buffered solution.
  • 66. A composition of claim 65, wherein the oligonucleotide is conjugated to the carrier.
  • 67. The composition of claim 66, wherein the carrier is a peptide.
  • 68. The composition of claim 66, wherein the carrier is a steroid.
  • 69. A pharmaceutical composition comprising a composition of any one of claims 61 to 65 and a pharmaceutically acceptable carrier.
  • 70. A kit comprising a container housing the composition of any one of claims 64 to 69.
  • 71. A method of modulating expression of a target gene in cells, the method comprising: i) determining presence of a low-abundance non-coding RNA (lancRNA) in cells; andii) based on the determination made in i), delivering to the cells a single-stranded oligonucleotide of 8 to 50 nucleotides in length that comprises a region of complementarity that is complementary with at least 5 contiguous nucleotides of a lancRNA that modulates expression of a target gene in the cells, wherein the at least 5 contiguous nucleotides of the lancRNA are transcribed from a chromosomal region within 5 kb of a transcriptional boundary of the target gene.
  • 72. The method of claim 71, wherein in step i) the lancRNA is determined to be present at a level of less than 0.01 fragments per kilobase per million mapped reads (FPKM) based sequencing of RNA of the cells.
  • 73. The method of claim 71, wherein in step i) the lancRNA is determined to be present at an average copy number of less than 10 transcripts per cell.
  • 74. The method of claim 73, wherein in step i) the lancRNA is determined to be present at an average copy number of less than 0.1 transcripts per cell.
  • 75. The method of claim 74, wherein in step i) the lancRNA is determined to be present at an average copy number of less than 0.0001 transcripts per cell.
  • 76. The method of claim 71, wherein in step i) the lancRNA is determined to be present at less than 1% of the average copy number of transcripts expressed from the target gene in the cells.
  • 77. A method of modulating expression of a target gene in cells, the method comprising: delivering to the cells a single-stranded oligonucleotide of 8 to 50 nucleotides in length that comprises a region of complementarity that is complementary with at least 5 contiguous nucleotides of a chromosomal region that corresponds to a 3′ UTR of the target gene, wherein the at least 5 contiguous nucleotides are antisense to the target gene.
  • 78. The method of claim 77, wherein the target gene is ABCA1, APOA1, ATP2A2, BDNF, FXN, HBA2, HBB, HBD, HBE1, HBG1, HBG2, SMN, UTRN, PTEN, MECP2, FOXP3, NFE2L2 (NRF2), THRB, NR1H4 (FXR), HAMP, ADIPOQ, PRKAA1, PRKAA2, PRKAB1, PRKAB2, PRKAG1, PRKAG2, or PRKAG3.
  • 79. The method of claim 78, wherein the target gene is FXN.
  • 80. The method of any one of claims 77-79, wherein the oligonucleotide does not comprise three or more consecutive guanosine nucleotides.
  • 81. The method of any one of claims 77-81, wherein the oligonucleotide does not comprise four or more consecutive guanosine nucleotides.
  • 82. The method of any one of claims 77-81, wherein the oligonucleotide is 8 to 30 nucleotides in length.
  • 83. The method of any one of claims 77-82, wherein the oligonucleotide is 8 to 10 nucleotides in length and all but 1, 2, or 3 of the nucleotides of the complementary sequence of the lancRNA are cytosine or guanosine nucleotides.
  • 84. The method of any one of claims 77-83, wherein at least one nucleotide of the oligonucleotide is a nucleotide analogue.
  • 85. The method of any one of claims 77-84, wherein the at least one nucleotide analogue results in an increase in Tm of the oligonucleotide in a range of 1 to 5° C. compared with an oligonucleotide that does not have the at least one nucleotide analogue.
  • 86. The method of any one of claims 77-85, wherein at least one nucleotide of the oligonucleotide comprises a 2′ O-methyl.
  • 87. The method of any one of claims 77-86, wherein the oligonucleotide comprises at least one ribonucleotide, at least one deoxyribonucleotide, or at least one bridged nucleotide.
  • 88. The method of claim 87, wherein the bridged nucleotide is a LNA nucleotide, a cEt nucleotide or a ENA modified nucleotide.
  • 89. The method of any one of claims 77-88, further comprising phosphorothioate internucleotide linkages between at least two nucleotides.
  • 90. The single stranded oligonucleotide of claim 89, further comprising phosphorothioate internucleotide linkages between all nucleotides.
  • 91. The method of any one of claims 77-90, wherein the nucleotide at the 3′ position of the oligonucleotide has a 3′ hydroxyl group.
  • 92. The method of any one of claims 77-90, wherein the nucleotide at the 3′ position of the oligonucleotide has a 3′ thiophosphate.
CROSS-REFERENCE TO RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C. §119(e) of U.S. Provisional Application No. U.S. 62/115,739, entitled “TARGETING OLIGONUCLEOTIDES AND USES THEREOF TO MODULATE GENE EXPRESSION”, filed on Feb. 13, 2015, the contents of which are incorporated herein by reference in its entirety.

PCT Information
Filing Document Filing Date Country Kind
PCT/US16/17773 2/12/2016 WO 00
Provisional Applications (1)
Number Date Country
62115739 Feb 2015 US