COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF THE ALAS1 GENE (2024)

This application is a continuation of U.S. patent application Ser. No. 16/893,253, filed Jun. 4, 2020, which is a divisional of U.S. application Ser. No. 16/142,953, filed Sep. 26, 2018, now abandoned, which is a divisional of U.S. application Ser. No. 14/814,911, filed Jul. 31, 2015, now U.S. Pat. No. 10,125,364, issued Nov. 13, 2018, which is a divisional of U.S. application Ser. No. 13/835,613, filed Mar. 15, 2013, now U.S. Pat. No. 9,133,461, issued Sep. 15, 2015, which claims priority to U.S. Provisional Application No. 61/622,288, filed Apr. 10, 2012. The entire contents of each of the foregoing applications are hereby incorporated herein.

The instant application contains a Sequence Listing which has been submitted electronically in XML format and is hereby incorporated by reference in its entirety. Said XML copy, created on Nov. 6, 2023, is named 121301-77901.xml and is 7,561,921 bytes in size. The sequence listing is part of the specification and is incorporated in its entirety by reference herein.

The invention relates to the specific inhibition of the expression of the ALAS1 gene.

The inherited porphyrias are a family of disorders resulting from the deficient activity of specific enzymes in the heme biosynthetic pathway, also referred to herein as the porphyrin pathway. Deficiency in the enzymes of the porphyrin pathway leads to insufficient heme production and to an accumulation of porphyrins, which are toxic to tissue in high concentrations.

Of the inherited porphyrias, acute intermittent porphyria (AIP, e.g., autosomal dominant AIP), variegate porphyria (VP, e.g., autosomal dominant VP), hereditary coproporphyria (copropophyria or HCP, e.g., autosomal dominant HCP), and 5′ aminolevulinic acid (also known as δ-aminolevulinic acid or ALA) dehydratase deficiency porphyria (ADP, e.g., autosomal recessive ADP) are classified as acute hepatic porphyrias and are manifested by acute neurological attacks that can be life threatening. The acute attacks are characterized by autonomic, peripheral, and central nervous symptoms, including severe abdominal pain, hypertension, tachycardias, constipation, motor weakness, paralysis, and seizures. If not treated properly, quadriplegia, respiratory impairment, and death may ensue. Various factors, including cytrochrome P450-inducing drugs, dieting, and hormonoal changes can precipitate acute attacks by increasing the activity of hepatic 5′-aminolevulinic acid synthase 1 (ALAS1), the first and rate-limiting enzyme of the heme biosynthetic pathway. In the acute porphyrias, e.g., AIP, VP, HCP and ADP, the respective enzyme deficiencies result in hepatic production and accumulation of one or more substances (e.g., porphyrins and/or porphyrin precursors, e.g., ALA and/or PBG) that can be neurotoxic and can result in the occurrence of acute attacks. See, e.g., Balwani, M and Desnick, R. J., Blood, 120:4496-4504, 2012.

The current therapy for the actute neuroloigcal attacks in the intravenous administration of hemin (Panhematin®, Lundbeck or Normosang®, Orphan Europe), which provides exogenous heme for the negative feedback inhibition of ALAS1, and thereby, decreases production of ALA and PBG. Hemin is used for the treatment during an acute attack and for prevention of attacks, particularly in women with the acute porphyrias who experience frequent attacks with the hormonal changes during their menstrual cycles. While patients generally respond well, its effect is slow, typically taking two to four days or longer to normalize urinary ALA and PBG concentrations towards normal levels. As the intravenous hemin is rapidly metabolized, three to four infusions are usually necessary to effectively treat or prevent an acute attack. In addition, repeated infusions may cause iron overload and phlebitis, which may compromise peripheral venous access. Although orthotrophic liver transplantation is curative, this procedure has significant morbidity and mortality and the availability of liver donors is limited. Therefore, an alternative therapeutic approach that is more effective, fast-acting, and safe is needed. It would be particularly advantageous if such treatment could be delivered by subcutaneous administration, as this would preclude the need for infusions and prolonged hospitalization.

AIP, also referred to as porphobilinogen deaminase deficiency (PBGD), or hydroxymethylbilane synthase (HMBS) deficiency, is the most common of the acute hepatic prophyrias. It is an autosomal dominant disorder caused by mutations in the HMB-synthase (HMBS) gene that result in reduced, e.g., half-normal activity of the enzyme. Previously, a mouse model of AIP that has ˜30% of wildtype HMBS activity was generated by hom*ologous recombination Like human patients, these mice increase hepatic ALAS1 activity and accumulate large quantities of plasma and urinary ALA and PBG when administered porphyrinogenic drugs, such as phenobarbital. Thus, they serve as an excellent model to evaluate the efficacy of novel therapeutics for the acute hepatic porphyrias.

The present invention describes methods and iRNA compositions for modulating the expression of an ALAS1 gene. In certain embodiments, expression of an ALAS1 gene is reduced or inhibited using an ALAS1-specific iRNA. Such inhibition can be useful in treating disorders related to ALAS1 expression, such as porphyrias.

Accordingly, described herein are compositions and methods that effect the RNA-induced silencing complex (RISC)-mediated cleavage of RNA transcripts of the ALAS1 gene, such as in a cell or in a subject (e.g., in a mammal, such as a human subject). Also described are compositions and methods for treating a disorder related to expression of an ALAS1 gene, such as a porphyria, e.g., X-linked sideroblastic anemia (XLSA), ALA deyhdratase deficiency porphyria (Doss porphyria or ADP), acute intermittent porphyria (AIP), congenital erythropoietic porphyria (CEP), Porphyria cutanea tarda (PCT), hereditary coproporphyria (coproporphyria, or HCP), variegate porphyria (VP), erythropoietic protoporphyria (EPP), or transient erythroporphyria of infancy. In some embodiments, the disorder is an acute hepatic porphyria, e.g., ALA deyhdratase deficiency porphyria (ADP), AIP, HCP, or VP. In certain embodiments, the disorder is ALA deyhdratase deficiency porphyria (ADP) or AIP.

In embodiments, the porphyria is a hepatic porphyria, e.g., a porphyria selected from acute intermittent porphyria (AIP) hereditary coproporphyria (HCP), variegate porphyria (VP), ALA deyhdratase deficiency porphyria (ADP), and hepatoerythropoietic porphyria. In embodiments, the porphyria is a hom*ozygous dominant hepatic porphyria (e.g., hom*ozygous dominant AIP, HCP, or VP) or hepatoerythropoietic porphyria, In embodiments, the porphyria is a dual porphyria.

As used herein, the term “iRNA,” “RNAi”, “iRNA agent,” or “RNAi agent” refers to an agent that contains RNA as that term is defined herein, and which mediates the targeted cleavage of an RNA transcript, e.g., via an RNA-induced silencing complex (RISC) pathway. In one embodiment, an iRNA as described herein effects inhibition of ALAS1 expression in a cell or mammal.

The iRNAs included in the compositions featured herein encompass a dsRNA having an RNA strand (the antisense strand) having a region, e.g., a region that is 30 nucleotides or less, generally 19-24 nucleotides in length, that is substantially complementary to at least part of an mRNA transcript of an ALAS1 gene (e.g., a mouse or human ALAS1 gene) (also referred to herein as an “ALAS1-specific iRNA”). Alternatively, or in combination, iRNAs encompass a dsRNA having an RNA strand (the antisense strand) having a region that is 30 nucleotides or less, generally 19-24 nucleotides in length, that is substantially complementary to at least part of an mRNA transcript of an ALAS1 gene (e.g., a human variant 1 or 2 of an ALAS1 gene) (also referred to herein as a “ALAS1-specific iRNA”).

In embodiments, the iRNA (e.g., dsRNA) described herein comprises an antisense strand having a region that is substantially complementary to a region of a human ALAS1. In embodiments, the human ALAS1 has the sequence of NM_000688.4 (SEQ ID NO:1) or NM_000688.5 (SEQ ID NO:382).

In other embodiments, an iRNA encompasses a dsRNA having an RNA strand (the antisense strand) having a region that is substantially complementary to a portion of an ALAS1 mRNA according to any one of Tables 2, 3, 6, 7, 8, 9, 14, or 15. In one embodiment, the iRNA encompasses a dsRNA having an RNA strand (the antisense strand) having a region that is substantially complementary to a portion of an ALAS1 mRNA, e.g., a human ALAS1 mRNA (e.g., a human ALAS1 mRNA as provided in SEQ ID NO:1 or SEQ ID NO:382).

In one embodiment, an iRNA for inhibiting expression of an ALAS1 gene includes at least two sequences that are complementary to each other. The iRNA includes a sense strand having a first sequence and an antisense strand having a second sequence. The antisense strand includes a nucleotide sequence that is substantially complementary to at least part of an mRNA encoding an ALAS1 transcript, and the region of complementarity is 30 nucleotides or less, and at least 15 nucleotides in length. Generally, the iRNA is 19 to 24 nucleotides in length.

In some embodiments, the iRNA is 19-21 nucleotides in length. In some embodiments, the iRNA is 19-21 nucleotides in length and is in a lipid formulation, e.g. a lipid nanoparticle (LNP) formulation (e.g., an LNP11 formulation).

In some embodiments, the iRNA is 21-23 nucleotides in length. In some embodiments, the iRNA is 21-23 nucleotides in length and is in the form of a conjugate, e.g., conjugated to one or more GalNAc derivatives as described herein.

In some embodiments the iRNA is from about 15 to about 25 nucleotides in length, and in other embodiments the iRNA is from about 25 to about 30 nucleotides in length. An iRNA targeting ALAS1, upon contact with a cell expressing ALAS1, inhibits the expression of an ALAS1 gene by at least 10%, at least 20%, at least 25%, at least 30%, at least 35% or at least 40% or more, such as when assayed by a method as described herein. In one embodiment, the iRNA targeting ALAS1 is formulated in a stable nucleic acid lipid particle (SNALP).

In one embodiment, an iRNA (e.g., a dsRNA) featured herein includes a first sequence of a dsRNA that is selected from the group consisting of the sense sequences of Tables 2, 3, 6, 7, 8, 9, 14, and 15 and a second sequence that is selected from the group consisting of the corresponding antisense sequences of Tables 2, 3, 6, 7, 8, 9, 14 and 15.

In one embodiment, an iRNA (e.g., a dsRNA) featured herein has sense and/or antisense sequences selected from those of AD-58882, AD-58878, AD-58886, AD-58877, AD-59115, AD-58856, AD-59129, AD-59124, AD-58874, AD-59125, AD-59105, AD-59120, AD-59122, AD-59106, AD-59126, and AD-59107 as disclosed herein in the Examples. In embodiments, the iRNA (e.g., dsRNA) has sense and/or antisense sequences selected from those of AD-58882, AD-58878, AD-58886, AD-58877, AD-59115, AD-58856, and AD-59129.

The iRNA molecules featured herein can include naturally occurring nucleotides or can include at least one modified nucleotide, including, but not limited to a 2′-O-methyl modified nucleotide, a nucleotide having a 5′-phosphorothioate group, and a terminal nucleotide linked to a cholesteryl derivative. Alternatively, the modified nucleotide may be chosen from the group of: a 2′-deoxy-2′-fluoro modified nucleotide, a 2′-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, 2′-amino-modified nucleotide, 2′-alkyl-modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base comprising nucleotide. Such a modified sequence can be based, e.g., on a first sequence of said iRNA selected from the group consisting of the sense sequences of Table 2, and a second sequence selected from the group consisting of the corresponding antisense sequences of Table 2.

In one embodiment, an iRNA (e.g., a dsRNA) featured herein comprises a sense strand comprising a sequence selected from the group consisting of SEQ ID NO:330, SEQ ID NO:334, SEQ ID NO:342, SEQ ID NO:344, SEQ ID NO:346, SEQ ID NO:356, SEQ ID NO:358, SEQ ID NO:362, SEQ ID NO:366, SEQ ID NO:376, and SEQ ID NO:380.

In one embodiment, an iRNA (e.g., a dsRNA) featured herein comprises an antisense strand comprising a sequence selected from the group consisting of SEQ ID NO:331, SEQ ID NO:335, SEQ ID NO:343, SEQ ID NO:345, SEQ ID NO:347, SEQ ID NO:357, SEQ ID NO:359, SEQ ID NO:363, SEQ ID NO:367, SEQ ID NO:377, and SEQ ID NO:381.

In one embodiment, an iRNA (e.g., a dsRNA) featured herein comprises a sense strand comprising a sequence selected from the group consisting of SEQ ID NO:140, SEQ ID NO:144, SEQ ID NO:152, SEQ ID NO:154, SEQ ID NO: 156, SEQ ID NO:166, SEQ ID NO:168, SEQ ID NO:172, SEQ ID NO:176, SEQ ID NO:186, and SEQ ID NO:190. In one embodiment, an iRNA (e.g., a dsRNA) featured herein comprises an antisense strand comprising a sequence selected from the group consisting of SEQ ID NO:141, SEQ ID NO:145, SEQ ID NO: 153, SEQ ID NO:155, SEQ ID NO:157, SEQ ID NO:167, SEQ ID NO:169, SEQ ID NO:173, SEQ ID NO:177, SEQ ID NO:187, and SEQ ID NO:191.

In one embodiment, an iRNA as described herein targets a wildtype ALAS1 RNA transcript variant, and in another embodiment, the iRNA targets a mutant transcript (e.g., an ALAS1 RNA carrying an allelic variant). For example, an iRNA featured in the invention can target a polymorphic variant, such as a single nucleotide polymorphism (SNP), of ALAS1. In another embodiment, the iRNA targets both a wildtype and a mutant ALAS1 transcript. In yet another embodiment, the iRNA targets a particular transcript variant of ALAS1 (e.g., human ALAS1 variant 1). In yet another embodiment, the iRNA agent targets multiple transcript variants (e.g., both variant 1 and variant 2 of human ALAS1).

In one embodiment, an iRNA featured in the invention targets a non-coding region of an ALAS1 RNA transcript, such as the 5′ or 3′ untranslated region of a transcript.

In some embodiments, an iRNA as described herein is in the form of a conjugate, e.g., a carbohydrate conjugate, which may serve as a targeting moiety and/or ligand, as described herein. In one embodiment, the conjugate is attached to the 3′ end of the sense strand of the dsRNA. In some embodiments, the conjugate is attached via a linker, e.g., via a bivalent or trivalent branched linker.

In some embodiments, the conjugate comprises one or more N-acetylgalactosamine (GalNAc) derivatives. Such a conjugate is also referred to herein as a GalNAc conjugate. In some embodiments, the conjugate targets the RNAi agent to a particular cell, e.g., a liver cell, e.g., a hepatocyte. The GalNAc derivatives can be attached via a linker, e.g., a bivalent or trivalent branched linker. In particular embodiments, the conjugate is

In some embodiments, the RNAi agent is attached to the carbohydrate conjugate via a linker, e.g., a linker as shown in the following schematic, wherein X is O or S

In some embodiments, X is O. In some embodiments, X is S.

In some embodiments, the RNAi agent is conjugated to L96 as defined in Table 1 and shown below

In an aspect provided herein is a pharmaceutical composition for inhibiting the expression of an ALAS1 gene in an organism, generally a human subject. The composition typically includes one or more of the iRNAs described herein and a pharmaceutically acceptable carrier or delivery vehicle. In one embodiment, the composition is used for treating a porphyria, e.g., AIP.

In one aspect, an iRNA provided herein is a double-stranded ribonucleic acid (dsRNA) for inhibiting expression of ALAS1, wherein said dsRNA comprises a sense strand and an antisense strand 15-30 base pairs in length and the antisense strand is complementary to at least 15 contiguous nucleotides of SEQ ID NO: 1 or 382.

In a further aspect, an iRNA provided herein is a double stranded RNAi (dsRNA) comprising a sense strand complementary to an antisense strand, wherein said antisense strand comprises a region of complementarity to an ALAS1 RNA transcript, wherein each strand has about 14 to about 30 nucleotides, wherein said double stranded RNAi agent is represented by formula (III):

    • sense: 5′np-Na-(XXX)i-Nb-YYY-Nb -(ZZZ)j -Na - nq 3′
    • antisense: 3′np′-Na′-(X′X′X′)k-Nb′-Y′Y′Y′-Nb′-(Z′Z′Z′)I-Na′- nq′5′
    • (III)
    • wherein:
    • i, j, k, and 1 are each independently 0 or 1;
    • p, p′, q, and q′ are each independently 0-6;
    • each Na and Na′ independently represents an oligonucleotide sequence comprising 0-25 nucleotides which are either modified or unmodified or combinations thereof, each sequence comprising at least two differently modified nucleotides;
    • each Nb and Nb′ independently represents an oligonucleotide sequence comprising 0-10 nucleotides which are either modified or unmodified or combinations thereof;
    • each np, np′, nq, and nq′ independently represents an overhang nucleotide;
    • XXX, YYY, ZZZ, X′X′X′, Y′Y′Y′, and Z′Z′Z′ each independently represent one motif of three identical modifications on three consecutive nucleotides;
    • modifications on Nb differ from the modification on Y and modifications on Nb′ differ from the modification on Y′.

In embodiments, the sense strand is conjugated to at least one ligand.

In embodiments, i is 1; j is 1; or both i and j are 1.

In embodiments, k is 1; 1 is 1; or both k and 1 are 1.

In embodiments, XXX is complementary to X′X′X′, YYY is complementary to Y′Y′Y′, and ZZZ is complementary to Z′Z′Z′.

In embodiments, the Y′Y′Y′ motif occurs at the 11, 12 and 13 positions of the antisense strand from the 5′-end.

In embodiments, the Y′ is 2′-O-methyl.

In embodiments, the duplex region is 15-30 nucleotide pairs in length.

In embodiments, the duplex region is 17-23 nucleotide pairs in length.

In embodiments, the duplex region is 19-21 nucleotide pairs in length.

In embodiments, the duplex region is 21-23 nucleotide pairs in length.

In embodiments, the modifications on the nucleotides are selected from the group consisting of LNA, HNA, CeNA, 2′-methoxyethyl, 2′-O-alkyl, 2′-O-allyl, 2′-C— allyl, 2′-fluoro, 2′-deoxy, 2′-hydroxyl, and combinations thereof.

In embodiments, the modifications on the nucleotides are 2′-O-methyl, 2′-fluoro or both.

In embodiments, the ligand comprises a carbohydrate.

In embodiments, the ligand is attached via a linker.

In embodiments, the linker is a bivalent or trivalent branched linker.

In embodiments, the ligand is

In embodiments, the ligand and linker are as shown in Formula XXIV:

In embodiments, the ligand is attached to the 3′ end of the sense strand.

In embodiments, the dsRNA has a nucleotide sequence selected from the group of sequences provided in Tables 2 and 3. In embodiments, the dsRNA has a nucleotide sequence selected from the group of sequences provided in Tables 2, 3, 6, 7, 8 and 9. In embodiments, the dsRNA has a nucleotide sequence selected from the group of sequences provided in Tables 2, 3, 6, 7, 8, 9, 14, and 15. In embodiments, the dsRNA has a nucleotide sequence selected from the group of sequences provided in Tables 14 and 15.

In embodiments, dsRNA has a nucleotide sequence selected from the group of sequences provided in Tables 3 and 8.

In a further aspect, an iRNA provided herein is a double-stranded ribonucleic acid (dsRNA) for inhibiting expression of ALAS1, wherein said dsRNA comprises a sense strand and an antisense strand, the antisense strand comprising a region of complementarity to an ALAS1 RNA transcript, which antisense strand comprises at least 15 contiguous nucleotides differing by no more than 3 nucleotides from one of the antisense sequences listed in any one of Tables 2, 3, 6, 7, 8, 9, 14, or 15. In some such embodiments, the sense and antisense sequences are selected from those of the duplexes AD-58882, AD-58878, AD-58886, AD-58877, AD-59115, AD-58856, AD-59129, AD-59124, AD-58874, AD-59125, AD-59105, AD-59120, AD-59122, AD-59106, AD-59126, and AD-59107 as disclosed herein in the Examples. In embodiments, the sense and antisense sequences are selected from those of the duplexes AD-58882, AD-58878, AD-58886, AD-58877, AD-59115, AD-58856, and AD-59129.

In some embodiments, the dsRNA comprises at least one modified nucleotide.

In some embodiments, at least one of the modified nucleotides is chosen from the group consisting of: a 2′-O-methyl modified nucleotide, a nucleotide comprising a 5′-phosphorothioate group, and a terminal nucleotide linked to a cholesteryl derivative or dodecanoic acid bisdecylamide group.

In some embodiments, the modified nucleotide is chosen from the group consisting of: a 2′-deoxy-2′-fluoro modified nucleotide, a 2′-deoxy-modified nucleotide, a locked nucleotide, an abasic nucleotide, 2′-amino-modified nucleotide, 2′-alkyl-modified nucleotide, morpholino nucleotide, a phosphoramidate, and a non-natural base comprising nucleotide.

In some embodiments, the region of complementarity is at least 17 nucleotides in length.

In some embodiments, the region of complementarity is between 19 and 21 nucleotides in length.

In some embodiments, the region of complementarity is 19 nucleotides in length.

In some embodiments, each strand is no more than 30 nucleotides in length.

In some embodiments, at least one strand comprises a 3′ overhang of at least 1 nucleotide.

In some embodiments, at least one strand comprises a 3′ overhang of at least 2 nucleotides.

In some embodiments, a dsRNA described herein further comprises a ligand.

In some embodiments, the ligand is a GalNAc ligand.

In some embodiments, the ligand targets the dsRNA to hepatocytes.

In some embodiments, the ligand is conjugated to the 3′ end of the sense strand of the dsRNA.

In some embodiments, the region of complementarity consists of an antisense sequence selected from Table 2 or Table 3. In embodiments, the region of complementarity consists of an antisense sequence selected from Tables 2, 3, 6, 7, 8, 9, 14, or 15. In some embodiments, the region of complementarity consists of an antisense sequence selected from that of AD-58882, AD-58878, AD-58886, AD-58877, AD-59115, AD-58856, AD-59129, AD-59124, AD-58874, AD-59125, AD-59105, AD-59120, AD-59122, AD-59106, AD-59126, or AD-59107 as disclosed herein in the Examples.

In some embodiments, the dsRNA comprises a sense strand consisting of a sense strand sequence selected from Table 2 or Table 3, and an antisense strand consisting of an antisense sequence selected from Table 2 or Table 3.

In some embodiments, the dsRNA comprises a sense strand consisting of a sense strand sequence selected from Tables 2, 3, 6, 7, 8, 9, 14, or 15, and an antisense strand consisting of an antisense sequence selected from Tables 2, 3, 6, 7, 8, 9, 14, or 15. In embodiments, the dsRNA comprises a pair of corresponding sense and antisense sequences selected from those of the duplexes disclosed in Tables 2, 3, 6, 7, 8, 9, 14, and 15.

In one aspect, the invention provides a cell containing at least one of the iRNAs (e.g., dsRNAs) featured herein. The cell is generally a mammalian cell, such as a human cell. In some embodiments, the cell is an erythroid cell. In other embodiments, the cell is a liver cell (e.g., a hepatocyte).

In an aspect provided herein is a pharmaceutical composition for inhibiting expression of an ALAS1 gene, the composition comprising an iRNA (e.g., a dsRNA) described herein.

In embodiments of the pharmaceutical compositions described herein, the iRNA (e.g., dsRNA) is administered in an unbuffered solution. In embodiments, the unbuffered solution is saline or water.

In embodiments of the pharmaceutical compositions described herein, the iRNA (e.g., dsRNA is administered with a buffer solution. In embodiments, the buffer solution comprises acetate, citrate, prolamine, carbonate, or phosphate or any combination thereof. In embodiments, the buffer solution is phosphate buffered saline (PBS).

In embodiments of the pharmaceutical compositions described herein, the iRNA (e.g., dsRNA) is targeted to hepatocytes.

In embodiments of the pharmaceutical compositions described herein, the composition is administered intravenously.

In embodiments of the pharmaceutical compositions described herein, the composition is administered subcutaneously.

In embodiments, a pharmaceutical composition comprises an iRNA (e.g., a dsRNA) described herein that comprises a ligand (e.g., a GalNAc ligand) that targets the iRNA (e.g., dsRNA) to hepatocytes.

In embodiments, a pharmaceutical composition comprises an iRNA (e.g., a dsRNA) described herein that comprises a ligand (e.g., a GalNAc ligand), and the pharmaceutical composition is administered subcutaneously. In embodiments, the ligand targets the iRNA (e.g., dsRNA) to hepatocytes.

In certain embodiments, a pharmaceutical composition, e.g., a composition described herein, includes a lipid formulation. In some embodiments, the RNAi agent is in a LNP formulation, e.g., a MC3 formulation. In some embodiments, the LNP formulation targets the RNAi agent to a particular cell, e.g., a liver cell, e.g., a hepatocyte. In embodiments, the lipid formulation is a LNP11 formulation. In embodiments, the composition is administered intravenously.

In another embodiment, the pharmaceutical composition is formulated for administration according to a dosage regimen described herein, e.g., not more than once every four weeks, not more than once every three weeks, not more than once every two weeks, or not more than once every week. In another embodiment, the administration of the pharmaceutical composition can be maintained for a month or longer, e.g., one, two, three, or six months, or one year or longer.

In another embodiment, a composition containing an iRNA featured in the invention, e.g., a dsRNA targeting ALAS1, is administered with a non-iRNA therapeutic agent, such as an agent known to treat a porphyria (e.g., AIP), or a symptom of a porphyria (e.g., pain). In another embodiment, a composition containing an iRNA featured in the invention, e.g., a dsRNA targeting AIP, is administered along with a non-iRNA therapeutic regimen, such as hemin or glucose (e.g., glucose infusion (e.g., IV glucose)). For example, an iRNA featured in the invention can be administered before, after, or concurrent with glucose, dextrose, or a similar treatment that serves to restore energy balance (e.g., total parenteral nutrition). An iRNA featured in the invention can also be administered before, after, or concurrent with the administration of a heme product (e.g., hemin, heme arginate, or heme albumin), and optionally also in combination with a glucose (e.g. IV glucose) or the like.

Typically, glucose administered for the treatment of a porphyria is administered intravenously (IV). Administration of glucose intravenously is referred to herein as “IV glucose.” However, alternative embodiments in which glucose is administered by other means are also encompassed.

In one embodiment, an ALAS1 iRNA is administered to a patient, and then the non-iRNA agent or therapeutic regimen (e.g., glucose and/or a heme product) is administered to the patient (or vice versa). In another embodiment, an ALAS1 iRNA and the non-iRNA therapeutic agent or therapeutic regimen are administered at the same time.

In an aspect provided herein is a method of inhibiting ALAS1 expression in a cell, the method comprising: (a) introducing into the cell an iRNA (e.g. a dsRNA) described herein and (b) maintaining the cell of step (a) for a time sufficient to obtain degradation of the mRNA transcript of an ALAS1 gene, thereby inhibiting expression of the ALAS1 gene in the cell.

In an aspect provided herein is a method for reducing or inhibiting the expression of an ALAS1 gene in a cell (e.g., an erythroid cell or a liver cell, such as, e.g., a hepatocyte). The method includes:

    • (a) introducing into the cell a double-stranded ribonucleic acid (dsRNA), wherein the dsRNA includes at least two sequences that are complementary to each other. The dsRNA has a sense strand having a first sequence and an antisense strand having a second sequence; the antisense strand has a region of complementarity that is substantially complementary to at least a part of an mRNA encoding ALAS1, and where the region of complementarity is 30 nucleotides or less, i.e., 15-30 nucleotides in length, and generally 19-24 nucleotides in length, and where the dsRNA upon contact with a cell expressing ALAS1, inhibits expression of an ALAS1 gene by at least 10%, e.g., at least 20%, at least 30%, at least 40% or more; and
    • (b) maintaining the cell of step (a) for a time sufficient to obtain degradation of the mRNA transcript of the ALAS1 gene, thereby reducing or inhibiting expression of an ALAS1gene in the cell.

In embodiments of the foregoing methods of inhibiting ALAS1 expression in a cell, the cell is treated ex vivo, in vitro, or in vivo. In embodiments, the cell is a hepatocyte.

In embodiments, the cell is present in a subject in need of treatment, prevention and/or management of a disorder related to ALAS1 expression.

In embodiments, the disorder is a porphyria. In embodiments, the porphyria is acute intermittent porphyria or ALA-dehydratase deficiency porphyria.

In embodiments, the porphyria is a hepatic porphyria, e.g., a porphyria selected from acute intermittent porphyria (AIP) hereditary coproporphyria (HCP), variegate porphyria (VP), ALA deyhdratase deficiency porphyria (ADP), and hepatoerythropoietic porphyria. In embodiments, the porphyria is a hom*ozygous dominant hepatic porphyria (e.g., hom*ozygous dominant AIP, HCP, or VP) or hepatoerythropoietic porphyria, In embodiments, the porphyria is a dual porphyria.

In embodiments, the expression of ALAS1 is inhibited by at least 30%.

In embodiments, the iRNA (e.g., dsRNA) has an IC50 in the range of 0.01-1 nM.

In certain embodiments, the cell (e.g., the hepatocyte) is a mammalian cell (e.g., a human, non-human primate, or rodent cell).

In one embodiment, the cell is treated ex vivo, in vitro, or in vivo (e.g., the cell is present in a subject (e.g., a patient in need of treatment, prevention and/or management of a disorder related to ALAS1 expression).

In one embodiment, the subject is a mammal (e.g., a human) at risk, or diagnosed with a porphyria, e.g., X-linked sideroblastic anemia (XLSA), ALA deyhdratase deficiency porphyria (ADP or Doss porphyria), acute intermittent porphyria (AIP), congenital erythropoietic porphyria (CEP), Prophyria cutanea tarda (PCT), hereditary coproporphyria (coproporphyria, or HCP), variegate porphyria (VP), erythropoietic protoporphyria (EPP), or transient erythroporphyria of infancy. In some embodiments, the disorder is an acute hepatic porphyria, e.g., ALA deyhdratase deficiency porphyria (ADP), AIP, HCP, or VP. In specific embodiments, the disorder is ALA deyhdratase deficiency porphyria (ADP) or AlP.

In embodiments, the porphyria is a hepatic porphyria, e.g., a porphyria selected from acute intermittent porphyria (AIP) hereditary coproporphyria (HCP), variegate porphyria (VP), ALA deyhdratase deficiency porphyria (ADP), and hepatoerythropoietic porphyria. In embodiments, the porphyria is a hom*ozygous dominant hepatic porphyria (e.g., hom*ozygous dominant AIP, HCP, or VP) or hepatoerythropoietic porphyria, In embodiments, the porphyria is a dual porphyria.

In one embodiment, the dsRNA introduced reduces or inhibits expression of an ALAS1 gene in the cell.

In one embodiment, the dsRNA introduced reduces or inhibits expression of an ALAS1 gene, or the level of one or more porphyrins or porphyrin precursors (e.g., δS-aminolevulinic acid (ALA), porphopilinogen (PBG), hydroxymethylbilane (HMB), uroporphyrinogen I or III, coproporphyrinogen I or III, protoporphrinogen IX, and protoporphyrin IX) or porphyrin products or metabolites, by at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50% or more compared to a reference, (e.g., an untreated cell or a cell treated with a non-targeting control dsRNA). Without being bound by theory, ALAS1 is the first enzyme of the porphyrin pathway. Thus, reducing expression of the ALAS1 gene is likely to reduce the level of one or more porphyrin precursors, porphyrins or porphyrin products or metabolites.

In other aspects, the invention provides methods for treating, preventing or managing pathological processes related to ALAS1 expression (e.g., pathological processes involving porphyrins, porphyrin precuorsors, or defects in the porphyrin pathway, such as, for example, porphyrias). In one embodiment, the method includes administering to a subject, e.g., a patient in need of such treatment, prevention or management, an effective (e.g., a therapeutically or prophylactically effective) amount of one or more of the iRNAs featured herein.

In an aspect provided herein is a method of treating and/or preventing a disorder related to ALAS1 expression comprising administering to a subject in need of such treatment a therapeutically effective amount of an iRNA (e.g., a dsRNA) described herein, or a composition comprising an iRNA (e.g., a dsRNA) described herein.

In an aspect provided herein is a method of treating and/or preventing a porphyria comprising administering to a subject in need of such treatment a double-stranded ribonucleic acid (dsRNA), wherein said dsRNA comprises a sense strand and an antisense strand 15-30 base pairs in length and the antisense strand is complementary to at least 15 contiguous nucleotides of SEQ ID NO:1 or SEQ ID NO:382.

In one embodiment, subject (e.g., the patient) has a porphyria. In another embodiment, the subject (e.g., patient) is at risk for developing a porphyria. In some embodiments, administration of the iRNA targeting ALAS1 alleviates or relieves the severity of at least one symptom of a disorder related to ALAS1 in the patient.

In one embodiment, the subject is a mammal (e.g., a human) at risk, or that has been diagnosed with, a disorder related to ALAS1 expression, e.g., a porphyria, e.g., X-linked sideroblastic anemia (XLSA), ALA deyhdratase deficiency porphyria (Doss porphyria), acute intermittent porphyria (AIP), congenital erythropoietic porphyria (CEP), prophyria cutanea tarda (PCT), hereditary coproporphyria (coproporphyria, or HCP), variegate porphyria (VP), erythropoietic protoporphyria (EPP), or transient erythroporphyria of infancy. In a further embodiment, the porphyria is an acute hepatic porphyria, e.g., ALA deyhdratase deficiency porphyria (ADP), AIP, HCP, or VP. In some such embodiments, the disorder is ALA deyhdratase deficiency porphyria (ADP) or AIP.

In embodiments the subject has, or is at risk for developing, a porphyria. In embodiments, the porphyria is a hepatic porphyria, e.g., a porphyria selected from acute intermittent porphyria (AIP) hereditary coproporphyria (HCP), variegate porphyria (VP), ALA deyhdratase deficiency porphyria (ADP), and hepatoerythropoietic porphyria. In embodiments, the porphyria is a hom*ozygous dominant hepatic porphyria (e.g., hom*ozygous dominant AIP, HCP, or VP) or hepatoerythropoietic porphyria, In embodiments, the porphyria is a dual porphyria.

In embodiments, a porphyria, a symptom of porphyria, a prodrome, or an attack of porphyria is induced by exposure to a precipitating factor, as described herein. In some embodiments, the precipitating factor is a chemical exposure. In some embodiments, the precipitating factor is a drug, e.g., a prescription drug or an over the counter drug. In some embodiments, the precipitating factor is the menstrual cycle, e.g., a particular phase of the menstrual cycle, e.g., the luteal phase.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered after an acute attack of porphyria.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered during an acute attack of porphyria.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered prophylactically to prevent an acute attack of porphyria.

In embodiments, the iRNA (e.g., dsRNA) is formulated as an LNP formulation.

In embodiments, the iRNA (e.g., dsRNA) is in the form of a GalNAc conjugate.

In embodiments, iRNA (e.g., dsRNA) is administered at a dose of 0.05-50 mg/kg.

In embodiments, the iRNA (e.g., dsRNA) is administered at a concentration of 0.01 mg/kg-5 mg/kg bodyweight of the subject.

In embodiments, the iRNA (e.g., dsRNA) is formulated as an LNP formulation and is administered at a dose of 0.05-5 mg/kg.

In embodiments, the iRNA (e.g., dsRNA) is in the form of a GalNAc conjugate and is administered at a dose of 0.5-50 mg/kg.

In embodiments, the method decreases a level of a porphyrin or a porphyrin precursor in the subject.

In embodiments, the level is decreased by at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90%. In an embodiment, the level is decreased by at least 30%.

In embodiments, the porphyrin precursor is δ-aminolevulinic acid (ALA) or porphopilinogen (PBG).

In embodiments, the iRNA (e.g., dsRNA) has an IC50 in the range of 0.01-1 nM.

In embodiments, a method described herein

    • (i) ameliorates a symptom associated with an ALAS1 related disorder (e.g., a porphyria)
    • (ii) inhibits ALAS1 expression in the subject,
    • (iii) decreases a level of a porphyrin precursor (e.g., ALA or PBG) or a porphyrin in the subject,
    • (iv) decreases frequency of acute attacks of symptoms associated with a porphyria in the subject, or
    • (v) decreases incidence of acute attacks of symptoms associated with a porphyria in the subject when the subject is exposed to a precipitating factor (e.g., the premenstrual phase or the luteal phase).

In embodiments, the method ameliorates pain and/or progressive neuropathy.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered according to a dosing regimen.

In some embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered before or during an acute attack of porphyria. In some embodiments, the iRNA is administered before an acute attack of porphyria.

In some embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered during a prodrome. In embodiments, the prodrome is characterized by abdominal pain, nausea, psychological symptoms (e.g., anxiety), restlessness and/or insomnia.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered during a particular phase of the menstrual cycle, e.g., during the luteal phase.

In embodiments, the method ameliorates or prevents cyclical attacks of porphyria, e.g., by reducing the severity, duration, or frequency of attacks. In embodiments, the cyclical attacks are associated with a precipitating factor. In embodiments, the precipitating factor is the menstrual cycle, e.g., a particular phase of the menstrual cycle, e.g., the luteal phase.

In embodiments, the subject has an elevated level of ALA and/or PBG. In embodiments, the subject has or is at risk for developing a porphyria, e.g., a hepatic porphyria. In embodiments, the subject is asymptomatic. In embodiments, the subject carries a genetic alteration (e.g., a gene mutation) associated with a porphyria, as described herein.

In embodiments, the subject has or is at risk for developing a porphyria and suffers from pain (e.g., chronic pain, e.g., chronic neuropathic pain) and/or neuropathy (e.g., progressive neuropathy). In embodiments, the subject does not suffer from acute attacks but suffers from pain (e.g., chronic pain, e.g., chronic neuropathic pain) and/or neuropathy (e.g., progressive neuropathy). In embodiments, the pain is abdominal pain.

In embodiments, the subject (a) has an elevated level of ALA and/or PBG and (b) suffers from pain (e.g., chronic pain, e.g., chronic neuropathic pain) and/or neuropathy (e.g., progressive neuropathy). In embodiments, the pain is abdominal pain.

In embodiments, the subject has a plasma level and/or a urine level of ALA and/or PBG that is elevated. In embodiments, the elevated level of ALA and/or PBG is accompanied by other symptoms, e.g., pain (e.g., chronic pain, e.g., chronic neuropathic pain) or neuropathy (e.g., progressive neuropathy). In embodiments, the pain is abdominal pain. In embodiments, the subject is asymptomatic. In embodiments, the subject has a genetic mutation associated with a porphyria, e.g., a mutation as described herein.

In embodiments, the subject has a level (e.g., a plasma level or a urine level) of a porphyrin precursor, e.g., ALA and/or PBG, that is elevated, e.g., the level is greater than, or greater than or equal to, a reference value. In embodiments, the level is greater than the reference value. In embodiments, the reference value is two standard deviations above the mean level in a sample of healthy individuals. In embodiments, the reference value is an upper reference limit.

In embodiments, the subject has a plasma level and/or a urine level of ALA and/or PBG that is greater than, or greater than or or equal to, 2 times, 3 times, 4 times, or 5 times that of an upper reference limit. As used herein, an “upper reference limit” refers to a level that is the upper limit of the 95% confidence interval for a reference sample, e.g., a sample of normal (e.g., wild type) or healthy individuals, e.g., individuals who do not carry a genetic mutation associated with a porphyria and/or individuals who do not suffer from a porphyria. In embodiments, the subject has a urine level of ALA and/or PBG that is greater than 2 to 4 times that of an upper reference limit. In embodiments, the subject has a urine level of ALA and/or PBG that is greater than 4 times that of an upper reference limit.

In embodiments, the reference value for plasma PBG is 0.12 μmol/L. In embodiments, the subject is a human and has a plasma PBG level that is greater than, or greater than or equal to, 0.12 μmol/L, 0.24 μmol/L, 0.36 μmol/L, 0.48 μmol/L, or 0.60 μmol/L. In embodiments, the subject is a human and has a plasma level of PBG that is greater than, or greater than or equal to, 0.48 μmol/L.

In embodiments, the reference value for urine PBG is 1.2 mmol/mol creatinine. In embodiments, the subject is a human and has a urine PBG level that is greater than, or greater than or equal to, 1.2 mmol/mol creatinine, 2.4 mmol/mol creatinine, 3.6 mmol/mol creatinine, 4.8 mmol/mol creatinine, or 6.0 mmol/mol creatinine. In embodiments, the subject is a human and has a urine level of PBG that is greater than, or greater than or equal to, 4.8 mmol/mol creatinine.

In embodiments, the reference value for plasma ALA is 0.12 μmol/L. In embodiments, the subject is a human and has a plasma ALA level that is greater than, or greater than or equal to, 0.12 μmol/L, 0.24 μmol/L, 0.36 μmol/L, 0.48 μmol/L, or 0.60 μmol/L. In embodiments, the subject is a human and has a plasma ALA level that is greater than, or greater than or equal to 0.48 μmol/L.

In embodiments, the reference value for urine ALA is 3.1 mmol/mol creatinine. In embodiments, the subject is a human and has a urine ALA level that is greater than, or greater than or equal to, 3.1 mmol/mol creatinine, 6.2 mmol/mol creatinine, 9.3 mmol/mol creatinine, 12.4 mmol/mol creatinine, or 15.5 mmol/mol creatinine.

In embodiments, the method decreases an elevated level of ALA and/or PBG. In embodiments, the method decreases pain (e.g., chronic pain, e.g. chronic neuropathic pain) and/or neuropathy (e.g., progressive neuropathy). In embodiments, the pain is abdominal pain. In embodiments, the pain is neuropathic pain (e.g., pain associated with the progressive neuropathy of acute porphyrias). The decrease in pain can include, e.g., prevention of pain, delay in the onset of pain, reduction in the frequency of pain, and/or reduction in severity of pain.

In embodiments, the method ameliorates or prevents acute attacks of porphyria, e.g., by reducing the severity, duration, or frequency of attacks.

In embodiments, the method decreases or prevents nerve damage.

In embodiments, the method prevents deterioration (e.g., prevents development of abnormalities) of or results in an improvement of clinical measures, e.g., clinical measures of muscle and/or nerve function, e.g., EMG and/or nerve conduction velocities.

In embodiments, the method is effective to reduce a level of ALA and/or PBG (e.g., a plasma or urine level of ALA and/or PBG). In embodiments, the method is effective to produce a predetermined reduction in the elevated level of ALA and/or PBG.

In embodiments, the predetermined reduction is a reduction to a value that is less than or equal to a reference value. In some embodiments, the reference value is an upper reference limit.

In some embodiments, the reference value is the value that is two standard deviations above the mean level in a reference sample.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered repeatedly, e.g., according to a dosing regimen.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered prophylactically to a subject who is at risk for developing a porphyria. In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered prophylactically beginning at puberty. In embodiments, the subject carries a genetic mutation associated with a porphyria and/or has an elevated level of ALA and/or PBG (e.g., an elevated plasma or urine level of ALA and/or PBG). In embodiments, the mutation makes an individual susceptible to an acute attack (e.g., upon exposure to a precipitating factor, e.g., a drug, dieting or other precipitating factor, e.g., a precipitating factor as disclosed herein). In embodiments, the mutation is associated with elevated levels of a porphyrin or a porphyrin precursor (e.g., ALA and/or PBG). In embodiments, the mutation is associated with chronic pain (e.g., chronic neuropathic pain) and/or neuropathy (e.g., progressive neuropathy).

In embodiments, the mutation is a mutation in the ALAS1 gene. In embodiments, the mutation is a mutation in the ALAS1 gene promoter, or in regions upstream or downstream from the ALAS1 gene. In embodiments, the mutation is a mutation in transcription factors or other genes that interact with ALAS1. In embodiments, the mutation is a mutation in a gene that encodes an enzyme in the heme biosynthetic pathway.

In embodiments, the iRNA (e.g., dsRNA) or composition comprising the iRNA is administered subcutaneously. In embodiments, the iRNA is in the form of a GalNAc conjugate. In embodiments, the iRNA (e.g., the dsRNA) is administered at a dose of 0.5-50 mg/kg.

In one aspect provided herein is a method of treating a subject with an elevated level of ALA and/or PBG, the method comprising administering to the subject a double-stranded ribonucleic acid (dsRNA), wherein said dsRNA comprises a sense strand and an antisense strand 15-30 base pairs in length and the antisense strand is complementary to at least 15 contiguous nucleotides of SEQ ID NO:1 or SEQ ID NO:382.

In one aspect provided herein is a method of treating a subject with an elevated level of ALA and/or PBG, the method comprising administering to the subject a therapeutically effective amount of an dsRNA or a composition comprising a dsRNA, as described herein.

In some embodiments, the methods described herein are effective to decrease the level of ALA and/or PBG. In some embodiments, the level of ALA and/or PBG is decreased such that it is less than, or less than or equal to, a reference value, e.g., an upper reference limit. In another aspect, the invention provides methods for decreasing a level of a porphyrin or a porphyrin precursor in a cell (e.g., an erythroid cell or a liver cell, such as, e.g., a hepatocyte). In one embodiment, the cell is treated ex vivo, in vitro, or in vivo (e.g., the cell is present in a subject (e.g., a patient in need of treatment, prevention and/or management of a disorder related to ALAS1 expression). The method includes contacting the cell with an effective amount of one or more of the iRNAs targeting ALAS1, e.g., one or more of the iRNAs disclosed herein, thereby decreasing the level of a porphyrin or a porphyrin precursor in the cell; or decreasing the level of a porphyrin or a porphyrin precursor in other cells, tissues, or fluids within a subject in which the cell is located; relative to the level prior to contacting. Such methods can be used to treat (e.g., ameliorate the severity) of disorders related to ALAS1 expression, such as porphyrias, e.g., AIP or ALA dehydratase deficiency porphyria.

In one embodiment, the contacting step is effected ex vivo, in vitro, or in vivo. For example, the cell can be present in a subject, e.g., a mammal (e.g., a human) at risk, or that has been diagnosed with, a porphyria. In an embodiment, the porphyria is an acute hepatic porphyria. In embodiments, the porphyria is a hepatic porphyria, e.g., a porphyria selected from acute intermittent porphyria (AIP), hereditary coproporphyria (HCP), variegate porphyria (VP), ALA deyhdratase deficiency porphyria (ADP), and hepatoerythropoietic porphyria. In embodiments, the porphyria is a hom*ozygous dominant hepatic porphyria (e.g., hom*ozygous dominant AIP, HCP, or VP) or hepatoerythropoietic porphyria, In embodiments, the porphyria is a dual porphyria.

In an aspect provided herein is a method for decreasing a level of a porphyrin or a porphyrin precursor (e.g., ALA or PBG) in a cell, comprising contacting the cell with an iRNA (e.g. a dsRNA), as described herein, in an amount effective to decrease the level of the porphyrin or the porphyrin precursor in the cell. In embodiments, the cell is a hepatocyte. In embodiments, the porphyrin or porphyrin precursor is S-aminolevulinic acid (ALA), porphopilinogen (PBG), hydroxymethylbilane (HMB), uroporphyrinogen I or III, coproporphyrinogen I or III, protoporphrinogen IX, or protoporphyrin IX. In embodiments, the porphyrin precursor is ALA or PBG.

In one embodiment, the cell is an erythroid cell. In a further embodiment, the cell is a liver cell (e.g., a hepatocyte).

In an aspect provided herein is a vector encoding at least one strand of an iRNA (e.g., a dsRNA) as described herein.

In an aspect provided herein is a vector encoding at least one strand of a dsRNA, wherein said dsRNA comprises a region of complementarity to at least a part of an mRNA encoding ALAS1, wherein said dsRNA is 30 base pairs or less in length, and wherein said dsRNA targets said mRNA for cleavage.

In embodiments, the region of complementarity is at least 15 nucleotides in length.

In embodiments, the region of complementarity is 19 to 21 nucleotides in length. In one aspect, the invention provides a vector for inhibiting the expression of an ALAS1 gene in a cell.

In one embodiment, the vector comprises an iRNA as described herein. In one embodiment, the vector includes at least one regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of an iRNA as described herein. In one embodiment the vector comprises at least one strand of an ALAS1 iRNA.

In an aspect provided herein is a cell comprising a vector as described herein. In an aspect provided herein is a cell containing a vector for inhibiting the expression of an ALAS1 gene in a cell. The vector includes a regulatory sequence operably linked to a nucleotide sequence that encodes at least one strand of one of the iRNAs as described herein. In one embodiment, the cell is a liver cell (e.g., a hepatocyte). In another embodiment, the cell is an erythroid cell.

All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety.

The details of various embodiments of the invention are set forth in the description below. Other features, objects, and advantages of the invention will be apparent from the description and the drawings, and from the claims.

FIG. 1 depicts the heme biosynthetic pathway.

FIG. 2A is a table that summarizes certain porphyrias associated with genetic errors in heme metabolism.

FIG. 2B is a continuation of the table in FIG. 2A.

FIG. 3A depicts nucleotides 1-2280 of the sequence of a human ALAS1 mRNA sequence transcript variant 1 (Ref. Seq. NM_000688.4 (GI:40316942, record dated Nov. 19, 2011), SEQ ID NO: 1).

FIG. 3B depicts nucleotides 2281-2407 of the sequence of a human ALAS1 mRNA sequence transcript variant 1 (Ref. Seq. NM_000688.4 (GI:40316942, record dated Nov. 19, 2011), SEQ ID NO: 1).

FIG. 4A depicts nucleotides 1-2280 of the sequence of a human ALAS1 mRNA sequence transcript variant 2 (Ref. Seq. NM_000688.5 (GI: 362999011, record dated Apr. 1, 2012), SEQ ID NO: 382).

FIG. 4B depicts nucleotides 2281-2458 of the sequence of a human ALAS1 mRNA sequence transcript variant 2 (Ref. Seq. NM_000688.5 (GI: 362999011, record dated Apr. 1, 2012), SEQ ID NO: 382).

FIG. 5 shows the dose-response of the siRNA AD-53558 in suppressing mouse ALAS1 (mALAS1) mRNA relative to a PBS control. Results for a luciferase (LUC) AD-1955 control are also shown.

FIG. 6 shows the dose-response of the siRNA AD-53558 in suppressing ALAS1 mRNA in rats relative to a PBS control. Results for a luciferase (LUC) AD-1955 control are also shown.

FIG. 7 shows the durability of suppression of mouse ALAS1 (mALAS1) mRNA by the siRNA AD-53558 relative to a PBS control.

FIG. 8 shows means±standard deviations of plasma ALA levels (in RM) at baseline, and after phenobarbitol treatment in the experimental (ALAS1 siRNA) and control (LUC siRNA) groups.

FIG. 9 shows shows the plasma ALA levels (in RM) of individual animals at baseline, and after phenobarbitol treatment in animals that received ALAS1 siRNA and control (LUC siRNA) treatment.

FIG. 10 shows means±standard deviations of plasma PBG levels (in RM) at baseline, and after phenobarbitol treatment in animals that received ALAS1 siRNA and control (LUC siRNA) treatment.

FIG. 11 shows shows the plasma PBG levels (in RM) of individual animals at baseline, and after phenobarbitol treatment in animals that received ALAS1 siRNA and control (LUC siRNA) treatment.

FIG. 12 shows the relative mALAS1mRNA level in liver at baseline, and after phenobarbitol treatment in select representative experimental (ALAS1 siRNA) and control (PBS) animals.

FIG. 13 shows the effects of three GalNAc conjugated mALAS1 siRNAs on mALAS1 expression (relative to a PBS control) in mouse liver tissue.

FIG. 14 shows plasma ALA and PBG levels over time after phenobarbitol administration and treatment with ALAS1 siRNA or control LUC siRNA.

iRNA directs the sequence-specific degradation of mRNA through a process known as RNA interference (RNAi). Described herein are iRNAs and methods of using them for inhibiting the expression of an ALAS1 gene in a cell or a mammal where the iRNA targets an ALAS1 gene. Also provided are compositions and methods for disorders related to ALAS1 expression, such as porphyrias (e.g., ALA deyhdratase deficiency porphyria (ADP or Doss porphyria), acute intermittent porphyria, congenital erythropoietic porphyria, Prophyria cutanea tarda, hereditary coproporphyria (coproporphyria), variegate porphyria, erythropoietic protoporphyria (EPP), X-linked sideroblastic anemia (XLSA), and and transient erythroporphyria of infancy).

Porphyrias are inherited or acquired disorders that can be caused by decreased or enhanced activity of specific enzymes in the heme biosynthetic pathway, also referred to herein as the porphyrin pathway (See FIG. 1). Porphyrins are the main precursors of heme. Porphyrins and porphyrin precursors include S-aminolevulinic acid (ALA), porphopilinogen (PBG), hydroxymethylbilane (HMB), uroporphyrinogen I or III, coproporphyrinogen I or III, protoporphrinogen IX, and protoporphyrin IX. Heme is an essential part of hemoglobin, myoglobin, catalases, peroxidases, and cytochromes, the latter including the respiratory and P450 liver cytochromes. Heme is synthesized in most or all human cells. About 85% of heme is made in erythroid cells, primarily for hemoglobin. Most of the remaining heme is made in the liver, 80% of which is used for the synthesis of cytochromes. Deficiency of specific enzymes in the porphyrin pathway leads to insufficient heme production and also to an accumulation of porphyrin precursors and/or porphyrins, which can be toxic to cell or organ function in high concentrations.

Porphyrias may manifest with neurological complications (“acute”), skin problems (“cutaneous”) or both. Porphyrias may be classified by the primary site of the overproduction and accumulation of porphyrins or their precursors. In hepatic porphyrias, porphyrins and porphyrin precursors are overproduced predominantly in the liver, whereas in erythropoietic porphyrias, porphyrins are overproduced in the erythroid cells in the bone. The acute or hepatic porphyrias lead to dysfunction of the the nervous system and neurologic manifestations that can affect both the central and peripheral nervous system, resulting in symptoms such as, for example, pain (e.g., abdominal pain and/or chronic neuropathic pain), vomiting, neuropathy (e.g., acute neuropathy progressive neuropathy), muscle weakness, seizures, mental disturbances (e.g., hallucinations, depression anxiety, paranoia), cardiac arrhythmias, tachycardia, constipation, and diarrhea. The cutaneous or erythropoietic porphyrias primarily affect the skin, causing symptoms such as photosensitivity that can be painful, blisters, necrosis, itching, swelling, and increased hair growth on areas such as the forehead. Subsequent infection of skin lesions can lead to bone and tissue loss, as well as scarring, disfigurement, and loss of digits (e.g., fingers, toes). Most porphyrias are caused by mutations that encode enzymes in the heme biosynthetic pathway. A summary of porphyrias associated with genetic errors in heme metabolism is provided in FIG. 2.

Not all porphyrias are genetic. For example, patients with liver disease may develop porphyria as a result of liver dysfunction, and a transient form of erythroporphria (transient erythroporphyria of infancy) has been described in infancy (see Crawford, R. I. et al, J Am Acad Dermatol. 1995 August; 33(2 Pt 2):333-6.) Patients with PCT can acquire the deficient activity of uroporphyrinogen decarboxylase (URO-D), due to the formation of a ORO-D enzyme with lower than normal enzymatic activity (see Phillips et al. Blood, 98:3179-3185, 2001.)

Acute intermittent porphyria (AIP) (also be referred to as porphobilinogen (PBG) deaminase deficiency, or hydroxymethylbilane synthase (HMBS) deficiency), is the most common type of acute hepatic porphyria. Other types of acute hepatic porphyrias include hereditary coproporphyria (HCP), variegate porphyria (VP), and ALA deyhdratase deficiency porphyria (ADP). Acute hepatic porphyrias are described, e.g., in Balwani, M and Desnick, R. J., Blood, 120:4496-4504, 2012.

AIP is typically an autosomal dominant disease that is characterized by a deficiency of the enzyme porphobilinogen deaminase (PBG deaminase); this enzyme is also known as hydroxymethylbilane synthase (HMB synthase or HMBS). PBG deaminase is the third enzyme of the heme biosynthetic pathway (see FIG. 1) and catalyzes the head to tail condensation of four porphobilinogen molecules into the linear tetrapyrrole, hydroxymethylbilane (HMB). Alternatively spliced transcript variants encoding different isoforms of PBG deaminase have been described. Mutations in the PBG deaminase gene are associated with AIP. Such mutations may lead to decreased amounts of PBG deaminase and/or decreased activity of PBG deaminase (affected individuals typically have a ˜50% reduction in PBG deaminase activity).

There are at least two different models of the pathophysiology of AIP and other acute hepatic porphyrias (see, e.g., Lin CS-Y et al., Clinical Neurophysiology, 2011; 122:2336-44). According to one model, the decreased heme production resulting from PBG deaminase deficiency causes energy failure and axonal degeneration. According to the other, currently more favored model, the buildup of porphyrin precursors (e.g., ALA and PBG) results in neurotoxicity.

AIP has been found to have a prevalence as high as 1 in 10,000 in certain populations (e.g., in Northern Sweden; see Floderus Y, et al. Clin Genet. 2002; 62:288-97). The prevalence in the general population in United States and Europe, excluding the U. K., is estimated to be about 1 in 10,000 to 1 in 20,000. Clinical disease manifests itself in only approximately 10-15% of individuals who carry mutations that are known to be associated with AIP. However, the penetrance is as high as 40% in individuals with certain mutations (e.g., the W198X mutation). AIP is typically latent prior to puberty. Symptoms are more common in females than in males. The prevalence of the disease is probably underestimated due to its incomplete penetrance and long periods of latency. In the United States, it is estimated that there are about 2000 patients who have suffered at least one attack. It is estimated that there are about 150 active recurrent cases in France, Sweden, the U.K., and Poland; these patients are predominantly young women, with a median age of 30. See, e.g., Elder et al, J Inherit Metab Dis., published online Nov. 1, 2012.

AIP affects, for example, the visceral, peripheral, autonomic, and central nervous systems. Symptoms of AIP are variable and include gastrointestinal symptoms (e.g., severe and poorly localized abdominal pain, nausea/vomiting, constipation, diarrhea, ileus), urinary symptoms (dysuria, urinary retention/incontinence, or dark urine), neurologic symptoms (e.g., sensory neuropathy, motor neuropathy (e.g., affecting the cranial nerves and/or leading to weakness in the arms or legs), seizures, neuropathic pain (e.g., pain associated with progressive neuropathy, e.g., chronic neuropathic pain), neuropsychiatric symptoms (e.g., mental confusion, anxiety, agitation, hallucination, hysteria, delirium, apathy, depression, phobias, psychosis, insomnia, somnolence, coma), autonomic nervous system involvement (resulting e.g., in cardiovascular symptoms such as tachycardia, hypertension, and/or arrhythmias, as well as other symptoms, such as, e.g., increased circulating catecholamine levels, sweating, restlessness, and/or tremor), dehydration, and electrolyte abnormalities. The most common symptoms are abdominal pain and tachycardia. In addition, patients frequently have chronic neuropathic pain and develop a progressive neuropathy. Patients with recurring attacks often have a prodrome. Permanent paralysis may occur after a severe attack. Recovery from severe attacks that are not promptly treated may take weeks or months. An acute attack may be fatal, for example, due to paralysis of respiratory muscles or cardiovascular failure from electrolyte imbalance. (See, e.g., Thunell S. Hydroxymethylbilane Synthase Deficiency. 2005 Sep. 27 [Updated 2011 Sep. 1]. In: Pagon R A, Bird T D, Dolan C R, et al., editors. GeneReviews™ [Internet]. Seattle (WA): University of Washington, Seattle; 1993-(hereinafter Thunell (1993)), which is hereby incorporated by reference in its entirety.) Prior to the availability of Hemin treatments, up to 20% of patients with AIP died from the disease.

In individuals who carry genes for AIP, the risk of hepatocellular cancer is increased. In those with recurrent attacks, the risk of hepatocellular cancer is particularly grave: after the age of 50, the risk is nearly 100-fold greater than in the general population.

Attacks of acute porphyria may be precipitated by endogenous or exogenous factors. The mechanisms by which such factors induce attacks may include, for example, increased demand for hepatic P450 enzymes and/or induction of ALAS1 activity in the liver. Increased demand for hepatic P450 enzymes results in decreased hepatic free heme, thereby inducing the synthesis of hepatic ALAS1.

Precipitating factors include fasting (or other forms of reduced or inadequate caloric intake, due to crash diets, long-distance athletics, etc.), metabolic stresses (e.g., infections, surgery, international air travel, and psychological stress), endogenous hormones (e.g., progesterone), cigarette smoking, lipid-soluble foreign chemicals (including, e.g., chemicals present in tobacco smoke, certain prescription drugs, organic solvents, biocides, components in alcoholic beverages), endocrine factors (e.g., reproductive hormones (women may experience exacerbations during the premenstrual period), synthetic estrogens, progesterones, ovulation stimulants, and hormone replacement therapy). See, for example, Thunell (1993).

Over 1000 drugs are contraindicated in the acute hepatic porphyrias (e.g., AIP, HCP, ADP, and VP) including, for example, alcohol, barbiturates, Carbamazepine, Carisoprodol, Clonazepam (high doses), Danazol, Diclofenac and possibly other NSAIDS, Ergots, estrogens, Ethyclorvynol, Glutethimide, Griseofulvin, Mephenytoin, Meprobamate (also mebutamate and tybutamate), Methyprylon, Metodopramide, Phenytoin, Primidone, progesterone and synthetic progestins, Pyrazinamide, Pyrazolones (aminopyrine and antipyrine), Rifampin, Succinimides (ethosuximide and methsuximide), sulfonamide antibiotics, and Valproic acid.

Objective signs of AIP include discoloration of the urine during an acute attack (the urine may appear red or red-brown), and increased concentrations of PBG and ALA in urine during an acute attack. Molecular genetic testing identifies mutations in the PBG deaminase (also known as HMBS) gene in more than 98% of affected individuals. Thunell (1993).

The differential diagnosis of porphyrias may involve determining the type of porphyria by measuring individual levels of porphyrins or porphyrin precursors (e.g., ALA, PBG) in the urine, feces, and/or plasma (e.g., by chromatography and fluorometry) during an attack. The diagnosis of AIP can be confirmed by establishing that erythrocyte PBG deaminase activity is at 50% or less of the normal level. DNA testing for mutations may be carried out in patients and at-risk family members. The diagnosis of AIP is typically confirmed by DNA testing to identify a specific caustative gene mutation (e.g., an HMBS mutation).

Treatment of acute attacks typically requires hospitalization to control and treat acute symptoms, including, e.g., abdominal pain, seizures, dehydration/hyponatremia, nausea/vomiting, tachycardia/hypertension, urinary retention/ileus. For example, abdominal pain may be treated, e.g., with narcotic analgesics, seizures may be treated with seizure precautions and possibly medications (although many anti-seizure medications are contraindicated), nausea/vomiting may be treated, e.g., with phenothiazines, and tachycardia/hypertension may be treated, e.g., with beta blockers. Treatment may include withdrawal of unsafe medications, monitoring of respiratory function, as well as muscle strength and neurological status. Mild attacks (e.g., those with no paresis or hyponatremia) may be treated with at least 300 g intravenous 10% glucose per day, although increasingly hemin is provided immediately. Severe attacks should be treated as soon as possible with intravenous hemin (3-4 mg/kg daily for 4-14 days) and with IV glucose while waiting for the IV hemin to take effect. Typically, attacks are treated with IV hemin for 4 days and with IV glucose while waiting for administration of the IV hemin.

Hemin (Panhematin® or hemin for injection, previously known as hematin) is the only heme product approved for use in the United States and was the first drug approved under the Orphan Drug Act. Panhematin® is hemin derived from processed red blood cells (PRBCs), and is Protoporphyrin IX containing a ferric iron ion (Heme B) with a chloride ligand. Heme acts to limit the hepatic and/or marrow synthesis of porphyrin. The exact mechanism by which hemin produces symptomatic improvement in patients with acute episodes of the hepatic porphyrias has not been elucidated; however, its action is likely due to the (feedback) inhibition of S-aminolevulinic acid (ALA) synthase, the enzyme which limits the rate of the porphyrin/heme biosynthetic pathway. See Panhematin® product label, Lundbeck, Inc., October 2010. Inhibition of ALA synthase should result in reduced production of ALA and PBG as well as porphyrins and porphyrin intermediates.

Drawbacks of hemin include its delayed impact on clinical symptoms and its failure to prevent the recurrence of attacks. Adverse reactions associated with hemin administration may include thrombophlebitis, anticoagulation, thrombocytopenia, renal shut down, or iron overload, which is particularly likely in patients requiring multiple courses of hemin treatment for recurrent attacks. To prevent phlebitis, an indwelling venous catheter is needed for access in patients with recurrent attacks. Uncommonly reported side effects include fever, aching, malaise, hemolysis, anaphylaxis, and circulatory collapse. See Anderson, K. E., Approaches to Treatment and Prevention of Human Porphyrias, in The Porphyrin Handbook: Medical Aspects of Porphyrins, Edited by Karl M. Kadish, Kevin M. Smith, Roger Guilard (2003) (hereinafter Anderson).

Heme is difficult to prepare in a stable form for intravenous administration. It is insoluble at neutral pH but can be prepared as heme hydroxide at pH 8 or higher. Anderson. Panhematin is a lyophilized hemin preparation. When lyophilized hemin is solubilized for intravenous administration, degradation products form rapidly; these degradation products are responsible for a transient anticoagulant effect and for phlebitis at the site of infusion. Anderson. Heme albumin and heme arginate (Normosang, the European version of hemin) are more stable and may potentially cause less thrombophlebitis. However, heme arginate is not approved for use in the United States. Panhemin may be stabilized by solubilizing it for infusion in 30% human albumin rather than in sterile water; however, albumin adds intravascular volume-expanding effects and increases the cost of treatment as well as risk of pathogens since it is isolated from human blood. See, e.g., Anderson.

The successful treatment of an acute attack does not prevent or delay recurrence. There is a question of whether hemin itself can trigger recurring attacks due to induction of heme oxygenase. Nonetheless, in some areas (especially France), young women with multiply recurrent attacks are being treated with weekly hemin with the goal of achieving prophylaxis.

Limited experience with liver transplantation suggests that if successful, it is an effective treatment for AIP. There have been approximately 12 transplants in Europe in human patients, with curative or varying effects. Liver transplantation can restore normal excretion of ALA and PBG and prevent acute attacks. See, e.g., Dar, F. S. et al. Hepatobiliary Pancreat. Dis. Int., 9(1):93-96 (2010). Furthermore, if the liver of a patient with AIP is transplanted into another patient (“domino transplant”), the patient receiving the transplant may develop AIP.

Among the long-term clinical effects of acute porphyrias is chronic neuropathic pain that may result from a progressive neuropathy due to neurotoxic effects, e.g., of elevated porphyrin precursors (e.g., ALA and/or PBG). Patients may suffer from neuropathic pain prior to or during an acute attack. Older patients may experience increased neuropathic pain with age for which various narcotic drugs are typically prescribed. Electromyogram abnormalities and decreased conduction times have been documented in patients with acute hepatic porphyrias. Of note, untreated, uninduced mice with AIP (PBG deaminase deficiency) develop a progressive motor neuropathy that has been shown to cause progressive quadriceps nerve axon degeneration and loss presumably due to constitutively elevated porphyrin precursor (ALA & PBG) levels, porphyrins and/or heme deficiency (Lindberg et al., J. Clin. Invest., 103(8): 1127-1134, 1999). In patients with acute porphyria (e.g., ADP, AIP, HCP, or VP), levels of porphyrin precursors (ALA & PBG) are often elevated in asymptomatic patients and in symptomatic patients between attacks. Thus, reduction of the porphyrin precursors and resumption of normal heme biosynthesis by reducing the level of ALAS1 expression and/or activity is expected to prevent and/or minimize development of chronic and progressive neuropathy. Treatment, e.g., chronic treatment (e.g., periodic treatment with iRNA as described herein, e.g., treatment according to a dosing regimen as described herein, e.g., weekly or biweekly treatment) can continuously reduce the ALAS1 expression in acute porphyria patients who have elevated levels of porphyrin precursors, porphyrins, porphyrin products or their metabolites. Such treatment may be provided as needed to prevent or reduce the frequency or severity of an individual patient's symptoms (e.g., pain and/or neuropathy) and/or to reduce a level of a porphyrin precursor, porphyrin, porphyrin product or metabolite.

The need exists for identifying novel therapeutics that can be used for the treatment of porphyrias. As discussed above, existing treatments such as hemin have numerous drawbacks. For example, the impact of hemin on clinical symptoms is delayed, it is expensive, and it may have side effects (e.g., thrombophlebitis, anticoagulation, thrombocytopenia, iron overload, renal shutdown). Novel therapeutics such as those described herein can address these drawbacks and the unmet needs of patients by, for example, acting faster, not inducing phlebitis, providing the convenience of subcutaneous administration, successfully preventing recurrent attacks, preventing or ameliorating pain (e.g., chronic neuropathic pain) and/or progressive neuropathy, and/or not causing certain adverse effects associated with hemin (e.g., iron overload, increased risk of hepatocellular cancer).

The present disclosure provides methods and iRNA compositions for modulating the expression of an ALAS1 gene. In certain embodiments, expression of ALAS1 is reduced or inhibited using an ALAS1-specific iRNA, thereby leading to a decreased expression of an ALAS1 gene. Reduced expression of an ALAS1 gene may reduce the level of one or more porphyrin precursors, porphyrins, or porphyrin products or metabolites. Decreased expression of an ALAS1 gene, as well as related decreases in the level of one or more porphyrin precursors and/or porphyrins, can be useful in treating disorders related to ALAS1 expression, e.g., porphyrias.

The iRNAs of the compositions featured herein include an RNA strand (the antisense strand) having a region which is 30 nucleotides or less in length, i.e., 15-30 nucleotides in length, generally 19-24 nucleotides in length, which region is substantially complementary to at least part of an mRNA transcript of an ALAS1 gene (also referred to herein as an “ALAS1-specific iRNA”). The use of such an iRNA enables the targeted degradation of mRNAs of genes that are implicated in pathologies associated with ALAS1 expression in mammals, e.g., porphyrias such as ALA dehydratase deficiency porphyria (Doss porphyria) or acute intermittent porphyria. Very low dosages of ALAS1-specific iRNAs can specifically and efficiently mediate RNAi, resulting in significant inhibition of expression of an ALAS1 gene. iRNAs targeting ALAS1 can specifically and efficiently mediate RNAi, resulting in significant inhibition of expression of an ALAS1 gene, e.g., in cell based assays. Thus, methods and compositions including these iRNAs are useful for treating pathological processes related to ALAS1 expression, such as porphyrias (e.g., X-linked sideroblastic anemia (XLSA), ALA deyhdratase deficiency porphyria (Doss porphyria), acute intermittent porphyria (AIP), congenital erythropoietic porphyria, prophyria cutanea tarda, hereditary coproporphyria (coproporphyria), variegate porphyria, erythropoietic protoporphyria (EPP), and transient erythroporphyria of infancy).

The following description discloses how to make and use compositions containing iRNAs to inhibit the expression of an ALAS1 gene, as well as compositions and methods for treating diseases and disorders caused by or modulated by the expression of this gene. Embodiments of the pharmaceutical compositions featured in the invention include an iRNA having an antisense strand comprising a region which is 30 nucleotides or less in length, generally 19-24 nucleotides in length, which region is substantially complementary to at least part of an RNA transcript of an ALAS1 gene, together with a pharmaceutically acceptable carrier. Embodiments of compositions featured in the invention also include an iRNA having an antisense strand having a region of complementarity which is 30 nucleotides or less in length, generally 19-24 nucleotides in length, and is substantially complementary to at least part of an RNA transcript of an ALAS1 gene.

Accordingly, in some aspects, pharmaceutical compositions containing an ALAS1 iRNA and a pharmaceutically acceptable carrier, methods of using the compositions to inhibit expression of an ALAS1 gene, and methods of using the pharmaceutical compositions to treat disorders related to ALAS1 expression are featured in the invention.

For convenience, the meaning of certain terms and phrases used in the specification, examples, and appended claims, are provided below. If there is an apparent discrepancy between the usage of a term in other parts of this specification and its definition provided in this section, the definition in this section shall prevail.

“G,” “A,” “T” and “U” each generally stand for a nucleotide that contains guanine, cytosine, adenine, thymidine and uracil as a base, respectively. However, it will be understood that the term “ribonucleotide” or “nucleotide” can also refer to a modified nucleotide, as further detailed below, or a surrogate replacement moiety. The skilled person is well aware that guanine, cytosine, adenine, and uracil may be replaced by other moieties without substantially altering the base pairing properties of an oligonucleotide comprising a nucleotide bearing such replacement moiety. For example, without limitation, a nucleotide comprising inosine as its base may base pair with nucleotides containing adenine, cytosine, or uracil. Hence, nucleotides containing uracil, guanine, or adenine may be replaced in the nucleotide sequences of dsRNA featured in the invention by a nucleotide containing, for example, inosine. In another example, adenine and cytosine anywhere in the oligonucleotide can be replaced with guanine and uracil, respectively to form G-U Wobble base pairing with the target mRNA. Sequences containing such replacement moieties are suitable for the compositions and methods featured in the invention.

As used herein, “ALAS1” (also known as ALAS-1; δ-aminolevulinate synthase 1; S-ALA synthase 1; 5′-aminolevulinic acid synthase 1; ALAS-H; ALASH; ALAS-N; ALAS3; EC2.3.1.37; 5-aminolevulinate synthase, nonspecific, mitochondrial; ALAS; MIG4; OTTHUMP00000212619; OTTHUMP00000212620; OTTHUMP00000212621; OTTHUMP00000212622; migration-inducing protein 4; EC 2.3.1) refers to a nuclear-encoded mitochondrial enzyme that is the first and typically rate-limiting enzyme in the mammalian heme biosynthetic pathway. ALAS1 catalyzes the condensation of glycine with succinyl-CoA to form δ-aminolevulinic acid (ALA). The human ALAS1 gene is expressed ubiquitously, is found on chromosome 3p21.1 and typically encodes a sequence of 640 amino acids. In contrast, the ALAS-2 gene, which encodes an isozyme, is expressed only in erythrocytes, is found on chromoxome Xp11.21, and typically encodes a sequence of 550 amino acids. As used herein an “ALAS1 protein” means any protein variant of ALAS1 from any species (e.g., human, mouse, non-human primate), as well as any mutants and fragments thereof that retain an ALAS1 activity. Similarly, an “ALAS1 transcript” refers to any transcript variant of ALAS1, from any species (e.g., human, mouse, non-human primate). A sequence of a human ALAS1 variant 1 mRNA transcript can be found at NM_000688.4 (FIG. 3; SEQ ID NO:1). Another version, a human ALAS1 variant 2 mRNA transcript, can be found at NM_000688.5 (FIG. 4; SEQ ID NO:382). The level of the mature encoded ALAS1 protein is regulated by heme: high levels of heme down-regulate the mature enzyme in mitochondria while low heme levels up-regulate. Multiple alternatively spliced variants, encoding the same protein, have been identified.

As used herein, the term “iRNA,” “RNAi”, “iRNA agent,” or “RNAi agent” refers to an agent that contains RNA as that term is defined herein, and which mediates the targeted cleavage of an RNA transcript, e.g., via an RNA-induced silencing complex (RISC) pathway. In one embodiment, an iRNA as described herein effects inhibition of ALAS1 expression. Inhibition of ALAS1 expression may be assessed based on a reduction in the level of ALAS1 mRNA or a reduction in the level of the ALAS1 protein. As used herein, “target sequence” refers to a contiguous portion of the nucleotide sequence of an mRNA molecule formed during the transcription of an ALAS1 gene, including mRNA that is a product of RNA processing of a primary transcription product. The target portion of the sequence will be at least long enough to serve as a substrate for iRNA-directed cleavage at or near that portion. For example, the target sequence will generally be from 9-36 nucleotides in length, e.g., 15-30 nucleotides in length, including all sub-ranges therebetween. As non-limiting examples, the target sequence can be from 15-30 nucleotides, 15-26 nucleotides, 15-23 nucleotides, 15-22 nucleotides, 15-21 nucleotides, 15-20 nucleotides, 15-19 nucleotides, 15-18 nucleotides, 15-17 nucleotides, 18-30 nucleotides, 18-26 nucleotides, 18-23 nucleotides, 18-22 nucleotides, 18-21 nucleotides, 18-20 nucleotides, 19-30 nucleotides, 19-26 nucleotides, 19-23 nucleotides, 19-22 nucleotides, 19-21 nucleotides, 19-20 nucleotides, 20-30 nucleotides, 20-26 nucleotides, 20-25 nucleotides, 20-24 nucleotides, 20-23 nucleotides, 20-22 nucleotides, 20-21 nucleotides, 21-30 nucleotides, 21-26 nucleotides, 21-25 nucleotides, 21-24 nucleotides, 21-23 nucleotides, or 21-22 nucleotides.

As used herein, the term “strand comprising a sequence” refers to an oligonucleotide comprising a chain of nucleotides that is described by the sequence referred to using the standard nucleotide nomenclature.

As used herein, and unless otherwise indicated, the term “complementary,” when used to describe a first nucleotide sequence in relation to a second nucleotide sequence, refers to the ability of an oligonucleotide or polynucleotide comprising the first nucleotide sequence to hybridize and form a duplex structure under certain conditions with an oligonucleotide or polynucleotide comprising the second nucleotide sequence, as will be understood by the skilled person. Such conditions can, for example, be stringent conditions, where stringent conditions may include: 400 mM NaCl, 40 mM PIPES pH 6.4, 1 mM EDTA, 50° C. or 70° C. for 12-16 hours followed by washing. Other conditions, such as physiologically relevant conditions as may be encountered inside an organism, can apply. The skilled person will be able to determine the set of conditions most appropriate for a test of complementarity of two sequences in accordance with the ultimate application of the hybridized nucleotides.

Complementary sequences within an iRNA, e.g., within a dsRNA as described herein, include base-pairing of the oligonucleotide or polynucleotide comprising a first nucleotide sequence to an oligonucleotide or polynucleotide comprising a second nucleotide sequence over the entire length of one or both nucleotide sequences. Such sequences can be referred to as “fully complementary” with respect to each other herein. However, where a first sequence is referred to as “substantially complementary” with respect to a second sequence herein, the two sequences can be fully complementary, or they may form one or more, but generally not more than 5, 4, 3 or 2 mismatched base pairs upon hybridization for a duplex up to 30 base pairs, while retaining the ability to hybridize under the conditions most relevant to their ultimate application, e.g., inhibition of gene expression via a RISC pathway. However, where two oligonucleotides are designed to form, upon hybridization, one or more single stranded overhangs, such overhangs shall not be regarded as mismatches with regard to the determination of complementarity. For example, a dsRNA comprising one oligonucleotide 21 nucleotides in length and another oligonucleotide 23 nucleotides in length, wherein the longer oligonucleotide comprises a sequence of 21 nucleotides that is fully complementary to the shorter oligonucleotide, may yet be referred to as “fully complementary” for the purposes described herein.

“Complementary” sequences, as used herein, may also include, or be formed entirely from, non-Watson-Crick base pairs and/or base pairs formed from non-natural and modified nucleotides, in as far as the above requirements with respect to their ability to hybridize are fulfilled. Such non-Watson-Crick base pairs includes, but are not limited to, G:U Wobble or Hoogstein base pairing.

The terms “complementary,” “fully complementary” and “substantially complementary” herein may be used with respect to the base matching between the sense strand and the antisense strand of a dsRNA, or between the antisense strand of an iRNA agent and a target sequence, as will be understood from the context of their use.

As used herein, a polynucleotide that is “substantially complementary to at least part of” a messenger RNA (mRNA) refers to a polynucleotide that is substantially complementary to a contiguous portion of the mRNA of interest (e.g., an mRNA encoding an ALAS1 protein). For example, a polynucleotide is complementary to at least a part of an ALAS1 mRNA if the sequence is substantially complementary to a non-interrupted portion of an mRNA encoding ALAS1. As another example, a polynucleotide is complementary to at least a part of an ALAS1 mRNA if the sequence is substantially complementary to a non-interrupted portion of an mRNA encoding ALAS1.

The term “double-stranded RNA” or “dsRNA,” as used herein, refers to an iRNA that includes an RNA molecule or complex of molecules having a hybridized duplex region that comprises two anti-parallel and substantially complementary nucleic acid strands, which will be referred to as having “sense” and “antisense” orientations with respect to a target RNA. The duplex region can be of any length that permits specific degradation of a desired target RNA, e.g., through a RISC pathway, but will typically range from 9 to 36 base pairs in length, e.g., 15-30 base pairs in length. Considering a duplex between 9 and 36 base pairs, the duplex can be any length in this range, for example, 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, or 36 and any sub-range therein between, including, but not limited to 15-30 base pairs, 15-26 base pairs, 15-23 base pairs, 15-22 base pairs, 15-21 base pairs, 15-20 base pairs, 15-19 base pairs, 15-18 base pairs, 15-17 base pairs, 18-30 base pairs, 18-26 base pairs, 18-23 base pairs, 18-22 base pairs, 18-21 base pairs, 18-20 base pairs, 19-30 base pairs, 19-26 base pairs, 19-23 base pairs, 19-22 base pairs, 19-21 base pairs, 19-20 base pairs, 20-30 base pairs, 20-26 base pairs, 20-25 base pairs, 20-24 base pairs, 20-23 base pairs, 20-22 base pairs, 20-21 base pairs, 21-30 base pairs, 21-26 base pairs, 21-25 base pairs, 21-24 base pairs, 21-23 base pairs, or 21-22 base pairs. dsRNAs generated in the cell by processing with Dicer and similar enzymes are generally in the range of 19-22 base pairs in length. One strand of the duplex region of a dsDNA comprises a sequence that is substantially complementary to a region of a target RNA. The two strands forming the duplex structure can be from a single RNA molecule having at least one self-complementary region, or can be formed from two or more separate RNA molecules. Where the duplex region is formed from two strands of a single molecule, the molecule can have a duplex region separated by a single stranded chain of nucleotides (herein referred to as a “hairpin loop”) between the 3′-end of one strand and the 5′-end of the respective other strand forming the duplex structure. The hairpin loop can comprise at least one unpaired nucleotide; in some embodiments the hairpin loop can comprise at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 20, at least 23 or more unpaired nucleotides. Where the two substantially complementary strands of a dsRNA are comprised by separate RNA molecules, those molecules need not, but can be covalently connected. Where the two strands are connected covalently by means other than a hairpin loop, the connecting structure is referred to as a “linker.” The term “siRNA” is also used herein to refer to a dsRNA as described above.

In another embodiment, the iRNA agent may be a “single-stranded siRNA” that is introduced into a cell or organism to inhibit a target mRNA. Single-stranded RNAi agents bind to the RISC endonuclease Argonaute 2, which then cleaves the target mRNA. The single-stranded siRNAs are generally 15-30 nucleotides and are chemically modified. The design and testing of single-stranded siRNAs are described in U.S. Pat. No. 8,101,348 and in Lima et al., (2012) Cell 150: 883-894, the entire contents of each of which are hereby incorporated herein by reference. Any of the antisense nucleotide sequences described herein (e.g., sequences provided in Tables 2, 3, 6, 7, 8, 9, 14, and 15) may be used as a single-stranded siRNA as described herein or as chemically modified by the methods described in Lima et al., (2012) Cell 150;:883-894.

In another aspect, the RNA agent is a “single-stranded antisense RNA molecule”. An single-stranded antisense RNA molecule is complementary to a sequence within the target mRNA. Single-stranded antisense RNA molecules can inhibit translation in a stoichiometric manner by base pairing to the mRNA and physically obstructing the translation machinery, see Dias, N. et al., (2002) Mol Cancer Ther 1:347-355. Alternatively, the single-stranded antisense molecules inhibit a target mRNA by hydridizing to the target and cleaving the target through an RNaseH cleavage event. The single-stranded antisense RNA molecule may be about 10 to about 30 nucleotides in length and have a sequence that is complementary to a target sequence. For example, the single-stranded antisense RNA molecule may comprise a sequence that is at least about 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, or more contiguous nucleotides from any one of the antisense nucleotide sequences described herein, e.g., sequences provided in any one of Tables 2, 3, 6, 7, 8, 9, 14, and 15.

The skilled artisan will recognize that the term “RNA molecule” or “ribonucleic acid molecule” encompasses not only RNA molecules as expressed or found in nature, but also analogs and derivatives of RNA comprising one or more ribonucleotide/ribonucleoside analogs or derivatives as described herein or as known in the art. Strictly speaking, a “ribonucleoside” includes a nucleoside base and a ribose sugar, and a “ribonucleotide” is a ribonucleoside with one, two or three phosphate moieties. However, the terms “ribonucleoside” and “ribonucleotide” can be considered to be equivalent as used herein. The RNA can be modified in the nucleobase structure or in the ribose-phosphate backbone structure, e.g., as described herein below.

However, the molecules comprising ribonucleoside analogs or derivatives must retain the ability to form a duplex. As non-limiting examples, an RNA molecule can also include at least one modified ribonucleoside including but not limited to a 2′-O-methyl modified nucleostide, a nucleoside comprising a 5′ phosphorothioate group, a terminal nucleoside linked to a cholesteryl derivative or dodecanoic acid bisdecylamide group, a locked nucleoside, an abasic nucleoside, a 2′-deoxy-2′-fluoro modified nucleoside, a 2′-amino-modified nucleoside, 2′-alkyl-modified nucleoside, morpholino nucleoside, a phosphoramidate or a non-natural base comprising nucleoside, or any combination thereof. Alternatively, an RNA molecule can comprise at least two modified ribonucleosides, at least 3, at least 4, at least 5, at least 6, at least 7, at least 8, at least 9, at least 10, at least 15, at least 20 or more, up to the entire length of the dsRNA molecule. The modifications need not be the same for each of such a plurality of modified ribonucleosides in an RNA molecule. In one embodiment, modified RNAs contemplated for use in methods and compositions described herein are peptide nucleic acids (PNAs) that have the ability to form the required duplex structure and that permit or mediate the specific degradation of a target RNA, e.g., via a RISC pathway.

In one aspect, a modified ribonucleoside includes a deoxyribonucleoside. In such an instance, an iRNA agent can comprise one or more deoxynucleosides, including, for example, a deoxynucleoside overhang(s), or one or more deoxynucleosides within the double stranded portion of a dsRNA. However, it is self evident that under no circ*mstances is a double stranded DNA molecule encompassed by the term “iRNA.”

In one aspect, an RNA interference agent includes a single stranded RNA that interacts with a target RNA sequence to direct the cleavage of the target RNA. Without wishing to be bound by theory, long double stranded RNA introduced into cells is broken down into siRNA by a Type III endonuclease known as Dicer (Sharp et al., Genes Dev. 2001, 15:485). Dicer, a ribonuclease-III-like enzyme, processes the dsRNA into 19-23 base pair short interfering RNAs with characteristic two base 3′ overhangs (Bernstein, et al., (2001) Nature 409:363). The siRNAs are then incorporated into an RNA-induced silencing complex (RISC) where one or more helicases unwind the siRNA duplex, enabling the complementary antisense strand to guide target recognition (Nykanen, et al., (2001) Cell 107:309). Upon binding to the appropriate target mRNA, one or more endonucleases within the RISC cleaves the target to induce silencing (Elbashir, et al., (2001) Genes Dev. 15:188). Thus, in one aspect the invention relates to a single stranded RNA that promotes the formation of a RISC complex to effect silencing of the target gene.

As used herein, the term “nucleotide overhang” refers to at least one unpaired nucleotide that protrudes from the duplex structure of an iRNA, e.g., a dsRNA. For example, when a 3′-end of one strand of a dsRNA extends beyond the 5′-end of the other strand, or vice versa, there is a nucleotide overhang. A dsRNA can comprise an overhang of at least one nucleotide; alternatively the overhang can comprise at least two nucleotides, at least three nucleotides, at least four nucleotides, at least five nucleotides or more. A nucleotide overhang can comprise or consist of a nucleotide/nucleoside analog, including a deoxynucleotide/nucleoside. The overhang(s) may be on the sense strand, the antisense strand or any combination thereof. Furthermore, the nucleotide(s) of an overhang can be present on the 5′ end, 3′ end or both ends of either an antisense or sense strand of a dsRNA.

In one embodiment, the antisense strand of a dsRNA has a 1-10 nucleotide overhang at the 3′ end and/or the 5′ end. In one embodiment, the sense strand of a dsRNA has a 1-10 nucleotide overhang at the 3′ end and/or the 5′ end. In another embodiment, one or more of the nucleotides in the overhang is replaced with a nucleoside thiophosphate.

The terms “blunt” or “blunt ended” as used herein in reference to a dsRNA mean that there are no unpaired nucleotides or nucleotide analogs at a given terminal end of a dsRNA, i.e., no nucleotide overhang. One or both ends of a dsRNA can be blunt. Where both ends of a dsRNA are blunt, the dsRNA is said to be blunt ended. To be clear, a “blunt ended” dsRNA is a dsRNA that is blunt at both ends, i.e., no nucleotide overhang at either end of the molecule. Most often such a molecule will be double-stranded over its entire length.

The term “antisense strand” or “guide strand” refers to the strand of an iRNA, e.g., a dsRNA, which includes a region that is substantially complementary to a target sequence. As used herein, the term “region of complementarity” refers to the region on the antisense strand that is substantially complementary to a sequence, for example a target sequence, as defined herein. Where the region of complementarity is not fully complementary to the target sequence, the mismatches may be in the internal or terminal regions of the molecule. Generally, the most tolerated mismatches are in the terminal regions, e.g., within 5, 4, 3, or 2 nucleotides of the 5′ and/or 3′ terminus.

The term “sense strand,” or “passenger strand” as used herein, refers to the strand of an iRNA that includes a region that is substantially complementary to a region of the antisense strand as that term is defined herein.

As used herein, the term “SNALP” refers to a stable nucleic acid-lipid particle. A SNALP represents a vesicle of lipids coating a reduced aqueous interior comprising a nucleic acid such as an iRNA or a plasmid from which an iRNA is transcribed. SNALPs are described, e.g., in U.S. Patent Application Publication Nos. 20060240093, 20070135372, and in International Application No. WO 2009082817. These applications are incorporated herein by reference in their entirety.

“Introducing into a cell,” when referring to an iRNA, means facilitating or effecting uptake or absorption into the cell, as is understood by those skilled in the art. Absorption or uptake of an iRNA can occur through unaided diffusive or active cellular processes, or by auxiliary agents or devices. The meaning of this term is not limited to cells in vitro; an iRNA may also be “introduced into a cell,” wherein the cell is part of a living organism. In such an instance, introduction into the cell will include the delivery to the organism. For example, for in vivo delivery, iRNA can be injected into a tissue site or administered systemically. In vivo delivery can also be by a β-glucan delivery system, such as those described in U.S. Pat. Nos. 5,032,401 and 5,607,677, and U.S. Publication No. 2005/0281781, which are hereby incorporated by reference in their entirety. In vitro introduction into a cell includes methods known in the art such as electroporation and lipofection. Further approaches are described herein below or known in the art.

As used herein, the term “modulate the expression of,” refers to at an least partial “inhibition” or partial “activation” of an ALAS1 gene expression in a cell treated with an iRNA composition as described herein compared to the expression of ALAS1 in a control cell. A control cell includes an untreated cell, or a cell treated with a non-targeting control iRNA.

The terms “activate,” “enhance,” “up-regulate the expression of,” “increase the expression of,” and the like, in so far as they refer to an ALAS1 gene, herein refer to the at least partial activation of the expression of an ALAS1 gene, as manifested by an increase in the amount of ALAS1 mRNA, which may be isolated from or detected in a first cell or group of cells in which an ALAS1 gene is transcribed and which has or have been treated such that the expression of an ALAS1 gene is increased, as compared to a second cell or group of cells substantially identical to the first cell or group of cells but which has or have not been so treated (control cells).

In one embodiment, expression of an ALAS1 gene is activated by at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% by administration of an iRNA as described herein. In some embodiments, an ALAS1 gene is activated by at least about 60%, 70%, or 80% by administration of an iRNA featured in the invention. In some embodiments, expression of an ALAS1 gene is activated by at least about 85%, 90%, or 95% or more by administration of an iRNA as described herein. In some embodiments, the ALAS1 gene expression is increased by at least 1-fold, at least 2-fold, at least 5-fold, at least 10-fold, at least 50-fold, at least 100-fold, at least 500-fold, at least 1000 fold or more in cells treated with an iRNA as described herein compared to the expression in an untreated cell. Activation of expression by small dsRNAs is described, for example, in Li et al., 2006 Proc. Natl. Acad. Sci. U.S.A. 103:17337-42, and in US20070111963 and US2005226848, each of which is incorporated herein by reference.

The terms “silence,” “inhibit expression of,” “down-regulate expression of,” “suppress expression of,” and the like, in so far as they refer to an ALAS1 gene, herein refer to the at least partial suppression of the expression of an ALAS1 gene, as assessed, e.g., based on on ALAS1 mRNA expression, ALAS1 protein expression, or another parameter functionally linked to ALAS1 gene expression (e.g., ALA or PBG concentrations in plasma or urine). For example, inhibition of ALAS1 expression may be manifested by a reduction of the amount of ALAS1 mRNA which may be isolated from or detected in a first cell or group of cells in which an ALAS1 gene is transcribed and which has or have been treated such that the expression of an ALAS1 gene is inhibited, as compared to a control. The control may be a second cell or group of cells substantially identical to the first cell or group of cells, except that the second cell or group of cells have not been so treated (control cells). The degree of inhibition is usually expressed as a percentage of a control level, e.g.,

( mRNA in control cells ) - ( mRNA in treated cells ) ( mRNA in control cells ) · 100 %

Alternatively, the degree of inhibition may be given in terms of a reduction of a parameter that is functionally linked to ALAS1 gene expression, e.g., the amount of protein encoded by an ALAS1 gene, or the level of one or more porphyrins. The reduction of a parameter functionally linked to ALAS1 gene expression may similarly be expressed as a percentage of a control level. In principle, ALAS1 gene silencing may be determined in any cell expressing ALAS1, either constitutively or by genomic engineering, and by any appropriate assay. However, when a reference is needed in order to determine whether a given iRNA inhibits the expression of the ALAS1 gene by a certain degree and therefore is encompassed by the instant invention, the assays provided in the Examples below shall serve as such reference.

For example, in certain instances, expression of an ALAS1 gene is suppressed by at least about 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, or 50% by administration of an iRNA featured in the invention. In some embodiments, an ALAS1 gene is suppressed by at least about 60%, 65%, 70%, 75%, or 80% by administration of an iRNA featured in the invention. In some embodiments, an ALAS1 gene is suppressed by at least about 85%, 90%, 95%, 98%, 99%, or more by administration of an iRNA as described herein.

As used herein in the context of ALAS1 expression, the terms “treat,” “treating,” “treatment,” and the like, refer to relief from or alleviation of pathological processes related to ALAS1 expression (e.g., pathological processes involving porphyrins or defects in the porphyrin pathway, such as, for example, porphyrias). In the context of the present invention insofar as it relates to any of the other conditions recited herein below (other than pathological processes related to ALAS1 expression), the terms “treat,” “treatment,” and the like mean to prevent, relieve or alleviate at least one symptom associated with such condition, or to slow or reverse the progression or anticipated progression of such condition. For example, the methods featured herein, when employed to treat porphyria, may serve to reduce or prevent one or more symptoms associated with porphyria (e.g., pain), to reduce the severity or frequency of attacks associated with porphyria, to reduce the likelihood that an attack of one or more symptoms associated with porphyria will occur upon exposure to a precipitating condition, to shorten an attack associated with porphyria, and/or to reduce the risk of developing conditions associated with porphyria (e.g., hepatocellular cancer or neuropathy (e.g., progressive neuropathy),). Thus, unless the context clearly indicates otherwise, the terms “treat,” “treatment,” and the like are intended to encompass prophylaxis, e.g., prevention of disorders and/or symptoms of disorders related to ALAS1 expression.

By “lower” in the context of a disease marker or symptom is meant a statistically or clinically significant decrease in such level. The decrease can be, for example, at least 10%, at least 20%, at least 30%, at least 40% or more, and is typically down to a level accepted as within the range of normal for an individual without such disorder.

As used herein, the phrases “therapeutically effective amount” and “prophylactically effective amount” refer to an amount that provides a therapeutic benefit in the treatment, prevention, or management of pathological processes related to ALAS1 expression. The specific amount that is therapeutically effective can be readily determined by an ordinary medical practitioner, and may vary depending on factors known in the art, such as, for example, the type of pathological process, the patient's history and age, the stage of pathological process, and the administration of other agents.

As used herein, a “pharmaceutical composition” comprises a pharmacologically effective amount of an iRNA and a pharmaceutically acceptable carrier. As used herein, “pharmacologically effective amount,” “therapeutically effective amount” or simply “effective amount” refers to that amount of an iRNA effective to produce the intended pharmacological, therapeutic or preventive result. For example, in a method of treating a disorder related to ALAS1 expression (e.g., in a method of treating a porphyria), an effective amount includes an amount effective to reduce one or more symptoms associated with a porphyria, an amount effective to reduce the frequency of attacks, an amount effective to reduce the likelihood that an attack of one or more symptoms associated with porphyria will occur upon exposure to a precipitating factor, or an amount effective to reduce the risk of developing conditions associated with porphyria (e.g., neuropathy (e.g., progressive neuropathy), hepatocellular cancer). For example, if a given clinical treatment is considered effective when there is at least a 10% reduction in a measurable parameter associated with a disease or disorder, a therapeutically effective amount of a drug for the treatment of that disease or disorder is the amount necessary to effect at least a 10% reduction in that parameter. For example, a therapeutically effective amount of an iRNA targeting ALAS1 can reduce ALAS1 protein levels by any measurable amount, e.g., by at least 10%, 20%, 30%, 40% or 50%.

The term “pharmaceutically acceptable carrier” refers to a carrier for administration of a therapeutic agent. Such carriers include, but are not limited to, saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof. The term specifically excludes cell culture medium. For drugs administered orally, pharmaceutically acceptable carriers include, but are not limited to pharmaceutically acceptable excipients such as inert diluents, disintegrating agents, binding agents, lubricating agents, sweetening agents, flavoring agents, coloring agents and preservatives. Suitable inert diluents include sodium and calcium carbonate, sodium and calcium phosphate, and lactose, while corn starch and alginic acid are suitable disintegrating agents. Binding agents may include starch and gelatin, while the lubricating agent, if present, will generally be magnesium stearate, stearic acid or talc. If desired, the tablets may be coated with a material such as glyceryl monostearate or glyceryl distearate, to delay absorption in the gastrointestinal tract. Agents included in drug formulations are described further herein below.

The term “about” when referring to a number or a numerical range means that the number or numerical range referred to is an approximation within experimental variability (or within statistical experimental error), and thus the number or numerical range may vary from, for example, between 1% and 15% of the stated number or numerical range.

II. Double-Stranded Ribonucleic Acid (dsRNA)

Described herein are iRNA agents that inhibit the expression of an ALAS1gene. In one embodiment, the iRNA agent includes double-stranded ribonucleic acid (dsRNA) molecules for inhibiting the expression of an ALAS1 gene in a cell or in a subject (e.g., in a mammal, e.g., in a human having a porphyria), where the dsRNA includes an antisense strand having a region of complementarity which is complementary to at least a part of an mRNA formed in the expression of an ALAS1gene, and where the region of complementarity is 30 nucleotides or less in length, generally 19-24 nucleotides in length, and where the dsRNA, upon contact with a cell expressing the ALAS1 gene, inhibits the expression of the ALAS1 gene by at least 10% as assayed by, for example, a PCR or branched DNA (bDNA)-based method, or by a protein-based method, such as by Western blot. In one embodiment, the iRNA agent activates the expression of an ALAS1 gene in a cell or mammal. Expression of an ALAS1 gene in cell culture, such as in COS cells, HeLa cells, primary hepatocytes, HepG2 cells, primary cultured cells or in a biological sample from a subject can be assayed by measuring ALAS1 mRNA levels, such as by bDNA or TaqMan assay, or by measuring protein levels, such as by immunofluorescence analysis, using, for example, Western Blotting or flow cytometric techniques.

A dsRNA includes two RNA strands that are sufficiently complementary to hybridize to form a duplex structure under conditions in which the dsRNA will be used. One strand of a dsRNA (the antisense strand) includes a region of complementarity that is substantially complementary, and generally fully complementary, to a target sequence, derived from the sequence of an mRNA formed during the expression of an ALAS1 gene. The other strand (the sense strand) includes a region that is complementary to the antisense strand, such that the two strands hybridize and form a duplex structure when combined under suitable conditions. Generally, the duplex structure is between 15 and 30 inclusive, more generally between 18 and 25 inclusive, yet more generally between 19 and 24 inclusive, and most generally between 19 and 21 base pairs in length, inclusive. Similarly, the region of complementarity to the target sequence is between 15 and 30 inclusive, more generally between 18 and 25 inclusive, yet more generally between 19 and 24 inclusive, and most generally between 19 and 21 nucleotides in length, inclusive. In some embodiments, the dsRNA is between 15 and 20 nucleotides in length, inclusive, and in other embodiments, the dsRNA is between 25 and 30 nucleotides in length, inclusive. As the ordinarily skilled person will recognize, the targeted region of an RNA targeted for cleavage will most often be part of a larger RNA molecule, often an mRNA molecule. Where relevant, a “part” of an mRNA target is a contiguous sequence of an mRNA target of sufficient length to be a substrate for RNAi-directed cleavage (i.e., cleavage through a RISC pathway). dsRNAs having duplexes as short as 9 base pairs can, under some circ*mstances, mediate RNAi-directed RNA cleavage. Most often a target will be at least 15 nucleotides in length, e.g., 15-30 nucleotides in length.

One of skill in the art will also recognize that the duplex region is a primary functional portion of a dsRNA, e.g., a duplex region of 9 to 36, e.g., 15-30 base pairs. Thus, in one embodiment, to the extent that it becomes processed to a functional duplex of e.g., 15-30 base pairs that targets a desired RNA for cleavage, an RNA molecule or complex of RNA molecules having a duplex region greater than 30 base pairs is a dsRNA. Thus, an ordinarily skilled artisan will recognize that in one embodiment, then, an miRNA is a dsRNA. In another embodiment, a dsRNA is not a naturally occurring miRNA. In another embodiment, an iRNA agent useful to target ALAS1 expression is not generated in the target cell by cleavage of a larger dsRNA.

A dsRNA as described herein may further include one or more single-stranded nucleotide overhangs. The dsRNA can be synthesized by standard methods known in the art as further discussed below, e.g., by use of an automated DNA synthesizer, such as are commercially available from, for example, Biosearch, Applied Biosystems, Inc. In one embodiment, an ALAS1 gene is a human ALAS1 gene. In another embodiment the ALAS1 gene is a mouse or a rat ALAS1 gene. In specific embodiments, the first sequence is a sense strand of a dsRNA that includes a sense sequence from Table 2 or Table 3, and the second sequence is an antisense strand of a dsRNA that includes an antisense sequence from Table 2 or Table 3. In embodiments, the first sequence is a sense strand of a dsRNA that includes a sense sequence from Table 2, 3, 6, 7, 8, 9, 14, or 15, and the second sequence is an antisense strand of a dsRNA that includes an antisense sequence from Table 2, 3, 6, 7, 8, 9, 14, or 15. Alternative dsRNA agents that target sequences other than those of the dsRNAs of Table 2 or Table 3 can readily be determined using the target sequence and the flanking ALAS1 sequence.

In one aspect, a dsRNA will include at least sense and antisense nucleotide sequences, whereby the sense strand is selected from the groups of sequences provided in Tables 2 and 3, and the corresponding antisense strand of the sense strand is selected from Tables 2 and 3. In a further aspect, a dsRNA will include at least sense and antisense nucleotide sequences, whereby the sense strand is selected from the groups of sequences provided in Tables 2, 3, 6, 7, 8, 9, 14, and 15, and the corresponding antisense strand of the sense strand is selected from Tables 2, 3, 6, 7, 8, 9, 14, and 15. In these aspects, one of the two sequences is complementary to the other of the two sequences, with one of the sequences being substantially complementary to a sequence of an mRNA generated by the expression of an ALAS1 gene gene. As such, a dsRNA will include two oligonucleotides, where one oligonucleotide is described as the sense strand in Table 2, 3, 6, 7, 8, 9, 14, or 15, and the second oligonucleotide is described as the corresponding antisense strand of the sense strand from 2, 3, 6, 7, 8, 9, 14, or 15. As described elsewhere herein and as known in the art, the complementary sequences of a dsRNA can also be contained as self-complementary regions of a single nucleic acid molecule, as opposed to being on separate oligonucleotides.

The skilled person is well aware that dsRNAs having a duplex structure of between 20 and 23, but specifically 21, base pairs have been hailed as particularly effective in inducing RNA interference (Elbashir et al., EMBO 2001, 20:6877-6888). However, others have found that shorter or longer RNA duplex structures can be effective as well. In the embodiments described above, by virtue of the nature of the oligonucleotide sequences provided in Tables 2, 3, 6, 7, 8, 9, 14, and 15, dsRNAs described herein can include at least one strand of a length of minimally 21 nucleotides. It can be reasonably expected that shorter duplexes having one of the sequences of Table 2, 3, 6, 7, 8, 9, 14, or 15 minus only a few nucleotides on one or both ends may be similarly effective as compared to the dsRNAs described above. Hence, dsRNAs having a partial sequence of at least 15, 16, 17, 18, 19, 20, or more contiguous nucleotides from one of the sequences of Table 2, 3, 6, 7, 8, 9, 14, or 15, and differing in their ability to inhibit the expression of an ALAS1gene by not more than 5, 10, 15, 20, 25, or 30% inhibition from a dsRNA comprising the full sequence, are contemplated according to the invention.

In addition, the RNAs provided in Tables 2 and 3, as well as the RNAs provided in Tables 2, 3, 6, 7, 8, 9, 14, and 15, identify a site in an ALAS1 transcript that is susceptible to RISC-mediated cleavage. As such, the present invention further features iRNAs that target within one of such sequences. As used herein, an iRNA is said to target within a particular site of an RNA transcript if the iRNA promotes cleavage of the transcript anywhere within that particular site. Such an iRNA will generally include at least 15 contiguous nucleotides from one of the sequences provided in Tables 2, 3, 6, 7, 8, 9, 14, and 15 coupled to additional nucleotide sequences taken from the region contiguous to the selected sequence in an ALAS1gene.

While a target sequence is generally 15-30 nucleotides in length, there is wide variation in the suitability of particular sequences in this range for directing cleavage of any given target RNA. Various software packages and the guidelines set out herein provide guidance for the identification of optimal target sequences for any given gene target, but an empirical approach can also be taken in which a “window” or “mask” of a given size (as a non-limiting example, 21 nucleotides) is literally or figuratively (including, e.g., in silico) placed on the target RNA sequence to identify sequences in the size range that may serve as target sequences. By moving the sequence “window” progressively one nucleotide upstream or downstream of an initial target sequence location, the next potential target sequence can be identified, until the complete set of possible sequences is identified for any given target size selected. This process, coupled with systematic synthesis and testing of the identified sequences (using assays as described herein or as known in the art) to identify those sequences that perform optimally can identify those RNA sequences that, when targeted with an iRNA agent, mediate the best inhibition of target gene expression. Thus, while the sequences identified, for example, in Tables 2, 3, 6, 7, 8, 9, 14, and 15, represent effective target sequences, it is contemplated that further optimization of inhibition efficiency can be achieved by progressively “walking the window” one nucleotide upstream or downstream of the given sequences to identify sequences with equal or better inhibition characteristics.

Further, it is contemplated that for any sequence identified, e.g., in Tables 2, 3, 6, 7, 8, 9, 14, and 15, further optimization can be achieved by systematically either adding or removing nucleotides to generate longer or shorter sequences and testing those and sequences generated by walking a window of the longer or shorter size up or down the target RNA from that point. Again, coupling this approach to generating new candidate targets with testing for effectiveness of iRNAs based on those target sequences in an inhibition assay as known in the art or as described herein can lead to further improvements in the efficiency of inhibition. Further still, such optimized sequences can be adjusted by, e.g., the introduction of modified nucleotides as described herein or as known in the art, addition or changes in overhang, or other modifications as known in the art and/or discussed herein to further optimize the molecule (e.g., increasing serum stability or circulating half-life, increasing thermal stability, enhancing transmembrane delivery, targeting to a particular location or cell type, increasing interaction with silencing pathway enzymes, increasing release from endosomes, etc.) as an expression inhibitor.

An iRNA as described herein can contain one or more mismatches to the target sequence. In one embodiment, an iRNA as described herein contains no more than 3 mismatches. If the antisense strand of the iRNA contains mismatches to a target sequence, it is preferable that the area of mismatch not be located in the center of the region of complementarity. If the antisense strand of the iRNA contains mismatches to the target sequence, it is preferable that the mismatch be restricted to be within the last 5 nucleotides from either the 5′ or 3′ end of the region of complementarity. For example, for a 23 nucleotide iRNA agent RNA strand which is complementary to a region of an ALAS1 gene, the RNA strand generally does not contain any mismatch within the central 13 nucleotides. The methods described herein or methods known in the art can be used to determine whether an iRNA containing a mismatch to a target sequence is effective in inhibiting the expression of an ALAS1 gene. Consideration of the efficacy of iRNAs with mismatches in inhibiting expression of an ALAS1 gene is important, especially if the particular region of complementarity in an ALAS1 gene is known to have polymorphic sequence variation within the population.

In one embodiment, at least one end of a dsRNA has a single-stranded nucleotide overhang of 1 to 4, generally 1 or 2 nucleotides. dsRNAs having at least one nucleotide overhang have unexpectedly superior inhibitory properties relative to their blunt-ended counterparts. In yet another embodiment, the RNA of an iRNA, e.g., a dsRNA, is chemically modified to enhance stability or other beneficial characteristics. The nucleic acids featured in the invention may be synthesized and/or modified by methods well established in the art, such as those described in “Current protocols in nucleic acid chemistry,” Beaucage, S. L. et al. (Edrs.), John Wiley & Sons, Inc., New York, NY, USA, which is hereby incorporated herein by reference. Modifications include, for example, (a) end modifications, e.g., 5′ end modifications (phosphorylation, conjugation, inverted linkages, etc.) 3′ end modifications (conjugation, DNA nucleotides, inverted linkages, etc.), (b) base modifications, e.g., replacement with stabilizing bases, destabilizing bases, or bases that base pair with an expanded repertoire of partners, removal of bases (abasic nucleotides), or conjugated bases, (c) sugar modifications (e.g., at the 2′ position or 4′ position) or replacement of the sugar, as well as (d) backbone modifications, including modification or replacement of the phosphodiester linkages. Specific examples of RNA compounds useful in this invention include, but are not limited to RNAs containing modified backbones or no natural internucleoside linkages. RNAs having modified backbones include, among others, those that do not have a phosphorus atom in the backbone. For the purposes of this specification, and as sometimes referenced in the art, modified RNAs that do not have a phosphorus atom in their internucleoside backbone can also be considered to be oligonucleosides. In particular embodiments, the modified RNA will have a phosphorus atom in its internucleoside backbone.

Modified RNA backbones include, for example, phosphorothioates, chiral phosphorothioates, phosphorodithioates, phosphotriesters, aminoalkylphosphotriesters, methyl and other alkyl phosphonates including 3′-alkylene phosphonates and chiral phosphonates, phosphinates, phosphoramidates including 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′. Various salts, mixed salts and free acid forms are also included.

Representative U.S. patents that teach the preparation of the above phosphorus-containing linkages include, but are not limited to, U.S. Pat. Nos. 3,687,808; 4,469,863; 4,476,301; 5,023,243; 5,177,195; 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; 5,541,316; 5,550,111; 5,563,253; 5,571,799; 5,587,361; 5,625,050; 6,028,188; 6,124,445; 6,160,109; 6,169,170; 6,172,209; 6,239,265; 6,277,603; 6,326,199; 6,346,614; 6,444,423; 6,531,590; 6,534,639; 6,608,035; 6,683,167; 6,858,715; 6,867,294; 6,878,805; 7,015,315; 7,041,816; 7,273,933; 7,321,029; and U.S. Pat. RE39464, each of which is herein incorporated by reference.

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

Representative U.S. patents that teach the preparation of the above oligonucleosides include, but are not limited to, U.S. Pat. Nos. 5,034,506; 5,166,315; 5,185,444; 5,214,134; 5,216,141; 5,235,033; 5,64,562; 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; 5,608,046; 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.

In other RNA mimetics suitable or contemplated for use in iRNAs, both the sugar and the 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 RNA 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 RNA is replaced with an amide containing backbone, in particular an aminoethylglycine backbone. The nucleobases are retained and are bound directly or indirectly to aza nitrogen atoms of the amide portion of the backbone. Representative U.S. 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, for example, in Nielsen et al., Science, 1991, 254, 1497-1500.

Some embodiments featured in the invention include RNAs with phosphorothioate backbones and oligonucleosides with heteroatom backbones, and in particular —CH2—NH—CH2—, —CH2—N(CH3)—O—CH2—[known as a methylene (methylimino) or MMI backbone], —CH2—O—N(CH3)—CH2—, —CH2—N(CH3)—N(CH3)—CH2— and —N(CH3)—CH2—CH2—[wherein the native phosphodiester backbone is represented as —O—P—O—CH2—] of the above-referenced U.S. Pat. No. 5,489,677, and the amide backbones of the above-referenced U.S. Pat. No. 5,602,240. In some embodiments, the RNAs featured herein have morpholino backbone structures of the above-referenced U.S. Pat. No. 5,034,506.

Modified RNAs may also contain one or more substituted sugar moieties. The iRNAs, e.g., dsRNAs, featured herein can include one of the following at the 2′ position: OH; F; O—, δ—, or N-alkyl; O—, S—, or N-alkenyl; O—, S— or N-alkynyl; or O-alkyl-O-alkyl, wherein the alkyl, alkenyl and alkynyl may be substituted or unsubstituted C1 to C10 alkyl or C2 to C10 alkenyl and alkynyl. Exemplary suitable modifications include O[(CH2)nO]mCH3, O(CH2nOCH3, O(CH2)nNH2, O(CH2)nCH3, O(CH2), ONH2, and O(CH2)nON[(CH2)nCH3)]2, where n and m are from 1 to about 10. In other embodiments, dsRNAs include one of the following at the 2′ position: C1 to C10 lower alkyl, substituted lower alkyl, alkaryl, aralkyl, O-alkaryl or O-aralkyl, SH, SCH3, OCN, Cl, Br, CN, CF3, OCF3, SOCH3, SO2CH3, ONO2, NO2, N3, NH2, heterocycloalkyl, heterocycloalkaryl, aminoalkylamino, polyalkylamino, substituted silyl, an RNA cleaving group, a reporter group, an intercalator, a group for improving the pharmaco*kinetic properties of an iRNA, or a group for improving the pharmacodynamic properties of an iRNA, and other substituents having similar properties. In some embodiments, the modification includes a 2′-methoxyethoxy (2′-O—CH2CH2OCH3, also known as 2′-O—(2-methoxyethyl) or 2′-MOE) (Martin et al., Helv. Chim. Acta, 1995, 78:486-504) i.e., an alkoxy-alkoxy group. Another exemplary modification is 2′-dimethylaminooxyethoxy, i.e., a O(CH2)2ON(CH3)2 group, also known as 2′-DMAOE, as described in examples herein below, and 2′-dimethylaminoethoxyethoxy (also known in the art as 2′-O-dimethylaminoethoxyethyl or 2′-DMAEOE), i.e., 2′-O—CH2—O—CH2—N(CH2)2, also described in examples herein below.

Other modifications include 2′-methoxy (2′-OCH3), 2′-aminopropoxy (2′-OCH2CH2CH2NH2) and 2′-fluoro (2′-F). Similar modifications may also be made at other positions on the RNA of an iRNA, particularly the 3′ position of the sugar on the 3′ terminal nucleotide or in 2′-5′ linked dsRNAs and the 5′ position of 5′ terminal nucleotide. iRNAs may also have sugar mimetics such as cyclobutyl moieties in place of the pentofuranosyl sugar. Representative U.S. patents that teach the preparation of such modified sugar structures include, but are not limited to, U.S. Pat. Nos. 4,981,957; 5,118,800; 5,319,080; 5,359,044; 5,393,878; 5,446,137; 5,466,786; 5,514,785; 5,519,134; 5,567,811; 5,576,427; 5,591,722; 5,597,909; 5,610,300; 5,627,053; 5,639,873; 5,646,265; 5,658,873; 5,670,633; and 5,700,920, certain of which are commonly owned with the instant application, and each of which is herein incorporated by reference.

An iRNA may also include nucleobase (often referred to in the art simply as “base”) modifications or substitutions. As used herein, “unmodified” or “natural” nucleobases include the purine bases adenine (A) and guanine (G), and the pyrimidine bases thymine (T), cytosine (C) and uracil (U). Modified nucleobases include other synthetic and natural nucleobases such as 5-methylcytosine (5-me-C), 5-hydroxymethyl cytosine, xanthine, hypoxanthine, 2-aminoadenine, 6-methyl and other alkyl derivatives of adenine and guanine, 2-propyl and other alkyl derivatives of adenine and guanine, 2-thiouracil, 2-thiothymine and 2-thiocytosine, 5-halouracil and cytosine, 5-propynyl uracil and cytosine, 6-azo uracil, cytosine and thymine, 5-uracil (pseudouracil), 4-thiouracil, 8-halo, 8-amino, 8-thiol, 8-thioalkyl, 8-hydroxyl anal other 8-substituted adenines and guanines, 5-halo, particularly 5-bromo, 5-trifluoromethyl and other 5-substituted uracils and cytosines, 7-methylguanine and 7-methyladenine, 8-azaguanine and 8-azaadenine, 7-deazaguanine and 7-daazaadenine and 3-deazaguanine and 3-deazaadenine. Further nucleobases include those disclosed in U.S. Pat. No. 3,687,808, those disclosed in Modified Nucleosides in Biochemistry, Biotechnology and Medicine, Herdewijn, P. ed. Wiley-VCH, 2008; those disclosed in The Concise Encyclopedia Of Polymer Science And Engineering, pages 858-859, Kroschwitz, J. L, ed. John Wiley & Sons, 1990, these disclosed by Englisch et al., Angewandte Chemie, International Edition, 1991, 30, 613, and those disclosed by Sanghvi, Y S., Chapter 15, dsRNA Research and Applications, pages 289-302, Crooke, S. T. and Lebleu, B., Ed., CRC Press, 1993. Certain of these nucleobases are particularly useful for increasing the binding affinity of the oligomeric compounds featured in the invention. These include 5-substituted pyrimidines, 6-azapyrimidines and N-2, N-6 and 0-6 substituted purines, including 2-aminopropyladenine, 5-propynyluracil and 5-propynylcytosine. 5-methylcytosine substitutions have been shown to increase nucleic acid duplex stability by 0.6-1.2° C. (Sanghvi, Y. S., Crooke, S. T. and Lebleu, B., Eds., dsRNA Research and Applications, CRC Press, Boca Raton, 1993, pp. 276-278) and are exemplary base substitutions, even more particularly when combined with 2′-O-methoxyethyl sugar modifications.

Representative U.S. patents that teach the preparation of certain of the above noted modified nucleobases as well as other modified nucleobases include, but are not limited to, the above noted U.S. Pat. No. 3,687,808, as well as U.S. Pat. Nos. 4,845,205; 5,130,30; 5,134,066; 5,175,273; 5,367,066; 5,432,272; 5,457,187; 5,459,255; 5,484,908; 5,502,177; 5,525,711; 5,552,540; 5,587,469; 5,594,121, 5,596,091; 5,614,617; 5,681,941; 6,015,886; 6,147,200; 6,166,197; 6,222,025; 6,235,887; 6,380,368; 6,528,640; 6,639,062; 6,617,438; 7,045,610; 7,427,672; and 7,495,088, each of which is herein incorporated by reference, and U.S. Pat. No. 5,750,692, also herein incorporated by reference.

The RNA of an iRNA can also be modified to include one or more locked nucleic acids (LNA). A locked nucleic acid is a nucleotide having a modified ribose moiety in which the ribose moiety comprises an extra bridge connecting the 2′ and 4′ carbons. This structure effectively “locks” the ribose in the 3′-endo structural conformation. The addition of locked nucleic acids to siRNAs has been shown to increase siRNA stability in serum, and to reduce off-target effects (Elmen, J. et al., (2005) Nucleic Acids Research 33(1):439-447; Mook, O R. et al., (2007) Mol Canc Ther 6(3):833-843; Grunweller, A. et al., (2003) Nucleic Acids Research 31(12):3185-3193).

Representative U.S. Patents that teach the preparation of locked nucleic acid nucleotides include, but are not limited to, the following: U.S. Pat. Nos. 6,268,490; 6,670,461; 6,794,499; 6,998,484; 7,053,207; 7,084,125; and 7,399,845, each of which is herein incorporated by reference in its entirety.

Potentially stabilizing modifications to the ends of RNA molecules can include N-(acetylaminocaproyl)-4-hydroxyprolinol (Hyp-C6-NHAc), N-(caproyl-4-hydroxyprolinol (Hyp-C6), N-(acetyl-4-hydroxyprolinol (Hyp-NHAc), thymidine-2′-O-deoxythymidine (ether), N-(aminocaproyl)-4-hydroxyprolinol (Hyp-C6-amino), 2-docosanoyl-uridine-3″-phosphate, inverted base dT(idT) and others. Disclosure of this modification can be found in PCT Publication No. WO 2011/005861.

iRNA Motifs

In one embodiment, the sense strand sequence may be represented by formula (I):

    • 5′np-Na -(XXX)i-Nb -Y Y Y -Nb -(ZZZ)j -Na - nq 3′(I)
    • wherein:
    • i and j are each independently 0 or 1;
    • p and q are each independently 0-6;
    • each Na independently represents an oligonucleotide sequence comprising 0-25 modified nucleotides, each sequence comprising at least two differently modified nucleotides;
    • each Nb independently represents an oligonucleotide sequence comprising 0-10 modified nucleotides;
    • each np and nq independently represent an overhang nucleotide;
    • wherein Nb and Y do not have the same modification; and
    • XXX, YYY and ZZZ each independently represent one motif of three identical modifications on three consecutive nucleotides. Preferably YYY is all 2′-F modified nucleotides.

In one embodiment, the Na and/or Nb comprise modifications of alternating pattern.

In one embodiment, the YYY motif occurs at or near the cleavage site of the sense strand. For example, when the RNAi agent has a duplex region of 17-23 nucleotides in length, the YYY motif can occur at or the vicinity of the cleavage site (e.g.: can occur at positions 6, 7, 8; 7, 8, 9; 8, 9, 10; 9, 10, 11; 10, 11,12 or 11, 12, 13) of—the sense strand, the count starting from the 1st nucleotide, from the 5′-end; or optionally, the count starting at the 1st paired nucleotide within the duplex region, from the 5′-end.

In one embodiment, i is 1 and j is 0, or i is 0 and j is 1, or both i and j are 1. The sense strand can therefore be represented by the following formulas:

    • 5′np-Na - YYY -Nb -ZZZ -Nb -ZZZ- na-nq3′(Ib);
    • 5′np-XXX-Nb -YYY-Na- nq 3′(Ic); or
    • 5′np-Na -XXX-Nb-YYY-Nb -ZZZ-Na - nq3′(Id).

When the sense strand is represented by formula (Ib), Nb represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or 10 modified nucleotides. Each Na independently can represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

When the sense strand is represented as formula (Ic), Nb represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or 0 modified nucleotides. Each Na can independently represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

When the sense strand is represented as formula (Id), each Nb independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or 0 modified nucleotides. Preferably, Nb is 0, 1, 2, 3, 4, 5 or 6. Each Na can independently represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

Each of X, Y and Z may be the same or different from each other.

In other embodiments, i is 0 and j is 0, and the sense strand may be represented by the formula:

    • 5′np-Na -YYY-Na- nq3′(Ia).

When the sense strand is represented by formula (Ia), each Na independently can represent an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

In one embodiment, the antisense strand sequence of the RNAi may be represented by formula (II):

    • 5′nq′-Nak-Nb′-Y′Y′Y′-Nb′-(X′X′X′)-l-N′a- np′3′(II)
    • wherein:
    • k and 1 are each independently 0 or 1;
    • p′ and q′ are each independently 0-6;
    • each Na′ independently represents an oligonucleotide sequence comprising 0-25 modified nucleotides, each sequence comprising at least two differently modified nucleotides;
    • each Nb′ independently represents an oligonucleotide sequence comprising 0-10 modified nucleotides;
    • each np′ and nq′ independently represent an overhang nucleotide;
    • wherein Nb‘ and Y’ do not have the same modification;
    • and
    • X′X′X′, Y′Y′Y′ and Z′Z′Z′ each independently represent one motif of three identical modifications on three consecutive nucleotides.

In one embodiment, the Na′ and/or Nb′ comprise modifications of alternating pattern.

The Y′Y′Y′ motif occurs at or near the cleavage site of the antisense strand. For example, when the RNAi agent has a duplex region of 17-23nucleotide in length, the Y′Y′Y′ motif can occur at positions 9, 10, 11;10, 11, 12; 11, 12, 13; 12, 13, 14; or 13, 14, 15 of the antisense strand, with the count starting from the 1st nucleotide, from the 5′-end; or optionally, the count starting at the 1st paired nucleotide within the duplex region, from the 5′-end.

Preferably, the Y′Y′Y′ motif occurs at positions 11, 12, 13.

In one embodiment, Y′Y′Y′ motif is all 2′-OMe modified nucleotides.

In one embodiment, k is 1 and 1 is 0, or k is 0 and 1 is 1, or both k and 1 are 1.

The antisense strand can therefore be represented by the following formulas:

    • 5′nq′-Na′-Z′Z′Z′-Nb′-Y′Y′Y′-Na′-np′3′(IIb);
    • 5′nq′-Y′Y′Y′-Nb -X′X′X′-Np′-3′(IIc); or
    • 5′nq′-Na′-Z′Z′Z′-Nb′-Y′Y′Y′-Nb -X′X′X′-Na′- np′3′(IId).

When the antisense strand is represented by formula (IIb), Nb′ represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or 10 modified nucleotides. Each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

When the antisense strand is represented as formula (IIc), Nb′ represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or 10 modified nucleotides. Each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

When the antisense strand is represented as formula (IId), each Nb′ independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or 0 modified nucleotides. Each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides. Preferably, Nb is 0, 1, 2, 3, 4, 5 or 6.

In other embodiments, k is 0 and 1 is 0 and the antisense strand may be represented by the formula:

    • 5′np′-Na′-Y′Y′Y′-Na′- nq3′(Ia).

When the antisense strand is represented as formula (IIa), each Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

Each of X′, Y′ and Z′ may be the same or different from each other.

Each nucleotide of the sense strand and antisense strand may be independently modified with LNA, HNA, CeNA, 2′-methoxyethyl, 2′-O-methyl, 2′-O-allyl, 2′-C— allyl, 2′-hydroxyl, or 2′-fluoro. For example, each nucleotide of the sense strand and antisense strand is independently modified with 2′-O-methyl or 2′-fluoro. Each X, Y, Z, X′, Y′ and Z′, in particular, may represent a 2′-O-methyl modification or a 2′-fluoro modification.

In one embodiment, the sense strand of the RNAi agent may contain YYY motif occurring at 9, 10 and 11 positions of the strand when the duplex region is 21 nt, the count starting from the 1st nucleotide from the 5′-end, or optionally, the count starting at the 1st paired nucleotide within the duplex region, from the 5′-end; and Y represents 2′-F modification. The sense strand may additionally contain XXX motif or ZZZ motifs as wing modifications at the opposite end of the duplex region; and XXX and ZZZ each independently represents a 2′-OMe modification or 2′-F modification.

In one embodiment the antisense strand may contain Y′Y′Y′ motif occurring at positions 11, 12, 13 of the strand, the count starting from the 1st nucleotide from the 5′-end, or optionally, the count starting at the 1st paired nucleotide within the duplex region, from the 5′-end; and Y′ represents 2′-O-methyl modification. The antisense strand may additionally contain X′X′X′ motif or Z′Z′Z′ motifs as wing modifications at the opposite end of the duplex region; and X′X′X′ and Z′Z′Z′ each independently represents a 2′-OMe modification or 2′-F modification.

The sense strand represented by any one of the above formulas (Ia), (Ib), (Ic), and (Id) forms a duplex with a antisense strand being represented by any one of formulas (IIa), (IIb), (IIc), and (IId), respectively.

Accordingly, the RNAi agents for use in the methods of the invention may comprise a sense strand and an antisense strand, each strand having 14 to 30 nucleotides, the RNAi duplex represented by formula (III):

    • sense: 5′np-Na-(XXX)i-Nb -YYY-Nb-(ZZZ)j-Na-nq3′
    • antisense: 3′np′-Na′-(X′X′X′)k-Nb′-Y′Y′Y′-Nb′-(Z′Z′Z′)l-Na′- nq′5′
    • wherein:
    • i, j, k, and 1 are each independently 0 or 1;
    • p, p′, q, and q′ are each independently 0-6;
    • each Na and Na′ independently represents an oligonucleotide sequence comprising 0-25 modified nucleotides, each sequence comprising at least two differently modified nucleotides;
    • each Nb and Nb′ independently represents an oligonucleotide sequence comprising 0-10 modified nucleotides;
    • wherein
    • each np′, np, nq′, and nq, each of which may or may not be present, independently represents an overhang nucleotide; and
    • XXX, YYY, ZZZ, X′X′X′, Y′Y′Y′, and Z′Z′Z′ each independently represent one motif of three identical modifications on three consecutive nucleotides.

In one embodiment, i is 0 and j is 0; or i is 1 and j is 0; or i is 0 and j is 1; or both i and j are 0; or both i and j are 1. In another embodiment, k is 0 and 1 is 0; or k is 1 and 1 is 0; k is 0 and 1 is 1; or both k and 1 are 0; or both k and 1 are 1.

Exemplary combinations of the sense strand and antisense strand forming a RNAi duplex include the formulas below:

    • 5′np-Na-YYY-Na-nq3′
    • 3′np′-Na′-Y′Y′Y′-Na′-nq′5′
    • (IIIa)
    • 5′np-Na-YYY-Nb-ZZZ-Na-na-nq3′
    • 3′np′-Na′-(Y′Y′Y′)-Nb′-Z′Z′Z′-Na′- nq′5′
    • (IIIb)
    • 5′np-Na-XXX-Nb-YYY-Na -nq3′
    • 3′np′-Na′-X′X′X′-Nb′-Y′Y′Y′-Na′- nq′5′
    • (IIIc)
    • 5′np-Na-XXX-Nb-YYY-Nb-ZZZ-Na- nq3′
    • 3′np′-Na′-X′X′X′-Nb′-Y′Y′Y′-Nb′-Z′Z′Z′-Na′- nq′5′
    • (IIId)

When the RNAi agent is represented by formula (IIIa), each Na independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

When the RNAi agent is represented by formula (IIIb), each Nb independently represents an oligonucleotide sequence comprising 1-10, 1-7, 1-5 or 1-4 modified nucleotides. Each Na independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

When the RNAi agent is represented as formula (IIIc), each Nb, Nb′ independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or Modified nucleotides. Each Na independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides.

When the RNAi agent is represented as formula (IIId), each Nb, Nb′ independently represents an oligonucleotide sequence comprising 0-10, 0-7, 0-5, 0-4, 0-2 or 0modified nucleotides. Each Na, Na′ independently represents an oligonucleotide sequence comprising 2-20, 2-15, or 2-10 modified nucleotides. Each of Na, Na′, Nb and Nb′ independently comprises modifications of alternating pattern.

Each of X, Y and Z in formulas (III), (IIIa), (IIIb), (IIIc), and (IIId) may be the same or different from each other.

When the RNAi agent is represented by formula (III), (IIIa), (IIIb), (IIc), and (IIId), at least one of the Y nucleotides may form a base pair with one of the Y′ nucleotides. Alternatively, at least two of the Y nucleotides form base pairs with the corresponding Y′ nucleotides; or all three of the Y nucleotides all form base pairs with the corresponding Y′ nucleotides.

When the RNAi agent is represented by formula (IIIb) or (IIId), at least one of the Z nucleotides may form a base pair with one of the Z′ nucleotides. Alternatively, at least two of the Z nucleotides form base pairs with the corresponding Z′ nucleotides; or all three of the Z nucleotides all form base pairs with the corresponding Z′ nucleotides.

When the RNAi agent is represented as formula (IIIc) or (IIId), at least one of the X nucleotides may form a base pair with one of the X′ nucleotides. Alternatively, at least two of the X nucleotides form base pairs with the corresponding X′ nucleotides; or all three of the X nucleotides all form base pairs with the corresponding X′ nucleotides.

In one embodiment, the modification on the Y nucleotide is different than the modification on the Y′ nucleotide, the modification on the Z nucleotide is different than the modification on the Z′ nucleotide, and/or the modification on the X nucleotide is different than the modification on the X′ nucleotide.

In one embodiment, when the RNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications. In another embodiment, when the RNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications and np′>0 and at least one np′ is linked to a neighboring nucleotide a via phosphorothioate linkage. In yet another embodiment, when the RNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications, np′>0 and at least one np′ is linked to a neighboring nucleotide via phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker. In another embodiment, when the RNAi agent is represented by formula (IIId), the Na modifications are 2′-O-methyl or 2′-fluoro modifications, np′>0 and at least one np′ is linked to a neighboring nucleotide via phosphorothioate linkage, the sense strand comprises at least one phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.

In one embodiment, when the RNAi agent is represented by formula (IIIa), the Na modifications are 2′-O-methyl or 2′-fluoro modifications, np′>0 and at least one np′ is linked to a neighboring nucleotide via phosphorothioate linkage, the sense strand comprises at least one phosphorothioate linkage, and the sense strand is conjugated to one or more GalNAc derivatives attached through a bivalent or trivalent branched linker.

In one embodiment, the RNAi agent is a multimer containing at least two duplexes represented by formula (III), (IIIa), (IIIb), (IIIc), and (IIId), wherein the duplexes are connected by a linker. The linker can be cleavable or non-cleavable. Optionally, the multimer further comprises a ligand. Each of the duplexes can target the same gene or two different genes; or each of the duplexes can target same gene at two different target sites.

In one embodiment, the RNAi agent is a multimer containing three, four, five, six or more duplexes represented by formula (III), (IIIa), (IIIb), (IIIc), and (IIId), wherein the duplexes are connected by a linker. The linker can be cleavable or non-cleavable. Optionally, the multimer further comprises a ligand. Each of the duplexes can target the same gene or two different genes; or each of the duplexes can target same gene at two different target sites.

In one embodiment, two RNAi agents represented by formula (III), (IIIa), (IIIb), (IIIc), and (IIId) are linked to each other at the 5′ end, and one or both of the 3′ ends and are optionally conjugated to to a ligand. Each of the agents can target the same gene or two different genes; or each of the agents can target same gene at two different target sites.

iRNA Conjugates

The iRNA agents disclosed herein can be in the form of conjugates. The conjugate may be attached at any suitable location in the iRNA molecule, e.g., at the 3′ end or the 5′ end of the sense or the antisense strand. The conjugates are optionally attached via a linker.

In some embodiments, an iRNA agent described herein is chemically linked to one or more ligands, moieties or conjugates, which may confer functionality, e.g., by affecting (e.g., enhancing) the activity, cellular distribution or cellular uptake of the iRNA. Such moieties include but are not limited to lipid moieties such as a cholesterol moiety (Letsinger et al., Proc. Natl. Acid. Sci. USA, 1989, 86: 6553-6556), cholic acid (Manoharan et al., Biorg. Med. Chem. Let., 1994, 4:1053-1060), a thioether, e.g., beryl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306-309; Manoharan et al., Biorg. 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 (Saison-Behmoaras et al., EMBO J, 1991, 10:1111-1118; 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 triethyl-ammonium 1,2-di-O-hexadecyl-rac-glycero-3-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 (Manoharan 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-carbonyloxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923-937).

In one embodiment, a ligand alters the distribution, targeting or lifetime of an iRNA agent into which it is incorporated. In some embodiments, a ligand provides an enhanced affinity for a selected target, e.g., molecule, cell or cell type, compartment, e.g., a cellular or organ compartment, tissue, organ or region of the body, as, e.g., compared to a species absent such a ligand. Typical ligands will not take part in duplex pairing in a duplexed nucleic acid.

Ligands can include a naturally occurring substance, such as a protein (e.g., human serum albumin (HSA), low-density lipoprotein (LDL), or globulin); carbohydrate (e.g., a dextran, pullulan, chitin, chitosan, inulin, cyclodextrin or hyaluronic acid); or a lipid. The ligand may also be a recombinant or synthetic molecule, such as a synthetic polymer, e.g., a synthetic polyamino acid. Examples of polyamino acids include polyamino acid is a polylysine (PLL), poly L-aspartic acid, poly L-glutamic acid, styrene-maleic acid anhydride copolymer, poly(L-lactide-co-glycolied) copolymer, divinyl ether-maleic anhydride copolymer, N-(2-hydroxypropyl)methacrylamide copolymer (HMPA), polyethylene glycol (PEG), polyvinyl alcohol (PVA), polyurethane, poly(2-ethylacryllic acid), N-isopropylacrylamide polymers, or polyphosphazine. Example of polyamines include: polyethylenimine, polylysine (PLL), spermine, spermidine, polyamine, pseudopeptide-polyamine, peptidomimetic polyamine, dendrimer polyamine, arginine, amidine, protamine, cationic lipid, cationic porphyrin, quaternary salt of a polyamine, or an a helical peptide.

Ligands can also include targeting groups, e.g., a cell or tissue targeting agent, e.g., a lectin, glycoprotein, lipid or protein, e.g., an antibody, that binds to a specified cell type such as a kidney cell. A targeting group can be a thyrotropin, melanotropin, lectin, glycoprotein, surfactant protein A, Mucin carbohydrate, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-gulucosamine multivalent mannose, multivalent fucose, glycosylated polyaminoacids, multivalent galactose, transferrin, bisphosphonate, polyglutamate, polyaspartate, a lipid, cholesterol, a steroid, bile acid, folate, vitamin B12, biotin, or an RGD peptide or RGD peptide mimetic.

In some embodiments, the ligand is a GalNAc ligand that comprises one or more N-acetylgalactosamine (GalNAc) derivatives. Additional description of GalNAc ligands is provided in the section titled Carbohydrate Conjugates.

Other examples of ligands include dyes, intercalating agents (e.g. acridines), cross-linkers (e.g. psoralene, mitomycin C), porphyrins (TPPC4, texaphyrin, Sapphyrin), polycyclic aromatic hydrocarbons (e.g., phenazine, dihydrophenazine), artificial endonucleases (e.g. EDTA), lipophilic molecules, e.g., cholesterol, cholic acid, adamantane acetic acid, 1-pyrene butyric acid, dihydrotestosterone, 1,3-Bis-O(hexadecyl)glycerol, geranyloxyhexyl group, hexadecylglycerol, borneol, menthol, 1,3-propanediol, heptadecyl group, palmitic acid, myristic acid, O3-(oleoyl)lithocholic acid, 03-(oleoyl)cholenic acid, dimethoxytrityl, or phenoxazine) and peptide conjugates (e.g., antennapedia peptide, Tat peptide), alkylating agents, phosphate, amino, mercapto, PEG (e.g., PEG-40K), MPEG, [MPEG]2, polyamino, alkyl, substituted alkyl, radiolabeled markers, enzymes, haptens (e.g. biotin), transport/absorption facilitators (e.g., aspirin, vitamin E, folic acid), synthetic ribonucleases (e.g., imidazole, bisimidazole, histamine, imidazole clusters, acridine-imidazole conjugates, Eu3+complexes of tetraazamacrocycles), dinitrophenyl, HRP, or AP.

Ligands can be proteins, e.g., glycoproteins, or peptides, e.g., molecules having a specific affinity for a co-ligand, or antibodies e.g., an antibody, that binds to a specified cell type such as a cancer cell, endothelial cell, or bone cell. Ligands may also include hormones and hormone receptors. They can also include non-peptidic species, such as lipids, lectins, carbohydrates, vitamins, cofactors, multivalent lactose, multivalent galactose, N-acetyl-galactosamine, N-acetyl-gulucosamine multivalent mannose, or multivalent fucose. The ligand can be, for example, a lipopolysaccharide, an activator of p38 MAP kinase, or an activator of NF-κB.

The ligand can be a substance, e.g., a drug, which can increase the uptake of the iRNA agent into the cell, for example, by disrupting the cell's cytoskeleton, e.g., by disrupting the cell's microtubules, microfilaments, and/or intermediate filaments. The drug can be, for example, taxon, vincristine, vinblastine, cytochalasin, nocodazole, japlakinolide, latrunculin A, phalloidin, swinholide A, indanocine, or myoservin.

In some embodiments, a ligand attached to an iRNA as described herein acts as a pharmaco*kinetic modulator (PK modulator). PK modulators include lipophiles, bile acids, steroids, phospholipid analogues, peptides, protein binding agents, PEG, vitamins etc. Exemplary PK modulators include, but are not limited to, cholesterol, fatty acids, cholic acid, lithocholic acid, dialkylglycerides, diacylglyceride, phospholipids, sphingolipids, naproxen, ibuprofen, vitamin E, biotin etc. Oligonucleotides that comprise a number of phosphorothioate linkages are also known to bind to serum protein, thus short oligonucleotides, e.g., oligonucleotides of about 5 bases, 10 bases, 15 bases or 20 bases, comprising multiple of phosphorothioate linkages in the backbone are also amenable to the present invention as ligands (e.g. as PK modulating ligands). In addition, aptamers that bind serum components (e.g. serum proteins) are also suitable for use as PK modulating ligands in the embodiments described herein.

Ligand-conjugated oligonucleotides of the invention may be synthesized by the use of an oligonucleotide that bears a pendant reactive functionality, such as that derived from the attachment of a linking molecule onto the oligonucleotide (described below). This reactive oligonucleotide may be reacted directly with commercially-available ligands, ligands that are synthesized bearing any of a variety of protecting groups, or ligands that have a linking moiety attached thereto.

The oligonucleotides used in the conjugates of the present invention may be conveniently and routinely made through the well-known technique of solid-phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is also known to use similar techniques to prepare other oligonucleotides, such as the phosphorothioates and alkylated derivatives.

In the ligand-conjugated oligonucleotides and ligand-molecule bearing sequence-specific linked nucleosides of the present invention, the oligonucleotides and oligonucleosides may be assembled on a suitable DNA synthesizer utilizing standard nucleotide or nucleoside precursors, or nucleotide or nucleoside conjugate precursors that already bear the linking moiety, ligand-nucleotide or nucleoside-conjugate precursors that already bear the ligand molecule, or non-nucleoside ligand-bearing building blocks.

When using nucleotide-conjugate precursors that already bear a linking moiety, the synthesis of the sequence-specific linked nucleosides is typically completed, and the ligand molecule is then reacted with the linking moiety to form the ligand-conjugated oligonucleotide. In some embodiments, the oligonucleotides or linked nucleosides of the present invention are synthesized by an automated synthesizer using phosphoramidites derived from ligand-nucleoside conjugates in addition to the standard phosphoramidites and non-standard phosphoramidites that are commercially available and routinely used in oligonucleotide synthesis.

In one embodiment, the ligand is a lipid or lipid-based molecule. Such a lipid or lipid-based molecule can typically bind a serum protein, such as human serum albumin (HSA). An HSA binding ligand allows for distribution of the conjugate to a target tissue, e.g., a non-kidney target tissue of the body. For example, the target tissue can be the liver, including parenchymal cells of the liver. Other molecules that can bind HSA can also be used as ligands. For example, neproxin or aspirin can be used. A lipid or lipid-based ligand can (a) increase resistance to degradation of the conjugate, (b) increase targeting or transport into a target cell or cell membrane, and/or (c) can be used to adjust binding to a serum protein, e.g., HSA.

A lipid based ligand can be used to modulate, e.g., control (e.g., inhibit) the binding of the conjugate to a target tissue. For example, a lipid or lipid-based ligand that binds to HSA more strongly will be less likely to be targeted to the kidney and therefore less likely to be cleared from the body. A lipid or lipid-based ligand that binds to HSA less strongly can be used to target the conjugate to the kidney.

In one embodiment, the lipid based ligand binds HSA. For example, the ligand can bind HSA with a sufficient affinity such that distribution of the conjugate to a non-kidney tissue is enhanced. However, the affinity is typically not so strong that the HSA-ligand binding cannot be reversed.

In another embodiment, the lipid based ligand binds HSA weakly or not at all, such that distribution of the conjugate to the kidney is enhanced. Other moieties that target to kidney cells can also be used in place of or in addition to the lipid based ligand.

In another aspect, the ligand is a moiety, e.g., a vitamin, which is taken up by a target cell, e.g., a proliferating cell. These are particularly useful for treating disorders characterized by unwanted cell proliferation, e.g., of the malignant or non-malignant type, e.g., cancer cells. Exemplary vitamins include vitamin A, E, and K. Other exemplary vitamins include are B vitamin, e.g., folic acid, B12, riboflavin, biotin, pyridoxal or other vitamins or nutrients taken up by cancer cells. Also included are HSA and low density lipoprotein (LDL).

In another aspect, the ligand is a cell-permeation agent, such as a helical cell-permeation agent. In one embodiment, the agent is amphipathic. An exemplary agent is a peptide such as tat or antennopedia. If the agent is a peptide, it can be modified, including a peptidylmimetic, invertomers, non-peptide or pseudo-peptide linkages, and use of D-amino acids. The helical agent is typically an α-helical agent, and can have a lipophilic and a lipophobic phase.

The ligand can be a peptide or peptidomimetic. A peptidomimetic (also referred to herein as an oligopeptidomimetic) is a molecule capable of folding into a defined three-dimensional structure similar to a natural peptide. The attachment of peptide and peptidomimetics to iRNA agents can affect pharmaco*kinetic distribution of the iRNA, such as by enhancing cellular recognition and absorption. The peptide or peptidomimetic moiety can be about 5-50 amino acids long, e.g., about 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 amino acids long.

A peptide or peptidomimetic can be, for example, a cell permeation peptide, cationic peptide, amphipathic peptide, or hydrophobic peptide (e.g., consisting primarily of Tyr, Trp or Phe). The peptide moiety can be a dendrimer peptide, constrained peptide or crosslinked peptide. In another alternative, the peptide moiety can include a hydrophobic membrane translocation sequence (MTS). An exemplary hydrophobic MTS-containing peptide is RFGF having the amino acid sequence AAVALLPAVLLALLAP (SEQ ID NO:3367). An RFGF analogue (e.g., amino acid sequence AALLPVLLAAP (SEQ ID NO:3368)) containing a hydrophobic MTS can also be a targeting moiety. The peptide moiety can be a “delivery” peptide, which can carry large polar molecules including peptides, oligonucleotides, and protein across cell membranes. For example, sequences from the HIV Tat protein (GRKKRRQRRRPPQ (SEQ ID NO:3369)) and the Drosophila Antennapedia protein (RQIKIWFQNRRMKWKK (SEQ ID NO: 3370)) have been found to be capable of functioning as delivery peptides. A peptide or peptidomimetic can be encoded by a random sequence of DNA, such as a peptide identified from a phage-display library, or one-bead-one-compound (OBOC) combinatorial library (Lam et al., Nature, 354:82-84, 1991). Typically, the peptide or peptidomimetic tethered to a dsRNA agent via an incorporated monomer unit is a cell targeting peptide such as an arginine-glycine-aspartic acid (RGD)-peptide, or RGD mimic. A peptide moiety can range in length from about 5 amino acids to about 40 amino acids. The peptide moieties can have a structural modification, such as to increase stability or direct conformational properties. Any of the structural modifications described below can be utilized.

An RGD peptide for use in the compositions and methods of the invention may be linear or cyclic, and may be modified, e.g., glycosylated or methylated, to facilitate targeting to a specific tissue(s). RGD-containing peptides and peptidiomimemtics may include D-amino acids, as well as synthetic RGD mimics. In addition to RGD, one can use other moieties that target the integrin ligand. Preferred conjugates of this ligand target PECAM-1 or VEGF.

An RGD peptide moiety can be used to target a particular cell type, e.g., a tumor cell, such as an endothelial tumor cell or a breast cancer tumor cell (Zitzmann et al., Cancer Res., 62:5139-43, 2002). An RGD peptide can facilitate targeting of an dsRNA agent to tumors of a variety of other tissues, including the lung, kidney, spleen, or liver (Aoki et al., Cancer Gene Therapy 8:783-787, 2001). Typically, the RGD peptide will facilitate targeting of an iRNA agent to the kidney. The RGD peptide can be linear or cyclic, and can be modified, e.g., glycosylated or methylated to facilitate targeting to specific tissues. For example, a glycosylated RGD peptide can deliver a iRNA agent to a tumor cell expressing avB3 (Haubner et al., Jour. Nucl. Med., 42:326-336, 2001).

A “cell permeation peptide” is capable of permeating a cell, e.g., a microbial cell, such as a bacterial or fungal cell, or a mammalian cell, such as a human cell. A microbial cell-permeating peptide can be, for example, an α-helical linear peptide (e.g., LL-37 or Ceropin P1), a disulfide bond-containing peptide (e.g., α-defensin, β-defensin or bactenecin), or a peptide containing only one or two dominating amino acids (e.g., PR-39 or indolicidin). A cell permeation peptide can also include a nuclear localization signal (NLS). For example, a cell permeation peptide can be a bipartite amphipathic peptide, such as MPG, which is derived from the fusion peptide domain of HIV-1 gp41 and the NLS of SV40 large T antigen (Simeoni et al., Nucl. Acids Res. 31:2717-2724, 2003).

In some embodiments of the compositions and methods of the invention, an iRNA oligonucleotide further comprises a carbohydrate. The carbohydrate conjugated iRNA are advantageous for the in vivo delivery of nucleic acids, as well as compositions suitable for in vivo therapeutic use, as described herein. As used herein, “carbohydrate” refers to a compound which is either a carbohydrate per se made up of one or more monosaccharide units having at least 6 carbon atoms (which can be linear, branched or cyclic) with an oxygen, nitrogen or sulfur atom bonded to each carbon atom; or a compound having as a part thereof a carbohydrate moiety made up of one or more monosaccharide units each having at least six carbon atoms (which can be linear, branched or cyclic), with an oxygen, nitrogen or sulfur atom bonded to each carbon atom. Representative carbohydrates include the sugars (mono-, di-, tri- and oligosaccharides containing from about 4, 5, 6, 7, 8, or 9 monosaccharide units), and polysaccharides such as starches, glycogen, cellulose and polysaccharide gums. Specific monosaccharides include C5 and above (e.g., C5, C6, C7, or C8) sugars; di- and trisaccharides include sugars having two or three monosaccharide units (e.g., C5, C6, C7, or C8).

In one embodiment, a carbohydrate conjugate comprises a monosaccharide. In one embodiment, the monosaccharide is an N-acetylgalactosamine (GalNAc). GalNAc conjugates are described, for example, in U.S. Pat. No. 8,106,022, the entire content of which is hereby incorporated herein by reference. In some embodiments, the GalNAc conjugate serves as a ligand that targets the iRNA to particular cells. In some embodiments, the GalNAc conjugate targets the iRNA to liver cells, e.g., by serving as a ligand for the asialoglycoprotein receptor of liver cells (e.g., hepatocytes).

In some embodiments, the carbohydrate conjugate comprises one or more GalNAc derivatives. The GalNAc derivatives may be attached via a linker, e.g., a bivalent or trivalent branched linker. In some embodiments the GalNAc conjugate is conjugated to the 3′ end of the sense strand. In some embodiments, the GalNAc conjugate is conjugated to the iRNA agent (e.g., to the 3′ end of the sense strand) via a linker, e.g., a linker as described herein.

In some embodiments, the GalNAc conjugate is

In some embodiments, the RNAi agent is attached to the carbohydrate conjugate via a linker as shown in the following schematic, wherein X is O or S

In some embodiments, the RNAi agent is conjugated to L96 as defined in Table 1 and shown below

In some embodiments, a carbohydrate conjugate for use in the compositions and methods of the invention is selected from the group consisting of:

Another representative carbohydrate conjugate for use in the embodiments described herein includes, but is not limited to,

when one of X or Y is an oligonucleotide, the other is a hydrogen.

In some embodiments, the carbohydrate conjugate further comprises one or more additional ligands as described above, such as, but not limited to, a PK modulator and/or a cell permeation peptide.

In one embodiment, an iRNA of the invention is conjugated to a carbohydrate through a linker. Non-limiting examples of iRNA carbohydrate conjugates with linkers of the compositions and methods of the invention include, but are not limited to,

when one of X or Y is an oligonucleotide, the other is a hydrogen.

In some embodiments, the conjugate or ligand described herein can be attached to an iRNA oligonucleotide with various linkers that can be cleavable or non-cleavable.

The term “linker” or “linking group” means an organic moiety that connects two parts of a compound, e.g., covalently attaches two parts of a compound. Linkers typically comprise a direct bond or an atom such as oxygen or sulfur, a unit such as NR8, C(O), C(O)NH, SO, SO2, SO2NH or a chain of atoms, such as, but not limited to, substituted or unsubstituted alkyl, substituted or unsubstituted alkenyl, substituted or unsubstituted alkynyl, arylalkyl, arylalkenyl, arylalkynyl, heteroarylalkyl, heteroarylalkenyl, heteroarylalkynyl, heterocyclylalkyl, heterocyclylalkenyl, heterocyclylalkynyl, aryl, heteroaryl, heterocyclyl, cycloalkyl, cycloalkenyl, alkylarylalkyl, alkylarylalkenyl, alkylarylalkynyl, alkenylarylalkyl, alkenylarylalkenyl, alkenylarylalkynyl, alkynylarylalkyl, alkynylarylalkenyl, alkynylarylalkynyl, alkylheteroarylalkyl, alkylheteroarylalkenyl, alkylheteroarylalkynyl, alkenylheteroarylalkyl, alkenylheteroarylalkenyl, alkenylheteroarylalkynyl, alkynylheteroarylalkyl, alkynylheteroarylalkenyl, alkynylheteroarylalkynyl, alkylheterocyclylalkyl, alkylheterocyclylalkenyl, alkylhererocyclylalkynyl, alkenylheterocyclylalkyl, alkenylheterocyclylalkenyl, alkenylheterocyclylalkynyl, alkynylheterocyclylalkyl, alkynylheterocyclylalkenyl, alkynylheterocyclylalkynyl, alkylaryl, alkenylaryl, alkynylaryl, alkylheteroaryl, alkenylheteroaryl, alkynylhereroaryl, which one or more methylenes can be interrupted or terminated by O, S, S(O), SO2, N(R8), C(O), substituted or unsubstituted aryl, substituted or unsubstituted heteroaryl, substituted or unsubstituted heterocyclic; where R8 is hydrogen, acyl, aliphatic or substituted aliphatic. In one embodiment, the linker is between about 1-24 atoms, 2-24, 3-24, 4-24, 5-24, 6-24, 6-18, 7-18, 8-18 atoms, 7-17, 8-17, 6-16, 7-16, or 8-16 atoms.

In one embodiment, a dsRNA of the invention is conjugated to a bivalent or trivalent branched linker selected from the group of structures shown in any of formula (XXXI)-(XXXIV):

wherein:

    • q2A, q2B, q3A, q3B, q4A, q4B, q5A, q5B and q5C represent independently for each occurrence 0-20 and wherein the repeating unit can be the same or different;
    • P2A, p2B, p3A, p3B, p4A, p4B, p5A, P5B, P5C, T2A, T2B, T3A, T3B, T4A, T4B, T4A, T5B, T5c are each independently for each occurrence absent, CO, NH, O, S, OC(O), NHC(O), CH2, CH2NH or CH2O;
    • Q2A, Q2B, Q3A, Q3B, Q4A, Q4B, Q5A, Q5B, Q5C are independently for each occurrence absent, alkylene, substituted alkylene wherein one or more methylenes can be interrupted or terminated by one or more of O, S, S(O), SO2, N(RN), C(R′)═C(R″), C≡C or C(O);
    • R2A, R2B, R3A, R3B, R4A, R4B, R5A, R5B, R5c are each independently for each occurrence absent, NH, O, S, CH2, C(O)O, C(O)NH, NHCH(Ra)C(O), —C(O)—CH(Ra)—NH—, CO, CH═N—O,

or

    • L2A, L2B, L3A L3B, L4A L4B L5A L5B and L5c represent the ligand; i.e. each independently for each occurrence a monosaccharide (such as GalNAc), disaccharide, trisaccharide, tetrasaccharide, oligosaccharide, or polysaccharide; and Ra is H or amino acid side chain. Trivalent conjugating GalNAc derivatives are particularly useful for use with RNAi agents for inhibiting the expression of a target gene, such as those of formula (XXXV):
    • wherein L5A, L5B and L5C represent a monosaccharide, such as GalNAc derivative.

Examples of suitable bivalent and trivalent branched linker groups conjugating GalNAc derivatives include, but are not limited to, the structures recited above as formulas II, VII, XI, X, and XIII.

A cleavable linking group is one which is sufficiently stable outside the cell, but which upon entry into a target cell is cleaved to release the two parts the linker is holding together. In a preferred embodiment, the cleavable linking group is cleaved at least about 10 times, 20, times, 30 times, 40 times, 50 times, 60 times, 70 times, 80 times, 90 times or more, or at least about 100 times faster in a target cell or under a first reference condition (which can, e.g., be selected to mimic or represent intracellular conditions) than in the blood of a subject, or under a second reference condition (which can, e.g., be selected to mimic or represent conditions found in the blood or serum).

Cleavable linking groups are susceptible to cleavage agents, e.g., pH, redox potential or the presence of degradative molecules. Generally, cleavage agents are more prevalent or found at higher levels or activities inside cells than in serum or blood. Examples of such degradative agents include: redox agents which are selected for particular substrates or which have no substrate specificity, including, e.g., oxidative or reductive enzymes or reductive agents such as mercaptans, present in cells, that can degrade a redox cleavable linking group by reduction; esterases; endosomes or agents that can create an acidic environment, e.g., those that result in a pH of five or lower; enzymes that can hydrolyze or degrade an acid cleavable linking group by acting as a general acid, peptidases (which can be substrate specific), and phosphatases.

A cleavable linkage group, such as a disulfide bond can be susceptible to pH. The pH of human serum is 7.4, while the average intracellular pH is slightly lower, ranging from about 7.1-7.3. Endosomes have a more acidic pH, in the range of 5.5-6.0, and lysosomes have an even more acidic pH at around 5.0. Some linkers will have a cleavable linking group that is cleaved at a preferred pH, thereby releasing a cationic lipid from the ligand inside the cell, or into the desired compartment of the cell.

A linker can include a cleavable linking group that is cleavable by a particular enzyme.

The type of cleavable linking group incorporated into a linker can depend on the cell to be targeted. For example, a liver-targeting ligand can be linked to a cationic lipid through a linker that includes an ester group. Liver cells are rich in esterases, and therefore the linker will be cleaved more efficiently in liver cells than in cell types that are not esterase-rich. Other cell-types rich in esterases include cells of the lung, renal cortex, and testis.

Linkers that contain peptide bonds can be used when targeting cell types rich in peptidases, such as liver cells and synoviocytes.

In general, the suitability of a candidate cleavable linking group can be evaluated by testing the ability of a degradative agent (or condition) to cleave the candidate linking group. It will also be desirable to also test the candidate cleavable linking group for the ability to resist cleavage in the blood or when in contact with other non-target tissue. Thus, one can determine the relative susceptibility to cleavage between a first and a second condition, where the first is selected to be indicative of cleavage in a target cell and the second is selected to be indicative of cleavage in other tissues or biological fluids, e.g., blood or serum. The evaluations can be carried out in cell free systems, in cells, in cell culture, in organ or tissue culture, or in whole animals. It can be useful to make initial evaluations in cell-free or culture conditions and to confirm by further evaluations in whole animals. In preferred embodiments, useful candidate compounds are cleaved at least about 2, 4, 10, 20, 30, 40, 50, 60, 70, 80, 90, or about 100 times faster in the cell (or under in vitro conditions selected to mimic intracellular conditions) as compared to blood or serum (or under in vitro conditions selected to mimic extracellular conditions).

In one embodiment, a cleavable linking group is a redox cleavable linking group that is cleaved upon reduction or oxidation. An example of reductively cleavable linking group is a disulphide linking group (—S—S—). To determine if a candidate cleavable linking group is a suitable “reductively cleavable linking group,” or for example is suitable for use with a particular iRNA moiety and particular targeting agent one can look to methods described herein. For example, a candidate can be evaluated by incubation with dithiothreitol (DTT), or other reducing agent using reagents know in the art, which mimic the rate of cleavage which would be observed in a cell, e.g., a target cell. The candidates can also be evaluated under conditions which are selected to mimic blood or serum conditions. In one, candidate compounds are cleaved by at most about 10% in the blood. In other embodiments, useful candidate compounds are degraded at least about 2, 4, 10, 20, 30, 40, 50, 60, 70, 80, 90, or about 100 times faster in the cell (or under in vitro conditions selected to mimic intracellular conditions) as compared to blood (or under in vitro conditions selected to mimic extracellular conditions). The rate of cleavage of candidate compounds can be determined using standard enzyme kinetics assays under conditions chosen to mimic intracellular media and compared to conditions chosen to mimic extracellular media.

In another embodiment, a cleavable linker comprises a phosphate-based cleavable linking group. A phosphate-based cleavable linking group is cleaved by agents that degrade or hydrolyze the phosphate group. An example of an agent that cleaves phosphate groups in cells are enzymes such as phosphatases in cells. Examples of phosphate-based linking groups are —O—P(O)(ORk)-O—, —O—P(S)(ORk)-O—, —O—P(S)(SRk)-O—, —S—P(O)(ORk)-O—, —O—P(O)(ORk)-S—, —S—P(O)(ORk)-S—, —O—P(S)(ORk)-S—, —S—P(S)(ORk)-O—, —O—P(O)(Rk)-O—, —O—P(S)(Rk)-O—, —S—P(O)(Rk)-O—, —S—P(S)(Rk)-O—, —S—P(O)(Rk)-S—, —O—P(S)(Rk)-S—. Preferred embodiments are —O—P(O)(OH)—O—, —O—P(S)(OH)—O—, —O—P(S)(SH)—O—, —S—P(O)(OH)—O—, —O—P(O)(OH)—S—, —S—P(O)(OH)—S—, —O—P(S)(OH)—S—, —S—P(S)(OH)—O—, —O—P(O)(H)—O—, —O—P(S)(H)—O—, —S—P(O)(H)—O, —S—P(S)(H)—O—, —S—P(O)(H)—S—, —O—P(S)(H)—S—. A preferred embodiment is —O—P(O)(OH)—O—. These candidates can be evaluated using methods analogous to those described above.

In another embodiment, a cleavable linker comprises an acid cleavable linking group. An acid cleavable linking group is a linking group that is cleaved under acidic conditions. In preferred embodiments acid cleavable linking groups are cleaved in an acidic environment with a pH of about 6.5 or lower (e.g., about 6.0, 5.75, 5.5, 5.25, 5.0, or lower), or by agents such as enzymes that can act as a general acid. In a cell, specific low pH organelles, such as endosomes and lysosomes can provide a cleaving environment for acid cleavable linking groups. Examples of acid cleavable linking groups include but are not limited to hydrazones, esters, and esters of amino acids. Acid cleavable groups can have the general formula —C═NN—, C(O)O, or —OC(O). A preferred embodiment is when the carbon attached to the oxygen of the ester (the alkoxy group) is an aryl group, substituted alkyl group, or tertiary alkyl group such as dimethyl pentyl or t-butyl. These candidates can be evaluated using methods analogous to those described above.

In another embodiment, a cleavable linker comprises an ester-based cleavable linking group. An ester-based cleavable linking group is cleaved by enzymes such as esterases and amidases in cells. Examples of ester-based cleavable linking groups include but are not limited to esters of alkylene, alkenylene and alkynylene groups. Ester cleavable linking groups have the general formula —C(O)O—, or —OC(O)—. These candidates can be evaluated using methods analogous to those described above.

In yet another embodiment, a cleavable linker comprises a peptide-based cleavable linking group. A peptide-based cleavable linking group is cleaved by enzymes such as peptidases and proteases in cells. Peptide-based cleavable linking groups are peptide bonds formed between amino acids to yield oligopeptides (e.g., dipeptides, tripeptides etc.) and polypeptides. Peptide-based cleavable groups do not include the amide group (—C(O)NH—). The amide group can be formed between any alkylene, alkenylene or alkynelene. A peptide bond is a special type of amide bond formed between amino acids to yield peptides and proteins. The peptide based cleavage group is generally limited to the peptide bond (i.e., the amide bond) formed between amino acids yielding peptides and proteins and does not include the entire amide functional group. Peptide-based cleavable linking groups have the general formula —NHCHRAC(O)NHCHRBC(O)—(SEQ ID NO: 13), where RA and RB are the R groups of the two adjacent amino acids. These candidates can be evaluated using methods analogous to those described above.

Representative U.S. patents that teach the preparation of RNA conjugates include, but are not limited to, U.S. Pat. Nos. 4,828,979; 4,948,882; 5,218,105; 5,525,465; 5,541,313; 5,545,730; 5,552,538; 5,578,717, 5,580,731; 5,591,584; 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; 6,294,664; 6,320,017; 6,576,752; 6,783,931; 6,900,297; 7,037,646; 8,106,022, the entire contents of each of which is herein incorporated by reference.

It is not necessary for all positions in a given compound to be uniformly modified, and in fact more than one of the aforementioned modifications may be incorporated in a single compound or even at a single nucleoside within an iRNA. The present invention also includes iRNA compounds that are chimeric compounds.

“Chimeric” iRNA compounds, or “chimeras,” in the context of the present invention, are iRNA compounds, e.g., dsRNAs, that contain two or more chemically distinct regions, each made up of at least one monomer unit, i.e., a nucleotide in the case of a dsRNA compound. These iRNAs typically contain at least one region wherein the RNA is modified so as to confer upon the iRNA increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the iRNA may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. By way of example, RNase H is a cellular endonuclease which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of iRNA inhibition of gene expression. Consequently, comparable results can often be obtained with shorter iRNAs when chimeric dsRNAs are used, compared to phosphorothioate deoxy dsRNAs hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.

In certain instances, the RNA of an iRNA can be modified by a non-ligand group. A number of non-ligand molecules have been conjugated to iRNAs in order to enhance the activity, cellular distribution or cellular uptake of the iRNA, and procedures for performing such conjugations are available in the scientific literature. Such non-ligand moieties have included lipid moieties, such as cholesterol (Kubo, T. et al., Biochem. Biophys. Res. Comm., 2007, 365(1):54-61; Letsinger et al., Proc. Natl. Acad. Sci. USA, 1989, 86:6553), cholic acid (Manoharan et al., Bioorg. Med. Chem. Lett., 1994, 4:1053), a thioether, e.g., hexyl-S-tritylthiol (Manoharan et al., Ann. N.Y. Acad. Sci., 1992, 660:306; Manoharan et al., Bioorg. Med. Chem. Let., 1993, 3:2765), a thiocholesterol (Oberhauser et al., Nucl. Acids Res., 1992, 20:533), an aliphatic chain, e.g., dodecandiol or undecyl residues (Saison-Behmoaras et al., EMBO J., 1991, 10:111; Kabanov et al., FEBS Lett., 1990, 259:327; Svinarchuk et al., Biochimie, 1993, 75:49), 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; Shea et al., Nucl. Acids Res., 1990, 18:3777), a polyamine or a polyethylene glycol chain (Manoharan et al., Nucleosides & Nucleotides, 1995, 14:969), or adamantane acetic acid (Manoharan et al., Tetrahedron Lett., 1995, 36:3651), a palmityl moiety (Mishra et al., Biochim. Biophys. Acta, 1995, 1264:229), or an octadecylamine or hexylamino-carbonyl-oxycholesterol moiety (Crooke et al., J. Pharmacol. Exp. Ther., 1996, 277:923). Representative United States patents that teach the preparation of such RNA conjugates have been listed above. Typical conjugation protocols involve the synthesis of an RNAs bearing an aminolinker at one or more positions of the sequence. The amino group is then reacted with the molecule being conjugated using appropriate coupling or activating reagents. The conjugation reaction may be performed either with the RNA still bound to the solid support or following cleavage of the RNA, in solution phase. Purification of the RNA conjugate by HPLC typically affords the pure conjugate.

Delivery of iRNA

The delivery of an iRNA to a subject in need thereof can be achieved in a number of different ways. In vivo delivery can be performed directly by administering a composition comprising an iRNA, e.g. a dsRNA, to a subject. Alternatively, delivery can be performed indirectly by administering one or more vectors that encode and direct the expression of the iRNA. These alternatives are discussed further below.

In general, any method of delivering a nucleic acid molecule can be adapted for use with an iRNA (see e.g., Akhtar S. and Julian R L. (1992) Trends Cell. Biol. 2(5):139-144 and WO94/02595, which are incorporated herein by reference in their entireties). However, there are three factors that are important to consider in order to successfully deliver an iRNA molecule in vivo: (a) biological stability of the delivered molecule, (2) preventing non-specific effects, and (3) accumulation of the delivered molecule in the target tissue. The non-specific effects of an iRNA can be minimized by local administration, for example by direct injection or implantation into a tissue (as a non-limiting example, a tumor) or topically administering the preparation. Local administration to a treatment site maximizes local concentration of the agent, limits the exposure of the agent to systemic tissues that may otherwise be harmed by the agent or that may degrade the agent, and permits a lower total dose of the iRNA molecule to be administered. Several studies have shown successful knockdown of gene products when an iRNA is administered locally. For example, intraocular delivery of a VEGF dsRNA by intravitreal injection in cynomolgus monkeys (Tolentino, M J., et al (2004) Retina 24:132-138) and subretinal injections in mice (Reich, S J., et al (2003) Mol. Vis. 9:210-216) were both shown to prevent neovascularization in an experimental model of age-related macular degeneration. In addition, direct intratumoral injection of a dsRNA in mice reduces tumor volume (Pille, J., et al (2005) Mol. Ther. 11:267-274) and can prolong survival of tumor-bearing mice (Kim, W J., et al (2006) Mol. Ther. 14:343-350; Li, S., et al (2007) Mol. Ther. 15:515-523). RNA interference has also shown success with local delivery to the CNS by direct injection (Dorn, G., et al. (2004) Nucleic Acids 32:e49; Tan, P H., et al (2005) Gene Ther. 12:59-66; Makimura, H., et al (2002) BMC Neurosci. 3:18; Shishkina, G T., et al (2004) Neuroscience 129:521-528; Thakker, E R., et al (2004) Proc. Natl. Acad. Sci. U.S.A. 101:17270-17275; Akaneya, Y., et al (2005) J. Neurophysiol. 93:594-602) and to the lungs by intranasal administration (Howard, K A., et al (2006) Mol. Ther. 14:476-484; Zhang, X., et al (2004) J. Biol. Chem. 279:10677-10684; Bitko, V., et al (2005) Nat. Med. 11:50-55). For administering an iRNA systemically for the treatment of a disease, the RNA can be modified or alternatively delivered using a drug delivery system; both methods act to prevent the rapid degradation of the dsRNA by endo- and exo-nucleases in vivo.

Modification of the RNA or the pharmaceutical carrier can also permit targeting of the iRNA composition to the target tissue and avoid undesirable off-target effects. iRNA molecules can be modified by chemical conjugation to other groups, e.g., a lipid or carbohydrate group as described herein. Such conjugates can be used to target iRNA to particular cells, e.g., liver cells, e.g., hepatocytes. For example, GalNAc conjugates or lipid (e.g., LNP) formulations can be used to target iRNA to particular cells, e.g., liver cells, e.g., hepatocytes.

Lipophilic groups such as cholesterol to enhance cellular uptake and prevent degradation. For example, an iRNA directed against ApoB conjugated to a lipophilic cholesterol moiety was injected systemically into mice and resulted in knockdown of apoB mRNA in both the liver and jejunum (Soutschek, J., et al (2004) Nature 432:173-178). Conjugation of an iRNA to an aptamer has been shown to inhibit tumor growth and mediate tumor regression in a mouse model of prostate cancer (McNamara, J O., et al (2006) Nat. Biotechnol. 24:1005-1015). In an alternative embodiment, the iRNA can be delivered using drug delivery systems such as a nanoparticle, a dendrimer, a polymer, liposomes, or a cationic delivery system. Positively charged cationic delivery systems facilitate binding of an iRNA molecule (negatively charged) and also enhance interactions at the negatively charged cell membrane to permit efficient uptake of an iRNA by the cell. Cationic lipids, dendrimers, or polymers can either be bound to an iRNA, or induced to form a vesicle or micelle (see e.g., Kim S H., et al (2008) Journal of Controlled Release 129(2):107-116) that encases an iRNA. The formation of vesicles or micelles further prevents degradation of the iRNA when administered systemically. Methods for making and administering cationic-iRNA complexes are well within the abilities of one skilled in the art (see e.g., Sorensen, D R., et al (2003) J. Mol. Biol 327:761-766; Verma, UN., et al (2003) Clin. Cancer Res. 9:1291-1300; Arnold, A S et al (2007) J. Hypertens. 25:197-205, which are incorporated herein by reference in their entirety). Some non-limiting examples of drug delivery systems useful for systemic delivery of iRNAs include DOTAP (Sorensen, D R., et al (2003), supra; Verma, UN., et al (2003), supra), Oligofectamine, “solid nucleic acid lipid particles” (Zimmermann, T S., et al (2006) Nature 441:111-114), cardiolipin (Chien, P Y., et al (2005) Cancer Gene Ther. 12:321-328; Pal, A., et al (2005) Int J. Oncol. 26:1087-1091), polyethyleneimine (Bonnet M E., et al (2008) Pharm. Res. August 16 Epub ahead of print; Aigner, A. (2006) J. Biomed. Biotechnol. 71659), Arg-Gly-Asp (RGD) peptides (Liu, S. (2006) Mol. Pharm. 3:472-487), and polyamidoamines (Tomalia, D A., et al (2007) Biochem. Soc. Trans. 35:61-67; Yoo, H., et al (1999) Pharm. Res. 16:1799-1804). In some embodiments, an iRNA forms a complex with cyclodextrin for systemic administration. Methods for administration and pharmaceutical compositions of iRNAs and cyclodextrins can be found in U.S. Patent No. 7, 427, 605, which is herein incorporated by reference in its entirety.

Vector Encoded iRNAs

In another aspect, iRNA targeting the ALAS1 gene can be expressed from transcription units inserted into DNA or RNA vectors (see, e.g., Couture, A, et al., TIG. (1996), 12:5-10; Skillern, A., et al., International PCT Publication No. WO 00/22113, Conrad, International PCT Publication No. WO 00/22114, and Conrad, U.S. Pat. No. 6,054,299). Expression can be transient (on the order of hours to weeks) or sustained (weeks to months or longer), depending upon the specific construct used and the target tissue or cell type. These transgenes can be introduced as a linear construct, a circular plasmid, or a viral vector, which can be an integrating or non-integrating vector. The transgene can also be constructed to permit it to be inherited as an extrachromosomal plasmid (Gassmann, et al., Proc. Natl. Acad. Sci. USA (1995) 92:1292).

The individual strand or strands of an iRNA can be transcribed from a promoter on an expression vector. Where two separate strands are to be expressed to generate, for example, a dsRNA, two separate expression vectors can be co-introduced (e.g., by transfection or infection) into a target cell. Alternatively each individual strand of a dsRNA can be transcribed by promoters both of which are located on the same expression plasmid. In one embodiment, a dsRNA is expressed as an inverted repeat joined by a linker polynucleotide sequence such that the dsRNA has a stem and loop structure.

An iRNA expression vector is typically a DNA plasmid or viral vector. An expression vector compatible with eukaryotic cells, e.g., with vertebrate cells, can be used to produce recombinant constructs for the expression of an iRNA as described herein. Eukaryotic cell expression vectors are well known in the art and are available from a number of commercial sources. Typically, such vectors contain convenient restriction sites for insertion of the desired nucleic acid segment. Delivery of iRNA expressing vectors can be systemic, such as by intravenous or intramuscular administration, by administration to target cells ex-planted from the patient followed by reintroduction into the patient, or by any other means that allows for introduction into a desired target cell.

An iRNA expression plasmid can be transfected into a target cell as a complex with a cationic lipid carrier (e.g., Oligofectamine) or a non-cationic lipid-based carrier (e.g., Transit-TKO™). Multiple lipid transfections for iRNA-mediated knockdowns targeting different regions of a target RNA over a period of a week or more are also contemplated by the invention. Successful introduction of vectors into host cells can be monitored using various known methods. For example, transient transfection can be signaled with a reporter, such as a fluorescent marker, such as Green Fluorescent Protein (GFP). Stable transfection of cells ex vivo can be ensured using markers that provide the transfected cell with resistance to specific environmental factors (e.g., antibiotics and drugs), such as hygromycin B resistance.

Viral vector systems which can be utilized with the methods and compositions described herein include, but are not limited to, (a) adenovirus vectors; (b) retrovirus vectors, including but not limited to lentiviral vectors, moloney murine leukemia virus, etc.; (c) adeno-associated virus vectors; (d) herpes simplex virus vectors; (e) SV40 vectors; (f) polyoma virus vectors; (g) papilloma virus vectors; (h) picornavirus vectors; (i) pox virus vectors such as an orthopox, e.g., vaccinia virus vectors or avipox, e.g. canary pox or fowl pox; and (j) a helper-dependent or gutless adenovirus. Replication-defective viruses can also be advantageous. Different vectors will or will not become incorporated into the cells' genome. The constructs can include viral sequences for transfection, if desired. Alternatively, the construct may be incorporated into vectors capable of episomal replication, e.g. EPV and EBV vectors. Constructs for the recombinant expression of an iRNA will generally require regulatory elements, e.g., promoters, enhancers, etc., to ensure the expression of the iRNA in target cells. Other aspects to consider for vectors and constructs are further described below.

Vectors useful for the delivery of an iRNA will include regulatory elements (promoter, enhancer, etc.) sufficient for expression of the iRNA in the desired target cell or tissue. The regulatory elements can be chosen to provide either constitutive or regulated/inducible expression.

Expression of the iRNA can be precisely regulated, for example, by using an inducible regulatory sequence that is sensitive to certain physiological regulators, e.g., circulating glucose levels, or hormones (Docherty et al., 1994, FASEB J. 8:20-24). Such inducible expression systems, suitable for the control of dsRNA expression in cells or in mammals include, for example, regulation by ecdysone, by estrogen, progesterone, tetracycline, chemical inducers of dimerization, and isopropyl-β-D1-thiogalactopyranoside (IPTG). A person skilled in the art would be able to choose the appropriate regulatory/promoter sequence based on the intended use of the iRNA transgene.

In a specific embodiment, viral vectors that contain nucleic acid sequences encoding an iRNA can be used. For example, a retroviral vector can be used (see Miller et al., Meth. Enzymol. 217:581-599 (1993)). These retroviral vectors contain the components necessary for the correct packaging of the viral genome and integration into the host cell DNA. The nucleic acid sequences encoding an iRNA are cloned into one or more vectors, which facilitates delivery of the nucleic acid into a patient. More detail about retroviral vectors can be found, for example, in Boesen et al., Biotherapy 6:291-302 (1994), which describes the use of a retroviral vector to deliver the mdr1 gene to hematopoietic stem cells in order to make the stem cells more resistant to chemotherapy. Other references illustrating the use of retroviral vectors in gene therapy are: Clowes et al., J. Clin. Invest. 93:644-651 (1994); Kiem et al., Blood 83:1467-1473 (1994); Salmons and Gunzberg, Human Gene Therapy 4:129-141 (1993); and Grossman and Wilson, Curr. Opin. in Genetics and Devel. 3:110-114 (1993). Lentiviral vectors contemplated for use include, for example, the HIV based vectors described in U.S. Pat. Nos. 6,143,520; 5,665,557; and 5,981,276, which are herein incorporated by reference. Adenoviruses are also contemplated for use in delivery of iRNAs. Adenoviruses are especially attractive vehicles, e.g., for delivering genes to respiratory epithelia. Adenoviruses naturally infect respiratory epithelia where they cause a mild disease. Other targets for adenovirus-based delivery systems are liver, the central nervous system, endothelial cells, and muscle. Adenoviruses have the advantage of being capable of infecting non-dividing cells. Kozarsky and Wilson, Current Opinion in Genetics and Development 3:499-503 (1993) present a review of adenovirus-based gene therapy. Bout et al., Human Gene Therapy 5:3-10 (1994) demonstrated the use of adenovirus vectors to transfer genes to the respiratory epithelia of rhesus monkeys. Other instances of the use of adenoviruses in gene therapy can be found in Rosenfeld et al., Science 252:431-434 (1991); Rosenfeld et al., Cell 68:143-155 (1992); Mastrangeli et al., J. Clin. Invest. 91:225-234 (1993); PCT Publication WO94/12649; and Wang, et al., Gene Therapy 2:775-783 (1995). A suitable AV vector for expressing an iRNA featured in the invention, a method for constructing the recombinant AV vector, and a method for delivering the vector into target cells, are described in Xia H et al. (2002), Nat. Biotech. 20: 1006-1010.

Use of Adeno-associated virus (AAV) vectors is also contemplated (Walsh et al., Proc. Soc. Exp. Biol. Med. 204:289-300 (1993); U.S. Pat. No. 5,436,146). In one embodiment, the iRNA can be expressed as two separate, complementary single-stranded RNA molecules from a recombinant AAV vector having, for example, either the U6 or H1 RNA promoters, or the cytomegalovirus (CMV) promoter. Suitable AAV vectors for expressing the dsRNA featured in the invention, methods for constructing the recombinant AV vector, and methods for delivering the vectors into target cells are described in Samulski R et al. (1987), J. Virol. 61: 3096-3101; Fisher K J et al. (1996), J. Virol, 70: 520-532; Samulski R et al. (1989), J. Virol. 63: 3822-3826; U.S. Pat. Nos. 5,252,479; 5,139,941; International Patent Application No. WO 94/13788; and International Patent Application No. WO 93/24641, the entire disclosures of which are herein incorporated by reference.

Another typical viral vector is a pox virus such as a vaccinia virus, for example an attenuated vaccinia such as Modified Virus Ankara (MVA) or NYVAC, an avipox such as fowl pox or canary pox.

The tropism of viral vectors can be modified by pseudotyping the vectors with envelope proteins or other surface antigens from other viruses, or by substituting different viral capsid proteins, as appropriate. For example, lentiviral vectors can be pseudotyped with surface proteins from vesicular stomatitis virus (VSV), rabies, Ebola, Mokola, and the like. AAV vectors can be made to target different cells by engineering the vectors to express different capsid protein serotypes; see, e.g., Rabinowitz J E et al. (2002), J Virol 76:791-801, the entire disclosure of which is herein incorporated by reference.

The pharmaceutical preparation of a vector can include the vector in an acceptable diluent, or can include a slow release matrix in which the gene delivery vehicle is imbedded. Alternatively, where the complete gene delivery vector can be produced intact from recombinant cells, e.g., retroviral vectors, the pharmaceutical preparation can include one or more cells which produce the gene delivery system.

III. Pharmaceutical Compositions Containing iRNA

In one embodiment, the invention provides pharmaceutical compositions containing an iRNA, as described herein, and a pharmaceutically acceptable carrier. The pharmaceutical composition containing the iRNA is useful for treating a disease or disorder related to the expression or activity of an ALAS1 gene (e.g., a disorder involving the porphyrin pathway). Such pharmaceutical compositions are formulated based on the mode of delivery. For example, compositions can be formulated for systemic administration via parenteral delivery, e.g., by intravenous (IV) delivery. In some embodiments, a composition provided herein (e.g., an LNP formulation) is formulated for intravenous delivery. In some embodiments, a composition provided herein (e.g., a composition comprising a GalNAc conjugate) is formulated for subcutaneous delivery.

The pharmaceutical compositions featured herein are administered in a dosage sufficient to inhibit expression of an ALAS1 gene. In general, a suitable dose of iRNA will be in the range of 0.01 to 200.0 milligrams per kilogram body weight of the recipient per day, generally in the range of 1 to 50 mg per kilogram body weight per day. For example, the dsRNA can be administered at 0.05 mg/kg, 0.5 mg/kg, 1 mg/kg, 1.5 mg/kg, 2 mg/kg, 3 mg/kg, 10 mg/kg, 20 mg/kg, 30 mg/kg, 40 mg/kg, or 50 mg/kg per single dose. The pharmaceutical composition may be administered once daily, or the iRNA may be administered as two, three, or more sub-doses at appropriate intervals throughout the day or even using continuous infusion or delivery through a controlled release formulation. In that case, the iRNA contained in each sub-dose must be correspondingly smaller in order to achieve the total daily dosage. The dosage unit can also be compounded for delivery over several days, e.g., using a conventional sustained release formulation which provides sustained release of the iRNA over a several day period. Sustained release formulations are well known in the art and are particularly useful for delivery of agents at a particular site, such as can be used with the agents of the present invention. In this embodiment, the dosage unit contains a corresponding multiple of the daily dose.

The effect of a single dose on ALAS1 levels can be long lasting, such that subsequent doses are administered at not more than 3, 4, or 5 day intervals, or at not more than 1, 2, 3, or 4 week intervals.

The skilled artisan will appreciate that certain factors may influence the dosage and timing 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 composition can include a single treatment or a series of treatments. Estimates of effective dosages and in vivo half-lives for the individual iRNAs encompassed by the invention can be made using conventional methodologies or on the basis of in vivo testing using an appropriate animal model, as described elsewhere herein.

Advances in mouse genetics have generated a number of mouse models for the study of various human diseases, such as pathological processes related to ALAS1 expression (e.g., pathological processes involving porphyrins or defects in the porphyrin pathway, such as, for example, porphyrias). Such models can be used for in vivo testing of iRNA, as well as for determining a therapeutically effective dose and/or an effective dosing regimen.

A suitable mouse model is, for example, a mouse containing a transgene expressing human ALAS1. Mice that have knock-in mutations (e.g., mutations that are associated with acute hepatic porphyrias in humans) can be used to determine the therapeutically effective dosage and/or duration of administration of ALAS1 siRNA. The present invention also includes pharmaceutical compositions and formulations that include the iRNA compounds featured in the invention. The pharmaceutical compositions of the present invention may be administered in a number of ways depending upon whether local or systemic treatment is desired and upon the area to be treated. Administration may be topical (e.g., by a transdermal patch), pulmonary, e.g., by inhalation or insufflation of powders or aerosols, including by nebulizer; intratracheal, intranasal, epidermal and transdermal, oral or parenteral. Parenteral administration includes intravenous, intraarterial, subcutaneous, intraperitoneal or intramuscular injection or infusion; subdermal, e.g., via an implanted device; or intracranial, e.g., by intraparenchymal, intrathecal or intraventricular, administration.

The iRNA can be delivered in a manner to target a particular tissue, such as a tissue that produces erythrocytes. For example, the iRNA can be delivered to bone marrow, liver (e.g., hepatocyes of liver), lymph glands, spleen, lungs (e.g., pleura of lungs) or spine. In one embodiment, the iRNA is delivered to bone marrow.

Pharmaceutical compositions and formulations for topical administration may include transdermal patches, ointments, lotions, creams, gels, drops, suppositories, sprays, liquids and powders. Conventional pharmaceutical carriers, aqueous, powder or oily bases, thickeners and the like may be necessary or desirable. Coated condoms, gloves and the like may also be useful. Suitable topical formulations include those in which the iRNAs featured in the invention are in admixture with a topical delivery agent such as lipids, liposomes, fatty acids, fatty acid esters, steroids, chelating agents and surfactants. Suitable lipids and liposomes include neutral (e.g., dioleoylphosphatidyl DOPE ethanolamine, dimyristoylphosphatidyl choline DMPC, distearolyphosphatidyl choline) negative (e.g., dimyristoylphosphatidyl glycerol DMPG) and cationic (e.g., dioleoyltetramethylaminopropyl DOTAP and dioleoylphosphatidyl ethanolamine DOTMA). iRNAs featured in the invention may be encapsulated within liposomes or may form complexes thereto, in particular to cationic liposomes. Alternatively, iRNAs may be complexed to lipids, in particular to cationic lipids. Suitable fatty acids and esters include but are not limited to arachidonic acid, oleic acid, eicosanoic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a C1-20 alkyl ester (e.g., isopropylmyristate IPM), monoglyceride, diglyceride or pharmaceutically acceptable salt thereof. Topical formulations are described in detail in U.S. Pat. No. 6,747,014, which is incorporated herein by reference.

There are many organized surfactant structures besides microemulsions that have been studied and used for the formulation of drugs. These include monolayers, micelles, bilayers and vesicles. Vesicles, such as liposomes, have attracted great interest because of their specificity and the duration of action they offer from the standpoint of drug delivery. As used in the present invention, the term “liposome” means a vesicle composed of amphiphilic lipids arranged in a spherical bilayer or bilayers.

Liposomes are unilamellar or multilamellar vesicles which have a membrane formed from a lipophilic material and an aqueous interior. The aqueous portion contains the composition to be delivered. Cationic liposomes possess the advantage of being able to fuse to the cell wall. Non-cationic liposomes, although not able to fuse as efficiently with the cell wall, are taken up by macrophages in vivo.

In order to traverse intact mammalian skin, lipid vesicles must pass through a series of fine pores, each with a diameter less than 50 nm, under the influence of a suitable transdermal gradient. Therefore, it is desirable to use a liposome which is highly deformable and able to pass through such fine pores.

Further advantages of liposomes include; liposomes obtained from natural phospholipids are biocompatible and biodegradable; liposomes can incorporate a wide range of water and lipid soluble drugs; liposomes can protect encapsulated drugs in their internal compartments from metabolism and degradation (Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245). Important considerations in the preparation of liposome formulations are the lipid surface charge, vesicle size and the aqueous volume of the liposomes.

Liposomes are useful for the transfer and delivery of active ingredients to the site of action. Because the liposomal membrane is structurally similar to biological membranes, when liposomes are applied to a tissue, the liposomes start to merge with the cellular membranes and as the merging of the liposome and cell progresses, the liposomal contents are emptied into the cell where the active agent may act.

Liposomal formulations have been the focus of extensive investigation as the mode of delivery for many drugs. There is growing evidence that for topical administration, liposomes present several advantages over other formulations. Such advantages include reduced side-effects related to high systemic absorption of the administered drug, increased accumulation of the administered drug at the desired target, and the ability to administer a wide variety of drugs, both hydrophilic and hydrophobic, into the skin.

Several reports have detailed the ability of liposomes to deliver agents including high-molecular weight DNA into the skin. Compounds including analgesics, antibodies, hormones and high-molecular weight DNAs have been administered to the skin. The majority of applications resulted in the targeting of the upper epidermis

Liposomes fall into two broad classes. Cationic liposomes are positively charged liposomes which interact with the negatively charged DNA molecules to form a stable complex. The positively charged DNA/liposome complex binds to the negatively charged cell surface and is internalized in an endosome. Due to the acidic pH within the endosome, the liposomes are ruptured, releasing their contents into the cell cytoplasm (Wang et al., Biochem. Biophys. Res. Commun., 1987, 147, 980-985).

Liposomes which are pH-sensitive or negatively-charged, entrap DNA rather than complex with it. Since both the DNA and the lipid are similarly charged, repulsion rather than complex formation occurs. Nevertheless, some DNA is entrapped within the aqueous interior of these liposomes. pH-sensitive liposomes have been used to deliver DNA encoding the thymidine kinase gene to cell monolayers in culture. Expression of the exogenous gene was detected in the target cells (Zhou et al., Journal of Controlled Release, 1992, 19, 269-274).

One major type of liposomal composition includes phospholipids other than naturally-derived phosphatidylcholine. Neutral liposome compositions, for example, can be formed from dimyristoyl phosphatidylcholine (DMPC) or dipalmitoyl phosphatidylcholine (DPPC). Anionic liposome compositions generally are formed from dimyristoyl phosphatidylglycerol, while anionic fusogenic liposomes are formed primarily from dioleoyl phosphatidylethanolamine (DOPE). Another type of liposomal composition is formed from phosphatidylcholine (PC) such as, for example, soybean PC, and egg PC. Another type is formed from mixtures of phospholipid and/or phosphatidylcholine and/or cholesterol.

Several studies have assessed the topical delivery of liposomal drug formulations to the skin. Application of liposomes containing interferon to guinea pig skin resulted in a reduction of skin herpes sores while delivery of interferon via other means (e.g., as a solution or as an emulsion) were ineffective (Weiner et al., Journal of Drug Targeting, 1992, 2, 405-410). Further, an additional study tested the efficacy of interferon administered as part of a liposomal formulation to the administration of interferon using an aqueous system, and concluded that the liposomal formulation was superior to aqueous administration (du Plessis et al., Antiviral Research, 1992, 18, 259-265).

Non-ionic liposomal systems have also been examined to determine their utility in the delivery of drugs to the skin, in particular systems comprising non-ionic surfactant and cholesterol. Non-ionic liposomal formulations comprising Novasome™ I (glyceryl dilaurate/cholesterol/polyoxyethylene-10-stearyl ether) and Novasome™ II (glyceryl distearate/cholesterol/polyoxyethylene-10-stearyl ether) were used to deliver cyclosporin-A into the dermis of mouse skin. Results indicated that such non-ionic liposomal systems were effective in facilitating the deposition of cyclosporin-A into different layers of the skin (Hu et al. S.T.P. Pharma. Sci., 1994, 4, 6, 466).

Liposomes also include “sterically stabilized” liposomes, a term which, as used herein, refers to liposomes comprising one or more specialized lipids that, when incorporated into liposomes, result in enhanced circulation lifetimes relative to liposomes lacking such specialized lipids. Examples of sterically stabilized liposomes are those in which part of the vesicle-forming lipid portion of the liposome (A) comprises one or more glycolipids, such as monosialoganglioside GM1, or (B) is derivatized with one or more hydrophilic polymers, such as a polyethylene glycol (PEG) moiety. While not wishing to be bound by any particular theory, it is thought in the art that, at least for sterically stabilized liposomes containing gangliosides, sphingomyelin, or PEG-derivatized lipids, the enhanced circulation half-life of these sterically stabilized liposomes derives from a reduced uptake into cells of the reticuloendothelial system (RES) (Allen et al., FEBS Letters, 1987, 223, 42; Wu et al., Cancer Research, 1993, 53, 3765).

Various liposomes comprising one or more glycolipids are known in the art. Papahadjopoulos et al. (Ann. N.Y. Acad. Sci., 1987, 507, 64) reported the ability of monosialoganglioside GM1, galactocerebroside sulfate and phosphatidylinositol to improve blood half-lives of liposomes. These findings were expounded upon by Gabizon et al. (Proc. Natl. Acad. Sci. U.S.A., 1988, 85, 6949). U.S. Pat. No. 4,837,028 and WO 88/04924, both to Allen et al., disclose liposomes comprising (1) sphingomyelin and (2) the ganglioside GM1 or a galactocerebroside sulfate ester. U.S. Pat. No. 5,543,152 (Webb et al.) discloses liposomes comprising sphingomyelin. Liposomes comprising 1,2-sn-dimyristoylphosphatidylcholine are disclosed in WO 97/13499 (Lim et al).

Many liposomes comprising lipids derivatized with one or more hydrophilic polymers, and methods of preparation thereof, are known in the art. Sunamoto et al. (Bull. Chem. Soc. Jpn., 1980, 53, 2778) described liposomes comprising a nonionic detergent, 2C1215G, that contains a PEG moiety. Illum et al. (FEBS Lett., 1984, 167, 79) noted that hydrophilic coating of polystyrene particles with polymeric glycols results in significantly enhanced blood half-lives. Synthetic phospholipids modified by the attachment of carboxylic groups of polyalkylene glycols (e.g., PEG) are described by Sears (U.S. Pat. Nos. 4,426,330 and 4,534,899). Klibanov et al. (FEBS Lett., 1990, 268, 235) described experiments demonstrating that liposomes comprising phosphatidylethanolamine (PE) derivatized with PEG or PEG stearate have significant increases in blood circulation half-lives. Blume et al. (Biochimica et Biophysica Acta, 1990, 1029, 91) extended such observations to other PEG-derivatized phospholipids, e.g., DSPE-PEG, formed from the combination of distearoylphosphatidylethanolamine (DSPE) and PEG. Liposomes having covalently bound PEG moieties on their external surface are described in European Patent No. EP 0 445 131 B1 and WO 90/04384 to Fisher. Liposome compositions containing 1-20 mole percent of PE derivatized with PEG, and methods of use thereof, are described by Woodle et al. (U.S. Pat. Nos. 5,013,556 and 5,356,633) and Martin et al. (U.S. Pat. No. 5,213,804 and European Patent No. EP 0 496 813 B1). Liposomes comprising a number of other lipid-polymer conjugates are disclosed in WO 91/05545 and U.S. Pat. No. 5,225,212 (both to Martin et al.) and in WO 94/20073 (Zalipsky et al.) Liposomes comprising PEG-modified ceramide lipids are described in WO 96/10391 (Choi et al). U.S. Pat. No. 5,540,935 (Miyazaki et al.) and U.S. Pat. No. 5,556,948 (Tagawa et al.) describe PEG-containing liposomes that can be further derivatized with functional moieties on their surfaces.

A number of liposomes comprising nucleic acids are known in the art. WO 96/40062 to Thierry et al. discloses methods for encapsulating high molecular weight nucleic acids in liposomes. U.S. Pat. No. 5,264,221 to Tagawa et al. discloses protein-bonded liposomes and asserts that the contents of such liposomes may include a dsRNA. U.S. Pat. No. 5,665,710 to Rahman et al. describes certain methods of encapsulating oligodeoxynucleotides in liposomes. WO 97/04787 to Love et al. discloses liposomes comprising dsRNAs targeted to the raf gene.

Transfersomes are yet another type of liposomes, and are highly deformable lipid aggregates which are attractive candidates for drug delivery vehicles. Transfersomes may be described as lipid droplets which are so highly deformable that they are easily able to penetrate through pores which are smaller than the droplet. Transfersomes are adaptable to the environment in which they are used, e.g., they are self-optimizing (adaptive to the shape of pores in the skin), self-repairing, frequently reach their targets without fragmenting, and often self-loading. To make transfersomes it is possible to add surface edge-activators, usually surfactants, to a standard liposomal composition. Transfersomes have been used to deliver serum albumin to the skin. The transfersome-mediated delivery of serum albumin has been shown to be as effective as subcutaneous injection of a solution containing serum albumin.

Surfactants find wide application in formulations such as emulsions (including microemulsions) and liposomes. The most common way of classifying and ranking the properties of the many different types of surfactants, both natural and synthetic, is by the use of the hydrophile/lipophile balance (HLB). The nature of the hydrophilic group (also known as the “head”) provides the most useful means for categorizing the different surfactants used in formulations (Rieger, in Pharmaceutical Dosage Forms, Marcel Dekker, Inc., New York, N.Y., 1988, p. 285).

If the surfactant molecule is not ionized, it is classified as a nonionic surfactant. Nonionic surfactants find wide application in pharmaceutical and cosmetic products and are usable over a wide range of pH values. In general their HLB values range from 2 to about 18 depending on their structure. Nonionic surfactants include nonionic esters such as ethylene glycol esters, propylene glycol esters, glyceryl esters, polyglyceryl esters, sorbitan esters, sucrose esters, and ethoxylated esters. Nonionic alkanolamides and ethers such as fatty alcohol ethoxylates, propoxylated alcohols, and ethoxylated/propoxylated block polymers are also included in this class. The polyoxyethylene surfactants are the most popular members of the nonionic surfactant class.

If the surfactant molecule carries a negative charge when it is dissolved or dispersed in water, the surfactant is classified as anionic. Anionic surfactants include carboxylates such as soaps, acyl lactylates, acyl amides of amino acids, esters of sulfuric acid such as alkyl sulfates and ethoxylated alkyl sulfates, sulfonates such as alkyl benzene sulfonates, acyl isethionates, acyl taurates and sulfosuccinates, and phosphates. The most important members of the anionic surfactant class are the alkyl sulfates and the soaps.

If the surfactant molecule carries a positive charge when it is dissolved or dispersed in water, the surfactant is classified as cationic. Cationic surfactants include quaternary ammonium salts and ethoxylated amines. The quaternary ammonium salts are the most used members of this class.

If the surfactant molecule has the ability to carry either a positive or negative charge, the surfactant is classified as amphoteric. Amphoteric surfactants include acrylic acid derivatives, substituted alkylamides, N-alkylbetaines and phosphatides.

The use of surfactants in drug products, formulations and in emulsions has been reviewed (Rieger, in Pharmaceutical Dosage Forms, Marcel Dekker, Inc., New York, N.Y., 1988, p. 285).

In one embodiment, an ALAS1 dsRNA featured in the invention is fully encapsulated in the lipid formulation, e.g., to form a SPLP, pSPLP, SNALP, or other nucleic acid-lipid particle. As used herein, the term “SNALP” refers to a stable nucleic acid-lipid particle, including SPLP. As used herein, the term “SPLP” refers to a nucleic acid-lipid particle comprising plasmid DNA encapsulated within a lipid vesicle. SNALPs and SPLPs typically contain a cationic lipid, a non-cationic lipid, and a lipid that prevents aggregation of the particle (e.g., a PEG-lipid conjugate). SNALPs and SPLPs are extremely useful for systemic applications, as they exhibit extended circulation lifetimes following intravenous (i.v.) injection and accumulate at distal sites (e.g., sites physically separated from the administration site). SPLPs include “pSPLP,” which include an encapsulated condensing agent-nucleic acid complex as set forth in PCT Publication No. WO 00/03683. The particles of the present invention typically have a mean diameter of about 50 nm to about 150 nm, more typically about 60 nm to about 130 nm, more typically about 70 nm to about 110 nm, most typically about 70 nm to about 90 nm, and are substantially nontoxic. In addition, the nucleic acids when present in the nucleic acid-lipid particles of the present invention are resistant in aqueous solution to degradation with a nuclease. Nucleic acid-lipid particles and their method of preparation are disclosed in, e.g., U.S. Pat. Nos. 5,976,567; 5,981,501; 6,534,484; 6,586,410; 6,815,432; and PCT Publication No. WO 96/40964.

In one embodiment, the lipid to drug ratio (mass/mass ratio) (e.g., lipid to dsRNA ratio) will be in the range of from about 1:1 to about 50:1, from about 1:1 to about 25:1, from about 3:1 to about 15:1, from about 4:1 to about 10:1, from about 5:1 to about 9:1, or about 6:1 to about 9:1.

The cationic lipid may be, for example, N,N-dioleyl-N,N-dimethylammonium chloride (DODAC), N,N-distearyl-N,N-dimethylammonium bromide (DDAB), N-(I-(2,3-dioleoyloxy)propyl)-N,N,N-trimethylammonium chloride (DOTAP), N-(I-(2,3-dioleyloxy)propyl)-N,N,N-trimethylammonium chloride (DOTMA), N,N-dimethyl-2,3-dioleyloxy)propylamine (DODMA), 1,2-DiLinoleyloxy-N,N-dimethylaminopropane (DLinDMA), 1,2-Dilinolenyloxy-N,N-dimethylaminopropane (DLenDMA), 1,2-Dilinoleylcarbamoyloxy-3-dimethylaminopropane (DLin-C-DAP), 1,2-Dilinoleyoxy-3-(dimethylamino)acetoxypropane (DLin-DAC), 1,2-Dilinoleyoxy-3-morpholinopropane (DLin-MA), 1,2-Dilinoleoyl-3-dimethylaminopropane (DLinDAP), 1,2-Dilinoleylthio-3-dimethylaminopropane (DLin-S-DMA), 1-Linoleoyl-2-linoleyloxy-3-dimethylaminopropane (DLin-2-DMAP), 1,2-Dilinoleyloxy-3-trimethylaminopropane chloride salt (DLin-TMA.Cl), 1,2-Dilinoleoyl-3-trimethylaminopropane chloride salt (DLin-TAP.Cl), 1,2-Dilinoleyloxy-3-(N-methylpiperazino)propane (DLin-MPZ), or 3-(N,N-Dilinoleylamino)-1,2-propanediol (DLinAP), 3-(N,N-Dioleylamino)-1,2-propanedio (DOAP), 1,2-Dilinoleyloxo-3-(2-N,N-dimethylamino)ethoxypropane (DLin-EG-DMA), 1,2-Dilinolenyloxy-N,N-dimethylaminopropane (DLinDMA), 2,2-Dilinoleyl-4-dimethylaminomethyl-[1,3]-dioxolane (DLin-K-DMA) or analogs thereof, (3aR,5s,6aS)-N,N-dimethyl-2,2-di((9Z,12Z)-octadeca-9,12-dienyl)tetrahydro-3aH-cyclopenta[d][1,3]dioxol-5-amine (ALN100), (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl 4-(dimethylamino)butanoate (MC3), 1,1′-(2-(4-(2-((2-(bis(2-hydroxydodecyl)amino)ethyl)(2-hydroxydodecyl)amino)ethyl)piperazin-1-yl)ethylazanediyl)didodecan-2-ol (Tech G1), or a mixture thereof. The cationic lipid may comprise from about 20 mol % to about 50 mol % or about 40 mol % of the total lipid present in the particle.

In another embodiment, the compound 2,2-Dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane can be used to prepare lipid-siRNA nanoparticles. Synthesis of 2,2-Dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane is described in U.S. provisional patent application No. 61/107,998 filed on Oct. 23, 2008, which is herein incorporated by reference.

In one embodiment, the lipid-siRNA particle includes 40% 2, 2-Dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane: 10% DSPC: 40% Cholesterol: 10% PEG-C-DOMG (mole percent) with a particle size of 63.0±20 nm and a 0.027 siRNA/Lipid Ratio.

The non-cationic lipid may be an anionic lipid or a neutral lipid including, but not limited to, distearoylphosphatidylcholine (DSPC), dioleoylphosphatidylcholine (DOPC), dipalmitoylphosphatidylcholine (DPPC), dioleoylphosphatidylglycerol (DOPG), dipalmitoylphosphatidylglycerol (DPPG), dioleoyl-phosphatidylethanolamine (DOPE), palmitoyloleoylphosphatidylcholine (POPC), palmitoyloleoylphosphatidylethanolamine (POPE), dioleoyl-phosphatidylethanolamine 4-(N-maleimidomethyl)-cyclohexane-l-carboxylate (DOPE-mal), dipalmitoyl phosphatidyl ethanolamine (DPPE), dimyristoylphosphoethanolamine (DMPE), distearoyl-phosphatidyl-ethanolamine (DSPE), 16-O-monomethyl PE, 16-O-dimethyl PE, 18-1-trans PE, 1-stearoyl-2-oleoyl-phosphatidyethanolamine (SOPE), cholesterol, or a mixture thereof. The non-cationic lipid may be from about 5 mol % to about 90 mol %, about 10 mol %, or about 58 mol % if cholesterol is included, of the total lipid present in the particle.

The conjugated lipid that inhibits aggregation of particles may be, for example, a polyethyleneglycol (PEG)-lipid including, without limitation, a PEG-diacylglycerol (DAG), a PEG-dialkyloxypropyl (DAA), a PEG-phospholipid, a PEG-ceramide (Cer), or a mixture thereof. The PEG-DAA conjugate may be, for example, a PEG-dilauryloxypropyl (Ci2), a PEG-dimyristyloxypropyl (Ci4), a PEG-dipalmityloxypropyl (Ci6), or a PEG-distearyloxypropyl (C]8). The conjugated lipid that prevents aggregation of particles may be from 0 mol % to about 20 mol % or about 2 mol % of the total lipid present in the particle.

In some embodiments, the nucleic acid-lipid particle further includes cholesterol at, e.g., about 10 mol % to about 60 mol % or about 48 mol % of the total lipid present in the particle.

In some embodiments, the iRNA is formulated in a lipid nanoparticle (LNP).

In one embodiment, the lipidoid ND98⋅4HCl (MW 1487) (see U.S. patent application Ser. No. 12/056,230, filed Mar. 26, 2008, which is herein incorporated by reference), Cholesterol (Sigma-Aldrich), and PEG-Ceramide C16 (Avanti Polar Lipids) can be used to prepare lipid-dsRNA nanoparticles (e.g., LNP01 particles). Stock solutions of each in ethanol can be prepared as follows: ND98, 133 mg/ml; Cholesterol, 25 mg/ml, PEG-Ceramide C16, 100 mg/ml. The ND98, Cholesterol, and PEG-Ceramide C16 stock solutions can then be combined in a, e.g., 42:48:10 molar ratio. The combined lipid solution can be mixed with aqueous dsRNA (e.g., in sodium acetate pH 5) such that the final ethanol concentration is about 35-45% and the final sodium acetate concentration is about 100-300 mM. Lipid-dsRNA nanoparticles typically form spontaneously upon mixing. Depending on the desired particle size distribution, the resultant nanoparticle mixture can be extruded through a polycarbonate membrane (e.g., 100 nm cut-off) using, for example, a thermobarrel extruder, such as Lipex Extruder (Northern Lipids, Inc). In some cases, the extrusion step can be omitted. Ethanol removal and simultaneous buffer exchange can be accomplished by, for example, dialysis or tangential flow filtration. Buffer can be exchanged with, for example, phosphate buffered saline (PBS) at about pH 7, e.g., about pH 6.9, about pH 7.0, about pH 7.1, about pH 7.2, about pH 7.3, or about pH 7.4.

LNP01 formulations are described, e.g., in International Application Publication No. WO 2008/042973, which is hereby incorporated by reference.

Additional exemplary lipid-dsRNA formulations are provided in the following table.

TABLE 10
Examplary lipid formulations
cationic lipid/non-cationic
lipid/cholesterol/PEG-lipid conjugate
Cationic LipidLipid:siRNA ratio
SNALP1,2-Dilinolenyloxy-N,N-DLinDMA/DPPC/Cholesterol/PEG-cDMA
dimethylaminopropane (DLinDMA)(57.1/7.1/34.4/1.4)
lipid: siRNA~7:1
S-XTC2,2-Dilinoleyl-4-dimethylaminoethyl-XTC/DPPC/Cholesterol/PEG-cDMA
[1,3]-dioxolane (XTC)57.1/7.1/34.4/1.4
lipid: siRNA~7:1
LNP052,2-Dilinoleyl-4-dimethylaminoethyl-XTC/DSPC/Cholesterol/PEG-DMG
[1,3]-dioxo1ane (XTC)57.5/7.5/31.5/3.5
lipid: siRNA~6:1
LNP062,2-Dilinoleyl-4-dimethylaminoethyl-XTC/DSPC/Cholesterol/PEG-DMG
[1,3]-dioxolane (XTC)57.5/7.5/31.5/3.5
lipid:siRNA~11:1
LNP072,2-Dilinoleyl-4-dimethylaminoethyl-XTC/DSPC/Cholesterol/PEG-DMG
[1,3]-dioxolane (XTC)60/7.5/31/1.5,
lipid: siRNA~6:1
LNP082,2-Dilinoleyl-4-dimethylaminoethyl-XTC/DSPC/Cholesterol/PEG-DMG
[1,3]-dioxolane (XTC)60/7.5/31/1.5,
lipid:siRNA~11:1
LNP092,2-Dilinoleyl-4-dimethylaminoethyl-XTC/DSPC/Cholesterol/PEG-DMG
[1,3]-dioxolane (XTC)50/10/38.5/1.5
Lipid:siRNA 10:1
LNP10(3aR,5s,6aS)-N,N-dimethyl-2,2-ALN100/DSPC/Cholesterol/PEG-DMG
di((9Z,12Z)-octadeca-9,12-50/10/38.5/1.5
dienyl)tetrahydro-3aH-Lipid: siRNA 10:1
cyclopenta[d][1,3]dioxo1-5-amine
(ALN100)
LNP11(6Z,9Z,28Z,31Z)-heptatriaconta-MC-3/DSPC/Cholesterol/PEG-DMG
6,9,28,31-tetraen-19-yl 4-50/10/38.5/1.5
(dimethylamino)butanoate (MC3)Lipid:siRNA 10:1
LNP121,1′-(2-(4-(2-((2-(bis(2-C12-200/DSPC/Cholesterol/PEG-DMG
hydroxydodecyl)amino)ethyl)(2-50/10/38.5/1.5
hydroxydodecyl)amino)ethyl)piperazin-Lipid:siRNA 10:1
1-yl)ethylazanediyl)didodecan-2-ol
(C12-200)
LNP13XTCXTC/DSPC/Chol/PEG-DMG
50/10/38.5/1.5
Lipid:siRNA: 33:1
LNP14MC3MC3/DSPC/Chol/PEG-DMG
40/15/40/5
Lipid:siRNA: 11:1
LNP15MC3MC3/DSPC/Chol/PEG-DSG/Ga1NAc-
PEG-DSG
50/10/35/4.5/0.5
Lipid:siRNA: 11:1
LNP16MC3MC3/DSPC/Chol/PEG-DMG
50/10/38.5/1.5
Lipid:siRNA: 7:1
LNP17MC3MC3/DSPC/Chol/PEG-DSG
50/10/38.5/1.5
Lipid:siRNA: 10:1
LNP18MC3MC3/DSPC/Chol/PEG-DMG
50/10/38.5/1.5
Lipid:siRNA: 12:1
LNP19MC3MC3/DSPC/Chol/PEG-DMG
50/10/35/5
Lipid:siRNA: 8:1
LNP20MC3MC3/DSPC/Chol/PEG-DPG
50/10/38.5/1.5
Lipid:siRNA: 10:1
LNP21C12-200C12-200/DSPC/Chol/PEG-DSG
50/10/38.5/1.5
Lipid:siRNA: 7:1
LNP22XTCXTC/DSPC/Chol/PEG-DSG
50/10/38.5/1.5
Lipid:siRNA: 10:1
DSPC: distearoylphosphatidylcholine
DPPC: dipalmitoylphosphatidylcholine
PEG-DMG: PEG-didimyristoyl glycerol (C14-PEG, or PEG-C14) (PEG with avg mol wt of 2000)
PEG-DSG: PEG-distyryl glycerol (C18-PEG, or PEG-C18) (PEG with avg mol wt of 2000)
PEG-cDMA: PEG-carbamoyl-1,2-dimyristyloxypropylamine (PEG with avg mol wt of 2000)
SNALP (1,2-Dilinolenyloxy-N,N-dimethylaminopropane (DLinDMA)) comprising formulations are described in International Publication No. W02009/127060, filed Apr. 15, 2009, which is hereby incorporated by reference.
XTC comprising formulations are described, e.g., in U.S. Provisional Serial No. 61/148,366, filed Jan. 29, 2009; U.S. Provisional Serial No. 61/156,851, filed Mar. 2, 2009; U.S. Provisional Serial No. filed Jun. 10, 2009; U.S. Provisional Serial No. 61/228,373, filed Jul. 24, 2009; U.S. Provisional Serial No. 61/239,686, filed Sep. 3, 2009, and International Application No. PCT/U52010/022614, filed Jan. 29, 2010, which are hereby incorporated by reference.
MC3 comprising formulations are described, e.g., in U.S. Provisional Serial No. 61/244,834, filed Sep. 22, 2009, U.S. Provisional Serial No. 61/185,800, filed Jun. 10, 2009, and International Application No. PCT/US10/28224, filed Jun. 10, 2010, which are hereby incorporated by reference.
ALNY-100 comprising formulations are described, e.g., International patent application number PCT/U509/63933, filed on Nov. 10, 2009, which is hereby incorporated by reference.
C12-200 comprising formulations are described in U.S. Provisional Serial No. 61/175,770, filed May 5, 2009 and International Application No. PCT/US10/33777, filed May 5, 2010, which are hereby incorporated by reference.

Any of the compounds, e.g., cationic lipids and the like, used in the nucleic acid-lipid particles featured in the invention may be prepared by known organic synthesis techniques, including the methods described in more detail in the Examples. All substituents are as defined below unless indicated otherwise.

“Alkyl” means a straight chain or branched, noncyclic or cyclic, saturated aliphatic hydrocarbon containing from 1 to 24 carbon atoms. Representative saturated straight chain alkyls include methyl, ethyl, n-propyl, n-butyl, n-pentyl, n-hexyl, and the like; while saturated branched alkyls include isopropyl, see-butyl, isobutyl, tert-butyl, isopentyl, and the like. Representative saturated cyclic alkyls include cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, and the like; while unsaturated cyclic alkyls include cyclopentenyl and cyclohexenyl, and the like.

“Alkenyl” means an alkyl, as defined above, containing at least one double bond between adjacent carbon atoms. Alkenyls include both cis and trans isomers. Representative straight chain and branched alkenyls include ethylenyl, propylenyl, 1-butenyl, 2-butenyl, isobutylenyl, 1-pentenyl, 2-pentenyl, 3-methyl-1-butenyl, 2-methyl-2-butenyl, 2,3-dimethyl-2-butenyl, and the like.

“Alkynyl” means any alkyl or alkenyl, as defined above, which additionally contains at least one triple bond between adjacent carbons. Representative straight chain and branched alkynyls include acetylenyl, propynyl, 1-butynyl, 2-butynyl, 1-pentynyl, 2-pentynyl, 3-methyl-1 butynyl, and the like.

“Acyl” means any alkyl, alkenyl, or alkynyl wherein the carbon at the point of attachment is substituted with an oxo group, as defined below. For example, —C(═O)alkyl, —C(═O)alkenyl, and —C(═O)alkynyl are acyl groups.

“Heterocycle” means a 5- to 7-membered monocyclic, or 7- to 10-membered bicyclic, heterocyclic ring which is either saturated, unsaturated, or aromatic, and which contains from 1 or 2 heteroatoms independently selected from nitrogen, oxygen and sulfur, and wherein the nitrogen and sulfur heteroatoms may be optionally oxidized, and the nitrogen heteroatom may be optionally quaternized, including bicyclic rings in which any of the above heterocycles are fused to a benzene ring. The heterocycle may be attached via any heteroatom or carbon atom. Heterocycles include heteroaryls as defined below. Heterocycles include morpholinyl, pyrrolidinonyl, pyrrolidinyl, piperidinyl, piperizynyl, hydantoinyl, valerolactamyl, oxiranyl, oxetanyl, tetrahydrofuranyl, tetrahydropyranyl, tetrahydropyridinyl, tetrahydroprimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, tetrahydropyrimidinyl, tetrahydrothiophenyl, tetrahydrothiopyranyl, and the like.

The terms “optionally substituted alkyl”, “optionally substituted alkenyl”, “optionally substituted alkynyl”, “optionally substituted acyl”, and “optionally substituted heterocycle” means that, when substituted, at least one hydrogen atom is replaced with a substituent. In the case of an oxo substituent (═O) two hydrogen atoms are replaced. In this regard, substituents include oxo, halogen, heterocycle, —CN, —ORx, —NRxRy, —NRxC(═O)Ry, —NRxSO2Ry, —C(═O)Rx, —C(═O)ORx, —C(═O)NRxRy, —SOnRx and —SOnNRxRy, wherein n is 0, 1 or 2, Rx and Ry are the same or different and independently hydrogen, alkyl or heterocycle, and each of said alkyl and heterocycle substituents may be further substituted with one or more of oxo, halogen, —OH, —CN, alkyl, —ORx, heterocycle, —NRxRy, —NRxC(═O)Ry, —NRxSO2Ry, —C(═O)Rx, —C(═O)ORx, —C(═O)NRxRy, —SOnRx and —SOnNRxRy.

“Halogen” means fluoro, chloro, bromo and iodo.

In some embodiments, the methods featured in the invention may require the use of protecting groups. Protecting group methodology is well known to those skilled in the art (see, for example, PROTECTIVE GROUPS IN ORGANIC SYNTHESIS, Green, T. W. et al., Wiley-Interscience, New York City, 1999). Briefly, protecting groups within the context of this invention are any group that reduces or eliminates unwanted reactivity of a functional group. A protecting group can be added to a functional group to mask its reactivity during certain reactions and then removed to reveal the original functional group. In some embodiments an “alcohol protecting group” is used. An “alcohol protecting group” is any group which decreases or eliminates unwanted reactivity of an alcohol functional group. Protecting groups can be added and removed using techniques well known in the art.

In one embodiments, nucleic acid-lipid particles featured in the invention are formulated using a cationic lipid of formula A:

where R1 and R2 are independently alkyl, alkenyl or alkynyl, each can be optionally substituted, and R3 and R4 are independently lower alkyl or R3 and R4 can be taken together to form an optionally substituted heterocyclic ring. In some embodiments, the cationic lipid is XTC (2,2-Dilinoleyl-4-dimethylaminoethyl-[1,3]-dioxolane). In general, the lipid of formula A above may be made by the following Reaction Schemes 1 or 2, wherein all substituents are as defined above unless indicated otherwise.

Lipid A, where R1 and R2 are independently alkyl, alkenyl or alkynyl, each can be optionally substituted, and R3 and R4 are independently lower alkyl or R3 and R4 can be taken together to form an optionally substituted heterocyclic ring, can be prepared according to Scheme 1. Ketone 1 and bromide 2 can be purchased or prepared according to methods known to those of ordinary skill in the art. Reaction of 1 and 2 yields ketal 3. Treatment of ketal 3 with amine 4 yields lipids of formula A. The lipids of formula A can be converted to the corresponding ammonium salt with an organic salt of formula 5, where X is anion counter ion selected from halogen, hydroxide, phosphate, sulfate, or the like.

Alternatively, the ketone 1 starting material can be prepared according to Scheme 2. Grignard reagent 6 and cyanide 7 can be purchased or prepared according to methods known to those of ordinary skill in the art. Reaction of 6 and 7 yields ketone 1. Conversion of ketone 1 to the corresponding lipids of formula A is as described in Scheme 1.

Preparation of DLin-M-C3-DMA (i.e., (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-yl 4-(dimethylamino)butanoate) was as follows. A solution of (6Z,9Z,28Z,31Z)-heptatriaconta-6,9,28,31-tetraen-19-ol (0.53 g), 4-N,N-dimethylaminobutyric acid hydrochloride (0.51 g), 4-N,N-dimethylaminopyridine (0.61 g) and 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide hydrochloride (0.53 g) in dichloromethane (5 mL) was stirred at room temperature overnight. The solution was washed with dilute hydrochloric acid followed by dilute aqueous sodium bicarbonate. The organic fractions were dried over anhydrous magnesium sulphate, filtered and the solvent removed on a rotovap. The residue was passed down a silica gel column (20 g) using a 1-5% methanol/dichloromethane elution gradient. Fractions containing the purified product were combined and the solvent removed, yielding a colorless oil (0.54 g).

Synthesis of ketal 519 [ALNY-100] was performed using the following scheme 3:

    • To a stirred suspension of LiAlH4 (3.74 g, 0.09852 mol) in 200 ml anhydrous THF in a two neck RBF (1L), was added a solution of 514 (10 g, 0.04926 mol) in 70 mL of THF slowly at 0° 0° C. under nitrogen atmosphere. After complete addition, reaction mixture was warmed to room temperature and then heated to reflux for 4 h. Progress of the reaction was monitored by TLC. After completion of reaction (by TLC) the mixture was cooled to 0° C. and quenched with careful addition of saturated Na2SO4 solution. Reaction mixture was stirred for 4 h at room temperature and filtered off. Residue was washed well with THF. The filtrate and washings were mixed and diluted with 400 mL dioxane and 26 mL conc. HCl and stirred for 20 minutes at room temperature. The volatilities were stripped off under vacuum to furnish the hydrochloride salt of 515 as a white solid. Yield: 7.12 g 1H-NMR (DMSO, 400 MHz): δ=9.34 (broad, 2H), 5.68 (s, 2H), 3.74 (m, 1H), 2.66-2.60 (m, 2H), 2.50-2.45 (m, 5H).
    • To a stirred solution of compound 515 in 100 mL dry DCM in a 250 mL two neck RBF, was added NEt3 (37.2 mL, 0.2669 mol) and cooled to 0° C. under nitrogen atmosphere. After a slow addition of N-(benzyloxy-carbonyloxy)-succinimide (20 g, 0.08007 mol) in 50 mL dry DCM, reaction mixture was allowed to warm to room temperature. After completion of the reaction (2-3 h by TLC) mixture was washed successively with 1N HCl solution (1×100 mL) and saturated NaHCO3 solution (1×50 mL). The organic layer was then dried over anhyd. Na2SO4 and the solvent was evaporated to give crude material which was purified by silica gel column chromatography to get 516 as sticky mass. Yield: 11 g (89%). 1H-NMR (CDCl3, 400 MHz): δ=7.36-7.27 (m, 5H), 5.69 (s, 2H), 5.12 (s, 2H), 4.96 (br., 1H) 2.74 (s, 3H), 2.60 (m, 2H), 2.30-2.25 (m, 2H). LC-MS [M+H]-232.3 (96.94%).
    • The cyclopentene 516 (5 g, 0.02164 mol) was dissolved in a solution of 220 mL acetone and water (10:1) in a single neck 500 mL RBF and to it was added N-methyl morpholine-N-oxide (7.6 g, 0.06492 mol) followed by 4.2 mL of 7.6% solution of 0.0 (0.275 g, 0.00108 mol) in tert-butanol at room temperature. After completion of the reaction (˜3 h), the mixture was quenched with addition of solid Na2SO3 and resulting mixture was stirred for 1.5 h at room temperature. Reaction mixture was diluted with DCM (300 mL) and washed with water (2×100 mL) followed by saturated NaHCO3(1×50 mL) solution, water (1×30 mL) and finally with brine (lx 50 mL). Organic phase was dried over an.Na2SO4 and solvent was removed in vacuum. Silica gel column chromatographic purification of the crude material was afforded a mixture of diastereomers, which were separated by prep HPLC. Yield:-6 g crude

517A-Peak-1 (white solid), 5.13 g (96%). 1H-NMR (DMSO, 400 MHz): δ=7.39-7.31 (m, 5H), 5.04 (s, 2H), 4.78-4.73 (m, 1H), 4.48-4.47 (d, 2H), 3.94-3.93 (m, 2H), 2.71 (s, 3H), 1.72-1.67 (m, 4H). LC-MS—[M+H]-266.3, [M+NH4+]-283.5 present, HPLC-97.86%. Stereochemistry confirmed by X-ray.

Using a procedure analogous to that described for the synthesis of compound 505, compound 518 (1.2 g, 41%) was obtained as a colorless oil. 1H-NMR (CDCl3, 400 MHz): δ=7.35-7.33 (m, 4H), 7.30-7.27 (m, 1H), 5.37-5.27 (m, 8H), 5.12 (s, 2H), 4.75 (m, 1H), 4.58-4.57 (m, 2H), 2.78-2.74 (m, 7H), 2.06-2.00 (m, 8H), 1.96-1.91 (m, 2H), 1.62 (m, 4H), 1.48 (m, 2H), 1.37-1.25 (br m, 36H), 0.87 (m, 6H). HPLC-98.65%.

A solution of compound 518 (1 eq) in hexane (15 mL) was added in a drop-wise fashion to an ice-cold solution of LAH in THF (1 M, 2 eq). After complete addition, the mixture was heated at 40° C. over 0.5 h then cooled again on an ice bath. The mixture was carefully hydrolyzed with saturated aqueous Na2SO4 then filtered through celite and reduced to an oil. Column chromatography provided the pure 519 (1.3 g, 68%) which was obtained as a colorless oil. 13C NMR=130.2, 130.1 (x2), 127.9 (x3), 112.3, 79.3, 64.4, 44.7, 38.3, 35.4, 31.5, 29.9 (x2), 29.7, 29.6 (x2), 29.5 (x3), 29.3 (x2), 27.2 (x3), 25.6, 24.5, 23.3, 226, 14.1; Electrospray MS (+ve): Molecular weight for C44H80NO2 (M+H)+Calc. 654.6, Found 654.6.

Formulations prepared by either the standard or extrusion-free method can be characterized in similar manners. For example, formulations are typically characterized by visual inspection. They should be whitish translucent solutions free from aggregates or sediment. Particle size and particle size distribution of lipid-nanoparticles can be measured by light scattering using, for example, a Malvern Zetasizer Nano ZS (Malvern, USA). Particles should be about 20-300 nm, such as 40-100 nm in size. The particle size distribution should be unimodal. The total dsRNA concentration in the formulation, as well as the entrapped fraction, is estimated using a dye exclusion assay. A sample of the formulated dsRNA can be incubated with an RNA-binding dye, such as Ribogreen (Molecular Probes) in the presence or absence of a formulation disrupting surfactant, e.g., 0.5% Triton-X100. The total dsRNA in the formulation can be determined by the signal from the sample containing the surfactant, relative to a standard curve. The entrapped fraction is determined by subtracting the “free” dsRNA content (as measured by the signal in the absence of surfactant) from the total dsRNA content. Percent entrapped dsRNA is typically >85%. For SNALP formulation, the particle size is at least 30 nm, at least 40 nm, at least 50 nm, at least 60 nm, at least 70 nm, at least 80 nm, at least 90 nm, at least 100 nm, at least 110 nm, and at least 120 nm. The suitable range is typically about at least 50 nm to about at least 110 nm, about at least 60 nm to about at least 100 nm, or about at least 80 nm to about at least 90 nm.

Compositions and formulations for oral administration include powders or granules, microparticulates, nanoparticulates, suspensions or solutions in water or non-aqueous media, capsules, gel capsules, sachets, tablets or minitablets. Thickeners, flavoring agents, diluents, emulsifiers, dispersing aids or binders may be desirable. In some embodiments, oral formulations are those in which dsRNAs featured in the invention are administered in conjunction with one or more penetration enhancers surfactants and chelators. Suitable surfactants include fatty acids and/or esters or salts thereof, bile acids and/or salts thereof. Suitable bile acids/salts include chenodeoxycholic acid (CDCA) and ursodeoxychenodeoxycholic acid (UDCA), cholic acid, dehydrocholic acid, deoxycholic acid, glucholic acid, glycholic acid, glycodeoxycholic acid, taurocholic acid, taurodeoxycholic acid, sodium tauro-24,25-dihydro-fusidate and sodium glycodihydrofusidate. Suitable fatty acids include arachidonic acid, undecanoic acid, oleic acid, lauric acid, caprylic acid, capric acid, myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein, dilaurin, glyceryl 1-monocaprate, 1-dodecylazacycloheptan-2-one, an acylcarnitine, an acylcholine, or a monoglyceride, a diglyceride or a pharmaceutically acceptable salt thereof (e.g., sodium). In some embodiments, combinations of penetration enhancers are used, for example, fatty acids/salts in combination with bile acids/salts. One exemplary combination is the sodium salt of lauric acid, capric acid and UDCA. Further penetration enhancers include polyoxyethylene-9-lauryl ether, polyoxyethylene-20-cetyl ether. DsRNAs featured in the invention may be delivered orally, in granular form including sprayed dried particles, or complexed to form micro or nanoparticles. DsRNA complexing agents include poly-amino acids; polyimines; polyacrylates; polyalkylacrylates, polyoxethanes, polyalkylcyanoacrylates; cationized gelatins, albumins, starches, acrylates, polyethyleneglycols (PEG) and starches; polyalkylcyanoacrylates; DEAE-derivatized polyimines, pollulans, celluloses and starches. Suitable complexing agents include chitosan, N-trimethylchitosan, poly-L-lysine, polyhistidine, polyornithine, polyspermines, protamine, polyvinylpyridine, polythiodiethylaminomethylethylene P(TDAE), polyaminostyrene (e.g., p-amino), poly(methylcyanoacrylate), poly(ethylcyanoacrylate), poly(butylcyanoacrylate), poly(isobutylcyanoacrylate), poly(isohexylcynaoacrylate), DEAE-methacrylate, DEAE-hexylacrylate, DEAE-acrylamide, DEAE-albumin and DEAE-dextran, polymethylacrylate, polyhexylacrylate, poly(D,L-lactic acid), poly(DL-lactic-co-glycolic acid (PLGA), alginate, and polyethyleneglycol (PEG). Oral formulations for dsRNAs and their preparation are described in detail in U.S. Pat. No. 6,887,906, US Publn. No. 20030027780, and U.S. Pat. No. 6,747,014, each of which is incorporated herein by reference.

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

Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, and liposome-containing formulations. These compositions may be generated from a variety of components that include, but are not limited to, preformed liquids, self-emulsifying solids and self-emulsifying semisolids.

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

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

The compositions of the present invention may be prepared and formulated as emulsions. Emulsions are typically heterogeneous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1 m in diameter (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., Volume 1, p. 245; Block in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 2, p. 335; Higuchi et al., in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 1985, p. 301). Emulsions are often biphasic systems comprising two immiscible liquid phases intimately mixed and dispersed with each other. In general, emulsions may be of either the water-in-oil (w/o) or the oil-in-water (o/w) variety. When an aqueous phase is finely divided into and dispersed as minute droplets into a bulk oily phase, the resulting composition is called a water-in-oil (w/o) emulsion. Alternatively, when an oily phase is finely divided into and dispersed as minute droplets into a bulk aqueous phase, the resulting composition is called an oil-in-water (o/w) emulsion. Emulsions may contain additional components in addition to the dispersed phases, and the active drug which may be present as a solution in either the aqueous phase, oily phase or itself as a separate phase. Pharmaceutical excipients such as emulsifiers, stabilizers, dyes, and anti-oxidants may also be present in emulsions as needed. Pharmaceutical emulsions may also be multiple emulsions that are comprised of more than two phases such as, for example, in the case of oil-in-water-in-oil (o/w/o) and water-in-oil-in-water (w/o/w) emulsions. Such complex formulations often provide certain advantages that simple binary emulsions do not. Multiple emulsions in which individual oil droplets of an o/w emulsion enclose small water droplets constitute a w/o/w emulsion. Likewise a system of oil droplets enclosed in globules of water stabilized in an oily continuous phase provides an o/w/o emulsion.

Emulsions are characterized by little or no thermodynamic stability. Often, the dispersed or discontinuous phase of the emulsion is well dispersed into the external or continuous phase and maintained in this form through the means of emulsifiers or the viscosity of the formulation. Either of the phases of the emulsion may be a semisolid or a solid, as is the case of emulsion-style ointment bases and creams. Other means of stabilizing emulsions entail the use of emulsifiers that may be incorporated into either phase of the emulsion. Emulsifiers may broadly be classified into four categories: synthetic surfactants, naturally occurring emulsifiers, absorption bases, and finely dispersed solids (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199).

Synthetic surfactants, also known as surface active agents, have found wide applicability in the formulation of emulsions and have been reviewed in the literature (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rieger, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), Marcel Dekker, Inc., New York, N.Y., 1988, volume 1, p. 199). Surfactants are typically amphiphilic and comprise a hydrophilic and a hydrophobic portion. The ratio of the hydrophilic to the hydrophobic nature of the surfactant has been termed the hydrophile/lipophile balance (HLB) and is a valuable tool in categorizing and selecting surfactants in the preparation of formulations. Surfactants may be classified into different classes based on the nature of the hydrophilic group: nonionic, anionic, cationic and amphoteric (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY Rieger, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 285).

Naturally occurring emulsifiers used in emulsion formulations include lanolin, beeswax, phosphatides, lecithin and acacia. Absorption bases possess hydrophilic properties such that they can soak up water to form w/o emulsions yet retain their semisolid consistencies, such as anhydrous lanolin and hydrophilic petrolatum. Finely divided solids have also been used as good emulsifiers especially in combination with surfactants and in viscous preparations. These include polar inorganic solids, such as heavy metal hydroxides, nonswelling clays such as bentonite, attapulgite, hectorite, kaolin, montmorillonite, colloidal aluminum silicate and colloidal magnesium aluminum silicate, pigments and nonpolar solids such as carbon or glyceryl tristearate.

A large variety of non-emulsifying materials are also included in emulsion formulations and contribute to the properties of emulsions. These include fats, oils, waxes, fatty acids, fatty alcohols, fatty esters, humectants, hydrophilic colloids, preservatives and antioxidants (Block, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 335; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199).

Hydrophilic colloids or hydrocolloids include naturally occurring gums and synthetic polymers such as polysaccharides (for example, acacia, agar, alginic acid, carrageenan, guar gum, karaya gum, and tragacanth), cellulose derivatives (for example, carboxymethylcellulose and carboxypropylcellulose), and synthetic polymers (for example, carbomers, cellulose ethers, and carboxyvinyl polymers). These disperse or swell in water to form colloidal solutions that stabilize emulsions by forming strong interfacial films around the dispersed-phase droplets and by increasing the viscosity of the external phase.

Since emulsions often contain a number of ingredients such as carbohydrates, proteins, sterols and phosphatides that may readily support the growth of microbes, these formulations often incorporate preservatives. Commonly used preservatives included in emulsion formulations include methyl paraben, propyl paraben, quaternary ammonium salts, benzalkonium chloride, esters of p-hydroxybenzoic acid, and boric acid. Antioxidants are also commonly added to emulsion formulations to prevent deterioration of the formulation. Antioxidants used may be free radical scavengers such as tocopherols, alkyl gallates, butylated hydroxyanisole, butylated hydroxytoluene, or reducing agents such as ascorbic acid and sodium metabisulfite, and antioxidant synergists such as citric acid, tartaric acid, and lecithin.

The application of emulsion formulations via dermatological, oral and parenteral routes and methods for their manufacture have been reviewed in the literature (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199). Emulsion formulations for oral delivery have been very widely used because of ease of formulation, as well as efficacy from an absorption and bioavailability standpoint (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245; Idson, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 199). Mineral-oil base laxatives, oil-soluble vitamins and high fat nutritive preparations are among the materials that have commonly been administered orally as o/w emulsions.

In one embodiment of the present invention, the compositions of iRNAs and nucleic acids are formulated as microemulsions. A microemulsion may be defined as a system of water, oil and amphiphile which is a single optically isotropic and thermodynamically stable liquid solution (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245). Typically microemulsions are systems that are prepared by first dispersing an oil in an aqueous surfactant solution and then adding a sufficient amount of a fourth component, generally an intermediate chain-length alcohol to form a transparent system. Therefore, microemulsions have also been described as thermodynamically stable, isotropically clear dispersions of two immiscible liquids that are stabilized by interfacial films of surface-active molecules (Leung and Shah, in: Controlled Release of Drugs: Polymers and Aggregate Systems, Rosoff, M., Ed., 1989, VCH Publishers, New York, pages 185-215). Microemulsions commonly are prepared via a combination of three to five components that include oil, water, surfactant, cosurfactant and electrolyte. Whether the microemulsion is of the water-in-oil (w/o) or an oil-in-water (o/w) type is dependent on the properties of the oil and surfactant used and on the structure and geometric packing of the polar heads and hydrocarbon tails of the surfactant molecules (Schott, in Remington's Pharmaceutical Sciences, Mack Publishing Co., Easton, Pa., 1985, p. 271).

The phenomenological approach utilizing phase diagrams has been extensively studied and has yielded a comprehensive knowledge, to one skilled in the art, of how to formulate microemulsions (see e.g., Ansel's Pharmaceutical Dosage Forms and Drug Delivery Systems, Allen, L V., Popovich N G., and Ansel H C., 2004, Lippincott Williams & Wilkins (8th ed.), New York, NY; Rosoff, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 245; Block, in Pharmaceutical Dosage Forms, Lieberman, Rieger and Banker (Eds.), 1988, Marcel Dekker, Inc., New York, N.Y., volume 1, p. 335). Compared to conventional emulsions, microemulsions offer the advantage of solubilizing water-insoluble drugs in a formulation of thermodynamically stable droplets that are formed spontaneously.

Surfactants used in the preparation of microemulsions include, but are not limited to, ionic surfactants, non-ionic surfactants, Brij 96, polyoxyethylene oleyl ethers, polyglycerol fatty acid esters, tetraglycerol monolaurate (ML310), tetraglycerol monooleate (MO310), hexaglycerol monooleate (PO310), hexaglycerol pentaoleate (PO500), decaglycerol monocaprate (MCA750), decaglycerol monooleate (MO750), decaglycerol sequioleate (S0750), decaglycerol decaoleate (DAO750), alone or in combination with cosurfactants. The cosurfactant, usually a short-chain alcohol such as ethanol, 1-propanol, and 1-butanol, serves to increase the interfacial fluidity by penetrating into the surfactant film and consequently creating a disordered film because of the void space generated among surfactant molecules. Microemulsions may, however, be prepared without the use of cosurfactants and alcohol-free self-emulsifying microemulsion systems are known in the art. The aqueous phase may typically be, but is not limited to, water, an aqueous solution of the drug, glycerol, PEG300, PEG400, polyglycerols, propylene glycols, and derivatives of ethylene glycol. The oil phase may include, but is not limited to, materials such as Captex 300, Captex 355, Capmul MCM, fatty acid esters, medium chain (C8-C12) mono, di, and tri-glycerides, polyoxyethylated glyceryl fatty acid esters, fatty alcohols, polyglycolized glycerides, saturated polyglycolized C8-C10 glycerides, vegetable oils and silicone oil.

Microemulsions are particularly of interest from the standpoint of drug solubilization and the enhanced absorption of drugs. Lipid based microemulsions (both o/w and w/o) have been proposed to enhance the oral bioavailability of drugs, including peptides (see e.g., U.S. Pat. Nos. 6,191,105; 7,063,860; 7,070,802; 7,157,099; Constantinides et al., Pharmaceutical Research, 1994, 11, 1385-1390; Ritschel, Meth. Find. Exp. Clin. Pharmacol., 1993, 13, 205). Microemulsions afford advantages of improved drug solubilization, protection of drug from enzymatic hydrolysis, possible enhancement of drug absorption due to surfactant-induced alterations in membrane fluidity and permeability, ease of preparation, ease of oral administration over solid dosage forms, improved clinical potency, and decreased toxicity (see e.g., U.S. Pat. Nos. 6,191,105; 7,063,860; 7,070,802; 7,157,099; Constantinides et al., Pharmaceutical Research, 1994, 11, 1385; Ho et al., J. Pharm. Sci., 1996, 85, 138-143). Often microemulsions may form spontaneously when their components are brought together at ambient temperature. This may be particularly advantageous when formulating thermolabile drugs, peptides or iRNAs. Microemulsions have also been effective in the transdermal delivery of active components in both cosmetic and pharmaceutical applications. It is expected that the microemulsion compositions and formulations of the present invention will facilitate the increased systemic absorption of iRNAs and nucleic acids from the gastrointestinal tract, as well as improve the local cellular uptake of iRNAs and nucleic acids.

Microemulsions of the present invention may also contain additional components and additives such as sorbitan monostearate (Grill 3), Labrasol, and penetration enhancers to improve the properties of the formulation and to enhance the absorption of the iRNAs and nucleic acids of the present invention. Penetration enhancers used in the microemulsions of the present invention may be classified as belonging to one of five broad categories—surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of these classes has been discussed above.

In one embodiment, the present invention employs various penetration enhancers to effect the efficient delivery of nucleic acids, particularly iRNAs, to the skin of animals. Most drugs are present in solution in both ionized and nonionized forms. However, usually only lipid soluble or lipophilic drugs readily cross cell membranes. It has been discovered that even non-lipophilic drugs may cross cell membranes if the membrane to be crossed is treated with a penetration enhancer. In addition to aiding the diffusion of non-lipophilic drugs across cell membranes, penetration enhancers also enhance the permeability of lipophilic drugs.

Penetration enhancers may be classified as belonging to one of five broad categories, i.e., surfactants, fatty acids, bile salts, chelating agents, and non-chelating non-surfactants (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, Informa Health Care, New York, NY, 2002; Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92). Each of the above mentioned classes of penetration enhancers are described below in greater detail.

Surfactants: In connection with the present invention, surfactants (or “surface-active agents”) are chemical entities which, when dissolved in an aqueous solution, reduce the surface tension of the solution or the interfacial tension between the aqueous solution and another liquid, with the result that absorption of iRNAs through the mucosa is enhanced. In addition to bile salts and fatty acids, these penetration enhancers include, for example, sodium lauryl sulfate, polyoxyethylene-9-lauryl ether and polyoxyethylene-20-cetyl ether) (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, Informa Health Care, New York, NY, 2002; Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92); and perfluorochemical emulsions, such as FC-43. Takahashi et al., J. Pharm. Pharmacol., 1988, 40, 252).

Fatty acids: Various fatty acids and their derivatives which act as penetration enhancers include, for example, oleic acid, lauric acid, capric acid (n-decanoic acid), myristic acid, palmitic acid, stearic acid, linoleic acid, linolenic acid, dicaprate, tricaprate, monoolein (1-monooleoyl-rac-glycerol), dilaurin, caprylic acid, arachidonic acid, glycerol 1-monocaprate, 1-dodecylazacycloheptan-2-one, acylcarnitines, acylcholines, C1-20alkyl esters thereof (e.g., methyl, isopropyl and t-butyl), and mono- and di-glycerides thereof (i.e., oleate, laurate, caprate, myristate, palmitate, stearate, linoleate, etc.) (see e.g., Touitou, E., et al. Enhancement in Drug Delivery, CRC Press, Danvers, M A, 2006; Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, p. 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; El Hariri et al., J. Pharm. Pharmacol., 1992, 44, 651-654).

Bile salts: The physiological role of bile includes the facilitation of dispersion and absorption of lipids and fat-soluble vitamins (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, Informa Health Care, New York, NY, 2002; Brunton, Chapter 38 in: Goodman & Gilman's The Pharmacological Basis of Therapeutics, 9th Ed., Hardman et al. Eds., McGraw-Hill, New York, 1996, pp. 934-935). Various natural bile salts, and their synthetic derivatives, act as penetration enhancers. Thus the term “bile salts” includes any of the naturally occurring components of bile as well as any of their synthetic derivatives. Suitable bile salts include, for example, cholic acid (or its pharmaceutically acceptable sodium salt, sodium cholate), dehydrocholic acid (sodium dehydrocholate), deoxycholic acid (sodium deoxycholate), glucholic acid (sodium glucholate), glycholic acid (sodium glycocholate), glycodeoxycholic acid (sodium glycodeoxycholate), taurocholic acid (sodium taurocholate), taurodeoxycholic acid (sodium taurodeoxycholate), chenodeoxycholic acid (sodium chenodeoxycholate), ursodeoxycholic acid (UDCA), sodium tauro-24,25-dihydro-fusidate (STDHF), sodium glycodihydrofusidate and polyoxyethylene-9-lauryl ether (POE) (see e.g., Malmsten, M. Surfactants and polymers in drug delivery, Informa Health Care, New York, NY, 2002; Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Swinyard, Chapter 39 In: Remington's Pharmaceutical Sciences, 18th Ed., Gennaro, ed., Mack Publishing Co., Easton, Pa., 1990, pages 782-783; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; Yamamoto et al., J. Pharm. Exp. Ther., 1992, 263, 25; Yamash*ta et al., J. Pharm. Sci., 1990, 79, 579-583).

Chelating Agents: Chelating agents, as used in connection with the present invention, can be defined as compounds that remove metallic ions from solution by forming complexes therewith, with the result that absorption of iRNAs through the mucosa is enhanced. With regards to their use as penetration enhancers in the present invention, chelating agents have the added advantage of also serving as DNase inhibitors, as most characterized DNA nucleases require a divalent metal ion for catalysis and are thus inhibited by chelating agents (Jarrett, J. Chromatogr., 1993, 618, 315-339). Suitable chelating agents include but are not limited to disodium ethylenediaminetetraacetate (EDTA), citric acid, salicylates (e.g., sodium salicylate, 5-methoxysalicylate and hom*ovanilate), N-acyl derivatives of collagen, laureth-9 and N-amino acyl derivatives of β-diketones (enamines)(see e.g., Katdare, A. et al., Excipient development for pharmaceutical, biotechnology, and drug delivery, CRC Press, Danvers, M A, 2006; Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92; Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33; Buur et al., J. Control Rel., 1990, 14, 43-51).

Non-chelating non-surfactants: As used herein, non-chelating non-surfactant penetration enhancing compounds can be defined as compounds that demonstrate insignificant activity as chelating agents or as surfactants but that nonetheless enhance absorption of iRNAs through the alimentary mucosa (see e.g., Muranishi, Critical Reviews in Therapeutic Drug Carrier Systems, 1990, 7, 1-33). This class of penetration enhancers include, for example, unsaturated cyclic ureas, 1-alkyl- and 1-alkenylazacyclo-alkanone derivatives (Lee et al., Critical Reviews in Therapeutic Drug Carrier Systems, 1991, page 92); and non-steroidal anti-inflammatory agents such as diclofenac sodium, indomethacin and phenylbutazone (Yamash*ta et al., J. Pharm. Pharmacol., 1987, 39, 621-626).

Agents that enhance uptake of iRNAs at the cellular level may also be added to the pharmaceutical and other compositions of the present invention. For example, cationic lipids, such as lipofectin (Junichi et al, U.S. Pat. No. 5,705,188), cationic glycerol derivatives, and polycationic molecules, such as polylysine (Lollo et al., PCT Application WO 97/30731), are also known to enhance the cellular uptake of dsRNAs. Examples of commercially available transfection reagents include, for example Lipofectamine™ (Invitrogen; Carlsbad, CA), Lipofectamine 2000™ (Invitrogen; Carlsbad, CA), 293Fectin™ (Invitrogen; Carlsbad, CA), Cellfectin™ (Invitrogen; Carlsbad, CA), DMRIE-C™ (Invitrogen; Carlsbad, CA), FreeStyle™ MAX (Invitrogen; Carlsbad, CA), Lipofectamine™ 2000 CD (Invitrogen; Carlsbad, CA), Lipofectamine™ (Invitrogen; Carlsbad, CA), RNAiMAX (Invitrogen; Carlsbad, CA), Oligofectamine™ (Invitrogen; Carlsbad, CA), Optifect™ (Invitrogen; Carlsbad, CA), X-tremeGENE Q2 Transfection Reagent (Roche; Grenzacherstrasse, Switzerland), DOTAP Liposomal Transfection Reagent (Grenzacherstrasse, Switzerland), DOSPER Liposomal Transfection Reagent (Grenzacherstrasse, Switzerland), or Fugene (Grenzacherstrasse, Switzerland), Transfectam® Reagent (Promega; Madison, WI), TransFast™ Transfection Reagent (Promega; Madison, WI), Tfx™-20 Reagent (Promega; Madison, WI), Tfx™_50 Reagent (Promega; Madison, WI), DreamFect™ (OZ Biosciences; Marseille, France), EcoTransfect (OZ Biosciences; Marseille, France), TransPassa D1 Transfection Reagent (New England Biolabs; Ipswich, MA, USA), LyoVec™/LipoGen™ (Invivogen; San Diego, CA, USA), PerFectin Transfection Reagent (Genlantis; San Diego, CA, USA), NeuroPORTER Transfection Reagent (Genlantis; San Diego, CA, USA), GenePORTER Transfection reagent (Genlantis; San Diego, CA, USA), GenePORTER 2 Transfection reagent (Genlantis; San Diego, CA, USA), Cytofectin Transfection Reagent (Genlantis; San Diego, CA, USA), BaculoPORTER Transfection Reagent (Genlantis; San Diego, CA, USA), TroganPORTER™ transfection Reagent (Genlantis; San Diego, CA, USA), RiboFect (Bioline; Taunton, MA, USA), PlasFect (Bioline; Taunton, MA, USA), UniFECTOR (B-Bridge International; Mountain View, CA, USA), SureFECTOR (B-Bridge International; Mountain View, CA, USA), or HiFect™ (B-Bridge International, Mountain View, CA, USA), among others.

Other agents may be utilized to enhance the penetration of the administered nucleic acids, including glycols such as ethylene glycol and propylene glycol, pyrrols such as 2-pyrrol, azones, and terpenes such as limonene and menthone.

Certain compositions of the present invention also incorporate carrier compounds in the formulation. As used herein, “carrier compound” or “carrier” can refer to a nucleic acid, or analog thereof, which is inert (i.e., does not possess biological activity per se) but is recognized as a nucleic acid by in vivo processes that reduce the bioavailability of a nucleic acid having biological activity by, for example, degrading the biologically active nucleic acid or promoting its removal from circulation. The coadministration of a nucleic acid and a carrier compound, typically with an excess of the latter substance, can result in a substantial reduction of the amount of nucleic acid recovered in the liver, kidney or other extracirculatory reservoirs, presumably due to competition between the carrier compound and the nucleic acid for a common receptor. For example, the recovery of a partially phosphorothioate dsRNA in hepatic tissue can be reduced when it is coadministered with polyinosinic acid, dextran sulfate, polycytidic acid or 4-acetamido-4′ isothiocyano-stilbene-2,2′-disulfonic acid (Miyao et al., DsRNA Res. Dev., 1995, 5, 115-121; Takakura et al., DsRNA & Nucl. Acid Drug Dev., 1996, 6, 177-183.

In contrast to a carrier compound, a “pharmaceutical carrier” or “excipient” is a pharmaceutically acceptable solvent, suspending agent or any other pharmacologically inert vehicle for delivering one or more nucleic acids to an animal. The excipient may be liquid or solid and is selected, with the planned manner of administration in mind, so as to provide for the desired bulk, consistency, etc., when combined with a nucleic acid and the other components of a given pharmaceutical composition. Typical pharmaceutical carriers include, but are not limited to, binding agents (e.g., pregelatinized maize starch, polyvinylpyrrolidone or hydroxypropyl methylcellulose, etc.); fillers (e.g., lactose and other sugars, microcrystalline cellulose, pectin, gelatin, calcium sulfate, ethyl cellulose, polyacrylates or calcium hydrogen phosphate, etc.); lubricants (e.g., magnesium stearate, talc, silica, colloidal silicon dioxide, stearic acid, metallic stearates, hydrogenated vegetable oils, corn starch, polyethylene glycols, sodium benzoate, sodium acetate, etc.); disintegrants (e.g., starch, sodium starch glycolate, etc.); and wetting agents (e.g., sodium lauryl sulphate, etc).

Pharmaceutically acceptable organic or inorganic excipients suitable for non-parenteral administration which do not deleteriously react with nucleic acids can also be used to formulate the compositions of the present invention. Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohols, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like.

Formulations for topical administration of nucleic acids may include sterile and non-sterile aqueous solutions, non-aqueous solutions in common solvents such as alcohols, or solutions of the nucleic acids in liquid or solid oil bases. The solutions may also contain buffers, diluents and other suitable additives. Pharmaceutically acceptable organic or inorganic excipients suitable for non-parenteral administration which do not deleteriously react with nucleic acids can be used.

Suitable pharmaceutically acceptable excipients include, but are not limited to, water, salt solutions, alcohol, polyethylene glycols, gelatin, lactose, amylose, magnesium stearate, talc, silicic acid, viscous paraffin, hydroxymethylcellulose, polyvinylpyrrolidone and the like.

The compositions of the present invention may additionally contain other adjunct components conventionally found in pharmaceutical compositions, at their art-established usage levels. Thus, for example, the compositions may contain additional, compatible, pharmaceutically-active materials such as, for example, antipruritics, astringents, local anesthetics or anti-inflammatory agents, or may contain additional materials useful in physically formulating various dosage forms of the compositions of the present invention, such as dyes, flavoring agents, preservatives, antioxidants, opacifiers, thickening agents and stabilizers. However, such materials, when added, should not unduly interfere with the biological activities of the components of the compositions of the present invention. The formulations can be sterilized and, if desired, mixed with auxiliary agents, e.g., lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, colorings, flavorings and/or aromatic substances and the like which do not deleteriously interact with the nucleic acid(s) of the formulation.

Aqueous suspensions may contain substances that increase the viscosity of the suspension including, for example, sodium carboxymethylcellulose, sorbitol and/or dextran. The suspension may also contain stabilizers.

In some embodiments, pharmaceutical compositions featured in the invention include (a) one or more iRNA compounds and (b) one or more biologic agents which function by a non-RNAi mechanism. Examples of such biologic agents include agents that interfere with an interaction of ALAS1 and at least one ALAS1 binding partner.

Toxicity and therapeutic efficacy of such compounds can be determined by standard pharmaceutical procedures in cell cultures or experimental animals, e.g., for determining the LD50 (the dose lethal to 50% of the population) and the ED50 (the dose therapeutically effective in 50% of the population). The dose ratio between toxic and therapeutic effects is the therapeutic index and it can be expressed as the ratio LD50/ED50. Compounds that exhibit high therapeutic indices are typical.

The data obtained from cell culture assays and animal studies can be used in formulating a range of dosage for use in humans. The dosage of compositions featured in the invention lies generally within a range of circulating concentrations that include the ED50 with little or no toxicity. The dosage may vary within this range depending upon the dosage form employed and the route of administration utilized. For any compound used in the methods featured in the invention, the therapeutically effective dose can be estimated initially from cell culture assays. A dose may be formulated in animal models to achieve a circulating plasma concentration range of the compound or, when appropriate, of the polypeptide product of a target sequence (e.g., achieving a decreased concentration of the polypeptide) that includes the IC50 (i.e., the concentration of the test compound which achieves a half-maximal inhibition of symptoms) as determined in cell culture. Such information can be used to more accurately determine useful doses in humans. Levels in plasma may be measured, for example, by high performance liquid chromatography.

In addition to their administration, as discussed above, the iRNAs featured in the invention can be administered in combination with other known agents effective in treatment of diseases or disorders related to ALAS1 expression. In any event, the administering physician can adjust the amount and timing of iRNA administration on the basis of results observed using standard measures of efficacy known in the art or described herein.

The invention relates in particular to the use of an iRNA targeting ALAS1 to inhibit ALAS1 expression and/or to treat a disease, disorder, or pathological process that is related to ALAS1 expression.

As used herein, “a disorder related to ALAS1 expression,” a “disease related to ALAS1 expression, a “pathological process related to ALAS1 expression,” or the like includes any condition, disorder, or disease in which ALAS1 expression is altered (e.g., elevated), the level of one or more porphyrins is altered (e.g., elevated), the level or activity of one or more enzymes in the heme biosynthetic pathway (porphyrin pathway) is altered, or other mechisms that lead to pathological changes in the heme biosynthetic pathway. For example, an iRNA targeting an ALAS1 gene, or a combination thereof, may be used for treatment of conditions in which levels of a porphyrin or a porphyrin precursor (e.g., ALA or PBG) are elevated (e.g., certain porphyrias), or conditions in which there are defects in the enzymes of the heme biosynthetic pathway (e.g., certain porphyrias). Disorders related to ALAS1 expression include, for example, X-linked sideroblastic anemia (XLSA), ALA deyhdratase deficiency porphyria (Doss porphyria), acute intermittent porphyria (AIP), congenital erythropoietic porphyria, Prophyria cutanea tarda, hereditary coproporphyria (coproporphyria), variegate porphyria, erythropoietic protoporphyria (EPP), and transient erythroporphyria of infancy.

As used herein, a “subject” to be treated according to the methods described herein, includes a human or non-human animal, e.g., a mammal. The mammal may be, for example, a rodent (e.g., a rat or mouse) or a primate (e.g., a monkey). In some embodiments, the subject is a human.

In some embodiments, the subject is suffering from a disorder related to ALAS1 expression (e.g., has been diagnosed with a porphyria or has suffered from one or more symptoms of porphyria and is a carrier of a mutation associated with porphyria) or is at risk of developing a disorder related to ALAS1 expression (e.g., a subject with a family history of porphyria, or a subject who is a carrier of a genetic mutation associated with porphyria).

Classifications of porphyrias, including acute hepatic porphyrias, are described, e.g., in Balwani, M. & Desnick, R. J., Blood, 120(23), published online as Blood First Edition paper, July 12, 102; DOI 10.1182/blood-2012-05-423186. As described in Balwain & Desnick, acute intermittent porphyria (AIP) hereditary coproporphyria (HCP), variegate porphyria (VP) are autosomal dominant porphyrias and ALA deyhdratase deficiency porphyria (ADP) is autosomal recessive. In rare cases, AIP, HCP, and VP occur as hom*ozygous dominant forms. In addition, there is a rare hom*ozygous recessive form of porphyria cutanea tarda (PCT), which is the single hepatic cutaneous porphyria, and is also known as hepatoerythropoietic porphyria. The clinical and laboratory features of these porphyrias are described in Table 11 below.

TABLE 11
Human hepatic porphyrias: clinical and laboratory features
Enzyme
Principalactivity,
Deficientsymptoms,% ofIncreased porphyrin precursors and/or porphyrins*
PorphyriaenzymeInheritanceNV or CPnormalErythrocytesUrineStool
Acute hepatic porphyrias
ADPALA-ARNV ~5Zn-protoporphyrinALA,
dehydratasecoproporphyrin
III
AIPHMB-ADNV~50ALA, PBG,
synthaseuroporphyrin
HCPCOPRO-ADNV and ~50ALA, PBG,coproporphyrin
oxidaseCPcoproporphyrinIII
III
VPPROTO-ADNV and ~50ALA, PBGcoproporphyrin
oxidaseCPcoproporphyrinIII,
IIIprotoporphyrin
Hepatic cutaneous porphyrias
PCTURO-Sporadic orCP<20uroporphyrin,uroporphyrin, 7-
decarboxylaseAD7-carboxylatecarboxylate
porphyrinporphyrin
AR indicates autosomal recessive;
AD, autosomal dominant;
NV, neurovisceral;
CP, cutaneous photosensitivity; and
—, not applicable.
*Increases that may be important for diagnosis.

In some embodiments, the subject has or is at risk for developing a porphyria, e.g., a hepatic porphyria, e.g., AIP, HCP, VP, ADP, or hepatoerythropoietic porphyria.

In some embodiments, the porphyria is an acute hepatic porphyria, e.g., an acute hepatic porphyria iselected from acute intermittent porphyria (AIP), hereditary coproporphyria (HCP), variegate porphyria (VP), and ALA deyhdratase deficiency porphyria (ADP).

In some embodiments, the porphyria is a dual porphyria, e.g., at least two porphyrias. In some embodiments, the dual porphyria comprises two or more porphyrias selected from acute intermittent porphyria (AIP) hereditary coproporphyria (HCP), variegate porphyria (VP), and ALA deyhdratase deficiency porphyria (ADP).

In some embodiments, the porphyria is a hom*ozygous dominant hepatic porphyria (e.g., hom*ozygous dominant AIP, HCP, or VP) or hepatoerythropoietic porphyria, In some embodiments, the porphyria is AIP, HCP, VP, or hepatoerythropoietic porphyria, or a combination thereof (e.g., a dual porphyria). In embodiments, the AIP, HCP, or VP is either heterozygous dominant or hom*ozygous dominant.

In embodiments, the subject has or is at risk for developing a porphyria, e.g., ADP, and shows an elevated level (e.g., an elevated urine level) of ALA and/or coproporphyrin III. In embodiments, the subject has or is at risk for developing a porphyria, e.g., ADP, and shows an elevated level of erythrocyte Zn-protoporphyrin.

In embodiments, the subject has or is at risk for developing a porphyria, e.g., AIP, and shows an elevated level (e.g., an elevated urine level) of ALA, PBG, and/or uroporphyrin.

In embodiments, the subject has or is at risk for developing a porphyria, e.g., HCP, and shows an elevated level (e.g., an elevated urine level) of ALA, PBG, and/or coproporphyrin III. In embodiments, the subject has or is at risk for developing a porphyria, e.g., HCP, and shows an elevated level (e.g., an elevated stool level) of coproporphyrin III.

In embodiments, the subject has or is at risk for developing a porphyria, e.g., VP, and shows an elevated level (e.g., an elevated urine level) of ALA, PBG, and/or coproporphyrin III.

In embodiments, the subject has or is at risk for developing a porphyria, e.g., HCP, and shows an elevated level (e.g., an elevated stool level) of coproporphyrin III and/or protoporphyrin.

In embodiments, the subject has or is at risk for developing a porphyria, e.g., PCT, (e.g., hepatoerythropoietic porphyria) and shows an elevated level (e.g., an elevated urine level) of uroporphyrin and/or 7-carboxylate porphyrin. In embodiments, the subject has or is at risk for developing a porphyria, e.g., PCT, (e.g., hepatoerythropoietic porphyria) and shows an elevated level (e.g., an elevated stool level) of uroporphyrin and/or 7-carboxylate porphyrin.

A mutation associated with porphyria includes any mutation in a gene encoding an enzyme in the heme biosynthetic pathway (porphyrin pathway) or a gene which alters the expression of a gene in the heme biosynthetic pathway. In many embodiments, the subject carries one or more mutations in an enzyme of the porphyrin pathway (e.g., a mutation in ALA deydratase or PBG deaminase). In some embodiments, the subject is suffereing from an acute porphyria (e.g., AIP, ALA deydratase deficiency porphyria).

In some cases, patients with an acute hepatic porphyria (e.g., AlP), or patients who carry mutations associated with an acute hepatic porphyria (e.g., AIP) but who are asymptomatic, have elevated ALA and/or PBG levels compared with healthy individuals. See, e.g., Floderus, Y. et al, Clinical Chemistry, 52(4): 701-707, 2006; Sardh et al., Clinical Pharmaco*kinetics, 46(4): 335-349, 2007. In such cases, the level of ALA and/or PBG can be elevated even when the patient is not having, or has never had, an attack. In some such cases, the patient is otherwise completely asymptomatic. In some such cases, the patient suffers from pain, e.g., neuropathic pain, which can be chronic pain (e.g., chronic neuropathic pain). In some cases, the patient has a neuropathy. In some cases, the patient has a progressive neuropathy.

In some embodiments, the subject to be treated according to the methods described herein has an elevated level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG. Levels of a porphyrin or a porphyrin precursor can be assessed using methods known in the art or methods described herein. For example, methods of assessing uring and plasma ALA and PBG levels, as well as urine and plasma porphyrin levels, are disclosed in Floderus, Y. et al, Clinical Chemistry, 52(4): 701-707, 2006; and Sardh et al., Clinical Pharmaco*kinetics, 46(4): 335-349, 2007, the entire contents of which are hereby incorporated in their entirety.

In some embodiments, the subject is an animal model of a porphyria, e.g., a mouse model of a porphyria (e.g., a mutant mouse as described in Lindberg et al. Nature Genetics, 12: 195-199, 1996). In some embodiments, the subject is a human, e.g., a human who has or is at risk for developing a porphyria, as described herein. In some embodiments, the subject is not having an acute attack of porphyria. In some embodiments, the subject has never had an attack. In some embodiments, the patient suffers from chronic pain. In some embodiments, the patient has nerve damage. In embodiments, the subject has EMG changes and/or changes in nerve conduction velocity. In some embodiments, the subject is asymptomatic. In some embodiments, the subject is at risk for developing a porphyria (e.g., carries a gene mutation associated with a porphyria) and is asymptomatic. In some embodiments, the subject has previously had an acute attack but is asymptomatic at the time of treatment.

In some embodiments, the subject is at risk for developing a porphyria and is treated prophylactically to prevent the development of a porphyria. In some embodiments the subject has an elevated level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG. In some embodiments, the prophylactic treatment begins at puberty. In some embodiments the treatment lowers the level (e.g., the plasma level or the urine level) of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG. In some embodiments, the treatment prevents the development of an elevated level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG. In some embodiments, the treatment prevents the development of, or decreases the frequency or severity of, a symptom associated with a porphyria, e.g., pain or nerve damage.

In some embodiments, the level of a porphyrin or a porphyrin precursor, e.g., ALA or PBG, is elevated, e.g., in a sample of plasma or urine from the subject. In some embodiments, the level of a porphyrin or a porphyrin precursor, e.g., ALA or PBG, in the subject is assessed based on the absolute level of the porphyrin or the porphyrin precursor, e.g., ALA or PBG in a sample from the subject. In some embodiments, the level of a porphyrin or a porphyrin precursor, e.g., ALA or PBG, in the subject is assessed based on the relative level of the porphyrin or porphyrin precursor, e.g., ALA or PBG, in a sample from the subject. In some embodiments, the relative level is relative to the level of another protein or compound, e.g., the level of creatinine, in a sample from the subject. In some embodiments, the sample is a urine sample. In some embodiments, the sample is a plasma sample. In some embodiments, the sample is a stool sample.

An elevated level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG, can be established, e.g., by showing that the subject has a level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG (e.g., a plasma or urine level of ALA and/or PBG) that is greater than, or greater than or equal to, a reference value. A physician with expertise in the treatment of porphyrias would be able to determine whether the level of a porphyrin or a porphyrin precursor, (e.g., ALA and/or PBG) is elevated, e.g., for the purpose of diagnosing a porphyria or for determining whether a subject is at risk for developing a porphyria, e.g., a subject may be predisposed to an acute attack or to pathology associated with a porphyria, such as, e.g., chronic pain (e.g., neuropathic pain) and neuropathy (e.g., progressive neuropathy).

As used herein, a “reference value” refers to a value from the subject when the subject is not in a disease state, or a value from a normal or healthy subject, or a value from a reference sample or population, e.g., a group of normal or healthy subjects (e.g., a group of subjects that does not carry a mutation associated with a porphyria and/or a group of subjects that does not suffer from symptoms associated with a porphyria).

In some embodiments, the reference value is a pre-disease level in the same individual. In some embodiments, the reference value is a level in a reference sample or population. In some embodiments, the reference value is the mean or median value in a reference sample or population. In some embodiments, the reference value the value that is is two standard deviations above the mean in a reference sample or population. In some embodiments, the reference value is the value that is 2.5, 3, 3.5, 4, 4.5, or 5 standard deviations above the mean in a reference sample or population.

In some embodiments, wherein the subject has an elevated level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG, the subject has a level of ALA and/or PBG that is at least 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, or 90% higher than a reference value. In some embodiments, the subject has a level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG, that is at least 2, 3, 4, 5, 6, 7, 8, 9, or 10 fold higher than a reference value.

In some embodiments, the reference value is an upper reference limit. As used herein, an “upper reference limit” refers to a level that is the upper limit of the 95% confidence interval for a reference sample or population, e.g., a group of normal (e.g., wild type) or healthy individuals, e.g., individuals who do not carry a genetic mutation associated with a porphyria and/or individuals who do not suffer from a porphyria. Accordingly, a lower reference limit refers to a level that is the lower limit of the same 95% confidence interval.

In some embodiments wherein the subject has an elevated level, e.g., a plasma level or a urine level, of a porphyrin or a porphyrin precursor, e.g., ALA or PBG, the level is greater than or equal to 2 times, 3 times, 4 times, or 5 times that of a reference value, e.g., an upper reference limit. In some embodiments, the subject has a urine level of a porphyrin or a porphyrin precursor, e.g., ALA or PBG, that is greater than 4 times that of an upper reference limit.

In some embodiments, the reference value is a value provided in Floderus, Y. et al, Clinical Chemistry, 52(4): 701-707, 2006 or Sardh et al., Clinical Pharmaco*kinetics, 46(4): 335-349, 2007. In some embodiments, the reference value is a value provided in Table 1 of Sardh et al.

In some embodiments, the subject is a human and has a urine level of PBG that is greater than or equal to 4.8 mmol/mol creatinine. In certain embodiments, the subject is a human and has a urine level of PBG that is greater than, or greater than or equal to, about 3, 4, 5, 6, 7, or 8 mmol/mol creatinine.

In embodiments, the reference value for plasma PBG is 0.12 μmol/L. In embodiments, the subject is a human and has a plasma PBG level that is greater than, or greater than or equal to, 0.10 μmol/L, 0.12 μmol/L, 0.24 μmol/L, 0.36 μmol/L, 0.48 μmol/L, or 0.60 μmol/L. In embodiments, the subject is a human and has a plasma level of PBG that is greater than, or greater than or equal to, 0.48 μmol/L.

In embodiments, the reference value for urine PBG is 1.2 mmol/mol creatinine. In embodiments, the subject is a human and has a urine PBG level that is greater than, or greater than or equal to, 1.0 mmol/mol creatinine, 1.2 mmol/mol creatinine, 2.4 mmol/mol creatinine, 3.6 mmol/mol creatinine, 4.8 mmol/mol creatinine, or 6.0 mmol/mol creatinine. In embodiments, the subject is a human and has a urine level of PBG that is greater than, or greater than or equal to, 4.8 mmol/mol creatinine.

In embodiments, the reference value for plasma ALA is 0.12 μmol/L. In embodiments, the subject is a human and has a plasma ALA level that is greater than, or greater than or equal to, 0.10 μmol/L, 0.12 μmol/L, 0.24 μmol/L, 0.36 μmol/L, 0.48 μmol/L, or 0.60 μmol/L. In embodiments, the subject is a human and has a plasma ALA level that is greater than, or greater than or equal to 0.48 μmol/L.

In embodiments, the reference value for urine ALA is 3.1 mmol/mol creatinine. In embodiments, the subject is a human and has a urine ALA level that is greater than, or greater than or equal to, 2.5 mmol/mol creatinine, 3.1 mmol/mol creatinine, 6.2 mmol/mol creatinine, 9.3 mmol/mol creatinine, 12.4 mmol/mol creatinine, or 15.5 mmol/mol creatinine.

In embodiments, the reference value for plasma porphyrin is 10 nmol/L. In embodiments, the subject is a human and has a plasma porphyrin level that is greater than, or greater than or equal to, 10 nmol/L. In embodiments, the subject is a human and has a plasma porphyrin level that is greater than, or greater than or equal to, 8, 10, 15, 20, 25, 30, 35, 40, 45, or 50 nmol/L. the subject is a human and has a plasma porphyrin level that is greater than, or greater than or equal to 40 nmol/L. In embodiments, the reference value for urine porphyrin is 25 μmol/mol creatinine. In embodiments, the subject is a human and has a urine porphyrin level that is greater than, or greater than or equal to, 25 μmol/mol creatinine. In embodiments, the subject is a human and has a urine porphyrin level that is greater than, or equal to, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, or 80 μmol/mol creatinine.

In some embodiments, the subject has a level, e.g., a plasma level or a urine level, of a porphyrin or a porphyrin precursor, e.g., ALA or PBG, that is greater than that of 99% of individuals in a sample of healthy individuals.

In some embodiments, the subject has a level, e.g., a plasma level or a urine level, of ALA or PBG that is greater than two standard deviations above the mean level in a sample of healthy individuals.

In some embodiments, the subject has a urine level of ALA that is 1.6 or more times that of the mean level in a normal subject (e.g., a subject that does not carry a mutation associated with a porphyria). In some embodiments, the subject has a plasma level of ALA that is 2 or 3 times that of the mean level in a normal subject. In some embodiments, the subject has a urine level of PBG that is four or more times that of the mean level in a normal subject. In some embodiments, the subject has a plasma level of PBG that is four or more times that of the mean level in a normal subject.

In some embodiments, the method is effective to decrease the level of a porphyrin or a porphyrin precursor, e.g., ALA and/or PBG. In embodiments, the method is effective to produce a predetermined reduction in the elevated level of the porphyrin or porphyrin precursor, e.g., ALA or PBG. In some embodiments, the predetermined reduction is a decrease of at least 10%, 20%, 30%, 40%, or 50%. In some embodiments, the predetermined reduction is a reduction that is effective to prevent or ameliorate symptoms, e.g., pain or recurring attacks.

In some embodiments, the predetermined reduction is a reduction that is at least 1, 2, 3, or more standard deviations, wherein the standard deviation is determined based on the values from a reference sample, e.g., a reference sample as described herein.

In some embodiments, the predetermined reduction is a reduction that brings the level of the porphyrin or porphyrin precursor to a level that is less than, or to a level that is less than or equal to, a reference value (e.g., a reference value as described herein).

In some embodiments, the subject to be treated according to the methods described suffers from pain, e.g., chronic pain. In some embodiments, the subject has or is at risk for developing a porphyria, e.g. an acute hepatic porphyria, e.g., AIP. In embodiments, the method is effective to treat the pain, e.g., by reducing the severity of the pain or curing the pain. In embodiments, the method is effective to decrease or prevent nerve damage.

In some embodiments, the subject to be treated according to the methods described herein (a) has an elevated level of ALA and/or PBG and (b) suffers from pain, e.g., chronic pain. In embodiments, the method is effective to decrease an elevated level of ALA and/or PBG and/or to treat the pain, e.g., by reducing the severity of the pain or curing the pain.

In some embodiments, the subject is an animal that serves as a model for a disorder related to ALAS1 expression.

In some embodiments the subject is an animal that serves as a model for porphyria (e.g., a genetically modified animal with one or more mutations. In some embodiments, the porphyria is AIP and the subject is an animal model of AIP. In one such embodiment, the subject is a genetically modified mouse that is deficient in porphobilinogen deaminase, such as, for example, the mouse described in Lindberg et al., Nature Genetics, 12:195-199, 1996, or the hom*ozygous R167Q mouse described in Yasuda, M., Yu, C. Zhang, J., Clavero, S., Edelmann, W., Gan, L., Phillips, J. D., & Desnick, R. J. Acute intermittent porphyria: A severely affected knock-in mouse that mimics the human hom*ozygous dominant phenotype. (Abstract of Presentation on Oct. 14, 2011 at the American Society of Human Genetics; Program No. 1308F; accessed online on Apr. 4, 2012 at ichg2011.org/cgi-bin/showdetail.pl?absno=21167); both of these references are hereby incorporated herein in their entirety. Several knock-in models for mutations causing hom*ozygous dominant AIP in humans have been generated. The mutations employed include, e.g., R167Q, R173Q, and R173W in PBG deaminase. Viable hom*ozygotes included the R167Q/R176Q and R167Q/R173Q, both of which exhibit constitutively elevated ALA and PBG levels analogous to the phenotype in human hom*ozygous dominant AIP; in some embodiments, such a viable hom*ozygous AIP mouse model is the subject.

In one embodiment, a subject to be treated according to the methods described herein, (e.g., a human subject or patient), is at risk of developing, or has been diagnosed, with a disorder related to ALAS1 expression, e.g. a porphyria. In some embodiments, the subject is a subject who has suffered one or more acute attacks of one or more porphyric symptoms. In other embodiments, the subject is a subject who has suffered chronically from one or more symptoms of porphyria (e.g., pain, e.g., neuropathic pain and or neuropathy, e.g., progressive neuropathy). In some embodiments, the subject carries a genetic alteration (e.g., a mutation) as described herein but is otherwise asymptomatic. In some embodiments, the subject has previously been treated with a heme product (e.g., hemin, heme arginate, or heme albumin), as described herein.

In some embodiments, a subject (e.g., a subject with a porphyria, such as, e.g., AIP) to be treated according to the methods described herein has recently experienced or is currently experiencing a prodrome. In some such embodiments, the subject is administered a combination treatment, e.g., an iRNA as described herein, and one or more additional treatments known to be effective against porphyria (e.g., glucose and/or a heme product such as hemin, as described herein) or its associated symptoms.

In one embodiment, an iRNA as described herein is administered in combination with glucose or dextrose. For example, 10-20% dextrose in normal saline may be provided intravenously. Typically, when glucose is administered, at least 300 g of 10% glucose is administered intravenously daily. The iRNA (e.g., an iRNA in an LNP formulation) may also be administered intravenously, as part of the same infusion that is used to administer the glucose or dextrose, or as a separate infusion that is administered before, concurrently, or after the administration of the glucose or dextrose. In some embodiments, the iRNA is administered via a different route of administration (e.g., subcutaneously). In yet another embodiment, the iRNA is administered in combination with total parenteral nutrition. The iRNA may be administered before, concurrent with, or after the administration of total parenteral nutrition.

In one embodiment, the iRNA is administered in combination with a heme product (e.g., hemin, heme arginate, or heme albumin). In a further embodiment, the iRNA is administered in combination with a heme product and glucose, a heme product and dextrose, or a heme product and total parenteral nutrition.

A “prodrome,” as used herein, includes any symptom that the individual subject has previously experienced immediately prior to developing an acute attack. Typical symptoms of a prodrome include, e.g., abdominal pain, nausea, headaches, psychological symptoms (e.g., anxiety), restlessness and/or insomnia. In some embodiments, the subject experiences pain (e.g., abdominal pain and/or a headache) during the prodrome. In some embodiments, the subject experiences nausea during the prodrome. In some embodiments, the subject experiences psychological symptoms (e.g., anxiety) during the prodrome. In some embodiments, the subject becomes restless and/or suffers from insomnia during the prodrome.

An acute “attack” of porphyria involves the onset of one or more symptoms of porphyria, typically in a patient who carries a mutation associated with porphyria (e.g., a mutation in a gene that encodes an enzyme in the porphyrin pathway).

In certain embodiments, administration of an ALAS1 iRNA results in a decrease in the level of one or more porphyrins or porphyrin precursors, as described herein (e.g., ALA and/or PBG). The decrease may be measured relative to any appropriate control or reference value. For example, the decrease in the level of one or more porphyrins or porphyrin precursors may be established in an individual subject, e.g., as a decrease of at least 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 45%, 50% or more compared with the level prior to treatment (e.g., immediately prior to treatment). A decrease in the level of a porphyrin precursor, a porphyrin, or or a porphyrin metabolite may be measured using any method known in the art. For example, the level of PBG and/or ALA in urine or plasma may be assessed, using the Watson-Schwartz test, ion exchange chromatography, or high-performance liquid chromatography-mass spectrometry. See, e.g., Thunell (1993).

In some embodiments, administration of an ALAS1 siRNA is effective to reduce the level of ALA and/or PBG in the subject. The level of ALA or PBG in the subject can be assessed, e.g., based on the absolute level of ALA or PBG, or based on the relative level of ALA or PBG (e.g., relative to the level of another protein or compound, e.g., the level of creatinine) in a sample from the subject. In some embodiments, the sample is a urine sample. In some embodiments, the sample is a plasma sample.

In certain embodiments, an iRNA that targets ALAS1 is administered in combination one or more additional treatments, e.g., another treatment known to be effective in treating porphyria or symptoms of porphyria. For example, the other treatment may be glucose (e.g., IV glucose) or a heme product (e.g., hemin, heme arginate, or heme albumin). The additional treatment(s) may be administered before, after, or concurrent with the administration of iRNA.

The iRNA and an additional therapeutic agent can be administered in combination in the same composition, e.g., intravenously, or the additional therapeutic agent can be administered as part of a separate composition or by another method described herein.

In some embodiments, administration of iRNA, or administration of iRNA in combination one or more additional treatments (e.g., glucose, dextrose or the like), decreases the frequency of acute attacks (e.g., by preventing acute attacks so that they no longer occur, or by reducing the number of attacks that occur in a certain time period, e.g., fewer attacks occur per year). In some such embodiments, the iRNA is administered according to a regular dosing regimen, e.g., daily, weekly, biweekly, or monthly.

In some embodiments, the iRNA is administered after an acute attack of porphyria. In some such embodiments, the iRNA is in a composition, e.g. a composition comprising a lipid formulation, e.g. an LNP formulation.

In some embodiments, the iRNA is administered during an acute attack of porphyria. In some such embodiments, the iRNA is in a composition, e.g. a composition comprising a lipid formulation (e.g., an LNP formulation) or a composition comprising a GalNAc conjugate.

In some embodiments, administration of an ALAS1 siRNA is effective to lessen the severity of the attack (e.g., by ameliorating one or more signs or symptoms associated with the attack). In some embodiments, administration of an ALAS1 siRNA is effective to shorten the duration of an attack. In some embodiments, administration of an ALAS1 siRNA is effective to stop an attack. In some embodiments, the iRNA is administered prophylactically to prevent an acute attack of porphyria. In some such embodiments, the iRNA is in the form of a GalNAc conjugate, e.g., in a composition comprising a GalNAc conjugate. In some embodiments, the prophylactic administration is before, during, or after exposure to or occurrence of a precipitating factor. In some embodiments, the subject is at risk of developing porphyria.

In some embodiments, the siRNA is administered during a prodrome. In some embodiments, the prodrome is characterized by pain (e.g., headache and/or abdominal pain), nausea, psychological symptoms (e.g., anxiety), restlessness and/or insomnia.

In some embodiments, the siRNA is administered during a particular phase of the menstrual cycle, e.g., during the luteal phase.

In some embodiments, administration of an ALAS1 siRNA is effective to prevent attacks (e.g., recurrent attacks that are associated with a prodrome and/or with a precipitating factor, e.g., with a particular phase of the menstrual cycle, e.g., the luteal phase). In some embodiments, administration of an ALAS1 siRNA is effective to reduce the frequency of attacks. In embodiments, administration of an ALAS1 siRNA is effective to lessen the severity of the attack (e.g., by ameliorating one or more signs or symptoms associated with the attack). In some embodiments, administration of an ALAS1 siRNA is effective to shorten the duration of an attack. In some embodiments, administration of an ALAS1 siRNA is effective to stop an attack.

In some embodiments administration of an ALAS1 siRNA is effective to prevent or decrease the frequency or severity of pain, e.g., neuropathic pain.

In some embodiments administration of an ALAS1 siRNA is effective to prevent or decrease the frequency or severity of neuropathy

Effects of administration of an ALAS1 siRNA can be established, for example, by comparison with an appropriate control. For example, a decrease in the frequency of acute attacks, as well as a decrease in the level of one or more porphyrins or porphyrin precursors, may be established, for example, in a group of patients with AIP, as a decreased frequency compared with an appropriate control group. A control group (e.g., a group of similar individuals or the same group of individuals in a crossover design) may include, for example, an untreated population, a population that has been treated with a conventional treatment for porphyria (e.g., a conventional treatment for AIP may include glucose, hemin, or both); a population that has been treated with placebo, or a non-targeting iRNA, optionally in combination with one or more conventional treatments for porphyria (e.g., glucose, e.g., IV glucose), and the like.

A subject “at risk” of developing porphyria, as used herein, includes a subject with a family history of porphyria and/or a history of one or more recurring or chronic porphyric symptoms, and/or a subject who carries a genetic alteration (e.g., a mutation) in a gene encoding an enzyme of the heme biosynthetic pathway, and a subject who carries a genetic alteration, e.g., a mutation. known to be associated with porphyria.

In embodiments, the alteration, e.g., the mutation, makes an individual susceptible to an acute attack (e.g., upon exposure to a precipitating factor, e.g., a drug, dieting or other precipitating factor, e.g., a precipitating factor as disclosed herein). In embodiments, the alteration, e.g., the mutation, is associated with elevated levels of a porphyrin or a porphyrin precursor (e.g., ALA and/or PBG). In embodiments, the alteration, e.g., the mutation, is associated with chronic pain (e.g., chronic neuropathic pain) and/or neuropathy (e.g., progressive neuropathy). In embodiments, the, the alteration, e.g., the mutation, is associated with changes in EMG and/or nerve conduction velocities.

In embodiments, the alteration is a mutation in the ALAS1 gene. In embodiments, the alteration is a mutation in the ALAS1 gene promoter, or in regions upstream or downstream from the ALAS1 gene. In embodiments, the alteration is a mutation in transcription factors or other genes that interact with ALAS1. In embodiments, the alteration is an alteration, e.g., a mutation, in a gene that encodes an enzyme in the heme biosynthetic pathway.

In some embodiments, the subject has an genetic alteration as described herein (e.g., a genetic mutation known to be associated with a porphyria). In some such embodiments, the subject has an elevated level (e.g., urine or plasma level) of ALA and/or PBG. In some such embodiments, the subject does not have an elevated level of ALA and/or PBG. In embodiments, the subject has a genetic alteration as described herein and has other symptoms, e.g., chronic pain, EMG changes, changes in nerve conduction velocity, and/or other symptoms associated with a porphyria. In embodiments, the subject has a genetic alteration but does not suffer from acute attacks.

In embodiments, the subject has a mutation associated with AIP, HCP, VP, or ADP.

In some embodiments, the porphyria is AIP. In some such embodiments, the subject has an alteration, e.g., at least one mutation, in the PBG deaminase gene. Many PBG deaminase mutations are known in the art, for example, as reported in Hrdinka, M. et al. Physiological Research, 55 (Suppl 2):S119-136 (2006). In some embodiments, the subject is heterozygous for a PBG deaminase mutation. In other embodiments, the subject is hom*ozygous for a PBG deaminase mutation. A hom*ozygous subject may carry two identical mutations or two different mutations in the PBG deaminase gene.

In some embodiments, the porphyria is HCP. In some such embodiments, the subject has an alteration, e.g., at least one mutation, in the gene that encodes the enzyme coproporphyrinogen III oxidase.

In some embodiments, the porphyria is VP. In some such embodiments, the subject has an alteration, e.g., at least one mutation, in the gene that encodes protoporphrinogen oxidase.

In embodiments, the porphyria is ADP, e.g., autosomal recessive ADP. In some such embodiments, the subject has an alteration, e.g., at least one mutation, in the gene that encodes ALA deydratase.

Methods of treatment provided herein may serve to ameliorate one or more symptoms associated with porphyria, to reduce the frequency of attacks associated with porphyria, to reduce the likelihood that an attack of one or more symptoms associated with porphyria will occur upon exposure to a precipitating factor, or to reduce the risk of developing conditions associated with porphyria (e.g., neuropathy (e.g., progressive neuropathy), hepatocellular cancer). Additionally, the methods provided herein may serve to decrease the level of one or more porphyrin precursors, porphyrins and/or related porphyrin products or metabolites. The level of a porphyrin precursor or a porhyrin may be measured in any biological sample, such as, e.g., urine, blood, feces, cerebrospinal fluid, or a tissue sample. The sample may be present within a subject or may be obtained or extracted from the subject. In some embodiments, the porphyria is AIP, and the level of PBG and/or ALA is decreased. In some embodiments, the porphyrin product or metabolite is porphobilin, porphobilinogen, or uroporphyrin. A decrease in the level of a porphyrin product or metabolite may be measured using any method known in the art. For example, the level of PBG and/or ALA in urine or plasma may be assessed, using the Watson-Schwartz test, ion exchange chromatography, or high-performance liquid chromatography-mass spectrometry. See, e.g., Thunell (1993).

Methods described herein may also serve to reduce chronically elevated levels of porphyrin precursors (e.g., ALA and/or PBG) in subjects suffering from a porphyria (e.g., an acute hepatic porphyria, e.g., AIP) or at risk for developing a porphyria. Methods for assessing plasma and urine levels (e.g., chronically elevated levels) of porphyrin precursors include, e.g., HPLC-mass spectrometry and ion-exchange chromatography. The levels of porphyrin precursors may be expressed as the level relative to another protein or compound, e.g., creatinine. See, e.g., Floderus, Y. et al, Clinical Chemistry, 52(4): 701-707, 2006; Sardh et al., Clinical Pharmaco*kinetics, 46(4): 335-349, 2007

A “precipitating factor” as used herein, refers to an endogenous or exogenous factor that may induce an acute attack of one or more symptoms associated with porphyria. Precipitating factors include fasting (or other forms of reduced or inadequate caloric intake, due to crash diets, long-distance athletics, etc.), metabolic stresses (e.g., infections, surgery, international air travel, and psychological stress), endogenous hormones (e.g., progesterone), cigarette smoking, lipid-soluble foreign chemicals (including, e.g., chemicals present in tobacco smoke, certain prescription drugs, organic solvents, biocides, components in alcoholic beverages), endocrine factors (e.g., reproductive hormones (women may experience exacerbations during the premenstrual period), synthetic estrogens, progesterones, ovulation stimulants, and hormone replacement therapy). See, for example, Thunell (1993). Common precipitating factors include cytochrome P450 inducing drugs and phenobarbitol.

Symptoms associated with porphyria may include abdominal pain or cramping, headaches, effects caused by nervous system abnormalities, and light sensitivity, causing rashes, blistering, and scarring of the skin (photodermatitis). In certain embodiments, the porphyria is AIP. Symptoms of AIP include gastrointestinal symptoms (e.g., severe and poorly localized abdominal pain, nausea/vomiting, constipation, diarrhea, ileus), urinary symptoms (dysuria, urinary retention/incontinence, or dark urine), neurologic symptoms (e.g., sensory neuropathy, motor neuropathy (e.g., affecting the cranial nerves and/or leading to weakness in the arms or legs), seizures, neuropathic pain, progressive neuropathy, headaches, neuropsychiatric symptoms (e.g., mental confusion, anxiety, agitation, hallucination, hysteria, delirium, apathy, depression, phobias, psychosis, insomnia, somnolence, coma), autonomic nervous system involvement (resulting e.g., in cardiovascular symptoms such as tachycardia, hypertension, and/or arrhythmias, as well as other symptoms, such as, e.g., increased circulating catecholamine levels, sweating, restlessness, and/or tremor), dehydration, and electrolyte abnormalities.

In some embodiments, an iRNA targeting ALAS1 is administered together with (e.g., before, after, or concurrent with) another treatment that may serve to alleviate one or more of the above symptoms. For example, abdominal pain may be treated, e.g., with narcotic analgesics, seizures may be treated, e.g., with anti-seizure medications, nausea/vomiting may be treated, e.g., with phenothiazines, and tachycardia/hypertension may be treated, e.g., with beta blockers.

The term “decrease” (or “increase”) is intended to refer to a measurable change, e.g., a statistically significant change. The change may be, for example, at least 5%, 10%, 20%, 30%, 40%, 50% or more change (e.g., decrease (or increase) relative to a reference value, e.g., a reference where no iRNA is provided).

The invention further relates to the use of an iRNA or a pharmaceutical composition thereof, e.g., for treating a disorder related to ALAS1 expression, in combination with other pharmaceuticals and/or other therapeutic methods, e.g., with known pharmaceuticals and/or known therapeutic methods, such as, for example, those which are currently employed for treating the disorder. In one embodiment, the iRNA or pharmaceutical composition thereof can be administered in conjunction with a heme product (e.g., hemin, heme arginate, or heme albumin, as described herein) and/or in conjunction with intravenous glucose infusions. In some embodiments, the iRNA or pharmaceutical composition thereof is used prophylactically, e.g., to prevent or ameliorate symptoms of an anticipated attack of acute porphyria. The prophylactic use may be timed according to the exposure or anticipated exposure of the subject to a precipitating factor. As described herein, a precipitating factor may be any endogenous or exogenous factor known to precipitate an acute attack. For example, the premenstrual phase is an endogenous precipitating factor, and a cytochrome P450 inducing drug is an exogenous precipitating factor.

The effective amount for the treatment of a disorder related to ALAS1 expression (e.g., a porphyria such as AIP) depends on the type of disorder to be treated, the severity of the symptoms, the subject being treated, the sex, age and general condition of the subject, the mode of administration and so forth. For any given case, an appropriate “effective amount” can be determined by one of ordinary skill in the art using routine experimentation. It is well within the ability of one skilled in the art to monitor efficacy of treatment or prevention by measuring any one of such parameters, or any combination of parameters. In connection with the administration of an iRNA targeting ALAS1 or pharmaceutical composition thereof, “effective against” a disorder related to ALAS1 expression indicates that administration in a clinically appropriate manner results in a beneficial effect, e.g., for an individual patient or for at least a fraction of patients, e.g., a statistically significant fraction of patients. Beneficial effects include, e.g., prevention of or reduction of symptoms or other effects. For example, beneficial effects include, e.g., an improvement (e.g., decrease in the severity or frequency) of symptoms, a reduction in the severity or frequency of attacks, a reduced risk of developing associated disease (e.g., neuropathy (e.g., progressive neuropathy), hepatocellular cancer), an improved ability to tolerate a precipitating factor, an improvement in quality of life, a reduction in the expression of ALAS1, a reduction in a level (e.g., a plasma or urine level) of a porphyrin or a porphyrin precursor (e.g., ALA and/or PBG) or other effect generally recognized as positive by medical doctors familiar with treating the particular type of disorder.

A treatment or preventive effect is evident when there is an improvement, e.g., a statistically significant improvement in one or more parameters of disease status, or by a failure to worsen or to develop symptoms where they would otherwise be anticipated. As an example, a favorable change of at least 10% in a measurable parameter of disease, e.g., at least 20%, 30%, 40%, 50% or more can be indicative of effective treatment. Efficacy for a given iRNA drug or formulation of that drug can also be judged using an experimental animal model for the given disease as known in the art. When using an experimental animal model, efficacy of treatment is evidenced when a statistically significant reduction in a marker (e.g., plasma or urinary ALA or PBG) or symptom is observed.

Patients can be administered a therapeutic amount of iRNA. The therapeutic amount can be, e.g., 0.05-50 mg/kg. For example, the therapeutic amount can be 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, 2.0, or 2.5, 3.0, 3.5, 4.0, 4.5, 5, 10, 15, 20, 25, 30, 35, 40, 45, or 50 mg/kg dsRNA.

In some embodiments, the iRNA is formulated as a lipid formulation, e.g., an LNP formulation as described herein. In some such embodiments, the therapeutic amount is 0.05-5 mg/kg, e.g., 0.05, 0.1, 0.2, 0.3, 0.4, 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, or 5.0 mg/kg dsRNA. In some embodiments, the lipid formulation, e.g., LNP formulation, is administered intravenously.

In some embodiments, the iRNA is administered by intravenous infusion over a period of time, such as over a 5 minute, 10 minute, 15 minute, 20 minute, or 25 minute period.

In some embodiments, the iRNA is in the form of a GalNAc conjugate as described herein. In some such embodiments, the therapeutic amount is 0.5-50 mg, e.g., 0.5, 0.6, 0.7, 0.8, 0.9, 1.0, 1.5, 2.0, 2.5, 3.0, 3.5, 4.0, 4.5, 5.0, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, or 50 mg/kg dsRNA. In some embodiments, the GalNAc conjugate is administered subcutaneously.

In some embodiments, the administration is repeated, for example, on a regular basis, such as, daily, biweekly (i.e., every two weeks) for one month, two months, three months, four months or longer. After an initial treatment regimen, the treatments can be administered on a less frequent basis. For example, after administration biweekly for three months, administration can be repeated once per month, for six months or a year or longer.

In some embodiments, the iRNA agent is administered in two or more doses. In some embodiments, the number or amount of subsequent doses is dependent on the achievement of a desired effect, e.g., suppression of a ALAS gene, reduction of a level of a porphyrin or porphyrin precursor (e.g., ALA and/or PBG), or the achievement of a therapeutic or prophylactic effect, e.g., reduction or prevention of one or more symptoms associated with porphyria (e.g., pain, e.g., neuropathic pain), and/or prevention of attacks or reduction in the frequency and/or severity of attacks associated with porphyria.

In some embodiments, the iRNA agent is administered according to a schedule. For example, the iRNA agent may be administered once per week, twice per week, three times per week, four times per week, or five times per week. In some embodiments, the schedule involves regularly spaced administrations, e.g., hourly, every four hours, every six hours, every eight hours, every twelve hours, daily, every 2 days, every 3 days, every 4 days, every 5 days, weekly, biweekly, or monthly. In embodiments, the iRNA agent is administered weekly or biweekly to achieve a desired effect, e.g., to decrease the level of ALA and/or PBG, to decrease pain, and/or to prevent acute attacks.

In embodiments, the schedule involves closely spaced administrations followed by a longer period of time during which the agent is not administered. For example, the schedule may involve an initial set of doses that are administered in a relatively short period of time (e.g., about every 6 hours, about every 12 hours, about every 24 hours, about every 48 hours, or about every 72 hours) followed by a longer time period (e.g., about 1 week, about 2 weeks, about 3 weeks, about 4 weeks, about 5 weeks, about 6 weeks, about 7 weeks, or about 8 weeks) during which the iRNA agent is not administered. In one embodiment, the iRNA agent is initially administered hourly and is later administered at a longer interval (e.g., daily, weekly, biweekly, or monthly). In another embodiment, the iRNA agent is initially administered daily and is later administered at a longer interval (e.g., weekly, biweekly, or monthly). In certain embodiments, the longer interval increases over time or is determined based on the achievement of a desired effect. In a specific embodiment, the iRNA agent is administered once daily during an acute attack, followed by weekly dosing starting on the eighth day of administration. In another specific embodiment, the iRNA agent is administered every other day during a first week followed by weekly dosing starting on the eighth day of administration.

In one embodiment, the iRNA agent is administered to prevent or reduce the severity or frequency of recurring attacks, e.g., cyclical attacks associated with a precipitating factor. In some embodiments, the precipitating factor is the menstrual cycle. In some embodiments, the iRNA is administered repeatedly, e.g., at regular intervals to prevent or reduce the severity or frequency of recurring attacks, e.g., cyclical attacks associated with a precipitating factor, e.g., the menstrual cycle, e.g., a particular phase of the menstrual cycle, e.g., the luteal phase. In some embodiments, the iRNA is administered during a particular phase of the menstrual cycle or based on hormone levels of the patient being treated (e.g., based on hormone levels that are associated with a particular phase of the menstrual cycle). In some embodiments, the iRNA is administered on one or more particular days of the menstrual cycle, e.g., on day 1, 2, 3, 4, 5, 6, 7, 8. 9. 10. 11. 12. 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, or on day 28 (or later day for subjects who have a longer menstrual cycle). In some embodiments, the iRNA is administered during the luteal phase, e.g., on one or more days between days 14-28 of the menstrual cycle (or later, in subjects who have a menstrual cycle longer than 28 days). In some embodiments, ovulation of the subject is assessed (e.g., using a blood or urine test that detects a hormone associated with ovulation, e.g., LH) and the iRNA is administered at a predetermined interval after ovulation. In some embodiments, the iRNA is administered immediately after ovulation. In some embodiments, the iRNA is administered 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, or 18 days after ovulation. Any of these schedules may optionally be repeated for one or more iterations. The number of iterations may depend on the achievement of a desired effect, e.g., the suppression of a ALAS1 gene and/or the achievement of a therapeutic or prophylactic effect, e.g., reduce or prevent one or more symptoms associated with porphyria, to reduce the frequency of attacks associated with porphyria.

In some embodiments, an initial dose of the iRNA agent is administered and the level of ALA or PBG is tested, e.g., 1-48 hours, e.g., 2, 4, 8, 12, or 24 hours following administration of the initial dose. In some embodiments, if the level of ALA and/or PBG has decreased (e.g., to achieve a predetermined reduction, e.g., a normalization), and/or if the symptoms associated with porphyria (e.g., pain) have improved (e.g., such that the patient is asymptomatic), no further dose is administered, whereas if the level of ALA and/or PBG has not decreased (e.g., has not achieved a predetermined reduction, e.g., has not normalized), a further dose of ALA or PBG is administered. In some embodiments, the further dose is administered 12, 24, 36, 48, 60, or 72 hours after the initial dose. In some embodiments, if the initial dose is not effective to decrease the level of ALA and/or PBG, the further dose is modified, e.g., increased to achieve a desired decrease (e.g., a predetermined reduction, e.g., a normalization) in ALA or PBG levels.

In some embodiments, the predetermined reduction is a decrease of at least 10%, 20%, 30%, 40%, or 50%. In some embodiments, the predetermined reduction is a reduction that is effective to prevent or ameliorate symptoms, e.g., pain, prodromal symptoms, or recurring attacks.

In some embodiments, the predetermined reduction is a reduction of at least 1, 2, 3, or more standard deviations, wherein the standard deviation is determined based on the values from a reference sample, e.g., a reference sample as described herein.

In some embodiments, the predetermined reduction is a reduction that brings the level of the porphyrin or porphyrin precursor to a level that is less than, or to a level that is less than or equal to, a reference value (e.g., a reference value as described herein).

As used herein, a “normalization” in ALA or PBG levels (or a “normal” or “normalized” level) refers to a level (e.g., a urine and/or plasma level) of either ALA, or PBG, or both, that is within the expected range for a healthy individual, an individual who is asymptomatic (e.g., an individual who does not experience pain and/or suffer from neuropathy), or an individual who does not have a mutation associated with a porphyria. For example, in some embodiments, a normalized level is within two standard deviations of the normal mean. In some embodiments, a normalized level is within normal reference limits, e.g., within the 95% confidence interval for an appropriate control sample, e.g., a sample of healthy individuals or individuals who do not carry a gene mutation associated with a porphyria. In some embodiments, the ALA and/or PBG level of the subject (e.g., the urine and/or plasma ALA and/or PBG level) is monitored at intervals, a further dose of the iRNA agent is administered when the level increases above the reference value

Administration of the iRNA may reduce ALAS1 mRNA or protein levels, e.g., in a cell, tissue, blood, urine or other compartment of the patient by at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80% or at least 90% or more. Administration of the iRNA may reduce levels of products associated with ALAS1 gene expression, e.g., levels of one or more porphyrins or porphyrin precursors (e.g., the level of ALA and/or PBG). Administration of the iRNA agent may also inhibit or prevent the upregulation of ALAS1 mRNA or protein levels during an acute attack of AIP.

Before administration of a full dose of the iRNA, patients can be administered a smaller dose, such as a 5% infusion dose, and monitored for adverse effects, such as an allergic reaction, or for elevated lipid levels or blood pressure. In another example, the patient can be monitored for unwanted effects.

In yet another aspect, the invention provides a method for modulating (e.g., inhibiting or activating) the expression of an ALAS1 gene, e.g., in a cell or in a subject. In some embodiments, the cell is ex vivo, in vitro, or in vivo. In some embodiments, the cell is an erythroid cell or a hepatocyte. In some embodiments, the cell is in a subject (e.g., a mammal, such as, for example, a human). In some embodiments, the subject (e.g., the human) is at risk, or is diagnosed with a disease related to ALAS1 expression, as described above.

In one embodiment, the method includes contacting the cell with an iRNA as described herein, in an amount effective to decrease the expression of an ALAS1 gene in the cell. “Contacting,” as used herein, includes directly contacting a cell, as well as indirectly contacting a cell. For example, a cell within a subject (e.g., an erythroid cell or a liver cell, such as a hepatocyte) may be contacted when a composition comprising an iRNA is administered (e.g., intravenously or subcutaneously) to the subject.

The expression of an ALAS1 gene may be assessed based on the level of expression of an ALAS1 mRNA, an ALAS1 protein, or the level of a parameter functionally linked to the level of expression of an ALAS1 gene (e.g., the level of a porphyrin or the incidence or severity of a symptom related to a porphyria). In some embodiments, the expression of ALAS1 is inhibited by at least 5%, at least 10%, at least 15%, at least 20%, at least 25%, at least 30%, at least 35%, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, at least 65%, at least 70%, at least 75%, at least 80%, at least 85%, at least 90%, or at least 95%. In some embodiments, the iRNA has an IC50 in the range of 0.001-0.01 nM, 0.001-0.10 nM, 0.001-1.0 nM, 0.001-10 nM, 0.01-0.05 nM, 0.01-0.50 nM, 0.02-0.60 nM, 0.01-1.0 nM, 0.01-1.5 nM, 0.01-10 nM. The IC50 value may be normalized relative to an appropriate control value, e.g., the IC50 of a non-targeting iRNA.

In some embodiments, the method includes introducing into the cell an iRNA as described herein and maintaining the cell for a time sufficient to obtain degradation of the mRNA transcript of an ALAS1 gene, thereby inhibiting the expression of the ALAS1 gene in the cell.

In one embodiment, the method includes administering a composition described herein, e.g., a composition comprising an iRNA that targets ALAS1, to the mammal such that expression of the target ALAS1 gene is decreased, such as for an extended duration, e.g., at least two, three, four days or more, e.g., one week, two weeks, three weeks, or four weeks or longer. In some embodiments, the decrease in expression of ALAS1 is detectable within 1 hour, 2 hours, 4 hours, 8 hours, 12 hours, or 24 hours of the first administration.

In another embodiment, the method includes administering a composition as described herein to a mammal such that expression of the target ALAS1 gene is increased by e.g., at least 10% compared to an untreated animal. In some embodiments, the activation of ALAS1 occurs over an extended duration, e.g., at least two, three, four days or more, e.g., one week, two weeks, three weeks, four weeks, or more. Without wishing to be bound by theory, an iRNA can activate ALAS1 expression by stabilizing the ALAS1 mRNA transcript, interacting with a promoter in the genome, and/or inhibiting an inhibitor of ALAS1 expression.

The iRNAs useful for the methods and compositions featured in the invention specifically target RNAs (primary or processed) of an ALAS1 gene. Compositions and methods for inhibiting the expression of an ALAS1 gene using iRNAs can be prepared and performed as described elsewhere herein.

In one embodiment, the method includes administering a composition containing an iRNA, where the iRNA includes a nucleotide sequence that is complementary to at least a part of an RNA transcript of the ALAS1 gene of the mammal to be treated. When the organism to be treated is a mammal such as a human, the composition may be administered by any means known in the art including, but not limited to oral, intraperitoneal, or parenteral routes, including intracranial (e.g., intraventricular, intraparenchymal and intrathecal), intravenous, intramuscular, subcutaneous, transdermal, airway (aerosol), nasal, rectal, and topical (including buccal and sublingual) administration.

In certain embodiments, the compositions are administered by intravenous infusion or injection. In some such embodiments, the compositions comprise a lipid formulated siRNA (e.g., an LNP formulation, such as an LNP11 formulation) for intravenous infusion. In particular embodiments, such compositions may be used to treat acute attacks of porphyria and/or for prophylaxis (e.g., to decrease the severity or frequency of attacks).

In other embodiments, the compositions are administered subcutaneously. In some such embodiments, the compositions comprise an iRNA conjugated to a GalNAc ligand. In particular embodiments, such compositions may be used to treat acute attacks of porphyria or for prophylaxis (e.g., to decrease the severity or frequency of attacks).

In another aspect, the invention provides a method for decreasing a level of a porphyrin or a porphyrin precursor, e.g., in a cell or in a subject.

In some embodiments, the cell is ex vivo, in vitro, or in vivo. In some embodiments, the cell is an erythroid cell or a hepatocyte. In some embodiments, the cell is a hepatocyte. In some embodiments, the cell is in a subject (e.g., a mammal, such as, for example, a human).

In some embodiments, the subject (e.g., the human) is at risk, or is diagnosed with a porphyria, as described herein. In some embodiments, the method is effective to treat a porphyria as described herein (e.g., by ameliorating one or more symptoms associated with a porphyria, reducing the frequency of attacks associated with a porphyria, reducing the likelihood that an attack of one or more symptoms associated with porphyria will occur upon exposure to a precipitating factor, or reducing the risk of developing conditions associated with a porphyria (e.g., neuropathy (e.g., progressive neuropathy), hepatocellular cancer). In one embodiment, the method includes contacting the cell with an RNAi, as described herein, in an amount sufficient to decrease the level of the porphyrin or porphyrin precursor (e.g., ALA or PBG) in the cell, or in another related cell or group of cells, or in the subject. “Contacting,” as used herein, includes directly contacting a cell, as well as indirectly contacting a cell. For example, a cell within a subject (e.g., an erythroid cell or a liver cell, such as a hepatocyte) may be contacted when a composition comprising an RNAi is administered (e.g., intravenously or subcutaneously) to the subject. “Another related cell or group of cells,” as used herein, includes any cell or group of cells in which the level of the porphyrin or porphyrin precursor decreases as a result of the contacting. For example, the cell may be part of a tissue present within a subject (e.g., a liver cell present within a subject), and contacting the cell within the subject (e.g., contacting one or more liver cells present within a subject) with the RNAi may result in a decrease in the level of the porphyrin or porphyrin precursor in another related cell or group of cells (e.g., nerve cells of the subject), or in a tissue or fluid of the subject (e.g., in the urine, blood, plasma, or cerebrospinal fluid of the subject).

In some embodiments, the porphyrin or porphyrin precursor is selected from the group consisting of S-aminolevulinic acid (ALA), porphopilinogen (PBG), hydroxymethylbilane (HMB), uroporphyrinogen III, coproporphyrinogen III, protoporphrinogen IX, and protoporphyrin IX In some embodiments the porphyrin precursor is ALA. In some embodiments, the porphyrin precursor is PBG. In some embodiments, the method decreases the level of ALA and PBG. The level of a porphyrin or a porphyrin precursor may be measured as described herein and as known in the art.

Unless otherwise defined, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this invention belongs. Although methods and materials similar or equivalent to those described herein can be used in the practice or testing of the iRNAs and methods featured in the invention, suitable methods and materials are described below. All publications, patent applications, patents, and other references mentioned herein are incorporated by reference in their entirety. In case of conflict, the present specification, including definitions, will control. In addition, the materials, methods, and examples are illustrative only and not intended to be limiting.

Where the source of a reagent is not specifically given herein, such reagent may be obtained from any supplier of reagents for molecular biology at a quality/purity standard for application in molecular biology.

All oligonucleotides are synthesized on an AKTAoligopilot synthesizer. Commercially available controlled pore glass solid support (dT-CPG, 500 Å, Prime Synthesis) and RNA phosphoramidites with standard protecting groups, 5′-O-dimethoxytrityl N6-benzoyl-2′-t-butyldimethylsilyl-adenosine-3′-O-N,N′-diisopropyl-2-cyanoethylphosphoramidite, 5′-O-dimethoxytrityl-N4-acetyl-2′-t-butyldimethylsilyl-cytidine-3′-0-N,N′-diisopropyl-2-cyanoethylphosphoramidite, 5′-O-dimethoxytrityl-N2-isobutryl-2′-t-butyldimethylsilyl-guanosine-3′-O-N,N′-diisopropyl-2-cyanoethylphosphoramidite, and 5′-O-dimethoxytrityl-2′-t-butyldimethylsilyl-uridine-3′-O-N,N′-diisopropyl-2-cyanoethylphosphoramidite (Pierce Nucleic Acids Technologies) were used for the oligonucleotide synthesis. The 2′-F phosphoramidites, 5′-O-dimethoxytrityl-N4-acetyl-2′-fluro-cytidine-3′-0-N,N′-diisopropyl-2-cyanoethyl-phosphoramidite and 5′-O-dimethoxytrityl-2′-fluro-uridine-3′-O-N,N′-diisopropyl-2-cyanoethyl-phosphoramidite are purchased from (Promega). All phosphoramidites are used at a concentration of 0.2M in acetonitrile (CH3CN) except for guanosine which is used at 0.2M concentration in 10% THF/ANC (v/v). Coupling/recycling time of 16 minutes is used. The activator is 5-ethyl thiotetrazole (0.75M, American International Chemicals); for the PO-oxidation iodine/water/pyridine is used and for the PS-oxidation PADS (2%) in 2,6-lutidine/ACN (1:1 v/v) is used.

3′-ligand conjugated strands are synthesized using solid support containing the corresponding ligand. For example, the introduction of cholesterol unit in the sequence is performed from a hydroxyprolinol-cholesterol phosphoramidite. Cholesterol is tethered to trans-4-hydroxyprolinol via a 6-aminohexanoate linkage to obtain a hydroxyprolinol-cholesterol moiety. 5′-end Cy-3 and Cy-5.5 (fluorophore) labeled iRNAs are synthesized from the corresponding Quasar-570 (Cy-3) phosphoramidite are purchased from Biosearch Technologies. Conjugation of ligands to 5′-end and or internal position is achieved by using appropriately protected ligand-phosphoramidite building block. An extended 15 min coupling of 0.1 M solution of phosphoramidite in anhydrous CH3CN in the presence of 5-(ethylthio)-1H-tetrazole activator to a solid-support-bound oligonucleotide. Oxidation of the internucleotide phosphite to the phosphate is carried out using standard iodine-water as reported (1) or by treatment with tert-butyl hydroperoxide/acetonitrile/water (10: 87: 3) with 10 min oxidation wait time conjugated oligonucleotide. Phosphorothioate is introduced by the oxidation of phosphite to phosphorothioate by using a sulfur transfer reagent such as DDTT (purchased from AM Chemicals), PADS and or Beaucage reagent. The cholesterol phosphoramidite is synthesized in house and used at a concentration of 0.1 M in dichloromethane. Coupling time for the cholesterol phosphoramidite is 16 minutes.

After completion of synthesis, the support is transferred to a 100 mL glass bottle (VWR). The oligonucleotide is cleaved from the support with simultaneous deprotection of base and phosphate groups with 80 mL of a mixture of ethanolic ammonia [ammonia: ethanol (3:1)] for 6.5 h at 55° C. The bottle is cooled briefly on ice and then the ethanolic ammonia mixture is filtered into a new 250-mL bottle. The CPG is washed with 2×40 mL portions of ethanol/water (1:1 v/v). The volume of the mixture is then reduced to ˜30 mL by roto-vap. The mixture is then frozen on dry ice and dried under vacuum on a speed vac.

Deprotection II (Removal of 2′-TBDMS group)

The dried residue is resuspended in 26 mL of triethylamine, triethylamine trihydrofluoride (TEA·3HF) or pyridine-HF and DMSO (3:4:6) and heated at 60° C. for 90 minutes to remove the tert-butyldimethylsilyl (TBDMS) groups at the 2′ position. The reaction is then quenched with 50 mL of 20 mM sodium acetate and the pH is adjusted to 6.5. Oligonucleotide is stored in a freezer until purification.

The oligonucleotides are analyzed by high-performance liquid chromatography (HPLC) prior to purification and selection of buffer and column depends on nature of the sequence and or conjugated ligand.

The ligand-conjugated oligonucleotides are purified by reverse-phase preparative HPLC. The unconjugated oligonucleotides are purified by anion-exchange HPLC on a TSK gel column packed in house. The buffers are 20 mM sodium phosphate (pH 8.5) in 10% CH3CN (buffer A) and 20 mM sodium phosphate (pH 8.5) in 10% CH3CN, 1M NaBr (buffer B). Fractions containing full-length oligonucleotides are pooled, desalted, and lyophilized. Approximately 0.15 OD of desalted oligonucleotidess are diluted in water to 150 μL and then pipetted into special vials for CGE and LC/MS analysis. Compounds are then analyzed by LC-ESMS and CGE.

siRNA Preparation

For the general preparation of siRNA, equimolar amounts of sense and antisense strand are heated in 1×PBS at 95° C. for 5 min and slowly cooled to room temperature. Integrity of the duplex is confirmed by HPLC analysis.

Nucleic acid sequences are represented below using standard nomenclature, and specifically the abbreviations of Table 1.

TABLE 1
Abbreviations of nucleotide monomers used in nucleic acid sequence
representation. It will be understood that these monomers, when present in an oligonucleotide,
are mutually linked by 5′-3′-phosphodiester bonds.
AbbreviationNucleotide(s)
AAdenosine-3′-phosphate
Abbeta-L-adenosine-3′-phosphate
Absbeta-L-adenosine-3′-phosphorothioate
Af2′-fluoroadenosine-3′-phosphate
Afs2′-fluoroadenosine-3′-phosphorothioate
Asadenosine-3′-phosphorothioate
Ccytidine-3′-phosphate
Cbbeta-L-cytidine-3′-phosphate
Cbsbeta-L-cytidine-3′-phosphorothioate
Cf2′-fluorocytidine-3′-phosphate
Cfs2′-fluorocytidine-3′-phosphorothioate
(Chd)2′-O-hexadecyl-cytidine-3′-phosphate
(Chds)2′-O-hexadecyl-cytidine-3′-phosphorothioate
Cscytidine-3′-phosphorothioate
Gguanosine-3′-phosphate
Gbbeta-L-guanosine-3′-phosphate
Gbsbeta-L-guanosine-3′-phosphorothioate
Gf2′-fluoroguanosine-3′-phosphate
Gfs2′-fluoroguanosine-3′-phosphorothioate
Gsguanosine-3′-phosphorothioate
T5′-methyluridine-3′-phosphate
Tbbeta-L-thymidine-3′-phosphate
Tbsbeta-L-thymidine-3′-phosphorothioate
Tf2′-fluoro-5-methyluridine-3′-phosphate
Tfs2′-fluoro-5-methyluridine-3′-phosphorothioate
Ts5-methyluridine-3′-phosphorothioate
UUridine-3′-phosphate
Ubbeta-L-uridine-3′-phosphate
Ubsbeta-L-uridine-3′-phosphorothioate
Uf2′-fluorouridine-3′-phosphate
Ufs2′-fluorouridine-3′-phosphorothioate
(Uhd)2′-O-hexadecyl-uridine-3′-phosphate
(Uhds)2′-O-hexadecyl-uridine-3′-phosphorothioate
Usuridine-3′-phosphorothioate
Nany nucleotide (G, A, C, T or U)
a2′-O-methyladenosine-3′-phosphate
as2′-O-methyladenosine-3′-phosphorothioate
c2′-O-methylcytidine-3′-phosphate
cs2′-O-methylcytidine-3′-phosphorothioate
g2′-O-methylguanosine-3′-phosphate
gs2′-O-methylguanosine-3′-phosphorothioate
t2′-O-methyl-5-methyluridine-3′-phosphate
ts2′-O-methyl-5-methyluridine-3′-phosphorothioate
u2′-O-methyluridine-3′-phosphate
us2′-O-methyluridine-3′-phosphorothioate
dA2′-deoxyadenosine-3′-phosphate
dAs2′-deoxyadenosine-3′-phosphorothioate
dC2′-deoxycytidine-3′-phosphate
dCs2′-deoxycytidine-3′-phosphorothioate
dG2′-deoxyguanosine-3′-phosphate
dGs2′-deoxyguanosine-3′-phosphorothioate
dT2′-deoxythymidine
dTs2′-deoxythymidine-3′-phosphorothioate
dU2′-deoxyuridine
sphosphorothioate linkage
L961N-[tris(GalNAc-alkyl)-amidodecanoyl)]-4-hydroxyprolinol Hyp-(GalNAc-alkyl)3
(Aeo)2′-O-methoxyethyladenosine-3′-phosphate
(Aeos)2′-O-methoxyethyladenosine-3′-phosphorothioate
(Geo)2′-O-methoxyethylguanosine-3′-phosphate
(Geos)2′-O-methoxyethylguanosine-3′-phosphorothioate
(Teo)2′-O-methoxyethyl-5-methyluridine-3′-phosphate
(Teos)2′-O-methoxyethyl-5-methyluridine-3′-phosphorothioate
(m5Ceo)2′-O-methoxyethyl-5-methylcytidine-3′-phosphate
(m5Ceos)2′-O-methoxyethyl-5-methylcytidine-3′-phosphorothioate
1The chemical structure of L96 is as follows:

siRNA design was carried out to identify siRNAs targeting human, rhesus (Macaca mulatta), mouse, and rat ALAS1 transcripts annotated in the NCBI Gene database (http://www.ncbi.nlm.nih.gov/gene/). Design used the following transcripts from the NCBI RefSeq collection: Human-NM_000688.4 (see FIG. 3), NM_199166.1; Rhesus-XM_001090440.2, XM_001090675.2; Mouse-NM_020559.2; Rat-NM_024484.2. Due to high primate/rodent sequence divergence, siRNA duplexes were designed in several separate batches, including but not limited to batches containing duplexes matching human and rhesus transcripts only; human, rhesus, mouse, and rat transcripts only; and mouse and rat transcripts only. Most siRNA duplexes were designed that shared 100% identity the listed human transcript and other species transcripts considered in each design batch (above). In some instances, (see Table 8) mismatches between duplex and mRNA target were allowed at the first antisense (last sense) position when the antisense strand:target mRNA complementary basepair was a GC or CG pair. In these cases, duplexes were designed with UA or AU pairs at the first antisense:last sense pair. Thus the duplexes maintained complementarity but were mismatched with respect to target (U:C, U:G, A:C, or A:G). Eighteen of these “UA-swap” duplexes were designed as part of the human/rhesus/mouse/rat set (see duplexes in Table 8 with “C19U”, “G19U”, “C19A”, or “G19A” labels in the Position column).

siRNA Design, Specificity, and Efficacy Prediction

The predicted specificity of all possible 19mers was predicted from each sequence. Candidate 19mers were then selected that lacked repeats longer than 7 nucleotides. These 1510 candidate human/rhesus, 114 human/rhesus/mouse/rat, and 717 mouse/rat siRNAs were used in comprehensive searches against the appropriate transcriptomes (defined as the set of NM_ and XM_records within the human, rhesus, dog, mouse, or rat NCBI Refseq sets) using an exhaustive “brute-force” algorithm implemented in the python script ‘BruteForce.py’. The script next parsed the transcript-oligo alignments to generate a score based on the position and number of mismatches between the siRNA and any potential ‘off-target’ transcript. The off-target score is weighted to emphasize differences in the ‘seed’ region of siRNAs, in positions 2-9 from the 5′ end of the molecule. Each oligo-transcript pair from the brute-force search was given a mismatch score by summing the individual mismatch scores; mismatches in the position 2-9 were counted as 2.8, mismatches in the cleavage site positions 10-11 were counted as 1.2, and mismatches in region 12-19 counted as 1.0. An additional off-target prediction was carried out by comparing the frequency of heptamers and octomers derived from 3 distinct, seed-derived hexamers of each oligo. The hexamers from positions 2-7 relative to the 5′ start is used to create 2 heptamers and one octomer. We create ‘heptamer1’ by adding a 3′ A to the hexamer; we create heptamer2 by adding a 5′ A to the hexamer; we create the octomer by adding an A to both 5′ and 3′ ends of the hexamer. The frequency of octomers and heptamers in the human, rhesus, mouse, or rat 3′UTRome (defined as the subsequence of the transcriptome from NCBI's Refseq database where the end of the coding region, the ‘CDS’, is clearly defined) was pre-calculated. The octomer frequency was normalized to the heptamer frequency using the median value from the range of octomer frequencies. A ‘mirSeedScore’ was then calculated by calculating the sum of ((3×normalized octomer count)+(2×heptamer2 count)+(1×heptamer1 count)).

Both siRNAs strands were assigned to a category of specificity according to the calculated scores: a score above 3 qualifies as highly specific, equal to 3 as specific and between 2.2 and 2.8 as moderately specific. We sorted by the specificity of the antisense strand. We then selected duplexes whose antisense oligos lacked GC at the first position, lacked G at both positions 13 and 14, and had 3 or more Us or As in the seed region (characteristics of duplexes with high predicted efficacy)

Candidate GalNac-conjugated duplexes, 21 and 23 nucleotides long on the sense and antisense strands respectively, were designed by extending antisense 19mers 4 additional nucleotides in the 3′ direction (preserving perfect complementarity with the target transcript). The sense strand was specified as the reverse complement of the first 21 nucleotides of the antisense 23mer. Duplexes were selected that maintained perfect matches to all selected species transcripts across all 23 nucleotides.

siRNA Sequence Selection

A total of 90 sense and 90 antisense derived human/rhesus, 40 sense and 40 antisense derived human/rhesus/mouse/mouse/rat, and 40 sense and 40 antisense derived mouse/rat siRNA 19mer oligos were synthesized and formed into duplexes. A total of 45 sense and 45 antisense derived human/rhesus 21/23mer oligos were synthesized to yield 45 GalNac-conjugated duplexes.

The sequences of the sense and antisense strands of the modified duplexes are shown in Table 2, and the sequences of the sense and antisense strands of the unmodified duplexes are shown in Table 3.

ALAS1 sequences were synthesized on MerMade 192 synthesizer at either 1 or 0.2 umol scale. Single strands were made with 2′O-methyl modifications for in vitro screening using transfection reagents. 3′ GalNAc conjugates were made with sequences containing 2′F and 2′-O-methyl modifications on the sense strand in the 21-23 mer designs for free uptake in cells. For all the 21mer sequences in the list, ‘endolight’ chemistry was applied as detailed below.

    • All pyrimidines (cytosine and uridine) in the sense strand contained 2′-O-Methyl bases (2′ O-Methyl C and 2′-O-Methyl U)
    • In the antisense strand, pyrimidines adjacent to (towards 5′ position) ribo A nucleoside were replaced with their corresponding 2-O-Methyl nucleosides
    • A two base dTsdT extension at 3′ end of both sense and anti sense sequences was introduced
    • The sequence file was converted to a text file to make it compatible for loading in the MerMade 192 synthesis software

For GalNAc conjugated sense strands and complementary antisense sequences, 2′F and other modified nucleosides were introduced in combination with ribo with 2′O-Methyl nucleosides. The synthesis was performed on a GalNAc modified CPG support for the sense strand and CPG modified with universal support on the antisense sequence.

The synthesis of ALAS1 sequences used solid supported oligonucleotide synthesis using phosphoramidite chemistry. For 21 mer endolight sequences, a deoxy thymidine CPG was used as the solid support while for the GalNAc conjugates, GalNAc solid support for sense strand and an universal CPG for the antisesense strand were used.

The synthesis of the above sequences was performed at either 1 or 0.2 um scale in 96 well plates. The amidite solutions were prepared at 0.1M concentration and ethyl thio tetrazole (0.6M in Acetonitrile) was used as activator.

The synthesized sequences were cleaved and deprotected in 96 well plates, using methylamine in the first step and fluoride reagent in the second step. For GalNAc and 2′F nucleoside containing sequences, deprotection conditions were modified. Sequences after cleavage and deprotection were precipitated using acetone:ethanol (80:20) mix and the pellet were re-suspended in 0.2M sodium acetate buffer. Samples from each sequence were analyzed by LC-MS to confirm the identity, UV for quantification and a selected set of samples by IEX chromatography to determine purity.

ALAS1 sequences were precipitated and purified on AKTA Purifier system using Sephadex column. The ALAS less was run at ambient temperature. Sample injection and collection was performed in 96 well (1.8 mL-deep well) plates. A single peak corresponding to the full length sequence was collected in the eluent. The desalted ALAS1 sequences were analyzed for concentration (by UV measurement at A260) and purity (by ion exchange HPLC). The complementary single strands were then combined in a 1:1 stoichiometric ratio to form siRNA duplexes.

TABLE 2
Human ALAS1 Modified Single Strands and Duplex Sequences
SEQ ID
SEQ IDNO:Position on
NO:(anti-transcriptDuplexSense SequenceAntisense Sequence
(sense)sense)NM_000688.4Name(5′-3′)(5′-3′)
23522-540AD-55078.2cuccGGccAGuGAGAAAGAdTsdTUCUUUCUcACUGGCCGGAGdTsdT
45669-687AD-55084.2uGGcAGcAcAGAuGAAucAdTsdTUGAUUcAUCUGUGCUGCcAdTsdT
67790-808AD-55090.2cAGuGuGGuuAGuGuGAAAdTsdTUUUcAcACuAACcAcACUGdTsdT
89853-871AD-55096.2cAucAuGcAAAAGcAAAGAdTsdTUCUUUGCUUUUGcAUGAUGdTsdT
1011876-894AD-55102.2AAAGAGuGucucAucuucudTsdTAGAAGAUGAGAcACUCUUUdTsdT
1213877-895AD-55106.2AAGAGuGucucAucuucuudTsdTAAGAAGAUGAGAcACUCUUdTsdT
1415914-932AD-55111.2ucuGuuuccAcuuuucAGudTsdTACUGAAAAGUGGAAAcAGAdTsdT
1617923-941AD-55073.2AcuuuucAGuAuGAucGuudTsdTAACGAUcAuACUGAAAAGUdTsdT
1819926-944AD-55079.2uuucAGuAuGAucGuuucudTsdTAGAAACGAUcAuACUGAAAdTsdT
2021927-945AD-55085.2uucAGuAuGAucGuuucuudTsdTAAGAAACGAUcAuACUGAAdTsdT
2223928-946AD-55091.2ucAGuAuGAucGuuucuuudTsdTAAAGAAACGAUcAuACUGAdTsdT
2425932-950AD-55097.2uAuGAucGuuucuuuGAGAdTsdTUCUcAAAGAAACGAUcAuAdTsdT
2627973-991AD-55103.2uGAccAcAccuAucGAGuudTsdTAACUCGAuAGGUGUGGUcAdTsdT
2829975-993AD-55107.2AccAcAccuAucGAGuuuudTsdTAAAACUCGAuAGGUGUGGUdTsdT
30311029-1047AD-55112.2uGGcAGAuGAcuAuucAGAdTsdTUCUGAAuAGUcAUCUGCcAdTsdT
32331077-1095AD-55074.2ucuGGuGcAGuAAuGAcuAdTsdTuAGUcAUuACUGcACcAGAdTsdT
34351124-1142AD-55080.2uGuGGGGcAGuuAuGGAcAdTsdTUGUCcAuAACUGCCCcAcAdTsdT
36371137-1155AD-55086.2uGGAcAcuuuGAAAcAAcAdTsdTUGUUGUUUcAAAGUGUCcAdTsdT
38391182-1200AD-55098.2AuAuuucuGGAAcuAGuAAdTsdTUuACuAGUUCcAGAAAuAUdTsdT
40411184-1202AD-55104.2AuuucuGGAAcuAGuAAAudTsdTAUUuACuAGUUCcAGAAAUdTsdT
42431185-1203AD-55108.2uuucuGGAAcuAGuAAAuudTsdTAAUUuACuAGUUCcAGAAAdTsdT
44451188-1206AD-55113.2cuGGAAcuAGuAAAuuccAdTsdTUGGAAUUuACuAGUUCcAGdTsdT
46471325-1343AD-55075.2uGuGAGAuuuAcucuGAuudTsdTAAUcAGAGuAAAUCUcAcAdTsdT
48491364-1382AD-55081.2AuccAAGGGAuucGAAAcAdTsdTUGUUUCGAAUCCCUUGGAUdTsdT
50511382-1400AD-55087.2AGccGAGuGccAAAGuAcAdTsdTUGuACUUUGGcACUCGGCUdTsdT
52531478-1496AD-55093.2uuuGAAAcuGuccAuucAAdTsdTUUGAAUGGAcAGUUUcAAAdTsdT
54551531-1549AD-55099.2uGAuGuGGcccAuGAGuuudTsdTAAACUcAUGGGCcAcAUcAdTsdT
56571631-1649AD-53573.3GucAuGccAAAAAuGGAcAdTsdTUGUCcAUUUUUGGcAUGACdTsdT
58591637-1655AD-55109.2ccAAAAAuGGAcAucAuuudTsdTAAAUGAUGUCcAUUUUUGGdTsdT
60611706-1724AD-55114.2AcGAGuucucuGAuuGAcAdTsdTUGUcAAUcAGAGAACUCGUdTsdT
62631962-1980AD-55076.2AAGucuGuGAuGAAcuAAudTsdTAUuAGUUcAUcAcAGACUUdTsdT
64651967-1985AD-55082.2uGuGAuGAAcuAAuGAGcAdTsdTUGCUcAUuAGUUcAUcAcAdTsdT
66671977-1995AD-55088.2uAAuGAGcAGAcAuAAcAudTsdTAUGUuAUGUCUGCUcAUuAdTsdT
68692189-2207AD-55094.2uuuGAAGuGAuGAGuGAAAdTsdTUUUcACUcAUcACUUcAAAdTsdT
70712227-2245AD-55100.2AGGcuuGAGcAAGuuGGuAdTsdTuACcAACUUGCUcAAGCCUdTsdT
72732313-2331AD-55105.2ucuucAGAGuuGucuuuAudTsdTAuAAAGAcAACUCUGAAGAdTsdT
74752317-2335AD-55110.2cAGAGuuGucuuuAuAuGudTsdTAcAuAuAAAGAcAACUCUGdTsdT
76772319-2337AD-55115.2GAGuuGucuuuAuAuGuGAdTsdTUcAcAuAuAAAGAcAACUCdTsdT
78792320-2338AD-55077.2AGuuGucuuuAuAuGuGAAdTsdTUUcAcAuAuAAAGAcAACUdTsdT
80812344-2362AD-55083.2uuAuAuuAAAuuuuAAucudTsdTAGAUuAAAAUUuAAuAuAAdTsdT
82832352-2370AD-55089.2AAuuuuAAucuAuAGuAAAdTsdTUUuACuAuAGAUuAAAAUUdTsdT
84852353-2371AD-55095.2AuuuuAAucuAuAGuAAAAdTsdTUUUuACuAuAGAUuAAAAUdTsdT
86872376-2394AD-55101.2AGuccuGGAAAuAAAuucudTsdTAGAAUUuAUUUCcAGGACUdTsdT
8889358-376AD-53511.1cuGcccAuucuuAucccGAdTsdTUCGGGAuAAGAAUGGGcAGdTsdT
9091789-807AD-53512.1ccAGuGuGGuuAGuGuGAAdTsdTUUcAcACuAACcAcACUGGdTsdT
92931076-1094AD-53513.1GucuGGuGcAGuAAuGAcudTsdTAGUcAUuACUGcACcAGACdTsdT
94951253-1271AD-53514.1GcAcucuuGuuuuccucGudTsdTACGAGGAAAAcAAGAGUGCdTsdT
96971544-1562AD-53515.1GAGuuuGGAGcAAucAccudTsdTAGGUGAUUGCUCcAAACUCdTsdT
98992228-2246AD-53516.1GGcuuGAGcAAGuuGGuAudTsdTAuACcAACUUGCUcAAGCCdTsdT
100101404-422AD-53517.1GGcAAAucucuGuuGuucudTsdTAGAAcAAcAGAGAUUUGCCdTsdT
102103404-422AD-53517.1GGcAAAucucuGuuGuucudTsdTAGAAcAAcAGAGAUUUGCCdTsdT
104105866-884AD-53518.1cAAAGAccAGAAAGAGuGudTsdTAcACUCUUUCUGGUCUUUGdTsdT
1061071080-1098AD-53519.1GGuGcAGuAAuGAcuAccudTsdTAGGuAGUcAUuACUGcACCdTsdT
1081091258-1276AD-53520.1cuuGuuuuccucGuGcuuudTsdTAAAGcACGAGGAAAAcAAGdTsdT
1101111616-1634AD-53521.1GGGGAucGGGAuGGAGucAdTsdTUGACUCcAUCCCGAUCCCCdTsdT
1121132230-2248AD-53522.1cuuGAGcAAGuuGGuAucudTsdTAGAuACcAACUUGCUcAAGdTsdT
114115436-454AD-53523.1ccccAAGAuGAuGGAAGuudTsdTAACUUCcAUcAUCUUGGGGdTsdT
116117436-454AD-53523.1ccccAAGAuGAuGGAAGuudTsdTAACUUCcAUcAUCUUGGGGdTsdT
118119885-903AD-53524.1cucAucuucuucAAGAuAAdTsdTUuAUCUUGAAGAAGAUGAGdTsdT
1201211127-1145AD-53525.1GGGGcAGuuAuGGAcAcuudTsdTAAGUGUCcAuAACUGCCCCdTsdT
1221231315-1333AD-53526.1GAuGccAGGcuGuGAGAuudTsdTAAUCUcAcAGCCUGGcAUCdTsdT
1241251870-1888AD-53527.1GAGAcAGAuGcuAAuGGAudTsdTAUCcAUuAGcAUCUGUCUCdTsdT
1261272286-2304AD-53528.1ccccAGGccAuuAucAuAudTsdTAuAUGAuAAUGGCCUGGGGdTsdT
128129489-507AD-53529.1cAGcAGuAcAcuAccAAcAdTsdTUGUUGGuAGUGuACUGCUGdTsdT
130131489-507AD-53529.1cAGcAGuAcAcuAccAAcAdTsdTUGUUGGuAGUGuACUGCUGdTsdT
132133915-933AD-53530.1cuGuuuccAcuuuucAGuAdTsdTuACUGAAAAGUGGAAAcAGdTsdT
1341351138-1156AD-53531.1GGAcAcuuuGAAAcAAcAudTsdTAUGUUGUUUcAAAGUGUCCdTsdT
1361371324-1342AD-53532.1cuGuGAGAuuuAcucuGAudTsdTAUcAGAGuAAAUCUcAcAGdTsdT
1381391927-1945AD-53533.1cccuGuGcGGGuuGcAGAudTsdTAUCUGcAACCCGcAcAGGGdTsdT
1401412312-2330AD-53534.1GucuucAGAGuuGucuuuAdTsdTuAAAGAcAACUCUGAAGACdTsdT
142143646-664AD-53535.1cAcuGcAAGcAAAuGcccudTsdTAGGGcAUUUGCUUGcAGUGdTsdT
144145922-940AD-53536.1cAcuuuucAGuAuGAucGudTsdTACGAUcAuACUGAAAAGUGdTsdT
1461471163-1181AD-53537.1GGGGcAGGuGGuAcuAGAAdTsdTUUCuAGuACcACCUGCCCCdTsdT
1481491347-1365AD-53538.1GGAAccAuGccuccAuGAudTsdTAUcAUGGAGGcAUGGUUCCdTsdT
1501511964-1982AD-53539.1GucuGuGAuGAAcuAAuGAdTsdTUcAUuAGUUcAUcAcAGACdTsdT
1521532321-2339AD-53540.1GuuGucuuuAuAuGuGAAudTsdTAUUcAcAuAuAAAGAcAACdTsdT
154155671-689AD-53541.1GcAGcAcAGAuGAAucAGAdTsdTUCUGAUUcAUCUGUGCUGCdTsdT
156157924-942AD-53542.1cuuuucAGuAuGAucGuuudTsdTAAACGAUcAuACUGAAAAGdTsdT
1581591164-1182AD-53543.1GGGcAGGuGGuAcuAGAAAdTsdTUUUCuAGuACcACCUGCCCdTsdT
1601611460-1478AD-53544.1GuccccAAGAuuGuGGcAudTsdTAUGCcAcAAUCUUGGGGACdTsdT
1621631976-1994AD-53545.1cuAAuGAGcAGAcAuAAcAdTsdTUGUuAUGUCUGCUcAUuAGdTsdT
164165786-804AD-53546.1GccccAGuGuGGuuAGuGudTsdTAcACuAACcAcACUGGGGCdTsdT
166167935-953AD-53547.1GAucGuuucuuuGAGAAAAdTsdTUUUUCUcAAAGAAACGAUCdTsdT
1681691165-1183AD-53548.1GGcAGGuGGuAcuAGAAAudTsdTAUUUCuAGuACcACCUGCCdTsdT
1701711530-1548AD-53549.1GuGAuGuGGcccAuGAGuudTsdTAACUcAUGGGCcAcAUcACdTsdT
1721732003-2021AD-53550.1cAAGcAAucAAuuAcccuAdTsdTuAGGGuAAUUGAUUGCUUGdTsdT
174175788-806AD-53551.1cccAGuGuGGuuAGuGuGAdTsdTUcAcACuAACcAcACUGGGdTsdT
176177974-992AD-53552.1GAccAcAccuAucGAGuuudTsdTAAACUCGAuAGGUGUGGUCdTsdT
1781791191-1209AD-53553.1GAAcuAGuAAAuuccAuGudTsdTAcAUGGAAUUuACuAGUUCdTsdT
1801811541-1559AD-53554.1cAuGAGuuuGGAGcAAucAdTsdTUGAUUGCUCcAAACUcAUGdTsdT
1821832075-2093AD-53555.1ccccAGAuGAuGAAcuAcudTsdTAGuAGUUcAUcAUCUGGGGdTsdT
184185360-378AD-53561.1GcccAuucuuAucccGAGudTsdTACUCGGGAuAAGAAUGGGCdTsdT
1861871356-1374AD-53567.1ccuccAuGAuccAAGGGAudTsdTAUCCCUUGGAUcAUGGAGGdTsdT
1881891631-1649AD-53573.1GucAuGccAAAAAuGGAcAdTsdTUGUCcAUUUUUGGcAUGACdTsdT
1901911634-1652AD-53579.1AuGccAAAAAuGGAcAucAdTsdTUGAUGUCcAUUUUUGGcAUdTsdT
TABLE 3
Human ALAS1 Unmodified Single Strands and Duplex Sequences
SEQ ID
SEQ IDNO:Position on
NO:(anti-transcriptDuplexSense SequenceAntisense Sequence
(sense)sense)NM_000688.4Name(5′-3′)(5'-3')
192193522-540AD-55078.2CUCCGGCCAGUGAGAAAGAUCUUUCUCACUGGCCGGAG
194195669-687AD-55084.2UGGCAGCACAGAUGAAUCAUGAUUCAUCUGUGCUGCCA
196197790-808AD-55090.2CAGUGUGGUUAGUGUGAAAUUUCACACUAACCACACUG
198199853-871AD-55096.2CAUCAUGCAAAAGCAAAGAUCUUUGCUUUUGCAUGAUG
200201876-894AD-55102.2AAAGAGUGUCUCAUCUUCUAGAAGAUGAGACACUCUUU
202203877-895AD-55106.2AAGAGUGUCUCAUCUUCUUAAGAAGAUGAGACACUCUU
204205914-932AD-55111.2UCUGUUUCCACUUUUCAGUACUGAAAAGUGGAAACAGA
206207923-941AD-55073.2ACUUUUCAGUAUGAUCGUUAACGAUCAUACUGAAAAGU
208209926-944AD-55079.2UUUCAGUAUGAUCGUUUCUAGAAACGAUCAUACUGAAA
210211927-945AD-55085.2UUCAGUAUGAUCGUUUCUUAAGAAACGAUCAUACUGAA
212213928-946AD-55091.2UCAGUAUGAUCGUUUCUUUAAAGAAACGAUCAUACUGA
214215932-950AD-55097.2UAUGAUCGUUUCUUUGAGAUCUCAAAGAAACGAUCAUA
216217973-991AD-55103.2UGACCACACCUAUCGAGUUAACUCGAUAGGUGUGGUCA
218219975-993AD-55107.2ACCACACCUAUCGAGUUUUAAAACUCGAUAGGUGUGGU
2202211029-1047AD-55112.2UGGCAGAUGACUAUUCAGAUCUGAAUAGUCAUCUGCCA
2222231077-1095AD-55074.2UCUGGUGCAGUAAUGACUAUAGUCAUUACUGCACCAGA
2242251124-1142AD-55080.2UGUGGGGCAGUUAUGGACAUGUCCAUAACUGCCCCACA
2262271137-1155AD-55086.2UGGACACUUUGAAACAACAUGUUGUUUCAAAGUGUCCA
2282291182-1200AD-55098.2AUAUUUCUGGAACUAGUAAUUACUAGUUCCAGAAAUAU
2302311184-1202AD-55104.2AUUUCUGGAACUAGUAAAUAUUUACUAGUUCCAGAAAU
2322331185-1203AD-55108.2UUUCUGGAACUAGUAAAUUAAUUUACUAGUUCCAGAAA
2342351188-1206AD-55113.2CUGGAACUAGUAAAUUCCAUGGAAUUUACUAGUUCCAG
2362371325-1343AD-55075.2UGUGAGAUUUACUCUGAUUAAUCAGAGUAAAUCUCACA
2382391364-1382AD-55081.2AUCCAAGGGAUUCGAAACAUGUUUCGAAUCCCUUGGAU
2402411382-1400AD-55087.2AGCCGAGUGCCAAAGUACAUGUACUUUGGCACUCGGCU
2422431478-1496AD-55093.2UUUGAAACUGUCCAUUCAAUUGAAUGGACAGUUUCAAA
2442451531-1549AD-55099.2UGAUGUGGCCCAUGAGUUUAAACUCAUGGGCCACAUCA
2462471631-1649AD-53573.3GUCAUGCCAAAAAUGGACAUGUCCAUUUUUGGCAUGAC
2482491637-1655AD-55109.2CCAAAAAUGGACAUCAUUUAAAUGAUGUCCAUUUUUGG
2502511706-1724AD-55114.2ACGAGUUCUCUGAUUGACAUGUCAAUCAGAGAACUCGU
2522531962-1980AD-55076.2AAGUCUGUGAUGAACUAAUAUUAGUUCAUCACAGACUU
2542551967-1985AD-55082.2UGUGAUGAACUAAUGAGCAUGCUCAUUAGUUCAUCACA
2562571977-1995AD-55088.2UAAUGAGCAGACAUAACAUAUGUUAUGUCUGCUCAUUA
2582592189-2207AD-55094.2UUUGAAGUGAUGAGUGAAAUUUCACUCAUCACUUCAAA
2602612227-2245AD-55100.2AGGCUUGAGCAAGUUGGUAUACCAACUUGCUCAAGCCU
2622632313-2331AD-55105.2UCUUCAGAGUUGUCUUUAUAUAAAGACAACUCUGAAGA
2642652317-2335AD-55110.2CAGAGUUGUCUUUAUAUGUACAUAUAAAGACAACUCUG
2662672319-2337AD-55115.2GAGUUGUCUUUAUAUGUGAUCACAUAUAAAGACAACUC
2682692320-2338AD-55077.2AGUUGUCUUUAUAUGUGAAUUCACAUAUAAAGACAACU
2702712344-2362AD-55083.2UUAUAUUAAAUUUUAAUCUAGAUUAAAAUUUAAUAUAA
2722732352-2370AD-55089.2AAUUUUAAUCUAUAGUAAAUUUACUAUAGAUUAAAAUU
2742752353-2371AD-55095.2AUUUUAAUCUAUAGUAAAAUUUUACUAUAGAUUAAAAU
2762772376-2394AD-55101.2AGUCCUGGAAAUAAAUUCUAGAAUUUAUUUCCAGGACU
278279358-376AD-53511.1CUGCCCAUUCUUAUCCCGAUCGGGAUAAGAAUGGGCAG
280281789-807AD-53512.1CCAGUGUGGUUAGUGUGAAUUCACACUAACCACACUGG
2822831076-1094AD-53513.1GUCUGGUGCAGUAAUGACUAGUCAUUACUGCACCAGAC
2842851253-1271AD-53514.1GCACUCUUGUUUUCCUCGUACGAGGAAAACAAGAGUGC
2862871544-1562AD-53515.1GAGUUUGGAGCAAUCACCUAGGUGAUUGCUCCAAACUC
2882892228-2246AD-53516.1GGCUUGAGCAAGUUGGUAUAUACCAACUUGCUCAAGCC
290291404-422AD-53517.1GGCAAAUCUCUGUUGUUCUAGAACAACAGAGAUUUGCC
292293404-422AD-53517.1GGCAAAUCUCUGUUGUUCUAGAACAACAGAGAUUUGCC
294295866-884AD-53518.1CAAAGACCAGAAAGAGUGUACACUCUUUCUGGUCUUUG
2962971080-1098AD-53519.1GGUGCAGUAAUGACUACCUAGGUAGUCAUUACUGCACC
2982991258-1276AD-53520.1CUUGUUUUCCUCGUGCUUUAAAGCACGAGGAAAACAAG
3003011616-1634AD-53521.1GGGGAUCGGGAUGGAGUCAUGACUCCAUCCCGAUCCCC
3023032230-2248AD-53522.1CUUGAGCAAGUUGGUAUCUAGAUACCAACUUGCUCAAG
304305436-454AD-53523.1CCCCAAGAUGAUGGAAGUUAACUUCCAUCAUCUUGGGG
306307436-454AD-53523.1CCCCAAGAUGAUGGAAGUUAACUUCCAUCAUCUUGGGG
308309885-903AD-53524.1CUCAUCUUCUUCAAGAUAAUUAUCUUGAAGAAGAUGAG
3103111127-1145AD-53525.1GGGGCAGUUAUGGACACUUAAGUGUCCAUAACUGCCCC
3123131315-1333AD-53526.1GAUGCCAGGCUGUGAGAUUAAUCUCACAGCCUGGCAUC
3143151870-1888AD-53527.1GAGACAGAUGCUAAUGGAUAUCCAUUAGCAUCUGUCUC
3163172286-2304AD-53528.1CCCCAGGCCAUUAUCAUAUAUAUGAUAAUGGCCUGGGG
318319489-507AD-53529.1CAGCAGUACACUACCAACAUGUUGGUAGUGUACUGCUG
320321489-507AD-53529.1CAGCAGUACACUACCAACAUGUUGGUAGUGUACUGCUG
322323915-933AD-53530.1CUGUUUCCACUUUUCAGUAUACUGAAAAGUGGAAACAG
3243251138-1156AD-53531.1GGACACUUUGAAACAACAUAUGUUGUUUCAAAGUGUCC
3263271324-1342AD-53532.1CUGUGAGAUUUACUCUGAUAUCAGAGUAAAUCUCACAG
3283291927-1945AD-53533.1CCCUGUGCGGGUUGCAGAUAUCUGCAACCCGCACAGGG
3303312312-2330AD-53534.1GUCUUCAGAGUUGUCUUUAUAAAGACAACUCUGAAGAC
332333646-664AD-53535.1CACUGCAAGCAAAUGCCCUAGGGCAUUUGCUUGCAGUG
334335922-940AD-53536.1CACUUUUCAGUAUGAUCGUACGAUCAUACUGAAAAGUG
3363371163-1181AD-53537.1GGGGCAGGUGGUACUAGAAUUCUAGUACCACCUGCCCC
3383391347-1365AD-53538.1GGAACCAUGCCUCCAUGAUAUCAUGGAGGCAUGGUUCC
3403411964-1982AD-53539.1GUCUGUGAUGAACUAAUGAUCAUUAGUUCAUCACAGAC
3423432321-2339AD-53540.1GUUGUCUUUAUAUGUGAAUAUUCACAUAUAAAGACAAC
344345671-689AD-53541.1GCAGCACAGAUGAAUCAGAUCUGAUUCAUCUGUGCUGC
346347924-942AD-53542.1CUUUUCAGUAUGAUCGUUUAAACGAUCAUACUGAAAAG
3483491164-1182AD-53543.1GGGCAGGUGGUACUAGAAAUUUCUAGUACCACCUGCCC
3503511460-1478AD-53544.1GUCCCCAAGAUUGUGGCAUAUGCCACAAUCUUGGGGAC
3523531976-1994AD-53545.1CUAAUGAGCAGACAUAACAUGUUAUGUCUGCUCAUUAG
354355786-804AD-53546.1GCCCCAGUGUGGUUAGUGUACACUAACCACACUGGGGC
356357935-953AD-53547.1GAUCGUUUCUUUGAGAAAAUUUUCUCAAAGAAACGAUC
3583591165-1183AD-53548.1GGCAGGUGGUACUAGAAAUAUUUCUAGUACCACCUGCC
3603611530-1548AD-53549.1GUGAUGUGGCCCAUGAGUUAACUCAUGGGCCACAUCAC
3623632003-2021AD-53550.1CAAGCAAUCAAUUACCCUAUAGGGUAAUUGAUUGCUUG
364365788-806AD-53551.1CCCAGUGUGGUUAGUGUGAUCACACUAACCACACUGGG
366367974-992AD-53552.1GACCACACCUAUCGAGUUUAAACUCGAUAGGUGUGGUC
3683691191-1209AD-53553.1GAACUAGUAAAUUCCAUGUACAUGGAAUUUACUAGUUC
3703711541-1559AD-53554.1CAUGAGUUUGGAGCAAUCAUGAUUGCUCCAAACUCAUG
3723732075-2093AD-53555.1CCCCAGAUGAUGAACUACUAGUAGUUCAUCAUCUGGGG
374375360-378AD-53561.1GCCCAUUCUUAUCCCGAGUACUCGGGAUAAGAAUGGGC
3763771356-1374AD-53567.1CCUCCAUGAUCCAAGGGAUAUCCCUUGGAUCAUGGAGG
3783791631-1649AD-53573.1GUCAUGCCAAAAAUGGACAUGUCCAUUUUUGGCAUGAC
3803811634-1652AD-53579.1AUGCCAAAAAUGGACAUCAUGAUGUCCAUUUUUGGCAU

ALAS1 siRNA duplexes were screened for the ability to knockdown ALAS1 expression in vitro.

Hep3B cells (ATCC, Manassas, VA) were grown to near confluence at 37° C. in an atmosphere of 5% CO2 in MEM (ATCC) supplemented with 10% FBS, before being released from the plate by trypsinization. Transfection was carried out by adding 14.8 μl of Opti-MEM plus 0.2 μl of Lipofectamine RNAiMax per well (Invitrogen, Carlsbad CA. cat #13778-150) to 5 μl of siRNA duplexes per well into a 96-well plate and incubated at room temperature for 15 minutes. 80 μl of complete growth media containing ˜2×104 Hep3B cells were then added to the siRNA mixture. Cells were incubated for either 24 or 120 hours prior to RNA purification. Single dose experiments were performed at 10 nM and 0.1 nM final duplex concentration and dose response experiments were done at 10, 1.67, 0.27, 0.046, 0.0077, 0.0013, 0.00021, 0.00004 nM final duplex concentration.

Total RNA Isolation Using DYNABEADS mRNA Isolation Kit (Invitrogen, Part #: 610-12)

Cells were harvested and lysed in 150 μl of Lysis/Binding Buffer then mixed for 5 minutes at 850 rpm using an Eppendorf Thermomixer (the mixing speed was the same throughout the process). Ten microliters of magnetic beads and 80 μl Lysis/Binding Buffer mixture were added to a round bottom plate and mixed for 1 minute. Magnetic beads were captured using magnetic stand and the supernatant was removed without disturbing the beads. After removing supernatant, the lysed cells were added to the remaining beads and mixed for 5 minutes. After removing supernatant, magnetic beads were washed 2 times with 150 μl Wash Buffer A and mixed for 1 minute. Beads were captured again and supernatant removed. Beads were then washed with 150 μl Wash Buffer B, captured and supernatant was removed. Beads were next washed with 150 μl Elution Buffer, captured and supernatant removed. Beads were allowed to dry for 2 minutes. After drying, 50 μl of Elution Buffer was added and mixed for 5 minutes at 70° C. Beads were captured on magnet for 5 minutes. 40 μl of supernatant was removed and added to another 96 well plate.

cDNA Synthesis Using ABI High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA, Cat #4368813)

A master mix of 2 μl 10×Buffer, 0.8 μl 25×dNTPs, 2 μl Random primers, 1 μl Reverse Transcriptase, 1 μl RNase inhibitor and 3.2 μl of H2O per reaction were added into 10 μl total RNA. cDNA was generated using a Bio-Rad C-1000 or S—1000 thermal cycler (Hercules, CA) through the following steps: 25° C. 10 min, 37° C. 120 min, 85° C. 5 see, 4° C. hold.

2 μl of cDNA were added to a master mix containing 0.5 μl GAPDH TaqMan Probe (Applied Biosystems Cat #4326317E), 0.5 μl ALAS1 TaqMan probe (Applied Biosystems cat #Hs00167441_ml) and 5 μl Lightcycler 480 probe master mix (Roche Cat #04887301001) per well in a 384 well plates (Roche cat #04887301001). Real time PCR was done in a Roche LC480 Real Time PCR system (Roche) using the ΔΔCt(RQ) assay. Each duplex was tested in two independent transfections with two biological replicates each, and each transfection was assayed in duplicate, unless otherwise noted in the summary tables.

To calculate relative fold change, real time data were analyzed using the ΔΔCt method and normalized to assays performed with cells transfected with 10 nM AD-1955, or mock transfected cells. IC50s were calculated using a 4 parameter fit model using XLFit and normalized to cells transfected with AD-1955 or naïve cells over the same dose range, or to its own lowest dose.

In Vitro Knockdown of Endogenous ALAS1 Expression by ALAS1 siRNA Duplexes

Table 4 illustrates the knockdown of ALAS1 in Hep3B cells by ALAS1 modified siRNA duplexes (See Table 2). Silencing is expressed as the fraction RNA message remaining relative to the negative (luciferase) control siRNA AD-1955. Data were generated as described above following transfection of 10 nM or 0.1 nM of each siRNA. qPCR was run using the ALAS1 TaqMan probe Hs00167441_ml.

TABLE 4
ALAS1 expression in Hep3B cells following transfection with ALAS1 siRNA
Duplex ID10 nM Avg0.1 nM Avg10 nM STDEV0.1 nM STDEV
AD-55078.20.70.870.0010.089
AD-55084.20.080.300.04
AD-55090.20.060.080.0020.003
AD-55096.20.610.920.1710.34
AD-55102.20.630.620.0050.069
AD-55106.20.070.080.0040.027
AD-55111.20.060.230.0130.062
AD-55073.20.210.40.0180.061
AD-55079.20.170.430.0330.089
AD-55085.20.130.210.0110.019
AD-55091.20.270.550.0330.009
AD-55097.20.310.380.0510.059
AD-55103.20.050.110.0170.006
AD-55107.20.120.240.0070.008
AD-55112.20.150.20.0360.025
AD-55074.20.160.450.0080.002
AD-55080.20.790.990.0950.304
AD-55086.20.090.220.0050.035
AD-55098.20.250.510.030.07
AD-55104.20.060.10.0170.001
AD-55108.20.470.650.030.015
AD-55113.20.380.620.0680.039
AD-55075.20.120.280.0070.051
AD-55081.20.210.510.0360.066
AD-55087.20.10.190.0170.02
AD-55093.20.240.560.0290.053
AD-55099.20.050.180.0010.038
AD-53573.30.671.070.160.153
AD-55109.20.070.230.0060.052
AD-55114.20.080.160.0040.017
AD-55076.20.050.140.0070.035
AD-55082.20.080.30.0190.016
AD-55088.20.060.120.0080.02
AD-55094.20.060.180.0050.023
AD-55100.20.450.830.020.05
AD-55105.20.020.050.0050.004
AD-55110.20.150.190.0310.016
AD-55115.20.350.580.0450.052
AD-55077.20.140.140.0060.019
AD-55083.20.560.980.240.188
AD-55089.20.620.790.0360.094
AD-55095.20.590.920.120.079
AD-55101.20.710.970.0740.097
AD-19551.001.010.030.04
AD-53511.10.841.080.0280.0515
AD-53512.10.150.650.0620.023
AD-53513.10.340.860.0550.011
AD-53514.10.120.610.0030.008
AD-53515.10.250.660.0050.004
AD-53516.11.051.020.0320.011
AD-53517.10.1450.7250.0250.0155
AD-53518.10.720.850.0450.028
AD-53519.10.180.660.0610.004
AD-53520.10.180.90.0410.001
AD-53521.10.971.070.010.003
AD-53522.10.871.10.0650.112
AD-53523.10.480.960.03050.0255
AD-53524.10.110.660.020.006
AD-53525.10.711.030.0160.01
AD-53526.10.230.850.0750.01
AD-53527.10.250.830.0150.017
AD-53528.10.440.930.0370.006
AD-53529.10.1850.730.0150.014
AD-53530.10.10.620.020.003
AD-53531.10.480.930.0190.045
AD-53532.10.060.1700.003
AD-53533.10.360.930.0250.034
AD-53534.10.10.360.0140.012
AD-53535.10.581.050.0360.071
AD-53536.10.120.450.0090.026
AD-53537.10.730.960.1010.015
AD-53538.10.741.0700.046
AD-53539.10.520.970.0570.032
AD-53540.10.10.470.0170.012
AD-53541.10.110.290.0260.015
AD-53542.10.080.230.0080.006
AD-53543.10.621.010.0270.014
AD-53544.10.81.040.0020.001
AD-53545.10.170.730.0070.007
AD-53546.10.270.930.0580.019
AD-53547.10.120.280.0080.01
AD-53548.10.10.340.0220.002
AD-53549.10.81.040.0110.026
AD-53550.10.050.540.020.003
AD-53551.10.961.160.0290.044
AD-53552.10.130.50.0020.009
AD-53553.10.921.10.0270.02
AD-53554.10.760.670.0050.004
AD-53555.10.110.530.0090.007
AD-53561.10.720.940.0140.001
AD-53567.10.160.660.0190.003
AD-53573.11.061.100.0190.037
AD-53579.10.190.760.0360.019

IC50s of Select ALAS1 siRNA Duplexes in In Vitro Screen

Table 5 illustrates the IC50s of select ALAS1 siRNA duplexes determined from the knockdown of endogenously expressed ALAS1 in the Hep3B cell line, by ALAS1 modified siRNA duplexes (see Table 2). Data were generated as described above, at 24 or 120 hours following transfection of each siRNA duplex. Silencing of ALAS1 is expressed as the fraction mRNA message remaining relative to the siRNA AD-1955, a non-targeting siRNA that was used as a negative control. Data from replicate transfection experiments were used to fit a single line to determine the IC50. Several of the duplexes (e.g., AD-53541.1, AD-53542.1, and AD-53547.1) had an IC50 as low as about 0.03 nM at 24 hours. Numerous duplexes had an IC50 of less than 0.1 nM (e.g., AD-53534.1, AD-53536.1, AD-53540.1, AD-53541.1, AD-53542.1, AD-53547.1, AD-53548.1, AD-53550.1, AD-53552.1) at 24 hours, and some of these also had an IC50 of less than 0.1 nM (e.g., AD-53534.1, AD-53540.1, AD-53541.1, AD-53542.1, AD-53547.1, AD-53552.1) at 120 hours.

TABLE 5
IC50S of select ALAS1 siRNA duplexes
normalized to AD-1955
IC50 (nM)
DUPLEX ID24 hrs120 hrs
AD-53534.10.0450.076
AD-53536.10.0490.105
AD-53540.10.0540.077
AD-53541.10.0320.062
AD-53542.10.0280.093
AD-53547.10.030.062
AD-53548.10.0440.101
AD-53550.10.0850.152
AD-53552.10.0770.063
AD-53567.10.2190.357
AD-53579.10.2170.566

The sequences of the modified duplex AD-53558 are shown in Table 6 below.

TABLE 6
Sequences of ALAS1 siRNA Duplex AD-53558.4
SEQ IDStart
SEQ IDNO:Position on
NO:(anti-transcript ofDuplexSense SequenceAntisense Sequence
(sense)sense)NM_020559.2Name(5′-3′)(5′-3′)
3833841184AD-53558cuGuGAAAuuuAcucuGAudTsdTAUcAGAGuAAAUUUcAcAGdTsdT

This duplex was formulated as a LNP11 formulation (see Table 10 above). The LNP-formulated AD-53558 siRNA was tested in in vivo in mice (N=25 animals; 5 animals per group) and rats (N=20 animals; 4 animals per group) and was confirmed to silence ALAS1 mRNA in vivo. The results are shown in FIG. 5 and FIG. 6.

FIG. 5 shows that the siRNA demonstrated a dose-response effect in mice. The expression of mouse ALAS1 (mALAS1) mRNA was reduced by about 78% when the siRNA was administered at 1 mg/kg; mouse ALAS1 mRNA was reduced by about 60% when the siRNA was administered at 0.3 mg/kg; and mouse ALAS1 mRNA was reduced by about 49% when the siRNA was administered at 0.1 mg/kg. These reductions are expressed relative to a PBS control. An AD-1955 LUC control was also employed, as shown in FIG. 5.

Similarly, FIG. 6 shows that the siRNA demonstrated a dose-response effect in rats. The expression of ALAS1 RNA was reduced by about 70% when the when the siRNA was administered at 1 mg/kg; ALAS1 mRNA was reduced by about 62% when the siRNA was administered at 0.3 mg/kg; and ALAS1 mRNA was reduced by about 34% when the siRNA was administered at 0.1 mg/kg.

The durability of silencing was also tested in mice (N=15; 3 animals per timepoint. The results are shown in FIG. 7, which shows that AD-53558 suppressed mALAS1 mRNA by about 80% for at least 9 days. Suppression of at least about 50% persisted for at least 14 days.

The effects of the AD-53558 LNP11 formulation (a mouse/rat ALAS1 siRNA described in the previous example) were investigated in a mouse model of AIP. The PBGD knockout is not viable (−/−, 0% activity). Heterozygous PBGD knockout mice (+/−, ˜50% activity) are available but do not have the full biochemical phenotype and thus do not recapitulate the human disease phenotype. Thus, a mouse model of AIP has been developed that is a compound heterozygote with T1/T2 alleles, including T1 (+/−) promoter disruption and T2 (−/−) splice-site alteration. These mice have been shown to have hepatic residual PBGD activity that is about ˜30% of the wild-type level and normal or slightly elevated baseline plasma ALA and PBG levels. The mice have been found to appear normal early in life and to become slightly slower and ataxic with age. By six months of age, the mice have been documented to develop impaired motor coordination and muscular performance and axonal degeneration on pathological examination. Investigation of the pathology of the mouse model has shown axonal degeneration, impaired motor coordination and muscular performance in older mice. Urinary and plasma ALA and PBG have been found to markedly increase with serial i.p. administration of phenobarbital (see Lindberg et al., (1996), Nature Genetics, 12:195-219 and Lindberg et al., (1999), Journal of Clinical Investigation, 103:1127-34). The mice were rescued by AAV-mediated expression of PBGD in the liver (Yasuda et al. (2010), Molecular Medicine, 1:17-22 and Unzu et al. (2011), Molecular Medicine, 2:243-50).

On day 1, the mice were administered 1 mg/kg ALAS1 siRNA (n=5) or LUC AD-1955 control (n=3) by i.v. injection. Three phenobarbitol injections were given (1 injection per day on days 2, 3, and 4) to induce hepatic ALAS1 ande the porphyrin precursors, ALA and PBG. Plasma and overnight urine specimens were collected on day 5 and metabolite levels were measured by LC-MS. Metabolite levels were measured in plasma by LC-MS and were also measured in urine. Baseline levels of metabolites were measured prior to the first treatment on day 1. The results are shown in FIGS. 8-12 and in Tables 12 and 13.

FIG. 8 and FIG. 9 show the plasma ALA levels in RM. Baseline ALA levels were low, (n=4), and phenobarbitol treatment induced significant increases in plasma ALA levels in the control LUC siRNA treated animals (n=3). Treatment with ALAS1 siRNA inhibited the induction of plasma ALA (n=5), as shown in FIG. 8. The ALAS1 siRNA was consistently effective in blocking the induction of plasma ALA in each of the individual animals studied (see FIG. 9). These results indicate that ALAS1 siRNA treatment was effective in preventing the increases in plasma ALA associated with the phenobarbital-induced acute attacks in this AlP animal model.

FIG. 10 and FIG. 11 show the plasma PBG levels in RM. Baseline PBG levels were low (n=4), and phenobarbitol treatment induced significant increases in plasma PBG levels in the control LUC siRNA treated animals (n=3) Treatment with ALAS1 siRNA inhibited the induction of plasma PBG (n=5), as shown in FIG. 10. The ALAS1 siRNA was consistently effective in blocking the induction of plasma PBG in each of the individual animals studied (see FIG. 11). These results indicate that ALAS1 siRNA treatment was effective in preventing the increases in plasma PBG associated with the phenobarbital-induced acute attacks in this AIP animal model.

Tables 12 and 13 shows urine ALA and PBG levels at baseline and after phenobarbitol treatment in LUC siRNA (n=2) control (CTR, which refers to a PBS buffer treated animal, n=1) and ALAS1 siRNA (n=5) treated animals.

TABLE 12
Urine data from individual animals showing prevention of induced acute
attack
ALAPBGALAPBG
(micro(microCreatinine(microM/mg(microM/mg
Mouse IDM/I)M/L)(mg/dl)creatinine)creatinine)siRNAPB
Ha-17-4-629.77.9Baseline
Ha-19-5-4/215.75.1Baseline
Ha-20-39-28.66.7Baseline
4/3
Ha-20-38-421.44.7Baseline
Ha-21-33-4934.92483.710.4205222.33115.03Luc+
Ha-21-36-9944.08563.530.5055186.76111.48Luc+
Ha-21-18-832.888.690.13324.726.53ALAS1;+
1 mg/kg
Ha-21-33-783.0723.280.42619.505.46ALAS1;+
1 mg/kg
Ha-21-34-559.1518.410.26322.497.00ALAS1;+
1 mg/kg
PB stands for phenobarbitol.
A “+” indicates that phenobarbitol was administered.
TABLE 13
Average Urine Data
Mean ALAMean PBG
(microM/mg creatinine)(microM/mg creatinine)
23.86.1 AIP Baseline
204.55113.26 Luc-siRNA
22.246.33 ALAS1-siRNA

Phenobarbitol treatment induced strong increases (˜25-30 fold increases) in urine ALA (˜9-fold over baseline levels) and PBG (˜19-fold over baseline levels) in the LUC siRNA treated mice, control, whereas such increases were not observed in the ALAS1 siRNA treated animals. Thus, ALAS1 siRNA blocked phenobarbitol-induced increases in urinary ALA and PBG. These results are consistent with the plasma measurements and show that ALAS1 siRNA treatment was effective in preventing increases in urinary metabolites (ALA and PBG) associated with the phenobarbital-induced acute attacks in this AIP animal model.

In further experiments (FIG. 12), it was found that phenobarbitol treatment induced large increases (˜25 fold) in ALAS1 mRNA expression in the liver of the mouse model. Administration of ALAS1 siRNA completely blocked this ALAS1 mRNA induction. These results provide further evidence that ALAS1 siRNA is effective in an animal model of AIP.

Collectively, the results provided in this Example show that ALAS1 siRNA was effective in treating acute attacks in an animal model of the acute hepatic porphyria AIP. Multiple outcome measures support this conclusion, including plasma ALA levels, plasma PBG levels, urine ALA levels, urine PBG levels, and liver ALAS1 mRNA expression levels.

The experiments described in this example investigated the in vivo efficacy of three GalNAc-conjugated siRNAs (see Table 7). These siRNAs were designed and produced with methods such as those described in Example 2.

TABLE 7
Sequences AD-57929
Position
SEQPositionof
SEQIDof senseantisense
IDNO:seq. onseq. on
NO:(anti-transcriptDuplexSense SequenceAntisense Sequencetranscript
(sense)sense)NM_020559.2Name(5′-3′)(5′-3′)NM_020559.2
385386775-795AD-Afa*gfuCfuGfuUfUfCfuUfgAfaAfa*gfuGfgaa773-795
56211cAfcUfuUfuCfaAfL96AfcAfgAfcUfusUfsg
3873882168-2188AD-AfcAfuAfgUfa*gfCfCfaGfaCfaAfuUfcUfggc2166-2188
56173aGfaAfuUfgUfcUfL96UfaCfuAfuGfusGfsg
389390775-795AD-AfsasGfuCfuGfuUfUfusUfsgAfaAfa*gfuGfg773-795
57929CfcAfcUfuUfuCfaAfLaaAfcAfgAfcUfususg
96

The mice (n+40; n+4 per experimental condition) were divided into groups that received PBS or doses of 3 mg/kg, 10 mg/kg, or 30 mg/kg of siRNA administered subcutaneously. The level of mALAS1/mGAPDH mRNA, relative to the PBS control, was determined in liver cells at 72 hours post-administration. The results are shown in FIG. 13. There was not a clear dose-response effect for the siRNAs AD-56211 and AD-56173. In contrast, the ALAS1 siRNA AD-57929 showed a dose-response effect in inhibiting mALAS1 expression. These results demonstrate that an ALAS1 GalNAc conjugate was effective in inhibiting expression of ALAS1 mRNA in vivo and showed a dose-response effect.

Additional human siRNAs were designed and produced as described in Example 2. The top 45 siRNAs were selected based on their predicted efficacy. The sequences of these 45 siRNAs are provided in Table 8.

TABLE 8
Human ALAS1 siRNA Sense and Antisense Sequences
SEQ IDPosition
SEQ IDNO:on
NO:(anti-transcriptSense SequenceAntisense Sequence
(sense)sense)NM_000688.4(5′-3′)(5′-3′)
3913921635-1657CAUGCCAAAAAUGGACAUCAUAUGAUGUCCAUUUUUGGCAUGAC
3933942352-2374UAAAUUUUAAUCUAUAGUAAAUUUACUAUAGAUUAAAAUUUAAU
3953961324-1346GGCUGUGAGAUUUACUCUGAUAUCAGAGUAAAUCUCACAGCCUG
3973981637-1659UGCCAAAAAUGGACAUCAUUUAAAUGAUGUCCAUUUUUGGCAUG
3994001363-1385AUGAUCCAAGGGAUUCGAAACGUUUCGAAUCCCUUGGAUCAUGG
401402925-947ACUUUUCAGUAUGAUCGUUUCGAAACGAUCAUACUGAAAAGUGG
403404790-812CCCAGUGUGGUUAGUGUGAAAUUUCACACUAACCACACUGGGGC
4054061531-1553UGUGAUGUGGCCCAUGAGUUUAAACUCAUGGGCCACAUCACACA
4074082189-2211AUUUUGAAGUGAUGAGUGAAAUUUCACUCAUCACUUCAAAAUGC
409410929-951UUCAGUAUGAUCGUUUCUUUGCAAAGAAACGAUCAUACUGAAAA
411412872-894GACCAGAAAGAGUGUCUCAUCGAUGAGACACUCUUUCUGGUCUU
413414706-728UUCUGCAAAGCCAGUCUUGAGCUCAAGACUGGCUUUGCAGAAGA
4154161362-1384CAUGAUCCAAGGGAUUCGAAAUUUCGAAUCCCUUGGAUCAUGGA
4174181634-1656UCAUGCCAAAAAUGGACAUCAUGAUGUCCAUUUUUGGCAUGACU
4194201325-1347GCUGUGAGAUUUACUCUGAUUAAUCAGAGUAAAUCUCACAGCCU
4214222208-2230AAGAGAGAAGUCCUAUUUCUCGAGAAAUAGGACUUCUCUCUUUC
4234242344-2366AGUUAUAUUAAAUUUUAAUCUAGAUUAAAAUUUAAUAUAACUUA
425426924-946CACUUUUCAGUAUGAUCGUUUAAACGAUCAUACUGAAAAGUGGA
427428873-895ACCAGAAAGAGUGUCUCAUCUAGAUGAGACACUCUUUCUGGUCU
429430759-781GAGGAAAGAGGUUGCUGAAACGUUUCAGCAACCUCUUUCCUCAC
431432871-893AGACCAGAAAGAGUGUCUCAUAUGAGACACUCUUUCUGGUCUUU
4334341183-1205AAUAUUUCUGGAACUAGUAAAUUUACUAGUUCCAGAAAUAUUUC
4354362229-2251AGGCUUGAGCAAGUUGGUAUCGAUACCAACUUGCUCAAGCCUGA
437438671-693UGGCAGCACAGAUGAAUCAGAUCUGAUUCAUCUGUGCUGCCAGG
4394402187-2209GCAUUUUGAAGUGAUGAGUGAUCACUCAUCACUUCAAAAUGCAG
441442913-935AAAUCUGUUUCCACUUUUCAGCUGAAAAGUGGAAACAGAUUUUG
4434441977-1999ACUAAUGAGCAGACAUAACAUAUGUUAUGUCUGCUCAUUAGUUC
4454461174-1196GGUACUAGAAAUAUUUCUGGAUCCAGAAAUAUUUCUAGUACCAC
4474481810-1832AUCCUGAAGAGCGCUGAGGGAUCCCUCAGCGCUCUUCAGGAUCC
449450892-914CUUCUUCAAGAUAACUUGCCAUGGCAAGUUAUCUUGAAGAAGAU
451452877-899GAAAGAGUGUCUCAUCUUCUUAAGAAGAUGAGACACUCUUUCUG
453454935-957AUGAUCGUUUCUUUGAGAAAAUUUUCUCAAAGAAACGAUCAUAC
4554561975-1997GAACUAAUGAGCAGACAUAACGUUAUGUCUGCUCAUUAGUUCAU
4574581478-1500CAUUUGAAACUGUCCAUUCAAUUGAAUGGACAGUUUCAAAUGCC
4594602366-2388UAGUAAAAACAUAGUCCUGGAUCCAGGACUAUGUUUUUACUAUA
461462853-875GACAUCAUGCAAAAGCAAAGAUCUUUGCUUUUGCAUGAUGUCCU
4634641966-1988GUCUGUGAUGAACUAAUGAGCGCUCAUUAGUUCAUCACAGACUU
465466928-950UUUCAGUAUGAUCGUUUCUUUAAAGAAACGAUCAUACUGAAAAG
4674681186-1208AUUUCUGGAACUAGUAAAUUCGAAUUUACUAGUUCCAGAAAUAU
4694701189-1211UCUGGAACUAGUAAAUUCCAUAUGGAAUUUACUAGUUCCAGAAA
471472973-995AAUGACCACACCUAUCGAGUUAACUCGAUAGGUGUGGUCAUUCU
473474983-1005CCUAUCGAGUUUUUAAAACUGCAGUUUUAAAAACUCGAUAGGUG
4754761185-1207UAUUUCUGGAACUAGUAAAUUAAUUUACUAGUUCCAGAAAUAUU
4774782353-2375AAAUUUUAAUCUAUAGUAAAAUUUUACUAUAGAUUAAAAUUUAA
479480875-897CAGAAAGAGUGUCUCAUCUUCGAAGAUGAGACACUCUUUCUGGU
481482360-378GCCCAUUCUUAUCCCGAGUACUCGGGAUAAGAAUGGGC
483484428-446CAAAACUGCCCCAAGAUGAUCAUCUUGGGGCAGUUUUG
485486873-891CAGAAAGAGUGUCUCAUCUAGAUGAGACACUCUUUCUG
487488874-892AGAAAGAGUGUCUCAUCUUAAGAUGAGACACUCUUUCU
489490877-895AAGAGUGUCUCAUCUUCUUAAGAAGAUGAGACACUCUU
4914921295-1313CUCUUCACCCUGGCUAAGAUCUUAGCCAGGGUGAAGAG
4934941296-1314UCUUCACCCUGGCUAAGAUAUCUUAGCCAGGGUGAAGA
4954961299-1317UCACCCUGGCUAAGAUGAUAUCAUCUUAGCCAGGGUGA
4974981347-1365GGAACCAUGCCUCCAUGAUAUCAUGGAGGCAUGGUUCC
4995001355-1373GCCUCCAUGAUCCAAGGGAUCCCUUGGAUCAUGGAGGC
5015021356-1374CCUCCAUGAUCCAAGGGAUAUCCCUUGGAUCAUGGAGG
5035041357-1375CUCCAUGAUCCAAGGGAUUAAUCCCUUGGAUCAUGGAG
5055061631-1649GUCAUGCCAAAAAUGGACAUGUCCAUUUUUGGCAUGAC
5075081634-1652AUGCCAAAAAUGGACAUCAUGAUGUCCAUUUUUGGCAU
5095101635-1653UGCCAAAAAUGGACAUCAUAUGAUGUCCAUUUUUGGCA
5115121791-1809CCCUGGAGUCUGUGCGGAUAUCCGCACAGACUCCAGGG
5135141794-1812UGGAGUCUGUGCGGAUCCUAGGAUCCGCACAGACUCCA
5155161921-1939CAUCAUCCCUGUGCGGGUUAACCCGCACAGGGAUGAUG
517518359-377UGCCCAUUCUUAUCCCGAAUUCGGGAUAAGAAUGGGCA
519520362-380CCAUUCUUAUCCCGAGUCAUGACUCGGGAUAAGAAUGG
521522363-381CAUUCUUAUCCCGAGUCCAUGGACUCGGGAUAAGAAUG
523524434-452UGCCCCAAGAUGAUGGAAUAUUCCAUCAUCUUGGGGCA
525526872-890CCAGAAAGAGUGUCUCAUAUAUGAGACACUCUUUCUGG
527528875-893GAAAGAGUGUCUCAUCUUAUAAGAUGAGACACUCUUUC
5295301112-1130CACCCACGGGUGUGUGGGAUCCCACACACCCGUGGGUG
5315321113-1131ACCCACGGGUGUGUGGGGAUCCCCACACACCCGUGGGU
5335341297-1315CUUCACCCUGGCUAAGAUAUAUCUUAGCCAGGGUGAAG
5355361300-1318CACCCUGGCUAAGAUGAUAUAUCAUCUUAGCCAGGGUG
5375381301-1319ACCCUGGCUAAGAUGAUGAUCAUCAUCUUAGCCAGGGU
5395401348-1366GAACCAUGCCUCCAUGAUAUAUCAUGGAGGCAUGGUUC
5415421481-1499GAAACUGUCCAUUCAAUGAUCAUUGAAUGGACAGUUUC
5435441786-1804UGGAGCCCUGGAGUCUGUAUACAGACUCCAGGGCUCCA
5455461795-1813GGAGUCUGUGCGGAUCCUAUAGGAUCCGCACAGACUCC
5475481919-1937CACAUCAUCCCUGUGCGGAUCCGCACAGGGAUGAUGUG
5495501922-1940AUCAUCCCUGUGCGGGUUAUAACCCGCACAGGGAUGAU
5515521923-1941UCAUCCCUGUGCGGGUUGAUCAACCCGCACAGGGAUGA

Additional 19mer human siRNAs were generated. The sequences of these siRNAs are provided in Table 9. These siRNAs can be tested for efficacy using methods described herein and/or methods known in the art.

TABLE 9
Human ALAS1 siRNA Sense and Antisense Sequences
SEQ
SEQID
IDNO:Position on
NO:(anti-transcriptSense SequenceAntisense Sequence
(sense)sense)NM_000688.4(5′-3′)(5′-3′)
553554 4-22UAUAUUAAGGCGCCGGCGAUCGCCGGCGCCUUAAUAUA
555556 5-23AUAUUAAGGCGCCGGCGAUAUCGCCGGCGCCUUAAUAU
557558 6-24UAUUAAGGCGCCGGCGAUCGAUCGCCGGCGCCUUAAUA
559560 7-25AUUAAGGCGCCGGCGAUCGCGAUCGCCGGCGCCUUAAU
561562 8-26UUAAGGCGCCGGCGAUCGCGCGAUCGCCGGCGCCUUAA
563564 9-27UAAGGCGCCGGCGAUCGCGCGCGAUCGCCGGCGCCUUA
56556610-28AAGGCGCCGGCGAUCGCGGCCGCGAUCGCCGGCGCCUU
56756811-29AGGCGCCGGCGAUCGCGGCGCCGCGAUCGCCGGCGCCU
56957012-30GGCGCCGGCGAUCGCGGCCGGCCGCGAUCGCCGGCGCC
57157213-31GCGCCGGCGAUCGCGGCCUAGGCCGCGAUCGCCGGCGC
57357414-32CGCCGGCGAUCGCGGCCUGCAGGCCGCGAUCGCCGGCG
57557681-99CUUGAGUGCCCGCCUCCUUAAGGAGGCGGGCACUCAAG
577578 82-100UUGAGUGCCCGCCUCCUUCGAAGGAGGCGGGCACUCAA
579580 83-101UGAGUGCCCGCCUCCUUCGCGAAGGAGGCGGGCACUCA
581582 84-102GAGUGCCCGCCUCCUUCGCGCGAAGGAGGCGGGCACUC
583584 85-103AGUGCCCGCCUCCUUCGCCGGCGAAGGAGGCGGGCACU
585586 86-104GUGCCCGCCUCCUUCGCCGCGGCGAAGGAGGCGGGCAC
587588 87-105UGCCCGCCUCCUUCGCCGCGCGGCGAAGGAGGCGGGCA
589590 88-106GCCCGCCUCCUUCGCCGCCGGCGGCGAAGGAGGCGGGC
591592 89-107CCCGCCUCCUUCGCCGCCGCGGCGGCGAAGGAGGCGGG
593594 90-108CCGCCUCCUUCGCCGCCGCGCGGCGGCGAAGGAGGCGG
595596 91-109CGCCUCCUUCGCCGCCGCCGGCGGCGGCGAAGGAGGCG
597598 92-110GCCUCCUUCGCCGCCGCCUAGGCGGCGGCGAAGGAGGC
599600 93-111CCUCCUUCGCCGCCGCCUCGAGGCGGCGGCGAAGGAGG
601602356-374CGCUGCCCAUUCUUAUCCCGGGAUAAGAAUGGGCAGCG
603604357-375GCUGCCCAUUCUUAUCCCGCGGGAUAAGAAUGGGCAGC
605606359-377UGCCCAUUCUUAUCCCGAGCUCGGGAUAAGAAUGGGCA
607608361-379CCCAUUCUUAUCCCGAGUCGACUCGGGAUAAGAAUGGG
609610362-380CCAUUCUUAUCCCGAGUCCGGACUCGGGAUAAGAAUGG
611612363-381CAUUCUUAUCCCGAGUCCCGGGACUCGGGAUAAGAAUG
613614364-382AUUCUUAUCCCGAGUCCCCGGGGACUCGGGAUAAGAAU
615616365-383UUCUUAUCCCGAGUCCCCCGGGGGACUCGGGAUAAGAA
617618366-384UCUUAUCCCGAGUCCCCCAUGGGGGACUCGGGAUAAGA
619620367-385CUUAUCCCGAGUCCCCCAGCUGGGGGACUCGGGAUAAG
621622368-386UUAUCCCGAGUCCCCCAGGCCUGGGGGACUCGGGAUAA
623624369-387UAUCCCGAGUCCCCCAGGCGCCUGGGGGACUCGGGAUA
625626370-388AUCCCGAGUCCCCCAGGCCGGCCUGGGGGACUCGGGAU
627628371-389UCCCGAGUCCCCCAGGCCUAGGCCUGGGGGACUCGGGA
629630372-390CCCGAGUCCCCCAGGCCUUAAGGCCUGGGGGACUCGGG
631632373-391CCGAGUCCCCCAGGCCUUUAAAGGCCUGGGGGACUCGG
633634374-392CGAGUCCCCCAGGCCUUUCGAAAGGCCUGGGGGACUCG
635636375-393GAGUCCCCCAGGCCUUUCUAGAAAGGCCUGGGGGACUC
637638376-394AGUCCCCCAGGCCUUUCUGCAGAAAGGCCUGGGGGACU
639640377-395GUCCCCCAGGCCUUUCUGCGCAGAAAGGCCUGGGGGAC
641642378-396UCCCCCAGGCCUUUCUGCAUGCAGAAAGGCCUGGGGGA
643644379-397CCCCCAGGCCUUUCUGCAGCUGCAGAAAGGCCUGGGGG
645646380-398CCCCAGGCCUUUCUGCAGAUCUGCAGAAAGGCCUGGGG
647648381-399CCCAGGCCUUUCUGCAGAAUUCUGCAGAAAGGCCUGGG
649650382-400CCAGGCCUUUCUGCAGAAAUUUCUGCAGAAAGGCCUGG
651652383-401CAGGCCUUUCUGCAGAAAGCUUUCUGCAGAAAGGCCUG
653654384-402AGGCCUUUCUGCAGAAAGCGCUUUCUGCAGAAAGGCCU
655656385-403GGCCUUUCUGCAGAAAGCAUGCUUUCUGCAGAAAGGCC
657658386-404GCCUUUCUGCAGAAAGCAGCUGCUUUCUGCAGAAAGGC
659660387-405CCUUUCUGCAGAAAGCAGGCCUGCUUUCUGCAGAAAGG
661662388-406CUUUCUGCAGAAAGCAGGCGCCUGCUUUCUGCAGAAAG
663664389-407UUUCUGCAGAAAGCAGGCAUGCCUGCUUUCUGCAGAAA
665666390-408UUCUGCAGAAAGCAGGCAAUUGCCUGCUUUCUGCAGAA
667668391-409UCUGCAGAAAGCAGGCAAAUUUGCCUGCUUUCUGCAGA
669670392-410CUGCAGAAAGCAGGCAAAUAUUUGCCUGCUUUCUGCAG
671672393-411UGCAGAAAGCAGGCAAAUCGAUUUGCCUGCUUUCUGCA
673674394-412GCAGAAAGCAGGCAAAUCUAGAUUUGCCUGCUUUCUGC
675676395-413CAGAAAGCAGGCAAAUCUCGAGAUUUGCCUGCUUUCUG
677678396-414AGAAAGCAGGCAAAUCUCUAGAGAUUUGCCUGCUUUCU
679680397-415GAAAGCAGGCAAAUCUCUGCAGAGAUUUGCCUGCUUUC
681682398-416AAAGCAGGCAAAUCUCUGUACAGAGAUUUGCCUGCUUU
683684399-417AAGCAGGCAAAUCUCUGUUAACAGAGAUUUGCCUGCUU
685686400-418AGCAGGCAAAUCUCUGUUGCAACAGAGAUUUGCCUGCU
687688401-419GCAGGCAAAUCUCUGUUGUACAACAGAGAUUUGCCUGC
689690402-420CAGGCAAAUCUCUGUUGUUAACAACAGAGAUUUGCCUG
691692403-421AGGCAAAUCUCUGUUGUUCGAACAACAGAGAUUUGCCU
693694405-423GCAAAUCUCUGUUGUUCUAUAGAACAACAGAGAUUUGC
695696406-424CAAAUCUCUGUUGUUCUAUAUAGAACAACAGAGAUUUG
697698407-425AAAUCUCUGUUGUUCUAUGCAUAGAACAACAGAGAUUU
699700408-426AAUCUCUGUUGUUCUAUGCGCAUAGAACAACAGAGAUU
701702409-427AUCUCUGUUGUUCUAUGCCGGCAUAGAACAACAGAGAU
703704410-428UCUCUGUUGUUCUAUGCCCGGGCAUAGAACAACAGAGA
705706411-429CUCUGUUGUUCUAUGCCCAUGGGCAUAGAACAACAGAG
707708412-430UCUGUUGUUCUAUGCCCAAUUGGGCAUAGAACAACAGA
709710413-431CUGUUGUUCUAUGCCCAAAUUUGGGCAUAGAACAACAG
711712414-432UGUUGUUCUAUGCCCAAAAUUUUGGGCAUAGAACAACA
713714415-433GUUGUUCUAUGCCCAAAACGUUUUGGGCAUAGAACAAC
715716416-434UUGUUCUAUGCCCAAAACUAGUUUUGGGCAUAGAACAA
717718417-435UGUUCUAUGCCCAAAACUGCAGUUUUGGGCAUAGAACA
719720418-436GUUCUAUGCCCAAAACUGCGCAGUUUUGGGCAUAGAAC
721722419-437UUCUAUGCCCAAAACUGCCGGCAGUUUUGGGCAUAGAA
723724420-438UCUAUGCCCAAAACUGCCCGGGCAGUUUUGGGCAUAGA
725726421-439CUAUGCCCAAAACUGCCCCGGGGCAGUUUUGGGCAUAG
727728422-440UAUGCCCAAAACUGCCCCAUGGGGCAGUUUUGGGCAUA
729730423-441AUGCCCAAAACUGCCCCAAUUGGGGCAGUUUUGGGCAU
731732424-442UGCCCAAAACUGCCCCAAGCUUGGGGCAGUUUUGGGCA
733734425-443GCCCAAAACUGCCCCAAGAUCUUGGGGCAGUUUUGGGC
735736426-444CCCAAAACUGCCCCAAGAUAUCUUGGGGCAGUUUUGGG
737738427-445CCAAAACUGCCCCAAGAUGCAUCUUGGGGCAGUUUUGG
739740429-447AAAACUGCCCCAAGAUGAUAUCAUCUUGGGGCAGUUUU
741742430-448AAACUGCCCCAAGAUGAUGCAUCAUCUUGGGGCAGUUU
743744431-449AACUGCCCCAAGAUGAUGGCCAUCAUCUUGGGGCAGUU
745746432-450ACUGCCCCAAGAUGAUGGAUCCAUCAUCUUGGGGCAGU
747748433-451CUGCCCCAAGAUGAUGGAAUUCCAUCAUCUUGGGGCAG
749750434-452UGCCCCAAGAUGAUGGAAGCUUCCAUCAUCUUGGGGCA
751752435-453GCCCCAAGAUGAUGGAAGUACUUCCAUCAUCUUGGGGC
753754437-455CCCAAGAUGAUGGAAGUUGCAACUUCCAUCAUCUUGGG
755756438-456CCAAGAUGAUGGAAGUUGGCCAACUUCCAUCAUCUUGG
757758439-457CAAGAUGAUGGAAGUUGGGCCCAACUUCCAUCAUCUUG
759760440-458AAGAUGAUGGAAGUUGGGGCCCCAACUUCCAUCAUCUU
761762441-459AGAUGAUGGAAGUUGGGGCGCCCCAACUUCCAUCAUCU
763764442-460GAUGAUGGAAGUUGGGGCCGGCCCCAACUUCCAUCAUC
765766443-461AUGAUGGAAGUUGGGGCCAUGGCCCCAACUUCCAUCAU
767768444-462UGAUGGAAGUUGGGGCCAAUUGGCCCCAACUUCCAUCA
769770445-463GAUGGAAGUUGGGGCCAAGCUUGGCCCCAACUUCCAUC
771772446-464AUGGAAGUUGGGGCCAAGCGCUUGGCCCCAACUUCCAU
773774447-465UGGAAGUUGGGGCCAAGCCGGCUUGGCCCCAACUUCCA
775776448-466GGAAGUUGGGGCCAAGCCAUGGCUUGGCCCCAACUUCC
777778449-467GAAGUUGGGGCCAAGCCAGCUGGCUUGGCCCCAACUUC
779780450-468AAGUUGGGGCCAAGCCAGCGCUGGCUUGGCCCCAACUU
781782451-469AGUUGGGGCCAAGCCAGCCGGCUGGCUUGGCCCCAACU
783784452-470GUUGGGGCCAAGCCAGCCCGGGCUGGCUUGGCCCCAAC
785786453-471UUGGGGCCAAGCCAGCCCCGGGGCUGGCUUGGCCCCAA
787788454-472UGGGGCCAAGCCAGCCCCUAGGGGCUGGCUUGGCCCCA
789790455-473GGGGCCAAGCCAGCCCCUCGAGGGGCUGGCUUGGCCCC
791792456-474GGGCCAAGCCAGCCCCUCGCGAGGGGCUGGCUUGGCCC
793794457-475GGCCAAGCCAGCCCCUCGGCCGAGGGGCUGGCUUGGCC
795796458-476GCCAAGCCAGCCCCUCGGGCCCGAGGGGCUGGCUUGGC
797798459-477CCAAGCCAGCCCCUCGGGCGCCCGAGGGGCUGGCUUGG
799800460-478CAAGCCAGCCCCUCGGGCAUGCCCGAGGGGCUGGCUUG
801802461-479AAGCCAGCCCCUCGGGCAUAUGCCCGAGGGGCUGGCUU
803804462-480AGCCAGCCCCUCGGGCAUUAAUGCCCGAGGGGCUGGCU
805806463-481GCCAGCCCCUCGGGCAUUGCAAUGCCCGAGGGGCUGGC
807808464-482CCAGCCCCUCGGGCAUUGUACAAUGCCCGAGGGGCUGG
809810465-483CAGCCCCUCGGGCAUUGUCGACAAUGCCCGAGGGGCUG
811812466-484AGCCCCUCGGGCAUUGUCCGGACAAUGCCCGAGGGGCU
813814467-485GCCCCUCGGGCAUUGUCCAUGGACAAUGCCCGAGGGGC
815816468-486CCCCUCGGGCAUUGUCCACGUGGACAAUGCCCGAGGGG
817818469-487CCCUCGGGCAUUGUCCACUAGUGGACAAUGCCCGAGGG
819820470-488CCUCGGGCAUUGUCCACUGCAGUGGACAAUGCCCGAGG
821822471-489CUCGGGCAUUGUCCACUGCGCAGUGGACAAUGCCCGAG
823824472-490UCGGGCAUUGUCCACUGCAUGCAGUGGACAAUGCCCGA
825826473-491CGGGCAUUGUCCACUGCAGCUGCAGUGGACAAUGCCCG
827828474-492GGGCAUUGUCCACUGCAGCGCUGCAGUGGACAAUGCCC
829830475-493GGCAUUGUCCACUGCAGCAUGCUGCAGUGGACAAUGCC
831832476-494GCAUUGUCCACUGCAGCAGCUGCUGCAGUGGACAAUGC
833834477-495CAUUGUCCACUGCAGCAGUACUGCUGCAGUGGACAAUG
835836478-496AUUGUCCACUGCAGCAGUAUACUGCUGCAGUGGACAAU
837838479-497UUGUCCACUGCAGCAGUACGUACUGCUGCAGUGGACAA
839840480-498UGUCCACUGCAGCAGUACAUGUACUGCUGCAGUGGACA
841842481-499GUCCACUGCAGCAGUACACGUGUACUGCUGCAGUGGAC
843844482-500UCCACUGCAGCAGUACACUAGUGUACUGCUGCAGUGGA
845846483-501CCACUGCAGCAGUACACUAUAGUGUACUGCUGCAGUGG
847848484-502CACUGCAGCAGUACACUACGUAGUGUACUGCUGCAGUG
849850485-503ACUGCAGCAGUACACUACCGGUAGUGUACUGCUGCAGU
851852486-504CUGCAGCAGUACACUACCAUGGUAGUGUACUGCUGCAG
853854487-505UGCAGCAGUACACUACCAAUUGGUAGUGUACUGCUGCA
855856488-506GCAGCAGUACACUACCAACGUUGGUAGUGUACUGCUGC
857858490-508AGCAGUACACUACCAACAGCUGUUGGUAGUGUACUGCU
859860491-509GCAGUACACUACCAACAGAUCUGUUGGUAGUGUACUGC
861862492-510CAGUACACUACCAACAGAUAUCUGUUGGUAGUGUACUG
863864493-511AGUACACUACCAACAGAUCGAUCUGUUGGUAGUGUACU
865866494-512GUACACUACCAACAGAUCAUGAUCUGUUGGUAGUGUAC
867868495-513UACACUACCAACAGAUCAAUUGAUCUGUUGGUAGUGUA
869870496-514ACACUACCAACAGAUCAAAUUUGAUCUGUUGGUAGUGU
871872497-515CACUACCAACAGAUCAAAGCUUUGAUCUGUUGGUAGUG
873874498-516ACUACCAACAGAUCAAAGAUCUUUGAUCUGUUGGUAGU
875876499-517CUACCAACAGAUCAAAGAAUUCUUUGAUCUGUUGGUAG
877878500-518UACCAACAGAUCAAAGAAAUUUCUUUGAUCUGUUGGUA
879880501-519ACCAACAGAUCAAAGAAACGUUUCUUUGAUCUGUUGGU
881882502-520CCAACAGAUCAAAGAAACCGGUUUCUUUGAUCUGUUGG
883884523-541UCCGGCCAGUGAGAAAGACGUCUUUCUCACUGGCCGGA
885886524-542CCGGCCAGUGAGAAAGACAUGUCUUUCUCACUGGCCGG
887888525-543CGGCCAGUGAGAAAGACAAUUGUCUUUCUCACUGGCCG
889890526-544GGCCAGUGAGAAAGACAAAUUUGUCUUUCUCACUGGCC
891892527-545GCCAGUGAGAAAGACAAAAUUUUGUCUUUCUCACUGGC
893894528-546CCAGUGAGAAAGACAAAACGUUUUGUCUUUCUCACUGG
895896529-547CAGUGAGAAAGACAAAACUAGUUUUGUCUUUCUCACUG
897898530-548AGUGAGAAAGACAAAACUGCAGUUUUGUCUUUCUCACU
899900531-549GUGAGAAAGACAAAACUGCGCAGUUUUGUCUUUCUCAC
901902570-588CUCCUGAUGGAUCCCAGCAUGCUGGGAUCCAUCAGGAG
903904571-589UCCUGAUGGAUCCCAGCAGCUGCUGGGAUCCAUCAGGA
905906572-590CCUGAUGGAUCCCAGCAGAUCUGCUGGGAUCCAUCAGG
907908573-591CUGAUGGAUCCCAGCAGAGCUCUGCUGGGAUCCAUCAG
909910574-592UGAUGGAUCCCAGCAGAGUACUCUGCUGGGAUCCAUCA
911912575-593GAUGGAUCCCAGCAGAGUCGACUCUGCUGGGAUCCAUC
913914576-594AUGGAUCCCAGCAGAGUCCGGACUCUGCUGGGAUCCAU
915916577-595UGGAUCCCAGCAGAGUCCAUGGACUCUGCUGGGAUCCA
917918578-596GGAUCCCAGCAGAGUCCAGCUGGACUCUGCUGGGAUCC
919920579-597GAUCCCAGCAGAGUCCAGAUCUGGACUCUGCUGGGAUC
921922580-598AUCCCAGCAGAGUCCAGAUAUCUGGACUCUGCUGGGAU
923924581-599UCCCAGCAGAGUCCAGAUGCAUCUGGACUCUGCUGGGA
925926582-600CCCAGCAGAGUCCAGAUGGCCAUCUGGACUCUGCUGGG
927928583-601CCAGCAGAGUCCAGAUGGCGCCAUCUGGACUCUGCUGG
929930584-602CAGCAGAGUCCAGAUGGCAUGCCAUCUGGACUCUGCUG
931932585-603AGCAGAGUCCAGAUGGCACGUGCCAUCUGGACUCUGCU
933934586-604GCAGAGUCCAGAUGGCACAUGUGCCAUCUGGACUCUGC
935936587-605CAGAGUCCAGAUGGCACACGUGUGCCAUCUGGACUCUG
937938588-606AGAGUCCAGAUGGCACACAUGUGUGCCAUCUGGACUCU
939940589-607GAGUCCAGAUGGCACACAGCUGUGUGCCAUCUGGACUC
941942590-608AGUCCAGAUGGCACACAGCGCUGUGUGCCAUCUGGACU
943944591-609GUCCAGAUGGCACACAGCUAGCUGUGUGCCAUCUGGAC
945946592-610UCCAGAUGGCACACAGCUUAAGCUGUGUGCCAUCUGGA
947948593-611CCAGAUGGCACACAGCUUCGAAGCUGUGUGCCAUCUGG
949950594-612CAGAUGGCACACAGCUUCCGGAAGCUGUGUGCCAUCUG
951952595-613AGAUGGCACACAGCUUCCGCGGAAGCUGUGUGCCAUCU
953954596-614GAUGGCACACAGCUUCCGUACGGAAGCUGUGUGCCAUC
955956597-615AUGGCACACAGCUUCCGUCGACGGAAGCUGUGUGCCAU
957958598-616UGGCACACAGCUUCCGUCUAGACGGAAGCUGUGUGCCA
959960599-617GGCACACAGCUUCCGUCUGCAGACGGAAGCUGUGUGCC
961962600-618GCACACAGCUUCCGUCUGGCCAGACGGAAGCUGUGUGC
963964601-619CACACAGCUUCCGUCUGGAUCCAGACGGAAGCUGUGUG
965966602-620ACACAGCUUCCGUCUGGACGUCCAGACGGAAGCUGUGU
967968603-621CACAGCUUCCGUCUGGACAUGUCCAGACGGAAGCUGUG
969970604-622ACAGCUUCCGUCUGGACACGUGUCCAGACGGAAGCUGU
971972605-623CAGCUUCCGUCUGGACACCGGUGUCCAGACGGAAGCUG
973974606-624AGCUUCCGUCUGGACACCCGGGUGUCCAGACGGAAGCU
975976607-625GCUUCCGUCUGGACACCCCGGGGUGUCCAGACGGAAGC
977978608-626CUUCCGUCUGGACACCCCUAGGGGUGUCCAGACGGAAG
979980609-627UUCCGUCUGGACACCCCUUAAGGGGUGUCCAGACGGAA
981982610-628UCCGUCUGGACACCCCUUGCAAGGGGUGUCCAGACGGA
983984611-629CCGUCUGGACACCCCUUGCGCAAGGGGUGUCCAGACGG
985986612-630CGUCUGGACACCCCUUGCCGGCAAGGGGUGUCCAGACG
987988613-631GUCUGGACACCCCUUGCCUAGGCAAGGGGUGUCCAGAC
989990614-632UCUGGACACCCCUUGCCUGCAGGCAAGGGGUGUCCAGA
991992615-633CUGGACACCCCUUGCCUGCGCAGGCAAGGGGUGUCCAG
993994616-634UGGACACCCCUUGCCUGCCGGCAGGCAAGGGGUGUCCA
995996617-635GGACACCCCUUGCCUGCCAUGGCAGGCAAGGGGUGUCC
997998618-636GACACCCCUUGCCUGCCACGUGGCAGGCAAGGGGUGUC
9991000619-637ACACCCCUUGCCUGCCACAUGUGGCAGGCAAGGGGUGU
10011002620-638CACCCCUUGCCUGCCACAAUUGUGGCAGGCAAGGGGUG
10031004621-639ACCCCUUGCCUGCCACAAGCUUGUGGCAGGCAAGGGGU
10051006622-640CCCCUUGCCUGCCACAAGCGCUUGUGGCAGGCAAGGGG
10071008623-641CCCUUGCCUGCCACAAGCCGGCUUGUGGCAGGCAAGGG
10091010624-642CCUUGCCUGCCACAAGCCAUGGCUUGUGGCAGGCAAGG
10111012625-643CUUGCCUGCCACAAGCCAGCUGGCUUGUGGCAGGCAAG
10131014626-644UUGCCUGCCACAAGCCAGGCCUGGCUUGUGGCAGGCAA
10151016627-645UGCCUGCCACAAGCCAGGGCCCUGGCUUGUGGCAGGCA
10171018628-646GCCUGCCACAAGCCAGGGCGCCCUGGCUUGUGGCAGGC
10191020629-647CCUGCCACAAGCCAGGGCAUGCCCUGGCUUGUGGCAGG
10211022630-648CUGCCACAAGCCAGGGCACGUGCCCUGGCUUGUGGCAG
10231024631-649UGCCACAAGCCAGGGCACUAGUGCCCUGGCUUGUGGCA
10251026632-650GCCACAAGCCAGGGCACUGCAGUGCCCUGGCUUGUGGC
10271028633-651CCACAAGCCAGGGCACUGCGCAGUGCCCUGGCUUGUGG
10291030634-652CACAAGCCAGGGCACUGCAUGCAGUGCCCUGGCUUGUG
10311032635-653ACAAGCCAGGGCACUGCAAUUGCAGUGCCCUGGCUUGU
10331034636-654CAAGCCAGGGCACUGCAAGCUUGCAGUGCCCUGGCUUG
10351036637-655AAGCCAGGGCACUGCAAGCGCUUGCAGUGCCCUGGCUU
10371038638-656AGCCAGGGCACUGCAAGCAUGCUUGCAGUGCCCUGGCU
10391040639-657GCCAGGGCACUGCAAGCAAUUGCUUGCAGUGCCCUGGC
10411042640-658CCAGGGCACUGCAAGCAAAUUUGCUUGCAGUGCCCUGG
10431044641-659CAGGGCACUGCAAGCAAAUAUUUGCUUGCAGUGCCCUG
10451046642-660AGGGCACUGCAAGCAAAUGCAUUUGCUUGCAGUGCCCU
10471048643-661GGGCACUGCAAGCAAAUGCGCAUUUGCUUGCAGUGCCC
10491050644-662GGCACUGCAAGCAAAUGCCGGCAUUUGCUUGCAGUGCC
10511052645-663GCACUGCAAGCAAAUGCCCGGGCAUUUGCUUGCAGUGC
10531054647-665ACUGCAAGCAAAUGCCCUUAAGGGCAUUUGCUUGCAGU
10551056648-666CUGCAAGCAAAUGCCCUUUAAAGGGCAUUUGCUUGCAG
10571058649-667UGCAAGCAAAUGCCCUUUCGAAAGGGCAUUUGCUUGCA
10591060650-668GCAAGCAAAUGCCCUUUCCGGAAAGGGCAUUUGCUUGC
10611062651-669CAAGCAAAUGCCCUUUCCUAGGAAAGGGCAUUUGCUUG
10631064652-670AAGCAAAUGCCCUUUCCUGCAGGAAAGGGCAUUUGCUU
10651066653-671AGCAAAUGCCCUUUCCUGGCCAGGAAAGGGCAUUUGCU
10671068654-672GCAAAUGCCCUUUCCUGGCGCCAGGAAAGGGCAUUUGC
10691070655-673CAAAUGCCCUUUCCUGGCAUGCCAGGAAAGGGCAUUUG
10711072656-674AAAUGCCCUUUCCUGGCAGCUGCCAGGAAAGGGCAUUU
10731074657-675AAUGCCCUUUCCUGGCAGCGCUGCCAGGAAAGGGCAUU
10751076658-676AUGCCCUUUCCUGGCAGCAUGCUGCCAGGAAAGGGCAU
10771078659-677UGCCCUUUCCUGGCAGCACGUGCUGCCAGGAAAGGGCA
10791080660-678GCCCUUUCCUGGCAGCACAUGUGCUGCCAGGAAAGGGC
10811082661-679CCCUUUCCUGGCAGCACAGCUGUGCUGCCAGGAAAGGG
10831084662-680CCUUUCCUGGCAGCACAGAUCUGUGCUGCCAGGAAAGG
10851086663-681CUUUCCUGGCAGCACAGAUAUCUGUGCUGCCAGGAAAG
10871088664-682UUUCCUGGCAGCACAGAUGCAUCUGUGCUGCCAGGAAA
10891090665-683UUCCUGGCAGCACAGAUGAUCAUCUGUGCUGCCAGGAA
10911092666-684UCCUGGCAGCACAGAUGAAUUCAUCUGUGCUGCCAGGA
10931094667-685CCUGGCAGCACAGAUGAAUAUUCAUCUGUGCUGCCAGG
10951096668-686CUGGCAGCACAGAUGAAUCGAUUCAUCUGUGCUGCCAG
10971098670-688GGCAGCACAGAUGAAUCAGCUGAUUCAUCUGUGCUGCC
10991100672-690CAGCACAGAUGAAUCAGAGCUCUGAUUCAUCUGUGCUG
11011102692-710GGCAGCAGUGUCUUCUGCAUGCAGAAGACACUGCUGCC
11031104693-711GCAGCAGUGUCUUCUGCAAUUGCAGAAGACACUGCUGC
11051106694-712CAGCAGUGUCUUCUGCAAAUUUGCAGAAGACACUGCUG
11071108695-713AGCAGUGUCUUCUGCAAAGCUUUGCAGAAGACACUGCU
11091110696-714GCAGUGUCUUCUGCAAAGCGCUUUGCAGAAGACACUGC
11111112697-715CAGUGUCUUCUGCAAAGCCGGCUUUGCAGAAGACACUG
11131114698-716AGUGUCUUCUGCAAAGCCAUGGCUUUGCAGAAGACACU
11151116699-717GUGUCUUCUGCAAAGCCAGCUGGCUUUGCAGAAGACAC
11171118700-718UGUCUUCUGCAAAGCCAGUACUGGCUUUGCAGAAGACA
11191120701-719GUCUUCUGCAAAGCCAGUCGACUGGCUUUGCAGAAGAC
11211122702-720UCUUCUGCAAAGCCAGUCUAGACUGGCUUUGCAGAAGA
11231124703-721CUUCUGCAAAGCCAGUCUUAAGACUGGCUUUGCAGAAG
11251126704-722UUCUGCAAAGCCAGUCUUGCAAGACUGGCUUUGCAGAA
11271128705-723UCUGCAAAGCCAGUCUUGAUCAAGACUGGCUUUGCAGA
11291130706-724CUGCAAAGCCAGUCUUGAGCUCAAGACUGGCUUUGCAG
11311132707-725UGCAAAGCCAGUCUUGAGCGCUCAAGACUGGCUUUGCA
11331134708-726GCAAAGCCAGUCUUGAGCUAGCUCAAGACUGGCUUUGC
11351136709-727CAAAGCCAGUCUUGAGCUUAAGCUCAAGACUGGCUUUG
11371138710-728AAAGCCAGUCUUGAGCUUCGAAGCUCAAGACUGGCUUU
11391140711-729AAGCCAGUCUUGAGCUUCAUGAAGCUCAAGACUGGCUU
11411142712-730AGCCAGUCUUGAGCUUCAGCUGAAGCUCAAGACUGGCU
11431144713-731GCCAGUCUUGAGCUUCAGGCCUGAAGCUCAAGACUGGC
11451146714-732CCAGUCUUGAGCUUCAGGAUCCUGAAGCUCAAGACUGG
11471148715-733CAGUCUUGAGCUUCAGGAGCUCCUGAAGCUCAAGACUG
11491150716-734AGUCUUGAGCUUCAGGAGGCCUCCUGAAGCUCAAGACU
11511152717-735GUCUUGAGCUUCAGGAGGAUCCUCCUGAAGCUCAAGAC
11531154718-736UCUUGAGCUUCAGGAGGAUAUCCUCCUGAAGCUCAAGA
11551156719-737CUUGAGCUUCAGGAGGAUGCAUCCUCCUGAAGCUCAAG
11571158720-738UUGAGCUUCAGGAGGAUGUACAUCCUCCUGAAGCUCAA
11591160721-739UGAGCUUCAGGAGGAUGUGCACAUCCUCCUGAAGCUCA
11611162722-740GAGCUUCAGGAGGAUGUGCGCACAUCCUCCUGAAGCUC
11631164723-741AGCUUCAGGAGGAUGUGCAUGCACAUCCUCCUGAAGCU
11651166724-742GCUUCAGGAGGAUGUGCAGCUGCACAUCCUCCUGAAGC
11671168725-743CUUCAGGAGGAUGUGCAGGCCUGCACAUCCUCCUGAAG
11691170726-744UUCAGGAGGAUGUGCAGGAUCCUGCACAUCCUCCUGAA
11711172727-745UCAGGAGGAUGUGCAGGAAUUCCUGCACAUCCUCCUGA
11731174728-746CAGGAGGAUGUGCAGGAAAUUUCCUGCACAUCCUCCUG
11751176729-747AGGAGGAUGUGCAGGAAAUAUUUCCUGCACAUCCUCCU
11771178730-748GGAGGAUGUGCAGGAAAUGCAUUUCCUGCACAUCCUCC
11791180731-749GAGGAUGUGCAGGAAAUGAUCAUUUCCUGCACAUCCUC
11811182732-750AGGAUGUGCAGGAAAUGAAUUCAUUUCCUGCACAUCCU
11831184733-751GGAUGUGCAGGAAAUGAAUAUUCAUUUCCUGCACAUCC
11851186734-752GAUGUGCAGGAAAUGAAUGCAUUCAUUUCCUGCACAUC
11871188735-753AUGUGCAGGAAAUGAAUGCGCAUUCAUUUCCUGCACAU
11891190755-773GUGAGGAAAGAGGUUGCUGCAGCAACCUCUUUCCUCAC
11911192756-774UGAGGAAAGAGGUUGCUGAUCAGCAACCUCUUUCCUCA
11931194757-775GAGGAAAGAGGUUGCUGAAUUCAGCAACCUCUUUCCUC
11951196758-776AGGAAAGAGGUUGCUGAAAUUUCAGCAACCUCUUUCCU
11971198759-777GGAAAGAGGUUGCUGAAACGUUUCAGCAACCUCUUUCC
11991200760-778GAAAGAGGUUGCUGAAACCGGUUUCAGCAACCUCUUUC
12011202761-779AAAGAGGUUGCUGAAACCUAGGUUUCAGCAACCUCUUU
12031204762-780AAGAGGUUGCUGAAACCUCGAGGUUUCAGCAACCUCUU
12051206763-781AGAGGUUGCUGAAACCUCAUGAGGUUUCAGCAACCUCU
12071208764-782GAGGUUGCUGAAACCUCAGCUGAGGUUUCAGCAACCUC
12091210765-783AGGUUGCUGAAACCUCAGCGCUGAGGUUUCAGCAACCU
12111212766-784GGUUGCUGAAACCUCAGCAUGCUGAGGUUUCAGCAACC
12131214787-805CCCCAGUGUGGUUAGUGUGCACACUAACCACACUGGGG
12151216791-809AGUGUGGUUAGUGUGAAAAUUUUCACACUAACCACACU
12171218792-810GUGUGGUUAGUGUGAAAACGUUUUCACACUAACCACAC
12191220812-830GAUGGAGGGGAUCCCAGUGCACUGGGAUCCCCUCCAUC
12211222813-831AUGGAGGGGAUCCCAGUGGCCACUGGGAUCCCCUCCAU
12231224833-851CUGCUGAAGAACUUCCAGGCCUGGAAGUUCUUCAGCAG
12251226834-852UGCUGAAGAACUUCCAGGAUCCUGGAAGUUCUUCAGCA
12271228835-853GCUGAAGAACUUCCAGGACGUCCUGGAAGUUCUUCAGC
12291230836-854CUGAAGAACUUCCAGGACAUGUCCUGGAAGUUCUUCAG
12311232837-855UGAAGAACUUCCAGGACAUAUGUCCUGGAAGUUCUUCA
12331234838-856GAAGAACUUCCAGGACAUCGAUGUCCUGGAAGUUCUUC
12351236839-857AAGAACUUCCAGGACAUCAUGAUGUCCUGGAAGUUCUU
12371238840-858AGAACUUCCAGGACAUCAUAUGAUGUCCUGGAAGUUCU
12391240841-859GAACUUCCAGGACAUCAUGCAUGAUGUCCUGGAAGUUC
12411242842-860AACUUCCAGGACAUCAUGCGCAUGAUGUCCUGGAAGUU
12431244843-861ACUUCCAGGACAUCAUGCAUGCAUGAUGUCCUGGAAGU
12451246844-862CUUCCAGGACAUCAUGCAAUUGCAUGAUGUCCUGGAAG
12471248845-863UUCCAGGACAUCAUGCAAAUUUGCAUGAUGUCCUGGAA
12491250846-864UCCAGGACAUCAUGCAAAAUUUUGCAUGAUGUCCUGGA
12511252847-865CCAGGACAUCAUGCAAAAGCUUUUGCAUGAUGUCCUGG
12531254848-866CAGGACAUCAUGCAAAAGCGCUUUUGCAUGAUGUCCUG
12551256849-867AGGACAUCAUGCAAAAGCAUGCUUUUGCAUGAUGUCCU
12571258850-868GGACAUCAUGCAAAAGCAAUUGCUUUUGCAUGAUGUCC
12591260851-869GACAUCAUGCAAAAGCAAAUUUGCUUUUGCAUGAUGUC
12611262852-870ACAUCAUGCAAAAGCAAAGCUUUGCUUUUGCAUGAUGU
12631264854-872AUCAUGCAAAAGCAAAGACGUCUUUGCUUUUGCAUGAU
12651266855-873UCAUGCAAAAGCAAAGACCGGUCUUUGCUUUUGCAUGA
12671268856-874CAUGCAAAAGCAAAGACCAUGGUCUUUGCUUUUGCAUG
12691270857-875AUGCAAAAGCAAAGACCAGCUGGUCUUUGCUUUUGCAU
12711272858-876UGCAAAAGCAAAGACCAGAUCUGGUCUUUGCUUUUGCA
12731274859-877GCAAAAGCAAAGACCAGAAUUCUGGUCUUUGCUUUUGC
12751276860-878CAAAAGCAAAGACCAGAAAUUUCUGGUCUUUGCUUUUG
12771278861-879AAAAGCAAAGACCAGAAAGCUUUCUGGUCUUUGCUUUU
12791280862-880AAAGCAAAGACCAGAAAGAUCUUUCUGGUCUUUGCUUU
12811282863-881AAGCAAAGACCAGAAAGAGCUCUUUCUGGUCUUUGCUU
12831284864-882AGCAAAGACCAGAAAGAGUACUCUUUCUGGUCUUUGCU
12851286865-883GCAAAGACCAGAAAGAGUGCACUCUUUCUGGUCUUUGC
12871288867-885AAAGACCAGAAAGAGUGUCGACACUCUUUCUGGUCUUU
12891290868-886AAGACCAGAAAGAGUGUCUAGACACUCUUUCUGGUCUU
12911292869-887AGACCAGAAAGAGUGUCUCGAGACACUCUUUCUGGUCU
12931294870-888GACCAGAAAGAGUGUCUCAUGAGACACUCUUUCUGGUC
12951296871-889ACCAGAAAGAGUGUCUCAUAUGAGACACUCUUUCUGGU
12971298872-890CCAGAAAGAGUGUCUCAUCGAUGAGACACUCUUUCUGG
12991300875-893GAAAGAGUGUCUCAUCUUCGAAGAUGAGACACUCUUUC
13011302878-896AGAGUGUCUCAUCUUCUUCGAAGAAGAUGAGACACUCU
13031304879-897GAGUGUCUCAUCUUCUUCAUGAAGAAGAUGAGACACUC
13051306880-898AGUGUCUCAUCUUCUUCAAUUGAAGAAGAUGAGACACU
13071308881-899GUGUCUCAUCUUCUUCAAGCUUGAAGAAGAUGAGACAC
13091310882-900UGUCUCAUCUUCUUCAAGAUCUUGAAGAAGAUGAGACA
13111312883-901GUCUCAUCUUCUUCAAGAUAUCUUGAAGAAGAUGAGAC
13131314884-902UCUCAUCUUCUUCAAGAUAUAUCUUGAAGAAGAUGAGA
13151316886-904UCAUCUUCUUCAAGAUAACGUUAUCUUGAAGAAGAUGA
13171318887-905CAUCUUCUUCAAGAUAACUAGUUAUCUUGAAGAAGAUG
13191320888-906AUCUUCUUCAAGAUAACUUAAGUUAUCUUGAAGAAGAU
13211322889-907UCUUCUUCAAGAUAACUUGCAAGUUAUCUUGAAGAAGA
13231324890-908CUUCUUCAAGAUAACUUGCGCAAGUUAUCUUGAAGAAG
13251326891-909UUCUUCAAGAUAACUUGCCGGCAAGUUAUCUUGAAGAA
13271328892-910UCUUCAAGAUAACUUGCCAUGGCAAGUUAUCUUGAAGA
13291330893-911CUUCAAGAUAACUUGCCAAUUGGCAAGUUAUCUUGAAG
13311332894-912UUCAAGAUAACUUGCCAAAUUUGGCAAGUUAUCUUGAA
13331334895-913UCAAGAUAACUUGCCAAAAUUUUGGCAAGUUAUCUUGA
13351336896-914CAAGAUAACUUGCCAAAAUAUUUUGGCAAGUUAUCUUG
13371338897-915AAGAUAACUUGCCAAAAUCGAUUUUGGCAAGUUAUCUU
13391340898-916AGAUAACUUGCCAAAAUCUAGAUUUUGGCAAGUUAUCU
13411342899-917GAUAACUUGCCAAAAUCUGCAGAUUUUGGCAAGUUAUC
13431344900-918AUAACUUGCCAAAAUCUGUACAGAUUUUGGCAAGUUAU
13451346901-919UAACUUGCCAAAAUCUGUUAACAGAUUUUGGCAAGUUA
13471348902-920AACUUGCCAAAAUCUGUUUAAACAGAUUUUGGCAAGUU
13491350903-921ACUUGCCAAAAUCUGUUUCGAAACAGAUUUUGGCAAGU
13511352904-922CUUGCCAAAAUCUGUUUCCGGAAACAGAUUUUGGCAAG
13531354905-923UUGCCAAAAUCUGUUUCCAUGGAAACAGAUUUUGGCAA
13551356906-924UGCCAAAAUCUGUUUCCACGUGGAAACAGAUUUUGGCA
13571358907-925GCCAAAAUCUGUUUCCACUAGUGGAAACAGAUUUUGGC
13591360908-926CCAAAAUCUGUUUCCACUUAAGUGGAAACAGAUUUUGG
13611362909-927CAAAAUCUGUUUCCACUUUAAAGUGGAAACAGAUUUUG
13631364910-928AAAAUCUGUUUCCACUUUUAAAAGUGGAAACAGAUUUU
13651366911-929AAAUCUGUUUCCACUUUUCGAAAAGUGGAAACAGAUUU
13671368912-930AAUCUGUUUCCACUUUUCAUGAAAAGUGGAAACAGAUU
13691370913-931AUCUGUUUCCACUUUUCAGCUGAAAAGUGGAAACAGAU
13711372916-934UGUUUCCACUUUUCAGUAUAUACUGAAAAGUGGAAACA
13731374917-935GUUUCCACUUUUCAGUAUGCAUACUGAAAAGUGGAAAC
13751376918-936UUUCCACUUUUCAGUAUGAUCAUACUGAAAAGUGGAAA
13771378919-937UUCCACUUUUCAGUAUGAUAUCAUACUGAAAAGUGGAA
13791380920-938UCCACUUUUCAGUAUGAUCGAUCAUACUGAAAAGUGGA
13811382921-939CCACUUUUCAGUAUGAUCGCGAUCAUACUGAAAAGUGG
13831384925-943UUUUCAGUAUGAUCGUUUCGAAACGAUCAUACUGAAAA
13851386929-947CAGUAUGAUCGUUUCUUUGCAAAGAAACGAUCAUACUG
13871388930-948AGUAUGAUCGUUUCUUUGAUCAAAGAAACGAUCAUACU
13891390931-949GUAUGAUCGUUUCUUUGAGCUCAAAGAAACGAUCAUAC
13911392933-951AUGAUCGUUUCUUUGAGAAUUCUCAAAGAAACGAUCAU
13931394934-952UGAUCGUUUCUUUGAGAAAUUUCUCAAAGAAACGAUCA
13951396936-954AUCGUUUCUUUGAGAAAAAUUUUUCUCAAAGAAACGAU
13971398937-955UCGUUUCUUUGAGAAAAAAUUUUUUCUCAAAGAAACGA
13991400938-956CGUUUCUUUGAGAAAAAAAUUUUUUUCUCAAAGAAACG
14011402939-957GUUUCUUUGAGAAAAAAAUAUUUUUUUCUCAAAGAAAC
14031404940-958UUUCUUUGAGAAAAAAAUUAAUUUUUUUCUCAAAGAAA
14051406941-959UUCUUUGAGAAAAAAAUUGCAAUUUUUUUCUCAAAGAA
14071408942-960UCUUUGAGAAAAAAAUUGAUCAAUUUUUUUCUCAAAGA
14091410943-961CUUUGAGAAAAAAAUUGAUAUCAAUUUUUUUCUCAAAG
14111412944-962UUUGAGAAAAAAAUUGAUGCAUCAAUUUUUUUCUCAAA
14131414945-963UUGAGAAAAAAAUUGAUGAUCAUCAAUUUUUUUCUCAA
14151416946-964UGAGAAAAAAAUUGAUGAGCUCAUCAAUUUUUUUCUCA
14171418947-965GAGAAAAAAAUUGAUGAGAUCUCAUCAAUUUUUUUCUC
14191420948-966AGAAAAAAAUUGAUGAGAAUUCUCAUCAAUUUUUUUCU
14211422949-967GAAAAAAAUUGAUGAGAAAUUUCUCAUCAAUUUUUUUC
14231424950-968AAAAAAAUUGAUGAGAAAAUUUUCUCAUCAAUUUUUUU
14251426951-969AAAAAAUUGAUGAGAAAAAUUUUUCUCAUCAAUUUUUU
14271428952-970AAAAAUUGAUGAGAAAAAGCUUUUUCUCAUCAAUUUUU
14291430953-971AAAAUUGAUGAGAAAAAGAUCUUUUUCUCAUCAAUUUU
14311432954-972AAAUUGAUGAGAAAAAGAAUUCUUUUUCUCAUCAAUUU
14331434955-973AAUUGAUGAGAAAAAGAAUAUUCUUUUUCUCAUCAAUU
14351436956-974AUUGAUGAGAAAAAGAAUGCAUUCUUUUUCUCAUCAAU
14371438957-975UUGAUGAGAAAAAGAAUGAUCAUUCUUUUUCUCAUCAA
14391440958-976UGAUGAGAAAAAGAAUGACGUCAUUCUUUUUCUCAUCA
14411442959-977GAUGAGAAAAAGAAUGACCGGUCAUUCUUUUUCUCAUC
14431444960-978AUGAGAAAAAGAAUGACCAUGGUCAUUCUUUUUCUCAU
14451446961-979UGAGAAAAAGAAUGACCACGUGGUCAUUCUUUUUCUCA
14471448962-980GAGAAAAAGAAUGACCACAUGUGGUCAUUCUUUUUCUC
14491450963-981AGAAAAAGAAUGACCACACGUGUGGUCAUUCUUUUUCU
14511452964-982GAAAAAGAAUGACCACACCGGUGUGGUCAUUCUUUUUC
14531454965-983AAAAAGAAUGACCACACCUAGGUGUGGUCAUUCUUUUU
14551456966-984AAAAGAAUGACCACACCUAUAGGUGUGGUCAUUCUUUU
14571458967-985AAAGAAUGACCACACCUAUAUAGGUGUGGUCAUUCUUU
14591460968-986AAGAAUGACCACACCUAUCGAUAGGUGUGGUCAUUCUU
14611462969-987AGAAUGACCACACCUAUCGCGAUAGGUGUGGUCAUUCU
14631464970-988GAAUGACCACACCUAUCGAUCGAUAGGUGUGGUCAUUC
14651466971-989AAUGACCACACCUAUCGAGCUCGAUAGGUGUGGUCAUU
14671468972-990AUGACCACACCUAUCGAGUACUCGAUAGGUGUGGUCAU
14691470976-994CCACACCUAUCGAGUUUUUAAAAACUCGAUAGGUGUGG
14711472977-995CACACCUAUCGAGUUUUUAUAAAAACUCGAUAGGUGUG
14731474978-996ACACCUAUCGAGUUUUUAAUUAAAAACUCGAUAGGUGU
14751476979-997CACCUAUCGAGUUUUUAAAUUUAAAAACUCGAUAGGUG
14771478980-998ACCUAUCGAGUUUUUAAAAUUUUAAAAACUCGAUAGGU
14791480981-999CCUAUCGAGUUUUUAAAACGUUUUAAAAACUCGAUAGG
14811482 982-1000CUAUCGAGUUUUUAAAACUAGUUUUAAAAACUCGAUAG
14831484 983-1001UAUCGAGUUUUUAAAACUGCAGUUUUAAAAACUCGAUA
14851486 984-1002AUCGAGUUUUUAAAACUGUACAGUUUUAAAAACUCGAU
14871488 985-1003UCGAGUUUUUAAAACUGUGCACAGUUUUAAAAACUCGA
14891490 986-1004CGAGUUUUUAAAACUGUGAUCACAGUUUUAAAAACUCG
14911492 987-1005GAGUUUUUAAAACUGUGAAUUCACAGUUUUAAAAACUC
14931494 988-1006AGUUUUUAAAACUGUGAACGUUCACAGUUUUAAAAACU
14951496 989-1007GUUUUUAAAACUGUGAACCGGUUCACAGUUUUAAAAAC
14971498 990-1008UUUUUAAAACUGUGAACCGCGGUUCACAGUUUUAAAAA
14991500 991-1009UUUUAAAACUGUGAACCGGCCGGUUCACAGUUUUAAAA
15011502 992-1010UUUAAAACUGUGAACCGGCGCCGGUUCACAGUUUUAAA
15031504 993-1011UUAAAACUGUGAACCGGCGCGCCGGUUCACAGUUUUAA
15051506 994-1012UAAAACUGUGAACCGGCGAUCGCCGGUUCACAGUUUUA
15071508 995-1013AAAACUGUGAACCGGCGAGCUCGCCGGUUCACAGUUUU
15091510 996-1014AAACUGUGAACCGGCGAGCGCUCGCCGGUUCACAGUUU
15111512 997-1015AACUGUGAACCGGCGAGCAUGCUCGCCGGUUCACAGUU
15131514 998-1016ACUGUGAACCGGCGAGCACGUGCUCGCCGGUUCACAGU
15151516 999-1017CUGUGAACCGGCGAGCACAUGUGCUCGCCGGUUCACAG
151715181000-1018UGUGAACCGGCGAGCACACGUGUGCUCGCCGGUUCACA
151915201001-1019GUGAACCGGCGAGCACACAUGUGUGCUCGCCGGUUCAC
152115221002-1020UGAACCGGCGAGCACACAUAUGUGUGCUCGCCGGUUCA
152315241003-1021GAACCGGCGAGCACACAUCGAUGUGUGCUCGCCGGUUC
152515261004-1022AACCGGCGAGCACACAUCUAGAUGUGUGCUCGCCGGUU
152715281005-1023ACCGGCGAGCACACAUCUUAAGAUGUGUGCUCGCCGGU
152915301006-1024CCGGCGAGCACACAUCUUCGAAGAUGUGUGCUCGCCGG
153115321007-1025CGGCGAGCACACAUCUUCCGGAAGAUGUGUGCUCGCCG
153315341008-1026GGCGAGCACACAUCUUCCCGGGAAGAUGUGUGCUCGCC
153515361028-1046AUGGCAGAUGACUAUUCAGCUGAAUAGUCAUCUGCCAU
153715381030-1048GGCAGAUGACUAUUCAGACGUCUGAAUAGUCAUCUGCC
153915401031-1049GCAGAUGACUAUUCAGACUAGUCUGAAUAGUCAUCUGC
154115421032-1050CAGAUGACUAUUCAGACUCGAGUCUGAAUAGUCAUCUG
154315441033-1051AGAUGACUAUUCAGACUCCGGAGUCUGAAUAGUCAUCU
154515461034-1052GAUGACUAUUCAGACUCCCGGGAGUCUGAAUAGUCAUC
154715481035-1053AUGACUAUUCAGACUCCCUAGGGAGUCUGAAUAGUCAU
154915501036-1054UGACUAUUCAGACUCCCUCGAGGGAGUCUGAAUAGUCA
155115521037-1055GACUAUUCAGACUCCCUCAUGAGGGAGUCUGAAUAGUC
155315541038-1056ACUAUUCAGACUCCCUCAUAUGAGGGAGUCUGAAUAGU
155515561039-1057CUAUUCAGACUCCCUCAUCGAUGAGGGAGUCUGAAUAG
155715581040-1058UAUUCAGACUCCCUCAUCAUGAUGAGGGAGUCUGAAUA
155915601041-1059AUUCAGACUCCCUCAUCACGUGAUGAGGGAGUCUGAAU
156115621042-1060UUCAGACUCCCUCAUCACCGGUGAUGAGGGAGUCUGAA
156315641043-1061UCAGACUCCCUCAUCACCAUGGUGAUGAGGGAGUCUGA
156515661044-1062CAGACUCCCUCAUCACCAAUUGGUGAUGAGGGAGUCUG
156715681045-1063AGACUCCCUCAUCACCAAAUUUGGUGAUGAGGGAGUCU
156915701046-1064GACUCCCUCAUCACCAAAAUUUUGGUGAUGAGGGAGUC
157115721047-1065ACUCCCUCAUCACCAAAAAUUUUUGGUGAUGAGGGAGU
157315741048-1066CUCCCUCAUCACCAAAAAGCUUUUUGGUGAUGAGGGAG
157515761049-1067UCCCUCAUCACCAAAAAGCGCUUUUUGGUGAUGAGGGA
157715781050-1068CCCUCAUCACCAAAAAGCAUGCUUUUUGGUGAUGAGGG
157915801070-1088GUGUCAGUCUGGUGCAGUAUACUGCACCAGACUGACAC
158115821071-1089UGUCAGUCUGGUGCAGUAAUUACUGCACCAGACUGACA
158315841072-1090GUCAGUCUGGUGCAGUAAUAUUACUGCACCAGACUGAC
158515861073-1091UCAGUCUGGUGCAGUAAUGCAUUACUGCACCAGACUGA
158715881074-1092CAGUCUGGUGCAGUAAUGAUCAUUACUGCACCAGACUG
158915901075-1093AGUCUGGUGCAGUAAUGACGUCAUUACUGCACCAGACU
159115921078-1096CUGGUGCAGUAAUGACUACGUAGUCAUUACUGCACCAG
159315941079-1097UGGUGCAGUAAUGACUACCGGUAGUCAUUACUGCACCA
159515961081-1099GUGCAGUAAUGACUACCUAUAGGUAGUCAUUACUGCAC
159715981082-1100UGCAGUAAUGACUACCUAGCUAGGUAGUCAUUACUGCA
159916001083-1101GCAGUAAUGACUACCUAGGCCUAGGUAGUCAUUACUGC
160116021084-1102CAGUAAUGACUACCUAGGAUCCUAGGUAGUCAUUACUG
160316041085-1103AGUAAUGACUACCUAGGAAUUCCUAGGUAGUCAUUACU
160516061086-1104GUAAUGACUACCUAGGAAUAUUCCUAGGUAGUCAUUAC
160716081087-1105UAAUGACUACCUAGGAAUGCAUUCCUAGGUAGUCAUUA
160916101088-1106AAUGACUACCUAGGAAUGAUCAUUCCUAGGUAGUCAUU
161116121089-1107AUGACUACCUAGGAAUGAGCUCAUUCCUAGGUAGUCAU
161316141090-1108UGACUACCUAGGAAUGAGUACUCAUUCCUAGGUAGUCA
161516161091-1109GACUACCUAGGAAUGAGUCGACUCAUUCCUAGGUAGUC
161716181092-1110ACUACCUAGGAAUGAGUCGCGACUCAUUCCUAGGUAGU
161916201093-1111CUACCUAGGAAUGAGUCGCGCGACUCAUUCCUAGGUAG
162116221094-1112UACCUAGGAAUGAGUCGCCGGCGACUCAUUCCUAGGUA
162316241095-1113ACCUAGGAAUGAGUCGCCAUGGCGACUCAUUCCUAGGU
162516261096-1114CCUAGGAAUGAGUCGCCACGUGGCGACUCAUUCCUAGG
162716281097-1115CUAGGAAUGAGUCGCCACCGGUGGCGACUCAUUCCUAG
162916301098-1116UAGGAAUGAGUCGCCACCCGGGUGGCGACUCAUUCCUA
163116321099-1117AGGAAUGAGUCGCCACCCAUGGGUGGCGACUCAUUCCU
163316341100-1118GGAAUGAGUCGCCACCCACGUGGGUGGCGACUCAUUCC
163516361101-1119GAAUGAGUCGCCACCCACGCGUGGGUGGCGACUCAUUC
163716381102-1120AAUGAGUCGCCACCCACGGCCGUGGGUGGCGACUCAUU
163916401103-1121AUGAGUCGCCACCCACGGGCCCGUGGGUGGCGACUCAU
164116421104-1122UGAGUCGCCACCCACGGGUACCCGUGGGUGGCGACUCA
164316441105-1123GAGUCGCCACCCACGGGUGCACCCGUGGGUGGCGACUC
164516461106-1124AGUCGCCACCCACGGGUGUACACCCGUGGGUGGCGACU
164716481107-1125GUCGCCACCCACGGGUGUGCACACCCGUGGGUGGCGAC
164916501108-1126UCGCCACCCACGGGUGUGUACACACCCGUGGGUGGCGA
165116521109-1127CGCCACCCACGGGUGUGUGCACACACCCGUGGGUGGCG
165316541110-1128GCCACCCACGGGUGUGUGGCCACACACCCGUGGGUGGC
165516561111-1129CCACCCACGGGUGUGUGGGCCCACACACCCGUGGGUGG
165716581112-1130CACCCACGGGUGUGUGGGGCCCCACACACCCGUGGGUG
165916601113-1131ACCCACGGGUGUGUGGGGCGCCCCACACACCCGUGGGU
166116621114-1132CCCACGGGUGUGUGGGGCAUGCCCCACACACCCGUGGG
166316641115-1133CCACGGGUGUGUGGGGCAGCUGCCCCACACACCCGUGG
166516661116-1134CACGGGUGUGUGGGGCAGUACUGCCCCACACACCCGUG
166716681117-1135ACGGGUGUGUGGGGCAGUUAACUGCCCCACACACCCGU
166916701118-1136CGGGUGUGUGGGGCAGUUAUAACUGCCCCACACACCCG
167116721119-1137GGGUGUGUGGGGCAGUUAUAUAACUGCCCCACACACCC
167316741120-1138GGUGUGUGGGGCAGUUAUGCAUAACUGCCCCACACACC
167516761121-1139GUGUGUGGGGCAGUUAUGGCCAUAACUGCCCCACACAC
167716781122-1140UGUGUGGGGCAGUUAUGGAUCCAUAACUGCCCCACACA
167916801123-1141GUGUGGGGCAGUUAUGGACGUCCAUAACUGCCCCACAC
168116821125-1143GUGGGGCAGUUAUGGACACGUGUCCAUAACUGCCCCAC
168316841126-1144UGGGGCAGUUAUGGACACUAGUGUCCAUAACUGCCCCA
168516861128-1146GGGCAGUUAUGGACACUUUAAAGUGUCCAUAACUGCCC
168716881129-1147GGCAGUUAUGGACACUUUGCAAAGUGUCCAUAACUGCC
168916901130-1148GCAGUUAUGGACACUUUGAUCAAAGUGUCCAUAACUGC
169116921131-1149CAGUUAUGGACACUUUGAAUUCAAAGUGUCCAUAACUG
169316941132-1150AGUUAUGGACACUUUGAAAUUUCAAAGUGUCCAUAACU
169516961133-1151GUUAUGGACACUUUGAAACGUUUCAAAGUGUCCAUAAC
169716981134-1152UUAUGGACACUUUGAAACAUGUUUCAAAGUGUCCAUAA
169917001135-1153UAUGGACACUUUGAAACAAUUGUUUCAAAGUGUCCAUA
170117021136-1154AUGGACACUUUGAAACAACGUUGUUUCAAAGUGUCCAU
170317041139-1157GACACUUUGAAACAACAUGCAUGUUGUUUCAAAGUGUC
170517061140-1158ACACUUUGAAACAACAUGGCCAUGUUGUUUCAAAGUGU
170717081141-1159CACUUUGAAACAACAUGGUACCAUGUUGUUUCAAAGUG
170917101142-1160ACUUUGAAACAACAUGGUGCACCAUGUUGUUUCAAAGU
171117121143-1161CUUUGAAACAACAUGGUGCGCACCAUGUUGUUUCAAAG
171317141144-1162UUUGAAACAACAUGGUGCUAGCACCAUGUUGUUUCAAA
171517161145-1163UUGAAACAACAUGGUGCUGCAGCACCAUGUUGUUUCAA
171717181146-1164UGAAACAACAUGGUGCUGGCCAGCACCAUGUUGUUUCA
171917201147-1165GAAACAACAUGGUGCUGGGCCCAGCACCAUGUUGUUUC
172117221148-1166AAACAACAUGGUGCUGGGGCCCCAGCACCAUGUUGUUU
172317241149-1167AACAACAUGGUGCUGGGGCGCCCCAGCACCAUGUUGUU
172517261150-1168ACAACAUGGUGCUGGGGCAUGCCCCAGCACCAUGUUGU
172717281151-1169CAACAUGGUGCUGGGGCAGCUGCCCCAGCACCAUGUUG
172917301152-1170AACAUGGUGCUGGGGCAGGCCUGCCCCAGCACCAUGUU
173117321153-1171ACAUGGUGCUGGGGCAGGUACCUGCCCCAGCACCAUGU
173317341154-1172CAUGGUGCUGGGGCAGGUGCACCUGCCCCAGCACCAUG
173517361155-1173AUGGUGCUGGGGCAGGUGGCCACCUGCCCCAGCACCAU
173717381156-1174UGGUGCUGGGGCAGGUGGUACCACCUGCCCCAGCACCA
173917401157-1175GGUGCUGGGGCAGGUGGUAUACCACCUGCCCCAGCACC
174117421158-1176GUGCUGGGGCAGGUGGUACGUACCACCUGCCCCAGCAC
174317441159-1177UGCUGGGGCAGGUGGUACUAGUACCACCUGCCCCAGCA
174517461160-1178GCUGGGGCAGGUGGUACUAUAGUACCACCUGCCCCAGC
174717481161-1179CUGGGGCAGGUGGUACUAGCUAGUACCACCUGCCCCAG
174917501162-1180UGGGGCAGGUGGUACUAGAUCUAGUACCACCUGCCCCA
175117521166-1184GCAGGUGGUACUAGAAAUAUAUUUCUAGUACCACCUGC
175317541167-1185CAGGUGGUACUAGAAAUAUAUAUUUCUAGUACCACCUG
175517561168-1186AGGUGGUACUAGAAAUAUUAAUAUUUCUAGUACCACCU
175717581169-1187GGUGGUACUAGAAAUAUUUAAAUAUUUCUAGUACCACC
175917601170-1188GUGGUACUAGAAAUAUUUCGAAAUAUUUCUAGUACCAC
176117621171-1189UGGUACUAGAAAUAUUUCUAGAAAUAUUUCUAGUACCA
176317641172-1190GGUACUAGAAAUAUUUCUGCAGAAAUAUUUCUAGUACC
176517661173-1191GUACUAGAAAUAUUUCUGGCCAGAAAUAUUUCUAGUAC
176717681174-1192UACUAGAAAUAUUUCUGGAUCCAGAAAUAUUUCUAGUA
176917701175-1193ACUAGAAAUAUUUCUGGAAUUCCAGAAAUAUUUCUAGU
177117721176-1194CUAGAAAUAUUUCUGGAACGUUCCAGAAAUAUUUCUAG
177317741177-1195UAGAAAUAUUUCUGGAACUAGUUCCAGAAAUAUUUCUA
177517761178-1196AGAAAUAUUUCUGGAACUAUAGUUCCAGAAAUAUUUCU
177717781179-1197GAAAUAUUUCUGGAACUAGCUAGUUCCAGAAAUAUUUC
177917801180-1198AAAUAUUUCUGGAACUAGUACUAGUUCCAGAAAUAUUU
178117821181-1199AAUAUUUCUGGAACUAGUAUACUAGUUCCAGAAAUAUU
178317841183-1201UAUUUCUGGAACUAGUAAAUUUACUAGUUCCAGAAAUA
178517861186-1204UUCUGGAACUAGUAAAUUCGAAUUUACUAGUUCCAGAA
178717881187-1205UCUGGAACUAGUAAAUUCCGGAAUUUACUAGUUCCAGA
178917901189-1207UGGAACUAGUAAAUUCCAUAUGGAAUUUACUAGUUCCA
179117921190-1208GGAACUAGUAAAUUCCAUGCAUGGAAUUUACUAGUUCC
179317941192-1210AACUAGUAAAUUCCAUGUGCACAUGGAAUUUACUAGUU
179517961193-1211ACUAGUAAAUUCCAUGUGGCCACAUGGAAUUUACUAGU
179717981194-1212CUAGUAAAUUCCAUGUGGAUCCACAUGGAAUUUACUAG
179918001195-1213UAGUAAAUUCCAUGUGGACGUCCACAUGGAAUUUACUA
180118021196-1214AGUAAAUUCCAUGUGGACUAGUCCACAUGGAAUUUACU
180318041197-1215GUAAAUUCCAUGUGGACUUAAGUCCACAUGGAAUUUAC
180518061198-1216UAAAUUCCAUGUGGACUUAUAAGUCCACAUGGAAUUUA
180718081199-1217AAAUUCCAUGUGGACUUAGCUAAGUCCACAUGGAAUUU
180918101200-1218AAUUCCAUGUGGACUUAGAUCUAAGUCCACAUGGAAUU
181118121201-1219AUUCCAUGUGGACUUAGAGCUCUAAGUCCACAUGGAAU
181318141202-1220UUCCAUGUGGACUUAGAGCGCUCUAAGUCCACAUGGAA
181518161222-1240GGAGCUGGCAGACCUCCAUAUGGAGGUCUGCCAGCUCC
181718181223-1241GAGCUGGCAGACCUCCAUGCAUGGAGGUCUGCCAGCUC
181918201224-1242AGCUGGCAGACCUCCAUGGCCAUGGAGGUCUGCCAGCU
182118221225-1243GCUGGCAGACCUCCAUGGGCCCAUGGAGGUCUGCCAGC
182318241226-1244CUGGCAGACCUCCAUGGGAUCCCAUGGAGGUCUGCCAG
182518261227-1245UGGCAGACCUCCAUGGGAAUUCCCAUGGAGGUCUGCCA
182718281228-1246GGCAGACCUCCAUGGGAAAUUUCCCAUGGAGGUCUGCC
182918301229-1247GCAGACCUCCAUGGGAAAGCUUUCCCAUGGAGGUCUGC
183118321230-1248CAGACCUCCAUGGGAAAGAUCUUUCCCAUGGAGGUCUG
183318341231-1249AGACCUCCAUGGGAAAGAUAUCUUUCCCAUGGAGGUCU
183518361232-1250GACCUCCAUGGGAAAGAUGCAUCUUUCCCAUGGAGGUC
183718381233-1251ACCUCCAUGGGAAAGAUGCGCAUCUUUCCCAUGGAGGU
183918401254-1272CACUCUUGUUUUCCUCGUGCACGAGGAAAACAAGAGUG
184118421255-1273ACUCUUGUUUUCCUCGUGCGCACGAGGAAAACAAGAGU
184318441256-1274CUCUUGUUUUCCUCGUGCUAGCACGAGGAAAACAAGAG
184518461257-1275UCUUGUUUUCCUCGUGCUUAAGCACGAGGAAAACAAGA
184718481259-1277UUGUUUUCCUCGUGCUUUGCAAAGCACGAGGAAAACAA
184918501260-1278UGUUUUCCUCGUGCUUUGUACAAAGCACGAGGAAAACA
185118521261-1279GUUUUCCUCGUGCUUUGUGCACAAAGCACGAGGAAAAC
185318541262-1280UUUUCCUCGUGCUUUGUGGCCACAAAGCACGAGGAAAA
185518561263-1281UUUCCUCGUGCUUUGUGGCGCCACAAAGCACGAGGAAA
185718581264-1282UUCCUCGUGCUUUGUGGCCGGCCACAAAGCACGAGGAA
185918601265-1283UCCUCGUGCUUUGUGGCCAUGGCCACAAAGCACGAGGA
186118621266-1284CCUCGUGCUUUGUGGCCAAUUGGCCACAAAGCACGAGG
186318641267-1285CUCGUGCUUUGUGGCCAAUAUUGGCCACAAAGCACGAG
186518661268-1286UCGUGCUUUGUGGCCAAUGCAUUGGCCACAAAGCACGA
186718681269-1287CGUGCUUUGUGGCCAAUGAUCAUUGGCCACAAAGCACG
186918701270-1288GUGCUUUGUGGCCAAUGACGUCAUUGGCCACAAAGCAC
187118721271-1289UGCUUUGUGGCCAAUGACUAGUCAUUGGCCACAAAGCA
187318741272-1290GCUUUGUGGCCAAUGACUCGAGUCAUUGGCCACAAAGC
187518761273-1291CUUUGUGGCCAAUGACUCAUGAGUCAUUGGCCACAAAG
187718781274-1292UUUGUGGCCAAUGACUCAAUUGAGUCAUUGGCCACAAA
187918801275-1293UUGUGGCCAAUGACUCAACGUUGAGUCAUUGGCCACAA
188118821276-1294UGUGGCCAAUGACUCAACCGGUUGAGUCAUUGGCCACA
188318841277-1295GUGGCCAAUGACUCAACCCGGGUUGAGUCAUUGGCCAC
188518861278-1296UGGCCAAUGACUCAACCCUAGGGUUGAGUCAUUGGCCA
188718881279-1297GGCCAAUGACUCAACCCUCGAGGGUUGAGUCAUUGGCC
188918901280-1298GCCAAUGACUCAACCCUCUAGAGGGUUGAGUCAUUGGC
189118921281-1299CCAAUGACUCAACCCUCUUAAGAGGGUUGAGUCAUUGG
189318941282-1300CAAUGACUCAACCCUCUUCGAAGAGGGUUGAGUCAUUG
189518961283-1301AAUGACUCAACCCUCUUCAUGAAGAGGGUUGAGUCAUU
189718981284-1302AUGACUCAACCCUCUUCACGUGAAGAGGGUUGAGUCAU
189919001285-1303UGACUCAACCCUCUUCACCGGUGAAGAGGGUUGAGUCA
190119021286-1304GACUCAACCCUCUUCACCCGGGUGAAGAGGGUUGAGUC
190319041287-1305ACUCAACCCUCUUCACCCUAGGGUGAAGAGGGUUGAGU
190519061288-1306CUCAACCCUCUUCACCCUGCAGGGUGAAGAGGGUUGAG
190719081289-1307UCAACCCUCUUCACCCUGGCCAGGGUGAAGAGGGUUGA
190919101290-1308CAACCCUCUUCACCCUGGCGCCAGGGUGAAGAGGGUUG
191119121291-1309AACCCUCUUCACCCUGGCUAGCCAGGGUGAAGAGGGUU
191319141292-1310ACCCUCUUCACCCUGGCUAUAGCCAGGGUGAAGAGGGU
191519161293-1311CCCUCUUCACCCUGGCUAAUUAGCCAGGGUGAAGAGGG
191719181294-1312CCUCUUCACCCUGGCUAAGCUUAGCCAGGGUGAAGAGG
191919201297-1315CUUCACCCUGGCUAAGAUGCAUCUUAGCCAGGGUGAAG
192119221298-1316UUCACCCUGGCUAAGAUGAUCAUCUUAGCCAGGGUGAA
192319241300-1318CACCCUGGCUAAGAUGAUGCAUCAUCUUAGCCAGGGUG
192519261301-1319ACCCUGGCUAAGAUGAUGCGCAUCAUCUUAGCCAGGGU
192719281302-1320CCCUGGCUAAGAUGAUGCCGGCAUCAUCUUAGCCAGGG
192919301303-1321CCUGGCUAAGAUGAUGCCAUGGCAUCAUCUUAGCCAGG
193119321304-1322CUGGCUAAGAUGAUGCCAGCUGGCAUCAUCUUAGCCAG
193319341305-1323UGGCUAAGAUGAUGCCAGGCCUGGCAUCAUCUUAGCCA
193519361306-1324GGCUAAGAUGAUGCCAGGCGCCUGGCAUCAUCUUAGCC
193719381307-1325GCUAAGAUGAUGCCAGGCUAGCCUGGCAUCAUCUUAGC
193919401308-1326CUAAGAUGAUGCCAGGCUGCAGCCUGGCAUCAUCUUAG
194119421309-1327UAAGAUGAUGCCAGGCUGUACAGCCUGGCAUCAUCUUA
194319441310-1328AAGAUGAUGCCAGGCUGUGCACAGCCUGGCAUCAUCUU
194519461311-1329AGAUGAUGCCAGGCUGUGAUCACAGCCUGGCAUCAUCU
194719481312-1330GAUGAUGCCAGGCUGUGAGCUCACAGCCUGGCAUCAUC
194919501313-1331AUGAUGCCAGGCUGUGAGAUCUCACAGCCUGGCAUCAU
195119521314-1332UGAUGCCAGGCUGUGAGAUAUCUCACAGCCUGGCAUCA
195319541316-1334AUGCCAGGCUGUGAGAUUUAAAUCUCACAGCCUGGCAU
195519561317-1335UGCCAGGCUGUGAGAUUUAUAAAUCUCACAGCCUGGCA
195719581318-1336GCCAGGCUGUGAGAUUUACGUAAAUCUCACAGCCUGGC
195919601319-1337CCAGGCUGUGAGAUUUACUAGUAAAUCUCACAGCCUGG
196119621320-1338CAGGCUGUGAGAUUUACUCGAGUAAAUCUCACAGCCUG
196319641321-1339AGGCUGUGAGAUUUACUCUAGAGUAAAUCUCACAGCCU
196519661322-1340GGCUGUGAGAUUUACUCUGCAGAGUAAAUCUCACAGCC
196719681323-1341GCUGUGAGAUUUACUCUGAUCAGAGUAAAUCUCACAGC
196919701326-1344GUGAGAUUUACUCUGAUUCGAAUCAGAGUAAAUCUCAC
197119721327-1345UGAGAUUUACUCUGAUUCUAGAAUCAGAGUAAAUCUCA
197319741328-1346GAGAUUUACUCUGAUUCUGCAGAAUCAGAGUAAAUCUC
197519761329-1347AGAUUUACUCUGAUUCUGGCCAGAAUCAGAGUAAAUCU
197719781330-1348GAUUUACUCUGAUUCUGGGCCCAGAAUCAGAGUAAAUC
197919801331-1349AUUUACUCUGAUUCUGGGAUCCCAGAAUCAGAGUAAAU
198119821332-1350UUUACUCUGAUUCUGGGAAUUCCCAGAAUCAGAGUAAA
198319841333-1351UUACUCUGAUUCUGGGAACGUUCCCAGAAUCAGAGUAA
198519861334-1352UACUCUGAUUCUGGGAACCGGUUCCCAGAAUCAGAGUA
198719881335-1353ACUCUGAUUCUGGGAACCAUGGUUCCCAGAAUCAGAGU
198919901336-1354CUCUGAUUCUGGGAACCAUAUGGUUCCCAGAAUCAGAG
199119921337-1355UCUGAUUCUGGGAACCAUGCAUGGUUCCCAGAAUCAGA
199319941338-1356CUGAUUCUGGGAACCAUGCGCAUGGUUCCCAGAAUCAG
199519961339-1357UGAUUCUGGGAACCAUGCCGGCAUGGUUCCCAGAAUCA
199719981340-1358GAUUCUGGGAACCAUGCCUAGGCAUGGUUCCCAGAAUC
199920001341-1359AUUCUGGGAACCAUGCCUCGAGGCAUGGUUCCCAGAAU
200120021342-1360UUCUGGGAACCAUGCCUCCGGAGGCAUGGUUCCCAGAA
200320041343-1361UCUGGGAACCAUGCCUCCAUGGAGGCAUGGUUCCCAGA
200520061344-1362CUGGGAACCAUGCCUCCAUAUGGAGGCAUGGUUCCCAG
200720081345-1363UGGGAACCAUGCCUCCAUGCAUGGAGGCAUGGUUCCCA
200920101346-1364GGGAACCAUGCCUCCAUGAUCAUGGAGGCAUGGUUCCC
201120121348-1366GAACCAUGCCUCCAUGAUCGAUCAUGGAGGCAUGGUUC
201320141349-1367AACCAUGCCUCCAUGAUCCGGAUCAUGGAGGCAUGGUU
201520161350-1368ACCAUGCCUCCAUGAUCCAUGGAUCAUGGAGGCAUGGU
201720181351-1369CCAUGCCUCCAUGAUCCAAUUGGAUCAUGGAGGCAUGG
201920201352-1370CAUGCCUCCAUGAUCCAAGCUUGGAUCAUGGAGGCAUG
202120221353-1371AUGCCUCCAUGAUCCAAGGCCUUGGAUCAUGGAGGCAU
202320241354-1372UGCCUCCAUGAUCCAAGGGCCCUUGGAUCAUGGAGGCA
202520261358-1376UCCAUGAUCCAAGGGAUUCGAAUCCCUUGGAUCAUGGA
202720281359-1377CCAUGAUCCAAGGGAUUCGCGAAUCCCUUGGAUCAUGG
202920301360-1378CAUGAUCCAAGGGAUUCGAUCGAAUCCCUUGGAUCAUG
203120321361-1379AUGAUCCAAGGGAUUCGAAUUCGAAUCCCUUGGAUCAU
203320341362-1380UGAUCCAAGGGAUUCGAAAUUUCGAAUCCCUUGGAUCA
203520361363-1381GAUCCAAGGGAUUCGAAACGUUUCGAAUCCCUUGGAUC
203720381365-1383UCCAAGGGAUUCGAAACAGCUGUUUCGAAUCCCUUGGA
203920401366-1384CCAAGGGAUUCGAAACAGCGCUGUUUCGAAUCCCUUGG
204120421367-1385CAAGGGAUUCGAAACAGCCGGCUGUUUCGAAUCCCUUG
204320441368-1386AAGGGAUUCGAAACAGCCGCGGCUGUUUCGAAUCCCUU
204520461369-1387AGGGAUUCGAAACAGCCGAUCGGCUGUUUCGAAUCCCU
204720481370-1388GGGAUUCGAAACAGCCGAGCUCGGCUGUUUCGAAUCCC
204920501371-1389GGAUUCGAAACAGCCGAGUACUCGGCUGUUUCGAAUCC
205120521372-1390GAUUCGAAACAGCCGAGUGCACUCGGCUGUUUCGAAUC
205320541373-1391AUUCGAAACAGCCGAGUGCGCACUCGGCUGUUUCGAAU
205520561374-1392UUCGAAACAGCCGAGUGCCGGCACUCGGCUGUUUCGAA
205720581375-1393UCGAAACAGCCGAGUGCCAUGGCACUCGGCUGUUUCGA
205920601376-1394CGAAACAGCCGAGUGCCAAUUGGCACUCGGCUGUUUCG
206120621377-1395GAAACAGCCGAGUGCCAAAUUUGGCACUCGGCUGUUUC
206320641378-1396AAACAGCCGAGUGCCAAAGCUUUGGCACUCGGCUGUUU
206520661379-1397AACAGCCGAGUGCCAAAGUACUUUGGCACUCGGCUGUU
206720681380-1398ACAGCCGAGUGCCAAAGUAUACUUUGGCACUCGGCUGU
206920701381-1399CAGCCGAGUGCCAAAGUACGUACUUUGGCACUCGGCUG
207120721383-1401GCCGAGUGCCAAAGUACAUAUGUACUUUGGCACUCGGC
207320741384-1402CCGAGUGCCAAAGUACAUCGAUGUACUUUGGCACUCGG
207520761385-1403CGAGUGCCAAAGUACAUCUAGAUGUACUUUGGCACUCG
207720781386-1404GAGUGCCAAAGUACAUCUUAAGAUGUACUUUGGCACUC
207920801387-1405AGUGCCAAAGUACAUCUUCGAAGAUGUACUUUGGCACU
208120821388-1406GUGCCAAAGUACAUCUUCCGGAAGAUGUACUUUGGCAC
208320841389-1407UGCCAAAGUACAUCUUCCGCGGAAGAUGUACUUUGGCA
208520861390-1408GCCAAAGUACAUCUUCCGCGCGGAAGAUGUACUUUGGC
208720881391-1409CCAAAGUACAUCUUCCGCCGGCGGAAGAUGUACUUUGG
208920901392-1410CAAAGUACAUCUUCCGCCAUGGCGGAAGAUGUACUUUG
209120921393-1411AAAGUACAUCUUCCGCCACGUGGCGGAAGAUGUACUUU
209320941394-1412AAGUACAUCUUCCGCCACAUGUGGCGGAAGAUGUACUU
209520961395-1413AGUACAUCUUCCGCCACAAUUGUGGCGGAAGAUGUACU
209720981396-1414GUACAUCUUCCGCCACAAUAUUGUGGCGGAAGAUGUAC
209921001397-1415UACAUCUUCCGCCACAAUGCAUUGUGGCGGAAGAUGUA
210121021398-1416ACAUCUUCCGCCACAAUGAUCAUUGUGGCGGAAGAUGU
210321041399-1417CAUCUUCCGCCACAAUGAUAUCAUUGUGGCGGAAGAUG
210521061400-1418AUCUUCCGCCACAAUGAUGCAUCAUUGUGGCGGAAGAU
210721081401-1419UCUUCCGCCACAAUGAUGUACAUCAUUGUGGCGGAAGA
210921101402-1420CUUCCGCCACAAUGAUGUCGACAUCAUUGUGGCGGAAG
211121121403-1421UUCCGCCACAAUGAUGUCAUGACAUCAUUGUGGCGGAA
211321141404-1422UCCGCCACAAUGAUGUCAGCUGACAUCAUUGUGGCGGA
211521161405-1423CCGCCACAAUGAUGUCAGCGCUGACAUCAUUGUGGCGG
211721181406-1424CGCCACAAUGAUGUCAGCCGGCUGACAUCAUUGUGGCG
211921201407-1425GCCACAAUGAUGUCAGCCAUGGCUGACAUCAUUGUGGC
212121221427-1445CUCAGAGAACUGCUGCAAAUUUGCAGCAGUUCUCUGAG
212321241428-1446UCAGAGAACUGCUGCAAAGCUUUGCAGCAGUUCUCUGA
212521261429-1447CAGAGAACUGCUGCAAAGAUCUUUGCAGCAGUUCUCUG
212721281430-1448AGAGAACUGCUGCAAAGAUAUCUUUGCAGCAGUUCUCU
212921301431-1449GAGAACUGCUGCAAAGAUCGAUCUUUGCAGCAGUUCUC
213121321432-1450AGAACUGCUGCAAAGAUCUAGAUCUUUGCAGCAGUUCU
213321341433-1451GAACUGCUGCAAAGAUCUGCAGAUCUUUGCAGCAGUUC
213521361434-1452AACUGCUGCAAAGAUCUGAUCAGAUCUUUGCAGCAGUU
213721381435-1453ACUGCUGCAAAGAUCUGACGUCAGAUCUUUGCAGCAGU
213921401436-1454CUGCUGCAAAGAUCUGACCGGUCAGAUCUUUGCAGCAG
214121421437-1455UGCUGCAAAGAUCUGACCCGGGUCAGAUCUUUGCAGCA
214321441457-1475UCAGUCCCCAAGAUUGUGGCCACAAUCUUGGGGACUGA
214521461458-1476CAGUCCCCAAGAUUGUGGCGCCACAAUCUUGGGGACUG
214721481459-1477AGUCCCCAAGAUUGUGGCAUGCCACAAUCUUGGGGACU
214921501461-1479UCCCCAAGAUUGUGGCAUUAAUGCCACAAUCUUGGGGA
215121521462-1480CCCCAAGAUUGUGGCAUUUAAAUGCCACAAUCUUGGGG
215321541463-1481CCCAAGAUUGUGGCAUUUGCAAAUGCCACAAUCUUGGG
215521561464-1482CCAAGAUUGUGGCAUUUGAUCAAAUGCCACAAUCUUGG
215721581465-1483CAAGAUUGUGGCAUUUGAAUUCAAAUGCCACAAUCUUG
215921601466-1484AAGAUUGUGGCAUUUGAAAUUUCAAAUGCCACAAUCUU
216121621467-1485AGAUUGUGGCAUUUGAAACGUUUCAAAUGCCACAAUCU
216321641468-1486GAUUGUGGCAUUUGAAACUAGUUUCAAAUGCCACAAUC
216521661469-1487AUUGUGGCAUUUGAAACUGCAGUUUCAAAUGCCACAAU
216721681470-1488UUGUGGCAUUUGAAACUGUACAGUUUCAAAUGCCACAA
216921701471-1489UGUGGCAUUUGAAACUGUCGACAGUUUCAAAUGCCACA
217121721472-1490GUGGCAUUUGAAACUGUCCGGACAGUUUCAAAUGCCAC
217321741473-1491UGGCAUUUGAAACUGUCCAUGGACAGUUUCAAAUGCCA
217521761474-1492GGCAUUUGAAACUGUCCAUAUGGACAGUUUCAAAUGCC
217721781475-1493GCAUUUGAAACUGUCCAUUAAUGGACAGUUUCAAAUGC
217921801476-1494CAUUUGAAACUGUCCAUUCGAAUGGACAGUUUCAAAUG
218121821477-1495AUUUGAAACUGUCCAUUCAUGAAUGGACAGUUUCAAAU
218321841479-1497UUGAAACUGUCCAUUCAAUAUUGAAUGGACAGUUUCAA
218521861480-1498UGAAACUGUCCAUUCAAUGCAUUGAAUGGACAGUUUCA
218721881481-1499GAAACUGUCCAUUCAAUGGCCAUUGAAUGGACAGUUUC
218921901482-1500AAACUGUCCAUUCAAUGGAUCCAUUGAAUGGACAGUUU
219121921483-1501AACUGUCCAUUCAAUGGAUAUCCAUUGAAUGGACAGUU
219321941484-1502ACUGUCCAUUCAAUGGAUGCAUCCAUUGAAUGGACAGU
219521961485-1503CUGUCCAUUCAAUGGAUGGCCAUCCAUUGAAUGGACAG
219721981486-1504UGUCCAUUCAAUGGAUGGGCCCAUCCAUUGAAUGGACA
219922001487-1505GUCCAUUCAAUGGAUGGGGCCCCAUCCAUUGAAUGGAC
220122021488-1506UCCAUUCAAUGGAUGGGGCGCCCCAUCCAUUGAAUGGA
220322041508-1526GUGUGCCCACUGGAAGAGCGCUCUUCCAGUGGGCACAC
220522061509-1527UGUGCCCACUGGAAGAGCUAGCUCUUCCAGUGGGCACA
220722081510-1528GUGCCCACUGGAAGAGCUGCAGCUCUUCCAGUGGGCAC
220922101511-1529UGCCCACUGGAAGAGCUGUACAGCUCUUCCAGUGGGCA
221122121512-1530GCCCACUGGAAGAGCUGUGCACAGCUCUUCCAGUGGGC
221322141513-1531CCCACUGGAAGAGCUGUGUACACAGCUCUUCCAGUGGG
221522161514-1532CCACUGGAAGAGCUGUGUGCACACAGCUCUUCCAGUGG
221722181515-1533CACUGGAAGAGCUGUGUGAUCACACAGCUCUUCCAGUG
221922201516-1534ACUGGAAGAGCUGUGUGAUAUCACACAGCUCUUCCAGU
222122221517-1535CUGGAAGAGCUGUGUGAUGCAUCACACAGCUCUUCCAG
222322241518-1536UGGAAGAGCUGUGUGAUGUACAUCACACAGCUCUUCCA
222522261519-1537GGAAGAGCUGUGUGAUGUGCACAUCACACAGCUCUUCC
222722281520-1538GAAGAGCUGUGUGAUGUGGCCACAUCACACAGCUCUUC
222922301521-1539AAGAGCUGUGUGAUGUGGCGCCACAUCACACAGCUCUU
223122321522-1540AGAGCUGUGUGAUGUGGCCGGCCACAUCACACAGCUCU
223322341523-1541GAGCUGUGUGAUGUGGCCCGGGCCACAUCACACAGCUC
223522361524-1542AGCUGUGUGAUGUGGCCCAUGGGCCACAUCACACAGCU
223722381525-1543GCUGUGUGAUGUGGCCCAUAUGGGCCACAUCACACAGC
223922401526-1544CUGUGUGAUGUGGCCCAUGCAUGGGCCACAUCACACAG
224122421527-1545UGUGUGAUGUGGCCCAUGAUCAUGGGCCACAUCACACA
224322441528-1546GUGUGAUGUGGCCCAUGAGCUCAUGGGCCACAUCACAC
224522461529-1547UGUGAUGUGGCCCAUGAGUACUCAUGGGCCACAUCACA
224722481532-1550GAUGUGGCCCAUGAGUUUGCAAACUCAUGGGCCACAUC
224922501533-1551AUGUGGCCCAUGAGUUUGGCCAAACUCAUGGGCCACAU
225122521534-1552UGUGGCCCAUGAGUUUGGAUCCAAACUCAUGGGCCACA
225322541535-1553GUGGCCCAUGAGUUUGGAGCUCCAAACUCAUGGGCCAC
225522561536-1554UGGCCCAUGAGUUUGGAGCGCUCCAAACUCAUGGGCCA
225722581537-1555GGCCCAUGAGUUUGGAGCAUGCUCCAAACUCAUGGGCC
225922601538-1556GCCCAUGAGUUUGGAGCAAUUGCUCCAAACUCAUGGGC
226122621539-1557CCCAUGAGUUUGGAGCAAUAUUGCUCCAAACUCAUGGG
226322641540-1558CCAUGAGUUUGGAGCAAUCGAUUGCUCCAAACUCAUGG
226522661542-1560AUGAGUUUGGAGCAAUCACGUGAUUGCUCCAAACUCAU
226722681543-1561UGAGUUUGGAGCAAUCACCGGUGAUUGCUCCAAACUCA
226922701545-1563AGUUUGGAGCAAUCACCUUAAGGUGAUUGCUCCAAACU
227122721546-1564GUUUGGAGCAAUCACCUUCGAAGGUGAUUGCUCCAAAC
227322741547-1565UUUGGAGCAAUCACCUUCGCGAAGGUGAUUGCUCCAAA
227522761548-1566UUGGAGCAAUCACCUUCGUACGAAGGUGAUUGCUCCAA
227722781549-1567UGGAGCAAUCACCUUCGUGCACGAAGGUGAUUGCUCCA
227922801550-1568GGAGCAAUCACCUUCGUGGCCACGAAGGUGAUUGCUCC
228122821551-1569GAGCAAUCACCUUCGUGGAUCCACGAAGGUGAUUGCUC
228322841552-1570AGCAAUCACCUUCGUGGAUAUCCACGAAGGUGAUUGCU
228522861553-1571GCAAUCACCUUCGUGGAUGCAUCCACGAAGGUGAUUGC
228722881554-1572CAAUCACCUUCGUGGAUGAUCAUCCACGAAGGUGAUUG
228922901555-1573AAUCACCUUCGUGGAUGAGCUCAUCCACGAAGGUGAUU
229122921556-1574AUCACCUUCGUGGAUGAGGCCUCAUCCACGAAGGUGAU
229322941557-1575UCACCUUCGUGGAUGAGGUACCUCAUCCACGAAGGUGA
229522961558-1576CACCUUCGUGGAUGAGGUCGACCUCAUCCACGAAGGUG
229722981559-1577ACCUUCGUGGAUGAGGUCCGGACCUCAUCCACGAAGGU
229923001560-1578CCUUCGUGGAUGAGGUCCAUGGACCUCAUCCACGAAGG
230123021561-1579CUUCGUGGAUGAGGUCCACGUGGACCUCAUCCACGAAG
230323041562-1580UUCGUGGAUGAGGUCCACGCGUGGACCUCAUCCACGAA
230523061563-1581UCGUGGAUGAGGUCCACGCGCGUGGACCUCAUCCACGA
230723081564-1582CGUGGAUGAGGUCCACGCAUGCGUGGACCUCAUCCACG
230923101565-1583GUGGAUGAGGUCCACGCAGCUGCGUGGACCUCAUCCAC
231123121566-1584UGGAUGAGGUCCACGCAGUACUGCGUGGACCUCAUCCA
231323141567-1585GGAUGAGGUCCACGCAGUGCACUGCGUGGACCUCAUCC
231523161568-1586GAUGAGGUCCACGCAGUGGCCACUGCGUGGACCUCAUC
231723181569-1587AUGAGGUCCACGCAGUGGGCCCACUGCGUGGACCUCAU
231923201570-1588UGAGGUCCACGCAGUGGGGCCCCACUGCGUGGACCUCA
232123221571-1589GAGGUCCACGCAGUGGGGCGCCCCACUGCGUGGACCUC
232323241572-1590AGGUCCACGCAGUGGGGCUAGCCCCACUGCGUGGACCU
232523261595-1613GGGGCUCGAGGCGGAGGGAUCCCUCCGCCUCGAGCCCC
232723281596-1614GGGCUCGAGGCGGAGGGAUAUCCCUCCGCCUCGAGCCC
232923301597-1615GGCUCGAGGCGGAGGGAUUAAUCCCUCCGCCUCGAGCC
233123321598-1616GCUCGAGGCGGAGGGAUUGCAAUCCCUCCGCCUCGAGC
233323341599-1617CUCGAGGCGGAGGGAUUGGCCAAUCCCUCCGCCUCGAG
233523361600-1618UCGAGGCGGAGGGAUUGGGCCCAAUCCCUCCGCCUCGA
233723381601-1619CGAGGCGGAGGGAUUGGGGCCCCAAUCCCUCCGCCUCG
233923401602-1620GAGGCGGAGGGAUUGGGGAUCCCCAAUCCCUCCGCCUC
234123421603-1621AGGCGGAGGGAUUGGGGAUAUCCCCAAUCCCUCCGCCU
234323441604-1622GGCGGAGGGAUUGGGGAUCGAUCCCCAAUCCCUCCGCC
234523461605-1623GCGGAGGGAUUGGGGAUCGCGAUCCCCAAUCCCUCCGC
234723481606-1624CGGAGGGAUUGGGGAUCGGCCGAUCCCCAAUCCCUCCG
234923501607-1625GGAGGGAUUGGGGAUCGGGCCCGAUCCCCAAUCCCUCC
235123521608-1626GAGGGAUUGGGGAUCGGGAUCCCGAUCCCCAAUCCCUC
235323541609-1627AGGGAUUGGGGAUCGGGAUAUCCCGAUCCCCAAUCCCU
235523561610-1628GGGAUUGGGGAUCGGGAUGCAUCCCGAUCCCCAAUCCC
235723581611-1629GGAUUGGGGAUCGGGAUGGCCAUCCCGAUCCCCAAUCC
235923601612-1630GAUUGGGGAUCGGGAUGGAUCCAUCCCGAUCCCCAAUC
236123621613-1631AUUGGGGAUCGGGAUGGAGCUCCAUCCCGAUCCCCAAU
236323641614-1632UUGGGGAUCGGGAUGGAGUACUCCAUCCCGAUCCCCAA
236523661615-1633UGGGGAUCGGGAUGGAGUCGACUCCAUCCCGAUCCCCA
236723681617-1635GGGAUCGGGAUGGAGUCAUAUGACUCCAUCCCGAUCCC
236923701618-1636GGAUCGGGAUGGAGUCAUGCAUGACUCCAUCCCGAUCC
237123721619-1637GAUCGGGAUGGAGUCAUGCGCAUGACUCCAUCCCGAUC
237323741620-1638AUCGGGAUGGAGUCAUGCCGGCAUGACUCCAUCCCGAU
237523761621-1639UCGGGAUGGAGUCAUGCCAUGGCAUGACUCCAUCCCGA
237723781622-1640CGGGAUGGAGUCAUGCCAAUUGGCAUGACUCCAUCCCG
237923801623-1641GGGAUGGAGUCAUGCCAAAUUUGGCAUGACUCCAUCCC
238123821624-1642GGAUGGAGUCAUGCCAAAAUUUUGGCAUGACUCCAUCC
238323841625-1643GAUGGAGUCAUGCCAAAAAUUUUUGGCAUGACUCCAUC
238523861626-1644AUGGAGUCAUGCCAAAAAUAUUUUUGGCAUGACUCCAU
238723881627-1645UGGAGUCAUGCCAAAAAUGCAUUUUUGGCAUGACUCCA
238923901628-1646GGAGUCAUGCCAAAAAUGGCCAUUUUUGGCAUGACUCC
239123921629-1647GAGUCAUGCCAAAAAUGGAUCCAUUUUUGGCAUGACUC
239323941630-1648AGUCAUGCCAAAAAUGGACGUCCAUUUUUGGCAUGACU
239523961632-1650UCAUGCCAAAAAUGGACAUAUGUCCAUUUUUGGCAUGA
239723981633-1651CAUGCCAAAAAUGGACAUCGAUGUCCAUUUUUGGCAUG
239924001636-1654GCCAAAAAUGGACAUCAUUAAUGAUGUCCAUUUUUGGC
240124021638-1656CAAAAAUGGACAUCAUUUCGAAAUGAUGUCCAUUUUUG
240324041639-1657AAAAAUGGACAUCAUUUCUAGAAAUGAUGUCCAUUUUU
240524061640-1658AAAAUGGACAUCAUUUCUGCAGAAAUGAUGUCCAUUUU
240724081641-1659AAAUGGACAUCAUUUCUGGCCAGAAAUGAUGUCCAUUU
240924101642-1660AAUGGACAUCAUUUCUGGAUCCAGAAAUGAUGUCCAUU
241124121643-1661AUGGACAUCAUUUCUGGAAUUCCAGAAAUGAUGUCCAU
241324141644-1662UGGACAUCAUUUCUGGAACGUUCCAGAAAUGAUGUCCA
241524161645-1663GGACAUCAUUUCUGGAACAUGUUCCAGAAAUGAUGUCC
241724181646-1664GACAUCAUUUCUGGAACACGUGUUCCAGAAAUGAUGUC
241924201647-1665ACAUCAUUUCUGGAACACUAGUGUUCCAGAAAUGAUGU
242124221648-1666CAUCAUUUCUGGAACACUUAAGUGUUCCAGAAAUGAUG
242324241649-1667AUCAUUUCUGGAACACUUGCAAGUGUUCCAGAAAUGAU
242524261650-1668UCAUUUCUGGAACACUUGGCCAAGUGUUCCAGAAAUGA
242724281651-1669CAUUUCUGGAACACUUGGCGCCAAGUGUUCCAGAAAUG
242924301652-1670AUUUCUGGAACACUUGGCAUGCCAAGUGUUCCAGAAAU
243124321653-1671UUUCUGGAACACUUGGCAAUUGCCAAGUGUUCCAGAAA
243324341654-1672UUCUGGAACACUUGGCAAAUUUGCCAAGUGUUCCAGAA
243524361655-1673UCUGGAACACUUGGCAAAGCUUUGCCAAGUGUUCCAGA
243724381656-1674CUGGAACACUUGGCAAAGCGCUUUGCCAAGUGUUCCAG
243924401657-1675UGGAACACUUGGCAAAGCCGGCUUUGCCAAGUGUUCCA
244124421658-1676GGAACACUUGGCAAAGCCUAGGCUUUGCCAAGUGUUCC
244324441659-1677GAACACUUGGCAAAGCCUUAAGGCUUUGCCAAGUGUUC
244524461660-1678AACACUUGGCAAAGCCUUUAAAGGCUUUGCCAAGUGUU
244724481661-1679ACACUUGGCAAAGCCUUUGCAAAGGCUUUGCCAAGUGU
244924501662-1680CACUUGGCAAAGCCUUUGGCCAAAGGCUUUGCCAAGUG
245124521682-1700UGUGUUGGAGGGUACAUCGCGAUGUACCCUCCAACACA
245324541683-1701GUGUUGGAGGGUACAUCGCGCGAUGUACCCUCCAACAC
245524561684-1702UGUUGGAGGGUACAUCGCCGGCGAUGUACCCUCCAACA
245724581685-1703GUUGGAGGGUACAUCGCCAUGGCGAUGUACCCUCCAAC
245924601686-1704UUGGAGGGUACAUCGCCAGCUGGCGAUGUACCCUCCAA
246124621687-1705UGGAGGGUACAUCGCCAGCGCUGGCGAUGUACCCUCCA
246324641688-1706GGAGGGUACAUCGCCAGCAUGCUGGCGAUGUACCCUCC
246524661689-1707GAGGGUACAUCGCCAGCACGUGCUGGCGAUGUACCCUC
246724681690-1708AGGGUACAUCGCCAGCACGCGUGCUGGCGAUGUACCCU
246924701691-1709GGGUACAUCGCCAGCACGAUCGUGCUGGCGAUGUACCC
247124721692-1710GGUACAUCGCCAGCACGAGCUCGUGCUGGCGAUGUACC
247324741693-1711GUACAUCGCCAGCACGAGUACUCGUGCUGGCGAUGUAC
247524761694-1712UACAUCGCCAGCACGAGUUAACUCGUGCUGGCGAUGUA
247724781695-1713ACAUCGCCAGCACGAGUUCGAACUCGUGCUGGCGAUGU
247924801696-1714CAUCGCCAGCACGAGUUCUAGAACUCGUGCUGGCGAUG
248124821697-1715AUCGCCAGCACGAGUUCUCGAGAACUCGUGCUGGCGAU
248324841698-1716UCGCCAGCACGAGUUCUCUAGAGAACUCGUGCUGGCGA
248524861699-1717CGCCAGCACGAGUUCUCUGCAGAGAACUCGUGCUGGCG
248724881700-1718GCCAGCACGAGUUCUCUGAUCAGAGAACUCGUGCUGGC
248924901701-1719CCAGCACGAGUUCUCUGAUAUCAGAGAACUCGUGCUGG
249124921702-1720CAGCACGAGUUCUCUGAUUAAUCAGAGAACUCGUGCUG
249324941703-1721AGCACGAGUUCUCUGAUUGCAAUCAGAGAACUCGUGCU
249524961704-1722GCACGAGUUCUCUGAUUGAUCAAUCAGAGAACUCGUGC
249724981705-1723CACGAGUUCUCUGAUUGACGUCAAUCAGAGAACUCGUG
249925001707-1725CGAGUUCUCUGAUUGACACGUGUCAAUCAGAGAACUCG
250125021727-1745GUACGGUCCUAUGCUGCUGCAGCAGCAUAGGACCGUAC
250325041728-1746UACGGUCCUAUGCUGCUGGCCAGCAGCAUAGGACCGUA
250525061729-1747ACGGUCCUAUGCUGCUGGCGCCAGCAGCAUAGGACCGU
250725081730-1748CGGUCCUAUGCUGCUGGCUAGCCAGCAGCAUAGGACCG
250925101731-1749GGUCCUAUGCUGCUGGCUUAAGCCAGCAGCAUAGGACC
251125121732-1750GUCCUAUGCUGCUGGCUUCGAAGCCAGCAGCAUAGGAC
251325141733-1751UCCUAUGCUGCUGGCUUCAUGAAGCCAGCAGCAUAGGA
251525161734-1752CCUAUGCUGCUGGCUUCAUAUGAAGCCAGCAGCAUAGG
251725181735-1753CUAUGCUGCUGGCUUCAUCGAUGAAGCCAGCAGCAUAG
251925201736-1754UAUGCUGCUGGCUUCAUCUAGAUGAAGCCAGCAGCAUA
252125221737-1755AUGCUGCUGGCUUCAUCUUAAGAUGAAGCCAGCAGCAU
252325241738-1756UGCUGCUGGCUUCAUCUUCGAAGAUGAAGCCAGCAGCA
252525261739-1757GCUGCUGGCUUCAUCUUCAUGAAGAUGAAGCCAGCAGC
252725281740-1758CUGCUGGCUUCAUCUUCACGUGAAGAUGAAGCCAGCAG
252925301741-1759UGCUGGCUUCAUCUUCACCGGUGAAGAUGAAGCCAGCA
253125321742-1760GCUGGCUUCAUCUUCACCAUGGUGAAGAUGAAGCCAGC
253325341743-1761CUGGCUUCAUCUUCACCACGUGGUGAAGAUGAAGCCAG
253525361744-1762UGGCUUCAUCUUCACCACCGGUGGUGAAGAUGAAGCCA
253725381745-1763GGCUUCAUCUUCACCACCUAGGUGGUGAAGAUGAAGCC
253925401746-1764GCUUCAUCUUCACCACCUCGAGGUGGUGAAGAUGAAGC
254125421747-1765CUUCAUCUUCACCACCUCUAGAGGUGGUGAAGAUGAAG
254325441748-1766UUCAUCUUCACCACCUCUCGAGAGGUGGUGAAGAUGAA
254525461749-1767UCAUCUUCACCACCUCUCUAGAGAGGUGGUGAAGAUGA
254725481750-1768CAUCUUCACCACCUCUCUGCAGAGAGGUGGUGAAGAUG
254925501751-1769AUCUUCACCACCUCUCUGCGCAGAGAGGUGGUGAAGAU
255125521752-1770UCUUCACCACCUCUCUGCCGGCAGAGAGGUGGUGAAGA
255325541753-1771CUUCACCACCUCUCUGCCAUGGCAGAGAGGUGGUGAAG
255525561754-1772UUCACCACCUCUCUGCCACGUGGCAGAGAGGUGGUGAA
255725581755-1773UCACCACCUCUCUGCCACCGGUGGCAGAGAGGUGGUGA
255925601756-1774CACCACCUCUCUGCCACCCGGGUGGCAGAGAGGUGGUG
256125621757-1775ACCACCUCUCUGCCACCCAUGGGUGGCAGAGAGGUGGU
256325641758-1776CCACCUCUCUGCCACCCAUAUGGGUGGCAGAGAGGUGG
256525661759-1777CACCUCUCUGCCACCCAUGCAUGGGUGGCAGAGAGGUG
256725681760-1778ACCUCUCUGCCACCCAUGCGCAUGGGUGGCAGAGAGGU
256925701761-1779CCUCUCUGCCACCCAUGCUAGCAUGGGUGGCAGAGAGG
257125721762-1780CUCUCUGCCACCCAUGCUGCAGCAUGGGUGGCAGAGAG
257325741763-1781UCUCUGCCACCCAUGCUGCGCAGCAUGGGUGGCAGAGA
257525761764-1782CUCUGCCACCCAUGCUGCUAGCAGCAUGGGUGGCAGAG
257725781765-1783UCUGCCACCCAUGCUGCUGCAGCAGCAUGGGUGGCAGA
257925801766-1784CUGCCACCCAUGCUGCUGGCCAGCAGCAUGGGUGGCAG
258125821767-1785UGCCACCCAUGCUGCUGGCGCCAGCAGCAUGGGUGGCA
258325841768-1786GCCACCCAUGCUGCUGGCUAGCCAGCAGCAUGGGUGGC
258525861769-1787CCACCCAUGCUGCUGGCUGCAGCCAGCAGCAUGGGUGG
258725881770-1788CACCCAUGCUGCUGGCUGGCCAGCCAGCAGCAUGGGUG
258925901771-1789ACCCAUGCUGCUGGCUGGAUCCAGCCAGCAGCAUGGGU
259125921772-1790CCCAUGCUGCUGGCUGGAGCUCCAGCCAGCAGCAUGGG
259325941773-1791CCAUGCUGCUGGCUGGAGCGCUCCAGCCAGCAGCAUGG
259525961774-1792CAUGCUGCUGGCUGGAGCCGGCUCCAGCCAGCAGCAUG
259725981775-1793AUGCUGCUGGCUGGAGCCCGGGCUCCAGCCAGCAGCAU
259926001776-1794UGCUGCUGGCUGGAGCCCUAGGGCUCCAGCCAGCAGCA
260126021777-1795GCUGCUGGCUGGAGCCCUGCAGGGCUCCAGCCAGCAGC
260326041778-1796CUGCUGGCUGGAGCCCUGGCCAGGGCUCCAGCCAGCAG
260526061779-1797UGCUGGCUGGAGCCCUGGAUCCAGGGCUCCAGCCAGCA
260726081780-1798GCUGGCUGGAGCCCUGGAGCUCCAGGGCUCCAGCCAGC
260926101781-1799CUGGCUGGAGCCCUGGAGUACUCCAGGGCUCCAGCCAG
261126121782-1800UGGCUGGAGCCCUGGAGUCGACUCCAGGGCUCCAGCCA
261326141783-1801GGCUGGAGCCCUGGAGUCUAGACUCCAGGGCUCCAGCC
261526161784-1802GCUGGAGCCCUGGAGUCUGCAGACUCCAGGGCUCCAGC
261726181785-1803CUGGAGCCCUGGAGUCUGUACAGACUCCAGGGCUCCAG
261926201786-1804UGGAGCCCUGGAGUCUGUGCACAGACUCCAGGGCUCCA
262126221787-1805GGAGCCCUGGAGUCUGUGCGCACAGACUCCAGGGCUCC
262326241788-1806GAGCCCUGGAGUCUGUGCGCGCACAGACUCCAGGGCUC
262526261789-1807AGCCCUGGAGUCUGUGCGGCCGCACAGACUCCAGGGCU
262726281790-1808GCCCUGGAGUCUGUGCGGAUCCGCACAGACUCCAGGGC
262926301792-1810CCUGGAGUCUGUGCGGAUCGAUCCGCACAGACUCCAGG
263126321793-1811CUGGAGUCUGUGCGGAUCCGGAUCCGCACAGACUCCAG
263326341795-1813GGAGUCUGUGCGGAUCCUGCAGGAUCCGCACAGACUCC
263526361796-1814GAGUCUGUGCGGAUCCUGAUCAGGAUCCGCACAGACUC
263726381797-1815AGUCUGUGCGGAUCCUGAAUUCAGGAUCCGCACAGACU
263926401798-1816GUCUGUGCGGAUCCUGAAGCUUCAGGAUCCGCACAGAC
264126421799-1817UCUGUGCGGAUCCUGAAGAUCUUCAGGAUCCGCACAGA
264326441800-1818CUGUGCGGAUCCUGAAGAGCUCUUCAGGAUCCGCACAG
264526461801-1819UGUGCGGAUCCUGAAGAGCGCUCUUCAGGAUCCGCACA
264726481802-1820GUGCGGAUCCUGAAGAGCGCGCUCUUCAGGAUCCGCAC
264926501803-1821UGCGGAUCCUGAAGAGCGCGCGCUCUUCAGGAUCCGCA
265126521804-1822GCGGAUCCUGAAGAGCGCUAGCGCUCUUCAGGAUCCGC
265326541805-1823CGGAUCCUGAAGAGCGCUGCAGCGCUCUUCAGGAUCCG
265526561806-1824GGAUCCUGAAGAGCGCUGAUCAGCGCUCUUCAGGAUCC
265726581807-1825GAUCCUGAAGAGCGCUGAGCUCAGCGCUCUUCAGGAUC
265926601808-1826AUCCUGAAGAGCGCUGAGGCCUCAGCGCUCUUCAGGAU
266126621809-1827UCCUGAAGAGCGCUGAGGGCCCUCAGCGCUCUUCAGGA
266326641810-1828CCUGAAGAGCGCUGAGGGAUCCCUCAGCGCUCUUCAGG
266526661811-1829CUGAAGAGCGCUGAGGGACGUCCCUCAGCGCUCUUCAG
266726681812-1830UGAAGAGCGCUGAGGGACGCGUCCCUCAGCGCUCUUCA
266926701813-1831GAAGAGCGCUGAGGGACGGCCGUCCCUCAGCGCUCUUC
267126721814-1832AAGAGCGCUGAGGGACGGGCCCGUCCCUCAGCGCUCUU
267326741815-1833AGAGCGCUGAGGGACGGGUACCCGUCCCUCAGCGCUCU
267526761816-1834GAGCGCUGAGGGACGGGUGCACCCGUCCCUCAGCGCUC
267726781817-1835AGCGCUGAGGGACGGGUGCGCACCCGUCCCUCAGCGCU
267926801818-1836GCGCUGAGGGACGGGUGCUAGCACCCGUCCCUCAGCGC
268126821819-1837CGCUGAGGGACGGGUGCUUAAGCACCCGUCCCUCAGCG
268326841820-1838GCUGAGGGACGGGUGCUUCGAAGCACCCGUCCCUCAGC
268526861821-1839CUGAGGGACGGGUGCUUCGCGAAGCACCCGUCCCUCAG
268726881822-1840UGAGGGACGGGUGCUUCGCGCGAAGCACCCGUCCCUCA
268926901823-1841GAGGGACGGGUGCUUCGCCGGCGAAGCACCCGUCCCUC
269126921824-1842AGGGACGGGUGCUUCGCCGCGGCGAAGCACCCGUCCCU
269326941825-1843GGGACGGGUGCUUCGCCGCGCGGCGAAGCACCCGUCCC
269526961826-1844GGACGGGUGCUUCGCCGCCGGCGGCGAAGCACCCGUCC
269726981827-1845GACGGGUGCUUCGCCGCCAUGGCGGCGAAGCACCCGUC
269927001828-1846ACGGGUGCUUCGCCGCCAGCUGGCGGCGAAGCACCCGU
270127021829-1847CGGGUGCUUCGCCGCCAGCGCUGGCGGCGAAGCACCCG
270327041830-1848GGGUGCUUCGCCGCCAGCAUGCUGGCGGCGAAGCACCC
270527061831-1849GGUGCUUCGCCGCCAGCACGUGCUGGCGGCGAAGCACC
270727081832-1850GUGCUUCGCCGCCAGCACCGGUGCUGGCGGCGAAGCAC
270927101833-1851UGCUUCGCCGCCAGCACCAUGGUGCUGGCGGCGAAGCA
271127121834-1852GCUUCGCCGCCAGCACCAGCUGGUGCUGGCGGCGAAGC
271327141835-1853CUUCGCCGCCAGCACCAGCGCUGGUGCUGGCGGCGAAG
271527161836-1854UUCGCCGCCAGCACCAGCGCGCUGGUGCUGGCGGCGAA
271727181837-1855UCGCCGCCAGCACCAGCGCGCGCUGGUGCUGGCGGCGA
271927201838-1856CGCCGCCAGCACCAGCGCAUGCGCUGGUGCUGGCGGCG
272127221839-1857GCCGCCAGCACCAGCGCAAUUGCGCUGGUGCUGGCGGC
272327241840-1858CCGCCAGCACCAGCGCAACGUUGCGCUGGUGCUGGCGG
272527261841-1859CGCCAGCACCAGCGCAACGCGUUGCGCUGGUGCUGGCG
272727281842-1860GCCAGCACCAGCGCAACGUACGUUGCGCUGGUGCUGGC
272927301865-1883CUCAUGAGACAGAUGCUAAUUAGCAUCUGUCUCAUGAG
273127321866-1884UCAUGAGACAGAUGCUAAUAUUAGCAUCUGUCUCAUGA
273327341867-1885CAUGAGACAGAUGCUAAUGCAUUAGCAUCUGUCUCAUG
273527361868-1886AUGAGACAGAUGCUAAUGGCCAUUAGCAUCUGUCUCAU
273727381869-1887UGAGACAGAUGCUAAUGGAUCCAUUAGCAUCUGUCUCA
273927401871-1889AGACAGAUGCUAAUGGAUGCAUCCAUUAGCAUCUGUCU
274127421872-1890GACAGAUGCUAAUGGAUGCGCAUCCAUUAGCAUCUGUC
274327441873-1891ACAGAUGCUAAUGGAUGCCGGCAUCCAUUAGCAUCUGU
274527461874-1892CAGAUGCUAAUGGAUGCCGCGGCAUCCAUUAGCAUCUG
274727481875-1893AGAUGCUAAUGGAUGCCGGCCGGCAUCCAUUAGCAUCU
274927501876-1894GAUGCUAAUGGAUGCCGGCGCCGGCAUCCAUUAGCAUC
275127521877-1895AUGCUAAUGGAUGCCGGCCGGCCGGCAUCCAUUAGCAU
275327541878-1896UGCUAAUGGAUGCCGGCCUAGGCCGGCAUCCAUUAGCA
275527561879-1897GCUAAUGGAUGCCGGCCUCGAGGCCGGCAUCCAUUAGC
275727581880-1898CUAAUGGAUGCCGGCCUCCGGAGGCCGGCAUCCAUUAG
275927601881-1899UAAUGGAUGCCGGCCUCCCGGGAGGCCGGCAUCCAUUA
276127621882-1900AAUGGAUGCCGGCCUCCCUAGGGAGGCCGGCAUCCAUU
276327641883-1901AUGGAUGCCGGCCUCCCUGCAGGGAGGCCGGCAUCCAU
276527661884-1902UGGAUGCCGGCCUCCCUGUACAGGGAGGCCGGCAUCCA
276727681885-1903GGAUGCCGGCCUCCCUGUUAACAGGGAGGCCGGCAUCC
276927701886-1904GAUGCCGGCCUCCCUGUUGCAACAGGGAGGCCGGCAUC
277127721887-1905AUGCCGGCCUCCCUGUUGUACAACAGGGAGGCCGGCAU
277327741888-1906UGCCGGCCUCCCUGUUGUCGACAACAGGGAGGCCGGCA
277527761889-1907GCCGGCCUCCCUGUUGUCCGGACAACAGGGAGGCCGGC
277727781890-1908CCGGCCUCCCUGUUGUCCAUGGACAACAGGGAGGCCGG
277927801891-1909CGGCCUCCCUGUUGUCCACGUGGACAACAGGGAGGCCG
278127821892-1910GGCCUCCCUGUUGUCCACUAGUGGACAACAGGGAGGCC
278327841893-1911GCCUCCCUGUUGUCCACUGCAGUGGACAACAGGGAGGC
278527861894-1912CCUCCCUGUUGUCCACUGCGCAGUGGACAACAGGGAGG
278727881895-1913CUCCCUGUUGUCCACUGCCGGCAGUGGACAACAGGGAG
278927901896-1914UCCCUGUUGUCCACUGCCCGGGCAGUGGACAACAGGGA
279127921897-1915CCCUGUUGUCCACUGCCCCGGGGCAGUGGACAACAGGG
279327941898-1916CCUGUUGUCCACUGCCCCAUGGGGCAGUGGACAACAGG
279527961899-1917CUGUUGUCCACUGCCCCAGCUGGGGCAGUGGACAACAG
279727981900-1918UGUUGUCCACUGCCCCAGCGCUGGGGCAGUGGACAACA
279928001901-1919GUUGUCCACUGCCCCAGCCGGCUGGGGCAGUGGACAAC
280128021902-1920UUGUCCACUGCCCCAGCCAUGGCUGGGGCAGUGGACAA
280328041903-1921UGUCCACUGCCCCAGCCACGUGGCUGGGGCAGUGGACA
280528061904-1922GUCCACUGCCCCAGCCACAUGUGGCUGGGGCAGUGGAC
280728081905-1923UCCACUGCCCCAGCCACAUAUGUGGCUGGGGCAGUGGA
280928101906-1924CCACUGCCCCAGCCACAUCGAUGUGGCUGGGGCAGUGG
281128121907-1925CACUGCCCCAGCCACAUCAUGAUGUGGCUGGGGCAGUG
281328141908-1926ACUGCCCCAGCCACAUCAUAUGAUGUGGCUGGGGCAGU
281528161909-1927CUGCCCCAGCCACAUCAUCGAUGAUGUGGCUGGGGCAG
281728181910-1928UGCCCCAGCCACAUCAUCCGGAUGAUGUGGCUGGGGCA
281928201911-1929GCCCCAGCCACAUCAUCCCGGGAUGAUGUGGCUGGGGC
282128221912-1930CCCCAGCCACAUCAUCCCUAGGGAUGAUGUGGCUGGGG
282328241913-1931CCCAGCCACAUCAUCCCUGCAGGGAUGAUGUGGCUGGG
282528261914-1932CCAGCCACAUCAUCCCUGUACAGGGAUGAUGUGGCUGG
282728281915-1933CAGCCACAUCAUCCCUGUGCACAGGGAUGAUGUGGCUG
282928301916-1934AGCCACAUCAUCCCUGUGCGCACAGGGAUGAUGUGGCU
283128321917-1935GCCACAUCAUCCCUGUGCGCGCACAGGGAUGAUGUGGC
283328341918-1936CCACAUCAUCCCUGUGCGGCCGCACAGGGAUGAUGUGG
283528361919-1937CACAUCAUCCCUGUGCGGGCCCGCACAGGGAUGAUGUG
283728381920-1938ACAUCAUCCCUGUGCGGGUACCCGCACAGGGAUGAUGU
283928401922-1940AUCAUCCCUGUGCGGGUUGCAACCCGCACAGGGAUGAU
284128421923-1941UCAUCCCUGUGCGGGUUGCGCAACCCGCACAGGGAUGA
284328441924-1942CAUCCCUGUGCGGGUUGCAUGCAACCCGCACAGGGAUG
284528461925-1943AUCCCUGUGCGGGUUGCAGCUGCAACCCGCACAGGGAU
284728481926-1944UCCCUGUGCGGGUUGCAGAUCUGCAACCCGCACAGGGA
284928501928-1946CCUGUGCGGGUUGCAGAUGCAUCUGCAACCCGCACAGG
285128521929-1947CUGUGCGGGUUGCAGAUGCGCAUCUGCAACCCGCACAG
285328541930-1948UGUGCGGGUUGCAGAUGCUAGCAUCUGCAACCCGCACA
285528561931-1949GUGCGGGUUGCAGAUGCUGCAGCAUCUGCAACCCGCAC
285728581932-1950UGCGGGUUGCAGAUGCUGCGCAGCAUCUGCAACCCGCA
285928601933-1951GCGGGUUGCAGAUGCUGCUAGCAGCAUCUGCAACCCGC
286128621934-1952CGGGUUGCAGAUGCUGCUAUAGCAGCAUCUGCAACCCG
286328641935-1953GGGUUGCAGAUGCUGCUAAUUAGCAGCAUCUGCAACCC
286528661936-1954GGUUGCAGAUGCUGCUAAAUUUAGCAGCAUCUGCAACC
286728681937-1955GUUGCAGAUGCUGCUAAAAUUUUAGCAGCAUCUGCAAC
286928701938-1956UUGCAGAUGCUGCUAAAAAUUUUUAGCAGCAUCUGCAA
287128721939-1957UGCAGAUGCUGCUAAAAACGUUUUUAGCAGCAUCUGCA
287328741940-1958GCAGAUGCUGCUAAAAACAUGUUUUUAGCAGCAUCUGC
287528761941-1959CAGAUGCUGCUAAAAACACGUGUUUUUAGCAGCAUCUG
287728781961-1979GAAGUCUGUGAUGAACUAAUUAGUUCAUCACAGACUUC
287928801963-1981AGUCUGUGAUGAACUAAUGCAUUAGUUCAUCACAGACU
288128821965-1983UCUGUGAUGAACUAAUGAGCUCAUUAGUUCAUCACAGA
288328841966-1984CUGUGAUGAACUAAUGAGCGCUCAUUAGUUCAUCACAG
288528861968-1986GUGAUGAACUAAUGAGCAGCUGCUCAUUAGUUCAUCAC
288728881969-1987UGAUGAACUAAUGAGCAGAUCUGCUCAUUAGUUCAUCA
288928901970-1988GAUGAACUAAUGAGCAGACGUCUGCUCAUUAGUUCAUC
289128921971-1989AUGAACUAAUGAGCAGACAUGUCUGCUCAUUAGUUCAU
289328941972-1990UGAACUAAUGAGCAGACAUAUGUCUGCUCAUUAGUUCA
289528961973-1991GAACUAAUGAGCAGACAUAUAUGUCUGCUCAUUAGUUC
289728981974-1992AACUAAUGAGCAGACAUAAUUAUGUCUGCUCAUUAGUU
289929001975-1993ACUAAUGAGCAGACAUAACGUUAUGUCUGCUCAUUAGU
290129021978-1996AAUGAGCAGACAUAACAUCGAUGUUAUGUCUGCUCAUU
290329041979-1997AUGAGCAGACAUAACAUCUAGAUGUUAUGUCUGCUCAU
290529061980-1998UGAGCAGACAUAACAUCUAUAGAUGUUAUGUCUGCUCA
290729082000-2018GUGCAAGCAAUCAAUUACCGGUAAUUGAUUGCUUGCAC
290929102001-2019UGCAAGCAAUCAAUUACCCGGGUAAUUGAUUGCUUGCA
291129122002-2020GCAAGCAAUCAAUUACCCUAGGGUAAUUGAUUGCUUGC
291329142004-2022AAGCAAUCAAUUACCCUACGUAGGGUAAUUGAUUGCUU
291529162024-2042GUGCCCCGGGGAGAAGAGCGCUCUUCUCCCCGGGGCAC
291729182025-2043UGCCCCGGGGAGAAGAGCUAGCUCUUCUCCCCGGGGCA
291929202026-2044GCCCCGGGGAGAAGAGCUCGAGCUCUUCUCCCCGGGGC
292129222027-2045CCCCGGGGAGAAGAGCUCCGGAGCUCUUCUCCCCGGGG
292329242028-2046CCCGGGGAGAAGAGCUCCUAGGAGCUCUUCUCCCCGGG
292529262029-2047CCGGGGAGAAGAGCUCCUAUAGGAGCUCUUCUCCCCGG
292729282030-2048CGGGGAGAAGAGCUCCUACGUAGGAGCUCUUCUCCCCG
292929302031-2049GGGGAGAAGAGCUCCUACGCGUAGGAGCUCUUCUCCCC
293129322032-2050GGGAGAAGAGCUCCUACGGCCGUAGGAGCUCUUCUCCC
293329342033-2051GGAGAAGAGCUCCUACGGAUCCGUAGGAGCUCUUCUCC
293529362034-2052GAGAAGAGCUCCUACGGAUAUCCGUAGGAGCUCUUCUC
293729382060-2078ACCCCUCACCACACACCCCGGGGUGUGUGGUGAGGGGU
293929402061-2079CCCCUCACCACACACCCCAUGGGGUGUGUGGUGAGGGG
294129422062-2080CCCUCACCACACACCCCAGCUGGGGUGUGUGGUGAGGG
294329442063-2081CCUCACCACACACCCCAGAUCUGGGGUGUGUGGUGAGG
294529462064-2082CUCACCACACACCCCAGAUAUCUGGGGUGUGUGGUGAG
294729482065-2083UCACCACACACCCCAGAUGCAUCUGGGGUGUGUGGUGA
294929502066-2084CACCACACACCCCAGAUGAUCAUCUGGGGUGUGUGGUG
295129522067-2085ACCACACACCCCAGAUGAUAUCAUCUGGGGUGUGUGGU
295329542068-2086CCACACACCCCAGAUGAUGCAUCAUCUGGGGUGUGUGG
295529562069-2087CACACACCCCAGAUGAUGAUCAUCAUCUGGGGUGUGUG
295729582070-2088ACACACCCCAGAUGAUGAAUUCAUCAUCUGGGGUGUGU
295929602071-2089CACACCCCAGAUGAUGAACGUUCAUCAUCUGGGGUGUG
296129622072-2090ACACCCCAGAUGAUGAACUAGUUCAUCAUCUGGGGUGU
296329642073-2091CACCCCAGAUGAUGAACUAUAGUUCAUCAUCUGGGGUG
296529662074-2092ACCCCAGAUGAUGAACUACGUAGUUCAUCAUCUGGGGU
296729682076-2094CCCAGAUGAUGAACUACUUAAGUAGUUCAUCAUCUGGG
296929702077-2095CCAGAUGAUGAACUACUUCGAAGUAGUUCAUCAUCUGG
297129722078-2096CAGAUGAUGAACUACUUCCGGAAGUAGUUCAUCAUCUG
297329742079-2097AGAUGAUGAACUACUUCCUAGGAAGUAGUUCAUCAUCU
297529762080-2098GAUGAUGAACUACUUCCUUAAGGAAGUAGUUCAUCAUC
297729782081-2099AUGAUGAACUACUUCCUUGCAAGGAAGUAGUUCAUCAU
297929802082-2100UGAUGAACUACUUCCUUGAUCAAGGAAGUAGUUCAUCA
298129822083-2101GAUGAACUACUUCCUUGAGCUCAAGGAAGUAGUUCAUC
298329842084-2102AUGAACUACUUCCUUGAGAUCUCAAGGAAGUAGUUCAU
298529862085-2103UGAACUACUUCCUUGAGAAUUCUCAAGGAAGUAGUUCA
298729882086-2104GAACUACUUCCUUGAGAAUAUUCUCAAGGAAGUAGUUC
298929902087-2105AACUACUUCCUUGAGAAUCGAUUCUCAAGGAAGUAGUU
299129922088-2106ACUACUUCCUUGAGAAUCUAGAUUCUCAAGGAAGUAGU
299329942089-2107CUACUUCCUUGAGAAUCUGCAGAUUCUCAAGGAAGUAG
299529962090-2108UACUUCCUUGAGAAUCUGCGCAGAUUCUCAAGGAAGUA
299729982091-2109ACUUCCUUGAGAAUCUGCUAGCAGAUUCUCAAGGAAGU
299930002117-2135UGGAAGCAAGUGGGGCUGGCCAGCCCCACUUGCUUCCA
300130022118-2136GGAAGCAAGUGGGGCUGGAUCCAGCCCCACUUGCUUCC
300330042119-2137GAAGCAAGUGGGGCUGGAAUUCCAGCCCCACUUGCUUC
300530062120-2138AAGCAAGUGGGGCUGGAACGUUCCAGCCCCACUUGCUU
300730082121-2139AGCAAGUGGGGCUGGAACUAGUUCCAGCCCCACUUGCU
300930102122-2140GCAAGUGGGGCUGGAACUGCAGUUCCAGCCCCACUUGC
301130122123-2141CAAGUGGGGCUGGAACUGAUCAGUUCCAGCCCCACUUG
301330142124-2142AAGUGGGGCUGGAACUGAAUUCAGUUCCAGCCCCACUU
301530162125-2143AGUGGGGCUGGAACUGAAGCUUCAGUUCCAGCCCCACU
301730182126-2144GUGGGGCUGGAACUGAAGCGCUUCAGUUCCAGCCCCAC
301930202127-2145UGGGGCUGGAACUGAAGCCGGCUUCAGUUCCAGCCCCA
302130222147-2165CAUUCCUCAGCUGAGUGCAUGCACUCAGCUGAGGAAUG
302330242148-2166AUUCCUCAGCUGAGUGCAAUUGCACUCAGCUGAGGAAU
302530262149-2167UUCCUCAGCUGAGUGCAACGUUGCACUCAGCUGAGGAA
302730282150-2168UCCUCAGCUGAGUGCAACUAGUUGCACUCAGCUGAGGA
302930302151-2169CCUCAGCUGAGUGCAACUUAAGUUGCACUCAGCUGAGG
303130322152-2170CUCAGCUGAGUGCAACUUCGAAGUUGCACUCAGCUGAG
303330342153-2171UCAGCUGAGUGCAACUUCUAGAAGUUGCACUCAGCUGA
303530362154-2172CAGCUGAGUGCAACUUCUGCAGAAGUUGCACUCAGCUG
303730382155-2173AGCUGAGUGCAACUUCUGCGCAGAAGUUGCACUCAGCU
303930402156-2174GCUGAGUGCAACUUCUGCAUGCAGAAGUUGCACUCAGC
304130422157-2175CUGAGUGCAACUUCUGCAGCUGCAGAAGUUGCACUCAG
304330442158-2176UGAGUGCAACUUCUGCAGGCCUGCAGAAGUUGCACUCA
304530462159-2177GAGUGCAACUUCUGCAGGAUCCUGCAGAAGUUGCACUC
304730482160-2178AGUGCAACUUCUGCAGGAGCUCCUGCAGAAGUUGCACU
304930502161-2179GUGCAACUUCUGCAGGAGGCCUCCUGCAGAAGUUGCAC
305130522162-2180UGCAACUUCUGCAGGAGGCGCCUCCUGCAGAAGUUGCA
305330542163-2181GCAACUUCUGCAGGAGGCCGGCCUCCUGCAGAAGUUGC
305530562164-2182CAACUUCUGCAGGAGGCCAUGGCCUCCUGCAGAAGUUG
305730582165-2183AACUUCUGCAGGAGGCCACGUGGCCUCCUGCAGAAGUU
305930602166-2184ACUUCUGCAGGAGGCCACUAGUGGCCUCCUGCAGAAGU
306130622167-2185CUUCUGCAGGAGGCCACUGCAGUGGCCUCCUGCAGAAG
306330642168-2186UUCUGCAGGAGGCCACUGCGCAGUGGCCUCCUGCAGAA
306530662169-2187UCUGCAGGAGGCCACUGCAUGCAGUGGCCUCCUGCAGA
306730682170-2188CUGCAGGAGGCCACUGCAUAUGCAGUGGCCUCCUGCAG
306930702171-2189UGCAGGAGGCCACUGCAUUAAUGCAGUGGCCUCCUGCA
307130722172-2190GCAGGAGGCCACUGCAUUUAAAUGCAGUGGCCUCCUGC
307330742173-2191CAGGAGGCCACUGCAUUUUAAAAUGCAGUGGCCUCCUG
307530762174-2192AGGAGGCCACUGCAUUUUGCAAAAUGCAGUGGCCUCCU
307730782175-2193GGAGGCCACUGCAUUUUGAUCAAAAUGCAGUGGCCUCC
307930802176-2194GAGGCCACUGCAUUUUGAAUUCAAAAUGCAGUGGCCUC
308130822177-2195AGGCCACUGCAUUUUGAAGCUUCAAAAUGCAGUGGCCU
308330842178-2196GGCCACUGCAUUUUGAAGUACUUCAAAAUGCAGUGGCC
308530862179-2197GCCACUGCAUUUUGAAGUGCACUUCAAAAUGCAGUGGC
308730882180-2198CCACUGCAUUUUGAAGUGAUCACUUCAAAAUGCAGUGG
308930902181-2199CACUGCAUUUUGAAGUGAUAUCACUUCAAAAUGCAGUG
309130922182-2200ACUGCAUUUUGAAGUGAUGCAUCACUUCAAAAUGCAGU
309330942183-2201CUGCAUUUUGAAGUGAUGAUCAUCACUUCAAAAUGCAG
309530962184-2202UGCAUUUUGAAGUGAUGAGCUCAUCACUUCAAAAUGCA
309730982185-2203GCAUUUUGAAGUGAUGAGUACUCAUCACUUCAAAAUGC
309931002186-2204CAUUUUGAAGUGAUGAGUGCACUCAUCACUUCAAAAUG
310131022187-2205AUUUUGAAGUGAUGAGUGAUCACUCAUCACUUCAAAAU
310331042188-2206UUUUGAAGUGAUGAGUGAAUUCACUCAUCACUUCAAAA
310531062190-2208UUGAAGUGAUGAGUGAAAGCUUUCACUCAUCACUUCAA
310731082191-2209UGAAGUGAUGAGUGAAAGAUCUUUCACUCAUCACUUCA
310931102192-2210GAAGUGAUGAGUGAAAGAGCUCUUUCACUCAUCACUUC
311131122193-2211AAGUGAUGAGUGAAAGAGAUCUCUUUCACUCAUCACUU
311331142194-2212AGUGAUGAGUGAAAGAGAGCUCUCUUUCACUCAUCACU
311531162195-2213GUGAUGAGUGAAAGAGAGAUCUCUCUUUCACUCAUCAC
311731182196-2214UGAUGAGUGAAAGAGAGAAUUCUCUCUUUCACUCAUCA
311931202197-2215GAUGAGUGAAAGAGAGAAGCUUCUCUCUUUCACUCAUC
312131222198-2216AUGAGUGAAAGAGAGAAGUACUUCUCUCUUUCACUCAU
312331242199-2217UGAGUGAAAGAGAGAAGUCGACUUCUCUCUUUCACUCA
312531262200-2218GAGUGAAAGAGAGAAGUCCGGACUUCUCUCUUUCACUC
312731282201-2219AGUGAAAGAGAGAAGUCCUAGGACUUCUCUCUUUCACU
312931302202-2220GUGAAAGAGAGAAGUCCUAUAGGACUUCUCUCUUUCAC
313131322203-2221UGAAAGAGAGAAGUCCUAUAUAGGACUUCUCUCUUUCA
313331342204-2222GAAAGAGAGAAGUCCUAUUAAUAGGACUUCUCUCUUUC
313531362205-2223AAAGAGAGAAGUCCUAUUUAAAUAGGACUUCUCUCUUU
313731382206-2224AAGAGAGAAGUCCUAUUUCGAAAUAGGACUUCUCUCUU
313931402207-2225AGAGAGAAGUCCUAUUUCUAGAAAUAGGACUUCUCUCU
314131422208-2226GAGAGAAGUCCUAUUUCUCGAGAAAUAGGACUUCUCUC
314331442209-2227AGAGAAGUCCUAUUUCUCAUGAGAAAUAGGACUUCUCU
314531462210-2228GAGAAGUCCUAUUUCUCAGCUGAGAAAUAGGACUUCUC
314731482211-2229AGAAGUCCUAUUUCUCAGGCCUGAGAAAUAGGACUUCU
314931502212-2230GAAGUCCUAUUUCUCAGGCGCCUGAGAAAUAGGACUUC
315131522213-2231AAGUCCUAUUUCUCAGGCUAGCCUGAGAAAUAGGACUU
315331542214-2232AGUCCUAUUUCUCAGGCUUAAGCCUGAGAAAUAGGACU
315531562215-2233GUCCUAUUUCUCAGGCUUGCAAGCCUGAGAAAUAGGAC
315731582216-2234UCCUAUUUCUCAGGCUUGAUCAAGCCUGAGAAAUAGGA
315931602217-2235CCUAUUUCUCAGGCUUGAGCUCAAGCCUGAGAAAUAGG
316131622218-2236CUAUUUCUCAGGCUUGAGCGCUCAAGCCUGAGAAAUAG
316331642219-2237UAUUUCUCAGGCUUGAGCAUGCUCAAGCCUGAGAAAUA
316531662220-2238AUUUCUCAGGCUUGAGCAAUUGCUCAAGCCUGAGAAAU
316731682221-2239UUUCUCAGGCUUGAGCAAGCUUGCUCAAGCCUGAGAAA
316931702222-2240UUCUCAGGCUUGAGCAAGUACUUGCUCAAGCCUGAGAA
317131722223-2241UCUCAGGCUUGAGCAAGUUAACUUGCUCAAGCCUGAGA
317331742224-2242CUCAGGCUUGAGCAAGUUGCAACUUGCUCAAGCCUGAG
317531762225-2243UCAGGCUUGAGCAAGUUGGCCAACUUGCUCAAGCCUGA
317731782226-2244CAGGCUUGAGCAAGUUGGUACCAACUUGCUCAAGCCUG
317931802229-2247GCUUGAGCAAGUUGGUAUCGAUACCAACUUGCUCAAGC
318131822231-2249UUGAGCAAGUUGGUAUCUGCAGAUACCAACUUGCUCAA
318331842232-2250UGAGCAAGUUGGUAUCUGCGCAGAUACCAACUUGCUCA
318531862233-2251GAGCAAGUUGGUAUCUGCUAGCAGAUACCAACUUGCUC
318731882234-2252AGCAAGUUGGUAUCUGCUCGAGCAGAUACCAACUUGCU
318931902235-2253GCAAGUUGGUAUCUGCUCAUGAGCAGAUACCAACUUGC
319131922236-2254CAAGUUGGUAUCUGCUCAGCUGAGCAGAUACCAACUUG
319331942237-2255AAGUUGGUAUCUGCUCAGGCCUGAGCAGAUACCAACUU
319531962238-2256AGUUGGUAUCUGCUCAGGCGCCUGAGCAGAUACCAACU
319731982239-2257GUUGGUAUCUGCUCAGGCCGGCCUGAGCAGAUACCAAC
319932002240-2258UUGGUAUCUGCUCAGGCCUAGGCCUGAGCAGAUACCAA
320132022241-2259UGGUAUCUGCUCAGGCCUGCAGGCCUGAGCAGAUACCA
320332042242-2260GGUAUCUGCUCAGGCCUGAUCAGGCCUGAGCAGAUACC
320532062243-2261GUAUCUGCUCAGGCCUGAGCUCAGGCCUGAGCAGAUAC
320732082244-2262UAUCUGCUCAGGCCUGAGCGCUCAGGCCUGAGCAGAUA
320932102245-2263AUCUGCUCAGGCCUGAGCAUGCUCAGGCCUGAGCAGAU
321132122246-2264UCUGCUCAGGCCUGAGCAUAUGCUCAGGCCUGAGCAGA
321332142247-2265CUGCUCAGGCCUGAGCAUGCAUGCUCAGGCCUGAGCAG
321532162248-2266UGCUCAGGCCUGAGCAUGAUCAUGCUCAGGCCUGAGCA
321732182249-2267GCUCAGGCCUGAGCAUGACGUCAUGCUCAGGCCUGAGC
321932202250-2268CUCAGGCCUGAGCAUGACCGGUCAUGCUCAGGCCUGAG
322132222251-2269UCAGGCCUGAGCAUGACCUAGGUCAUGCUCAGGCCUGA
322332242252-2270CAGGCCUGAGCAUGACCUCGAGGUCAUGCUCAGGCCUG
322532262253-2271AGGCCUGAGCAUGACCUCAUGAGGUCAUGCUCAGGCCU
322732282279-2297CACUUAACCCCAGGCCAUUAAUGGCCUGGGGUUAAGUG
322932302280-2298ACUUAACCCCAGGCCAUUAUAAUGGCCUGGGGUUAAGU
323132322281-2299CUUAACCCCAGGCCAUUAUAUAAUGGCCUGGGGUUAAG
323332342282-2300UUAACCCCAGGCCAUUAUCGAUAAUGGCCUGGGGUUAA
323532362283-2301UAACCCCAGGCCAUUAUCAUGAUAAUGGCCUGGGGUUA
323732382284-2302AACCCCAGGCCAUUAUCAUAUGAUAAUGGCCUGGGGUU
323932402285-2303ACCCCAGGCCAUUAUCAUAUAUGAUAAUGGCCUGGGGU
324132422287-2305CCCAGGCCAUUAUCAUAUCGAUAUGAUAAUGGCCUGGG
324332442288-2306CCAGGCCAUUAUCAUAUCCGGAUAUGAUAAUGGCCUGG
324532462289-2307CAGGCCAUUAUCAUAUCCAUGGAUAUGAUAAUGGCCUG
324732482290-2308AGGCCAUUAUCAUAUCCAGCUGGAUAUGAUAAUGGCCU
324932502291-2309GGCCAUUAUCAUAUCCAGAUCUGGAUAUGAUAAUGGCC
325132522292-2310GCCAUUAUCAUAUCCAGAUAUCUGGAUAUGAUAAUGGC
325332542314-2332CUUCAGAGUUGUCUUUAUAUAUAAAGACAACUCUGAAG
325532562315-2333UUCAGAGUUGUCUUUAUAUAUAUAAAGACAACUCUGAA
325732582316-2334UCAGAGUUGUCUUUAUAUGCAUAUAAAGACAACUCUGA
325932602318-2336AGAGUUGUCUUUAUAUGUGCACAUAUAAAGACAACUCU
326132622322-2340UUGUCUUUAUAUGUGAAUUAAUUCACAUAUAAAGACAA
326332642323-2341UGUCUUUAUAUGUGAAUUAUAAUUCACAUAUAAAGACA
326532662324-2342GUCUUUAUAUGUGAAUUAAUUAAUUCACAUAUAAAGAC
326732682325-2343UCUUUAUAUGUGAAUUAAGCUUAAUUCACAUAUAAAGA
326932702326-2344CUUUAUAUGUGAAUUAAGUACUUAAUUCACAUAUAAAG
327132722327-2345UUUAUAUGUGAAUUAAGUUAACUUAAUUCACAUAUAAA
327332742328-2346UUAUAUGUGAAUUAAGUUAUAACUUAAUUCACAUAUAA
327532762329-2347UAUAUGUGAAUUAAGUUAUAUAACUUAAUUCACAUAUA
327732782330-2348AUAUGUGAAUUAAGUUAUAUAUAACUUAAUUCACAUAU
327932802331-2349UAUGUGAAUUAAGUUAUAUAUAUAACUUAAUUCACAUA
328132822332-2350AUGUGAAUUAAGUUAUAUUAAUAUAACUUAAUUCACAU
328332842333-2351UGUGAAUUAAGUUAUAUUAUAAUAUAACUUAAUUCACA
328532862334-2352GUGAAUUAAGUUAUAUUAAUUAAUAUAACUUAAUUCAC
328732882335-2353UGAAUUAAGUUAUAUUAAAUUUAAUAUAACUUAAUUCA
328932902336-2354GAAUUAAGUUAUAUUAAAUAUUUAAUAUAACUUAAUUC
329132922337-2355AAUUAAGUUAUAUUAAAUUAAUUUAAUAUAACUUAAUU
329332942338-2356AUUAAGUUAUAUUAAAUUUAAAUUUAAUAUAACUUAAU
329532962339-2357UUAAGUUAUAUUAAAUUUUAAAAUUUAAUAUAACUUAA
329732982340-2358UAAGUUAUAUUAAAUUUUAUAAAAUUUAAUAUAACUUA
329933002341-2359AAGUUAUAUUAAAUUUUAAUUAAAAUUUAAUAUAACUU
330133022342-2360AGUUAUAUUAAAUUUUAAUAUUAAAAUUUAAUAUAACU
330333042343-2361GUUAUAUUAAAUUUUAAUCGAUUAAAAUUUAAUAUAAC
330533062345-2363UAUAUUAAAUUUUAAUCUAUAGAUUAAAAUUUAAUAUA
330733082346-2364AUAUUAAAUUUUAAUCUAUAUAGAUUAAAAUUUAAUAU
330933102347-2365UAUUAAAUUUUAAUCUAUAUAUAGAUUAAAAUUUAAUA
331133122348-2366AUUAAAUUUUAAUCUAUAGCUAUAGAUUAAAAUUUAAU
331333142349-2367UUAAAUUUUAAUCUAUAGUACUAUAGAUUAAAAUUUAA
331533162350-2368UAAAUUUUAAUCUAUAGUAUACUAUAGAUUAAAAUUUA
331733182351-2369AAAUUUUAAUCUAUAGUAAUUACUAUAGAUUAAAAUUU
331933202354-2372UUUUAAUCUAUAGUAAAAAUUUUUACUAUAGAUUAAAA
332133222355-2373UUUAAUCUAUAGUAAAAACGUUUUUACUAUAGAUUAAA
332333242356-2374UUAAUCUAUAGUAAAAACAUGUUUUUACUAUAGAUUAA
332533262357-2375UAAUCUAUAGUAAAAACAUAUGUUUUUACUAUAGAUUA
332733282358-2376AAUCUAUAGUAAAAACAUAUAUGUUUUUACUAUAGAUU
332933302359-2377AUCUAUAGUAAAAACAUAGCUAUGUUUUUACUAUAGAU
333133322360-2378UCUAUAGUAAAAACAUAGUACUAUGUUUUUACUAUAGA
333333342361-2379CUAUAGUAAAAACAUAGUCGACUAUGUUUUUACUAUAG
333533362362-2380UAUAGUAAAAACAUAGUCCGGACUAUGUUUUUACUAUA
333733382363-2381AUAGUAAAAACAUAGUCCUAGGACUAUGUUUUUACUAU
333933402364-2382UAGUAAAAACAUAGUCCUGCAGGACUAUGUUUUUACUA
334133422365-2383AGUAAAAACAUAGUCCUGGCCAGGACUAUGUUUUUACU
334333442366-2384GUAAAAACAUAGUCCUGGAUCCAGGACUAUGUUUUUAC
334533462367-2385UAAAAACAUAGUCCUGGAAUUCCAGGACUAUGUUUUUA
334733482368-2386AAAAACAUAGUCCUGGAAAUUUCCAGGACUAUGUUUUU
334933502369-2387AAAACAUAGUCCUGGAAAUAUUUCCAGGACUAUGUUUU
335133522370-2388AAACAUAGUCCUGGAAAUAUAUUUCCAGGACUAUGUUU
335333542371-2389AACAUAGUCCUGGAAAUAAUUAUUUCCAGGACUAUGUU
335533562372-2390ACAUAGUCCUGGAAAUAAAUUUAUUUCCAGGACUAUGU
335733582373-2391CAUAGUCCUGGAAAUAAAUAUUUAUUUCCAGGACUAUG
335933602374-2392AUAGUCCUGGAAAUAAAUUAAUUUAUUUCCAGGACUAU
336133622375-2393UAGUCCUGGAAAUAAAUUCGAAUUUAUUUCCAGGACUA
336333642377-2395GUCCUGGAAAUAAAUUCUUAAGAAUUUAUUUCCAGGAC
336533662378-2396UCCUGGAAAUAAAUUCUUGCAAGAAUUUAUUUCCAGGA

The AIP mouse model (see Example 5) was used to investigate whether ALAS1 siRNA would work an an acute treatment paradigm to lower already elevated levels of ALA and PBG, as would be present, for example, when a human porphyria patient suffers from an acute attack. Administration of the AD-53558 LNP11 formulation siRNA at a 1 mg/kg dose 12 hours after the last dose of phenobarbitol rapidly decreased the levels of both ALA and PBG in mouse plasma, whereas in Luc control treated animals the levels continued to rise (FIG. 14). These results indicate that ALAS siRNA is effective for treating an acute attack. The ALAS1 siRNA was effective to lower and prevent further increases in ALA and PBG levels.

Further unmodified and modified siRNA sequences that target ALAS1 siRNA were designed and produced as described in Example 2. The in vitro activity of the modified duplexes was tested as described below.

For Hep3B, PMH, and primary Cynomolgus hepatocytes, transfection was carried out by adding 14.8 μl of Opti-MEM plus 0.2 μl of Lipofectamine RNAiMax per well (Invitrogen, Carlsbad CA. catalog number 13778-150) to 5 μl of each siRNA duplex to an individual well in a 96-well plate. The mixture was then incubated at room temperature for 20 minutes. Eighty μl of complete growth media without antibiotic containing the appropriate cell number were then added to the siRNA mixture. Cells were incubated for 24 hours prior to RNA purification.

Single dose experiments were performed at 1 uM, 500 nM, 20 nM, 10 nM and 0.2 nM final duplex concentration for GalNAc modified.

Cryopreserved Primary Cynomolgus Hepatocytes (Celsis In Vitro Technologies, M003055-P) were thawed at 37° C. water bath immediately prior to usage and re-suspended at 0.26×106 cells/ml in InVitroGRO CP (plating) medium (Celsis In Vitro Technologies, catalog number Z99029). During transfections, cells were plated onto a BD BioCoat 96 well collagen plate (BD, 356407) at 25,000 cells per well and incubated at 37° C. in an atmosphere of 5% CO2. Free Uptake experiments were performed by adding 10 μl of siRNA duplexes in PBS per well into a 96 well (96w) plate. Ninety 1l of complete growth media containing appropriate cell number for the cell type was then added to the siRNA. Cells were incubated for 24 hours prior to RNA purification. Single dose experiments were performed at 1 uM, 500 nM, 20 nM and 10 nM final duplex.

Total RNA Isolation Using DYNABEADS mRNA Isolation Kit (Invitrogen, Part #: 610-12)

Cells were harvested and lysed in 150 μl of Lysis/Binding Buffer then mixed for 5 minutes at 850 rpm using an Eppendorf Thermomixer (the mixing speed was the same throughout the process). Ten microliters of magnetic beads and 80 μl Lysis/Binding Buffer mixture were added to a round bottom plate and mixed for 1 minute. Magnetic beads were captured using a magnetic stand and the supernatant was removed without disturbing the beads. After removing the supernatant, the lysed cells were added to the remaining beads and mixed for 5 minutes. After removing the supernatant, magnetic beads were washed 2 times with 150 μl Wash Buffer A and mixed for 1 minute. The beads were captured again and the supernatant was removed. The beads were then washed with 150 μl Wash Buffer B, captured and the supernatant was removed. The beads were next washed with 150 μl Elution Buffer, captured and the supernatant removed. Finally, the beads were allowed to dry for 2 minutes. After drying, 50 l of Elution Buffer was added and mixed for 5 minutes at 70° C. The beads were captured on magnet for 5 minutes. Forty-five μl of supernatant was removed and added to another 96 well plate.

cDNA Synthesis Using ABI High Capacity cDNA Reverse Transcription Kit (Applied Biosystems, Foster City, CA, Cat #4368813)

A master mix of 2 μl 10×Buffer, 0.8 1 25×dNTPs, 2 μl Random primers, 1 μl Reverse Transcriptase, 1 μl RNase inhibitor and 3.2 μl of H2O per reaction as prepared. Equal volumes master mix and RNA were mixed for a final volume of 12 μl for in vitro screened or 20 μl for in vivo screened samples. cDNA was generated using a Bio-Rad C-1000 or S—1000 thermal cycler (Hercules, CA) through the following steps: 25° C. for 10 minutes, 37° C. for 120 minutes, 85° C. for 5 seconds, and 4° C. hold.

Two μl of cDNA were added to a master mix containing 2 μl of H2O, 0.5 μl GAPDH TaqMan Probe (Life Technologies catalog number 4326317E for Hep3B cells, catalog number 352339E for primary mouse hepatocytes or custom probe for cynomolgus primary hepatocytes), 0.5 μl C5 TaqMan probe (Life Technologies catalog number Hs00167441_ml for Hep3B cells or Mm00457879_ml for Primary Mouse Hepatoctyes or custom probe for cynomolgus primary hepatocytes) and 5 μl Lightcycler 480 probe master mix (Roche catalog number 04887301001) per well in a 384 well (384 w) plates (Roche catalog number 04887301001). Real time PCR was performed in an Roche LC480 Real Time PCR system (Roche) using the ΔΔCt(RQ) assay. For in vitro screening, each duplex was tested with two biological replicates unless otherwise noted and each Real Time PCR was performed in duplicate technical replicates. For in vivo screening, each duplex was tested in one or more experiments (3 mice per group) and each Real Time PCR was run in duplicate technical replicates.

To calculate relative fold change in ALAS1 mRNA levels, real time data were analyzed using the ΔΔCt method and normalized to assays performed with cells transfected with 10 nM AD-1955, or mock transfected cells. IC50s were calculated using a 4 parameter fit model using XLFit and normalized to cells transfected with AD-1955 over the same dose range, or to its own lowest dose.

The sense and antisense sequences of AD-1955 are:

SENSE:
(SEQ ID NO: 3682)
cuuAcGcuGAGuAcuucGAdTsdT
ANTISENSE:
(SEQ ID NO: 3683)
UCGAAGuACUcAGCGuAAGdTsdT.

The single strand and duplex sequences of the modified and unmodified siRNAs are provided in Table 14 and Table 15, respectively.

TABLE 14
Human ALAS1 Modified Single Strands and Duplex Sequences
SEQTarget
IDsites of
SEQ IDNO:antisense
NO:(anti-Duplexsequence
on NM_
(sense)sense)NameSense Sequence (5′-3′)Antisense Sequence (5′-3′)000688.4
33713372AD-58848CfsasUfgCfcAfaAfAfAfuGfgAfcAasUfsgAfuGfuCfcAfuuuUfuGfgCfaU1635-1657
fuCfaUfL96fgsAfsc
33733374AD-58849AfsusUfuUfgAfa*gfUfGfaUfgAfgUusUfsuCfaCfuCfaUfcacUfuCfaAfaA2189-2211
fgAfaAfL96fusGfsc
33753376AD-58850AfsgsUfuAfuAfuUfAfAfaUfuUfuAasGfsaUfuAfaAfaUfuuaAfuAfuAfaC2344-2366
faUfcUfL96fusUfsa
33773378AD-58851GfscsAfuUfuUfgAfAfGfuGfaUfgAusCfsaCfuCfaUfcAfcuuCfaAfaAfuG2187-2209
fgUfgAfL96fcsAfsg
33793380AD-58852GfsasAfcUfaAfuGfAfGfcAfgAfcAgsUfsuAfuGfuCfuGfcucAfuUfa*gfuU1975-1997
fuAfaCfL96fcsAfsu
33813382AD-58853AfsasUfgAfcCfaCfAfCfcUfaUfcGasAfscUfcGfaUfa*gfgugUfgGfuCfaU 973-995
fa*gfuUfL96fusCfsu
33833384AD-58854UfsasAfaUfuUfuAfAfUfcUfaUfa*gusUfsuAfcUfaUfa*gfauuAfaAfaUfuU2352-2374
fuAfaAfL96fasAfsu
33853386AD-58855UfsusCfa*gfuAfuGfAfUfcGfuUfuCcsAfsaAfgAfaAfcGfaucAfuAfcUfgA 929-951
fuUfuGfL96fasAfsa
33873388AD-58856CfsasCfuUfuUfcAfGfUfaUfgAfuCasAfsaCfgAfuCfaUfacuGfaAfaAfgU 924-946
fgUfuUfL96fgsGfsa
33893390AD-58857AfsasAfuCfuGfuUfUfCfcAfcUfuUcsUfsgAfaAfa*gfuGfgaaAfcAfgAfuU 913-935
fuCfa*gfL96fusUfsg
33913392AD-58858CfsasUfuUfgAfaAfCfUfgUfcCfaUusUfsgAfaUfgGfaCfa*guUfuCfaAfaU1478-1500
fuCfaAfL96fgsCfsc
33933394AD-58859CfscsUfaUfcGfa*gfUfUfuUfuAfaAcsAfsgUfuUfuAfaAfaacUfcGfaUfa*g 983-1005
faCfuGfL96fgsUfsg
33953396AD-58861GfsasCfcAfgAfaAfGfAfgUfgUfcUgsAfsuGfa*gfaCfaCfucuUfuCfuGfgU 872-894
fcAfuCfL96fcsUfsu
33973398AD-58862AfscsCfa*gfaAfa*gfAfGfuGfuCfuCasGfsaUfgAfgAfcAfcucUfuUfcUfgG 873-895
faUfcUfL96fusCfsu
33993400AD-58863AfscsUfaAfuGfa*gfCfAfgAfcAfuAasUfsgUfuAfuGfuCfugcUfcAfuUfa*g1977-1999
faCfaUfL96fusUfsc
34013402AD-58864UfsasGfuAfaAfaAfCfAfuAfgUfcCusCfscAfgGfaCfuAfuguUfuUfuAfcU2366-2388
fuGfgAfL96fasUfsa
34033404AD-58865UfsasUfuUfcUfgGfAfAfcUfa*gfuAasAfsuUfuAfcUfa*gfuucCfa*gfaAfaU1185-1207
faAfuUfL96fasUfsu
34053406AD-58867UfsusCfuGfcAfaAfGfCfcAfgUfcUcsUfscAfa*gfaCfuGfgcuUfuGfcAfgA 706-728
fuGfa*gfL96fasGfsa
34073408AD-58868GfsasGfgAfaAfgAfGfGfuUfgCfuGgsUfsuUfcAfgCfaAfccuCfuUfuCfcU 759-781
faAfaCfL96fcsAfsc
34093410AD-58869GfsgsUfaCfuAfgAfAfAfuAfuUfuCusCfscAfgAfaAfuAfuuuCfuAfgUfaC1174-1196
fuGfgAfL96fcsAfsc
34113412AD-58870GfsasCfaUfcAfuGfCfAfaAfa*gfcAusCfsuUfuGfcUfuUfugcAfuGfaUfgU 853-875
faAfgAfL96fcsCfsu
34133414AD-58871AfsasAfuUfuUfaAfUfCfuAfuAfgUusUfsuUfaCfuAfuAfgauUfaAfaAfuU2353-2375
faAfaAfL96fusAfsa
34153416AD-58873CfsasUfgAfuCfcAfAfGfgGfaUfuCusUfsuCfgAfaUfcCfcuuGfgAfuCfaU1362-1384
fgAfaAfL96fgsGfsa
34173418AD-58874AfsgsAfcCfa*gfaAfAfGfa*gfuGfuCasUfsgAfgAfcAfcUfcuuUfcUfgGfuC 871-893
fuCfaUfL96fusUfsu
34193420AD-58875AfsusCfcUfgAfa*gfAfGfcGfcUfgAusCfscCfuCfa*gfcGfcucUfuCfa*gfgA1810-1832
fgGfgAfL96fusCfsc
34213422AD-58876GfsusCfuGfuGfaUfGfAfaCfuAfaUgsCfsuCfaUfuAfgUfucaUfcAfcAfgA1966-1988
fgAfgCfL96fcsUfsu
34233424AD-58877CfsasGfaAfa*gfa*gfUfGfuCfuCfaUgsAfsaGfaUfgAfgAfcacUfcUfuUfcU 875-897
fcUfuCfL96fgsGfsu
34253426AD-58878AfscsUfuUfuCfa*gfUfAfuGfaUfcGgsAfsaAfcGfaUfcAfuacUfgAfaAfa*g 925-947
fuUfuCfL96fusGfsg
34273428AD-58879UfscsAfuGfcCfaAfAfAfaUfgGfaCusGfsaUfgUfcCfaUfuuuUfgGfcAfuG1634-1656
faUfcAfL96fasCfsu
34293430AD-58880AfsasUfaUfuUfcUfGfGfaAfcUfa*gusUfsuAfcUfa*gfuUfccaGfaAfaUfaU1183-1205
fuAfaAfL96fusUfsc
34313432AD-58881CfsusUfcUfuCfaAfGfAfuAfaCfuUusGfsgCfaAfgUfuAfucuUfgAfa*gfaA 892-914
fgCfcAfL96fgsAfsu
34333434AD-58882UfsusUfcAfgUfaUfGfAfuCfgUfuUasAfsaGfaAfaCfgAfucaUfaCfuGfaA 928-950
fcUfuUfL96fasAfsg
34353436AD-58883CfscsCfa*gfuGfuGfGfUfuAfgUfgUusUfsuCfaCfaCfuAfaccAfcAfcUfgG 790-812
fgAfaAfL96fgsGfsc
34373438AD-58884GfscsUfgUfgAfgAfUfUfuAfcUfcUasAfsuCfa*gfa*gfuAfaauCfuCfaCfa*g1325-1347
fgAfuUfL96fcsCfsu
34393440AD-58885AfsgsGfcUfuGfa*gfCfAfa*gfuUfgGgsAfsuAfcCfaAfcUfugcUfcAfa*gfcC2229-2251
fuAfuCfL96fusGfsa
34413442AD-58886GfsasAfa*gfa*gfuGfUfCfuCfaUfcUasAfsgAfa*gfaUfgAfgacAfcUfcUfuU 877-899
fuCfuUfL96fcsUfsg
34433444AD-58887AfsusUfuCfuGfgAfAfCfuAfgUfaAgsAfsaUfuUfaCfuAfguuCfcAfgAfaA1186-1208
faUfuCfL96fusAfsu
34453446AD-58888UfsgsUfgAfuGfuGfGfCfcCfaUfgAasAfsaCfuCfaUfgGfgccAfcAfuCfaC1531-1553
fgUfuUfL96fasCfsa
34473448AD-58889AfsasGfa*gfa*gfaAfGfUfcCfuAfuUgsAfsgAfaAfuAfgGfacuUfcUfcUfcU2208-2230
fuCfuCfL96fusUfsc
34493450AD-58890UfsgsGfcAfgCfaCfAfGfaUfgAfaUusCfsuGfaUfuCfaUfcugUfgCfuGfcC 671-693
fcAfgAfL96fasGfsg
34513452AD-58891AfsusGfaUfcGfuUfUfCfuUfuGfa*gusUfsuUfcUfcAfaAfgaaAfcGfaUfcA 935-957
faAfaAfL96fusAfsc
34533454AD-58892UfscsUfgGfaAfcUfAfGfuAfaAfuUasUfsgGfaAfuUfuAfcuaGfuUfcCfa*g1189-1211
fcCfaUfL96fasAfsa
34553456AD-59095GfscsCfcAfuUfcUfUfAfuCfcCfgAasCfsuCfgGfgAfuAfa*gaAfuGfgsgsc 360-382
fgUfL96
34573458AD-59096GfsgsAfaCfcAfuGfCfCfuCfcAfuGasUfscAfuGfgAfgGfcauGfgUfuscsc1347-1369
faUfL96
34593460AD-59097UfsgsGfa*gfuCfuGfUfGfcGfgAfuCasGfsgAfuCfcGfcAfcagAfcUfcscsa1794-1816
fcUfL96
34613462AD-59098CfsasCfcCfaCfgGfGfUfgUfgUfgGusCfscCfaCfaCfaCfccgUfgGfgsusg1112-1134
fgAfL96
34633464AD-59099GfsgsAfgUfcUfgUfGfCfgGfaUfcCusAfsgGfaUfcCfgCfacaGfaCfuscsc1795-1817
fuAfL96
34653466AD-59100CfsasAfaAfcUfgCfCfCfcAfa*gfaUusCfsaUfcUfuGfgGfgcaGfuUfususg 428-450
fgAfL96
34673468AD-59101GfscsCfuCfcAfuGfAfUfcCfaAfgGusCfscCfuUfgGfaUfcauGfgAfgsgsc1355-1377
fgAfL96
34693470AD-59102CfsasUfcAfuCfcCfUfGfuGfcGfgGasAfscCfcGfcAfcAfgggAfuGfasusg1921-1943
fuUfL96
34713472AD-59103AfscsCfcAfcGfgGfUfGfuGfuGfgGusCfscCfcAfcAfcAfcccGfuGfgsgsu1113-1135
fgAfL96
34733474AD-59104CfsasCfaUfcAfuCfCfCfuGfuGfcGusCfscGfcAfcAfgGfgauGfaUfgsusg1919-1941
fgAfL96
34753476AD-59105CfsasGfaAfa*gfa*gfUfGfuCfuCfaUasGfsaUfgAfgAfcAfcucUfuUfcsusg 873-895
fcUfL96
34773478AD-59106CfscsUfcCfaUfgAfUfCfcAfa*gfgGasUfscCfcUfuGfgAfucaUfgGfasgsg1356-1378
faUfL96
34793480AD-59107UfsgsCfcCfaUfuCfUfUfaUfcCfcGusUfscGfgGfaUfaAfgaaUfgGfgscsa 359-381
faAfL96
34813482AD-59108CfsusUfcAfcCfcUfGfGfcUfaAfgAusAfsuCfuUfa*gfcCfa*ggGfuGfasasg1297-1319
fuAfL96
34833484AD-59109AfsusCfaUfcCfcUfGfUfgCfgGfgUusAfsaCfcCfgCfaCfa*ggGfaUfgsasu1922-1944
fuAfL96
34853486AD-59110AfsgsAfaAfgAfgUfGfUfcUfcAfuCasAfsgAfuGfa*gfaCfacuCfuUfuscsu 874-896
fuUfL96
34873488AD-59111CfsusCfcAfuGfaUfCfCfaAfgGfgAasAfsuCfcCfuUfgGfaucAfuGfgsasg1357-1379
fuUfL96
34893490AD-59112CfscsAfuUfcUfuAfUfCfcCfgAfgUusGfsaCfuCfgGfgAfuaaGfaAfusgsg 362-384
fcAfL96
34913492AD-59113CfsasCfcCfuGfgCfUfAfa*gfaUfgAusAfsuCfaUfcUfuAfgccAfgGfgsusg1300-1322
fuAfL96
34933494AD-59114UfscsAfuCfcCfuGfUfGfcGfgGfuUusCfsaAfcCfcGfcAfcagGfgAfusgsa1923-1945
fgAfL96
34953496AD-59115AfsasGfa*gfuGfuCfUfCfaUfcUfuCasAfsgAfa*gfaUfgAfgacAfcUfcsusu 877-899
fuUfL96
34973498AD-59116GfsusCfaUfgCfcAfAfAfaAfuGfgAusGfsuCfcAfuUfuUfuggCfaUfgsasc1631-1653
fcAfL96
34993500AD-59117CfsasUfuCfuUfaUfCfCfcGfa*gfuCusGfsgAfcUfcGfgGfauaAfgAfasusg 363-385
fcAfL96
35013502AD-59118AfscsCfcUfgGfcUfAfAfgAfuGfaUusCfsaUfcAfuCfuUfa*gcCfa*gfgsgsu1301-1323
fgAfL96
35033504AD-59119CfsusCfuUfcAfcCfCfUfgGfcUfaAusCfsuUfa*gfcCfa*gfgguGfaAfgsasg1295-1317
fgAfL96
35053506AD-59120AfsusGfcCfaAfaAfAfUfgGfaCfaUusGfsaUfgUfcCfaUfuuuUfgGfcsasu1634-1656
fcAfL96
35073508AD-59121UfsgsCfcCfcAfa*gfAfUfgAfuGfgAasUfsuCfcAfuCfaUfcuuGfgGfgscsa 434-456
faUfL96
35093510AD-59122GfsasAfcCfaUfgCfCfUfcCfaUfgAusAfsuCfaUfgGfa*gfgcaUfgGfususc1348-1370
fuAfL96
35113512AD-59123UfscsUfuCfaCfcCfUfGfgCfuAfa*gasUfscUfuAfgCfcAfgggUfgAfasgsa1296-1318
faUfL96
35133514AD-59124UfsgsCfcAfaAfaAfUfGfgAfcAfuCasUfsgAfuGfuCfcAfuuuUfuGfgscsa1635-1657
faUfL96
35153516AD-59125CfscsAfgAfaAfgAfGfUfgUfcUfcAusAfsuGfa*gfaCfaCfucuUfuCfusgsg 872-894
fuAfL96
35173518AD-59126GfsasAfaCfuGfuCfCfAfuUfcAfaUusCfsaUfuGfaAfuGfgacAfgUfususc1481-1503
fgAfL96
35193520AD-59127UfscsAfcCfcUfgGfCfUfaAfgAfuGasUfscAfuCfuUfa*gfccaGfgGfusgsa1299-1321
faUfL96
35213522AD-59128CfscsCfuGfgAfgUfCfUfgUfgCfgGasUfscCfgCfaCfa*gfacuCfcAfgsgsg1791-1813
faUfL96
35233524AD-59129GfsasAfa*gfa*gfuGfUfCfuCfaUfcUusAfsaGfaUfgAfgAfcacUfcUfususc 875-897
fuAfL96
35253526AD-59130UfsgsGfa*gfcCfcUfGfGfa*gfuCfuGusAfscAfgAfcUfcCfa*ggGfcUfcscsa1786-1808
fuAfL96
TABLE 15
Human ALASl Unmodified Single Strands and Duplex Sequences
Target
SEQsites of
SEQ IDID NO:antisense  
NO:(anti-DuplexSense SequenceAntisense Sequencesequence on
(sense)sense)Name(5′-3′)(5′-3′)NM_000688.4
36843527AD-58848CAUGCCAAAAAUGGACAUCAUAUGAUGUCCAUUUUUGGCAUGAC1635-1657
35283529AD-58849AUUUUGAAGUGAUGAGUGAAAUUUCACUCAUCACUUCAAAAUGC2189-2211
35303531AD-58850AGUUAUAUUAAAUUUUAAUCUAGAUUAAAAUUUAAUAUAACUUA2344-2366
35323533AD-58851GCAUUUUGAAGUGAUGAGUGAUCACUCAUCACUUCAAAAUGCAG2187-2209
35343535AD-58852GAACUAAUGAGCAGACAUAACGUUAUGUCUGCUCAUUAGUUCAU1975-1997
35363537AD-58853AAUGACCACACCUAUCGAGUUAACUCGAUAGGUGUGGUCAUUCU 973-995
35383539AD-58854UAAAUUUUAAUCUAUAGUAAAUUUACUAUAGAUUAAAAUUUAAU2352-2374
35403541AD-58855UUCAGUAUGAUCGUUUCUUUGCAAAGAAACGAUCAUACUGAAAA 929-951
35423543AD-58856CACUUUUCAGUAUGAUCGUUUAAACGAUCAUACUGAAAAGUGGA 924-946
35443545AD-58857AAAUCUGUUUCCACUUUUCAGCUGAAAAGUGGAAACAGAUUUUG 913-935
35463547AD-58858CAUUUGAAACUGUCCAUUCAAUUGAAUGGACAGUUUCAAAUGCC1478-1500
35483549AD-58859CCUAUCGAGUUUUUAAAACUGCAGUUUUAAAAACUCGAUAGGUG 983-1005
35503551AD-58861GACCAGAAAGAGUGUCUCAUCGAUGAGACACUCUUUCUGGUCUU 872-894
35523553AD-58862ACCAGAAAGAGUGUCUCAUCUAGAUGAGACACUCUUUCUGGUCU 873-895
35543555AD-58863ACUAAUGAGCAGACAUAACAUAUGUUAUGUCUGCUCAUUAGUUC1977-1999
35563557AD-58864UAGUAAAAACAUAGUCCUGGAUCCAGGACUAUGUUUUUACUAUA2366-2388
35583559AD-58865UAUUUCUGGAACUAGUAAAUUAAUUUACUAGUUCCAGAAAUAUU1185-1207
35603561AD-58867UUCUGCAAAGCCAGUCUUGAGCUCAAGACUGGCUUUGCAGAAGA 706-728
35623563AD-58868GAGGAAAGAGGUUGCUGAAACGUUUCAGCAACCUCUUUCCUCAC 759-781
35643565AD-58869GGUACUAGAAAUAUUUCUGGAUCCAGAAAUAUUUCUAGUACCAC1174-1196
35663567AD-58870GACAUCAUGCAAAAGCAAAGAUCUUUGCUUUUGCAUGAUGUCCU 853-875
35683569AD-58871AAAUUUUAAUCUAUAGUAAAAUUUUACUAUAGAUUAAAAUUUAA2353-2375
35703571AD-58873CAUGAUCCAAGGGAUUCGAAAUUUCGAAUCCCUUGGAUCAUGGA1362-1384
35723573AD-58874AGACCAGAAAGAGUGUCUCAUAUGAGACACUCUUUCUGGUCUUU 871-893
35743575AD-58875AUCCUGAAGAGCGCUGAGGGAUCCCUCAGCGCUCUUCAGGAUCC1810-1832
35763577AD-58876GUCUGUGAUGAACUAAUGAGCGCUCAUUAGUUCAUCACAGACUU1966-1988
35783579AD-58877CAGAAAGAGUGUCUCAUCUUCGAAGAUGAGACACUCUUUCUGGU 875-897
35803581AD-58878ACUUUUCAGUAUGAUCGUUUCGAAACGAUCAUACUGAAAAGUGG 925-947
35823583AD-58879UCAUGCCAAAAAUGGACAUCAUGAUGUCCAUUUUUGGCAUGACU1634-1656
35843585AD-58880AAUAUUUCUGGAACUAGUAAAUUUACUAGUUCCAGAAAUAUUUC1183-1205
35863587AD-58881CUUCUUCAAGAUAACUUGCCAUGGCAAGUUAUCUUGAAGAAGAU 892-914
35883589AD-58882UUUCAGUAUGAUCGUUUCUUUAAAGAAACGAUCAUACUGAAAAG 928-950
35903591AD-58883CCCAGUGUGGUUAGUGUGAAAUUUCACACUAACCACACUGGGGC 790-812
35923593AD-58884GCUGUGAGAUUUACUCUGAUUAAUCAGAGUAAAUCUCACAGCCU1325-1347
35943595AD-58885AGGCUUGAGCAAGUUGGUAUCGAUACCAACUUGCUCAAGCCUGA2229-2251
35963597AD-58886GAAAGAGUGUCUCAUCUUCUUAAGAAGAUGAGACACUCUUUCUG 877-899
35983599AD-58887AUUUCUGGAACUAGUAAAUUCGAAUUUACUAGUUCCAGAAAUAU1186-1208
36003601AD-58888UGUGAUGUGGCCCAUGAGUUUAAACUCAUGGGCCACAUCACACA1531-1553
36023603AD-58889AAGAGAGAAGUCCUAUUUCUCGAGAAAUAGGACUUCUCUCUUUC2208-2230
36043605AD-58890UGGCAGCACAGAUGAAUCAGAUCUGAUUCAUCUGUGCUGCCAGG 671-693
36063607AD-58891AUGAUCGUUUCUUUGAGAAAAUUUUCUCAAAGAAACGAUCAUAC 935-957
36083609AD-58892UCUGGAACUAGUAAAUUCCAUAUGGAAUUUACUAGUUCCAGAAA1189-1211
36103611AD-59095GCCCAUUCUUAUCCCGAGUACUCGGGAUAAGAAUGGGC 360-382
36123613AD-59096GGAACCAUGCCUCCAUGAUAUCAUGGAGGCAUGGUUCC1347-1369
36143615AD-59097UGGAGUCUGUGCGGAUCCUAGGAUCCGCACAGACUCCA1794-1816
36163617AD-59098CACCCACGGGUGUGUGGGAUCCCACACACCCGUGGGUG1112-1134
36183619AD-59099GGAGUCUGUGCGGAUCCUAUAGGAUCCGCACAGACUCC1795-1817
36203621AD-59100CAAAACUGCCCCAAGAUGAUCAUCUUGGGGCAGUUUUG 428-450
36223623AD-59101GCCUCCAUGAUCCAAGGGAUCCCUUGGAUCAUGGAGGC1355-1377
36243625AD-59102CAUCAUCCCUGUGCGGGUUAACCCGCACAGGGAUGAUG1921-1943
36263627AD-59103ACCCACGGGUGUGUGGGGAUCCCCACACACCCGUGGGU1113-1135
36283629AD-59104CACAUCAUCCCUGUGCGGAUCCGCACAGGGAUGAUGUG1919-1941
36303631AD-59105CAGAAAGAGUGUCUCAUCUAGAUGAGACACUCUUUCUG 873-895
36323633AD-59106CCUCCAUGAUCCAAGGGAUAUCCCUUGGAUCAUGGAGG1356-1378
36343635AD-59107UGCCCAUUCUUAUCCCGAAUUCGGGAUAAGAAUGGGCA 359-381
36363637AD-59108CUUCACCCUGGCUAAGAUAUAUCUUAGCCAGGGUGAAG1297-1319
36383639AD-59109AUCAUCCCUGUGCGGGUUAUAACCCGCACAGGGAUGAU1922-1944
36403641AD-59110AGAAAGAGUGUCUCAUCUUAAGAUGAGACACUCUUUCU 874-896
36423643AD-59111CUCCAUGAUCCAAGGGAUUAAUCCCUUGGAUCAUGGAG1357-1379
36443645AD-59112CCAUUCUUAUCCCGAGUCAUGACUCGGGAUAAGAAUGG 362-384
36463647AD-59113CACCCUGGCUAAGAUGAUAUAUCAUCUUAGCCAGGGUG1300-1322
36483649AD-59114UCAUCCCUGUGCGGGUUGAUCAACCCGCACAGGGAUGA1923-1945
36503651AD-59115AAGAGUGUCUCAUCUUCUUAAGAAGAUGAGACACUCUU 877-899
36523653AD-59116GUCAUGCCAAAAAUGGACAUGUCCAUUUUUGGCAUGAC1631-1653
36543655AD-59117CAUUCUUAUCCCGAGUCCAUGGACUCGGGAUAAGAAUG 363-385
36563657AD-59118ACCCUGGCUAAGAUGAUGAUCAUCAUCUUAGCCAGGGU1301-1323
36583659AD-59119CUCUUCACCCUGGCUAAGAUCUUAGCCAGGGUGAAGAG1295-1317
36603661AD-59120AUGCCAAAAAUGGACAUCAUGAUGUCCAUUUUUGGCAU1634-1656
36623663AD-59121UGCCCCAAGAUGAUGGAAUAUUCCAUCAUCUUGGGGCA 434-456
36643665AD-59122GAACCAUGCCUCCAUGAUAUAUCAUGGAGGCAUGGUUC1348-1370
36663667AD-59123UCUUCACCCUGGCUAAGAUAUCUUAGCCAGGGUGAAGA1296-1318
36683669AD-59124UGCCAAAAAUGGACAUCAUAUGAUGUCCAUUUUUGGCA1635-1657
36703671AD-59125CCAGAAAGAGUGUCUCAUAUAUGAGACACUCUUUCUGG 872-894
36723673AD-59126GAAACUGUCCAUUCAAUGAUCAUUGAAUGGACAGUUUC1481-1503
36743675AD-59127UCACCCUGGCUAAGAUGAUAUCAUCUUAGCCAGGGUGA1299-1321
36763677AD-59128CCCUGGAGUCUGUGCGGAUAUCCGCACAGACUCCAGGG1791-1813
36783679AD-59129GAAAGAGUGUCUCAUCUUAUAAGAUGAGACACUCUUUC 875-897
36803681AD-59130UGGAGCCCUGGAGUCUGUAUACAGACUCCAGGGCUCCA1786-1808

The results of the in vitro assays are provided in Table 16. Table 16 also notes the target species of each of the siRNAs.

TABLE 16
Results of Functional Assays
Cyno Free UptakeCyno TransfectionHep3b Transfection
Target1 uM20 nM20 nM0.2 nM10 nM0.1 nM
Duplex IDSpeciesTypeAvg500 nMAvg10 nMAvgAvgAvgAvg
AD-58848M/R/Rh/H21/23131.6176.0104.4128.043.544.825.376.8
AD-58849H/Rh21/2391.988.192.2105.029.435.411.547.1
AD-58850H/Rh21/2379.4103.480.0111.2NA62.231.372.0
AD-58851H/Rh21/2399.774.794.8104.7NA40.78.681.3
AD-58852H/Rh21/23108.191.8103.3111.9101.1128.843.4129.0
AD-58853H/Rh21/2374.867.784.293.524.752.914.161.2
AD-58854H/Rh21/23145.9124.1106.6115.3119.083.985.084.0
AD-58855H/Rh21/2381.597.992.7101.839.540.315.367.6
AD-58856H/Rh21/2374.190.684.682.622.430.78.733.3
AD-58857H/Rh21/2364.791.462.387.122.031.69.8106.3
AD-58858H/Rh21/2367.491.768.698.327.940.317.444.8
AD-58859H/Rh21/2371.277.292.490.119.134.313.139.7
AD-58861H/Rh21/23104.6107.2102.0100.625.935.118.069.8
AD-58862H/Rh21/2366.877.068.788.520.331.124.249.9
AD-58863H/Rh21/2370.866.876.898.521.529.78.754.9
AD-58864H/Rh21/2376.285.683.7100.860.461.056.487.3
AD-58865H/Rh21/2367.977.995.998.421.338.615.581.4
AD-58867H/Rh21/2395.993.3107.097.532.342.716.679.8
AD-58868H/Rh21/2395.292.1116.294.754.669.261.5105.9
AD-58869H/Rh21/2365.078.275.888.217.425.013.063.9
AD-58870H/Rh21/2369.492.381.088.129.243.833.779.1
AD-58871H/Rh21/2361.277.388.277.071.273.236.7110.3
AD-58873H/Rh21/2395.2100.983.394.654.252.836.673.3
AD-58874H/Rh21/2375.876.863.885.322.331.215.038.2
AD-58875H/Rh21/2380.788.778.697.948.673.661.290.6
AD-58876H/Rh21/2390.893.182.5100.241.156.921.258.7
AD-58877H/Rh21/2368.385.151.278.718.546.611.927.4
AD-58878H/Rh21/2378.368.381.291.224.123.46.237.1
AD-58879H/Rh21/2387.994.179.795.432.047.815.782.5
AD-58880H/Rh21/2374.972.288.988.120.127.514.060.7
AD-58881H/Rh21/2385.976.878.8118.022.236.727.671.6
AD-58882H/Rh21/2354.153.460.385.814.627.28.223.8
AD-58883H/Rh21/2380.469.975.780.331.825.812.363.0
AD-58884H/Rh21/2357.755.364.878.220.030.011.868.9
AD-58885H/Rh21/23101.891.8104.1101.585.971.961.871.2
AD-58886M/R/Rh/H21/2347.158.036.393.316.026.69.232.0
AD-58887H/Rh21/2373.698.782.695.228.533.512.865.2
AD-58888H/Rh21/2390.269.969.485.646.945.016.672.0
AD-58889H/Rh21/2383.698.682.492.236.540.331.699.4
AD-58890H/Rh21/2369.595.484.288.250.845.621.792.9
AD-58891H/Rh21/2362.875.775.4109.223.634.315.655.8
AD-58892H/Rh21/2360.292.989.892.922.843.320.275.6
AD-59095M/R/Rh/H19mer88.9NA132.8NA48.397.454.399.0
AD-59096M/R/Rh/H19mer95.5NA90.5NA105.7138.6131.4120.7
AD-59097M/R/Rh/H19mer92.5NA84.2NA75.0NA94.7108.5
AD-59098M/R/Rh/H19mer84.0NA87.7NA109.3NA130.087.3
AD-59099M/R/Rh/H19mer89.7NA90.0NA77.885.446.874.9
AD-59100M/R/Rh/H19mer84.8NA144.3NA70.6108.191.5117.6
AD-59101M/R/Rh/H19mer79.0NA103.8NA89.8102.9124.2107.0
AD-59102M/R/Rh/H19mer85.9NA100.6NA72.268.587.995.1
AD-59103M/R/Rh/H19mer86.0NA91.1NA93.081.3130.096.0
AD-59104M/R/Rh/H19mer92.6NA96.9NA94.991.4124.483.1
AD-59105M/R/Rh/H19mer48.9NA101.7NA18.448.917.034.7
AD-59106M/R/Rh/H19mer63.2NA76.7NA28.540.728.646.4
AD-59107M/R/Rh/H19mer71.4NA68.7NA37.145.326.863.6
AD-59108M/R/Rh/H19mer70.7NA85.1NA89.984.8139.2101.7
AD-59109M/R/Rh/H19mer86.1NA83.4NA84.996.2131.786.7
AD-59110M/R/Rh/H19mer70.8NA119.7NA38.560.467.480.3
AD-59111M/R/Rh/H19mer66.1NA76.5NA52.261.069.787.6
AD-59112M/R/Rh/H19mer71.2NA80.2NA91.283.4127.489.0
AD-59113M/R/Rh/H19mer67.0NA77.8NA49.159.066.891.4
AD-59114M/R/Rh/H19mer81.7NA79.3NA96.388.0129.672.4
AD-59115M/R/Rh/H19mer40.4NA69.6NA19.635.79.316.9
AD-59116M/R/Rh/H19mer72.2NA78.3NA53.577.870.1107.8
AD-59117M/R/Rh/H19mer70.7NA75.6NA75.874.9129.0103.5
AD-59118M/R/Rh/H19mer68.8NA75.9NA81.482.1114.189.7
AD-59119M/R/Rh/H19mer64.9NA86.5NA85.1125.1122.8124.8
AD-59120M/R/Rh/H19mer63.5NA75.1NA29.952.016.154.1
AD-59121M/R/Rh/H19mer67.6NA72.0NA88.877.4108.0103.1
AD-59122M/R/Rh/H19mer60.2NA62.3NA25.145.316.254.8
AD-59123M/R/Rh/H19mer68.6NA108.2NA59.284.680.097.7
AD-59124M/R/Rh/H19mer47.5NA56.5NA23.940.09.818.9
AD-59125M/R/Rh/H19mer45.4NA47.2NA15.240.714.715.1
AD-59126M/R/Rh/H19mer64.3NA74.6NA51.657.135.554.4
AD-59127M/R/Rh/H19mer103.4NA105.8NA94.0156.4135.9113.7
AD-59128M/R/Rh/H19mer102.4NA81.4NA66.389.360.274.9
AD-59129M/R/Rh/H19mer41.3NA38.8NA17.941.48.612.6
AD-59130M/R/Rh/H19mer58.3NA80.8NA94.978.3106.788.0

Table 17 illustrates the C50s of select ALAS1 siRNA duplexes. The IC50s were determined from the knockdown of endogenously expressed ALAS1 in the Hep3B cell line, at 24 hours following transfection of each ALAS1 modified siRNA duplex (see Table 14). At least seven duplexes, including AD-58882, AD-58878, AD-58886, AD-58877, AD-59115, AD-58856, and AD-59129, consistently demonstrated IC50s of less than 0.1 nm, indicating that these duplexes were particularly effective in suppressing ALAS1 expression.

TABLE 17
IC50S of select ALAS1 siRNA duplexes
Duplex ID384w IC50 (nM)96w IC50 (nM)
AD-588820.0080.014
AD-588780.0400.031
AD-588860.0370.033
AD-588770.0310.034
AD-591150.0930.052
AD-588560.0610.066
AD-591290.0850.071
AD-591240.5720.078
AD-588740.1400.102
AD-591250.1180.115
AD-591050.5110.144
AD-59120180.5920.498
AD-5912236.6460.646
AD-591067.9060.847
AD-59126n/a1.014
AD-59107n/a1.971

Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific embodiments of the invention described herein. Such equivalents are intended to be encompassed by the following claims.

COMPOSITIONS AND METHODS FOR INHIBITING EXPRESSION OF THE ALAS1 GENE (2024)
Top Articles
Imvu Historical Room Viewer
Ultimate Guide To Getting Ess Swissport Pay Stubs And W2s For A Current And Former Employee - Wikiaccounting
The Largest Banks - ​​How to Transfer Money With Only Card Number and CVV (2024)
Jackerman Mothers Warmth Part 3
Hannaford Weekly Flyer Manchester Nh
Cash4Life Maryland Winning Numbers
My Boyfriend Has No Money And I Pay For Everything
360 Training Alcohol Final Exam Answers
AB Solutions Portal | Login
Rubfinder
Jesus Revolution Showtimes Near Chisholm Trail 8
Max 80 Orl
Everything You Need to Know About Holly by Stephen King
Flower Mound Clavicle Trauma
Hoe kom ik bij mijn medische gegevens van de huisarts? - HKN Huisartsen
Finger Lakes Ny Craigslist
Jinx Chapter 24: Release Date, Spoilers & Where To Read - OtakuKart
使用 RHEL 8 时的注意事项 | Red Hat Product Documentation
Petco Vet Clinic Appointment
Faurot Field Virtual Seating Chart
Busted Newspaper Fauquier County Va
Iroquois Amphitheater Louisville Ky Seating Chart
Melendez Imports Menu
Putin advierte que si se permite a Ucrania usar misiles de largo alcance, los países de la OTAN estarán en guerra con Rusia - BBC News Mundo
Ticket To Paradise Showtimes Near Cinemark Mall Del Norte
Effingham Daily News Police Report
Tim Steele Taylorsville Nc
Puffin Asmr Leak
91 Octane Gas Prices Near Me
What are the 7 Types of Communication with Examples
Ravens 24X7 Forum
Sf Bay Area Craigslist Com
How To Make Infinity On Calculator
Blackstone Launchpad Ucf
Rocketpult Infinite Fuel
Unlock The Secrets Of "Skip The Game" Greensboro North Carolina
Ny Post Front Page Cover Today
ATM Near Me | Find The Nearest ATM Location | ATM Locator NL
Greater Keene Men's Softball
Myql Loan Login
Latest Nigerian Music (Next 2020)
2700 Yen To Usd
Why I’m Joining Flipboard
Lake Andes Buy Sell Trade
Carroll White Remc Outage Map
Sdn Fertitta 2024
Panolian Batesville Ms Obituaries 2022
The Great Brian Last
Powah: Automating the Energizing Orb - EnigmaticaModpacks/Enigmatica6 GitHub Wiki
The Ultimate Guide To 5 Movierulz. Com: Exploring The World Of Online Movies
Lux Nails & Spa
Latest Posts
Article information

Author: Geoffrey Lueilwitz

Last Updated:

Views: 6105

Rating: 5 / 5 (80 voted)

Reviews: 87% of readers found this page helpful

Author information

Name: Geoffrey Lueilwitz

Birthday: 1997-03-23

Address: 74183 Thomas Course, Port Micheal, OK 55446-1529

Phone: +13408645881558

Job: Global Representative

Hobby: Sailing, Vehicle restoration, Rowing, Ghost hunting, Scrapbooking, Rugby, Board sports

Introduction: My name is Geoffrey Lueilwitz, I am a zealous, encouraging, sparkling, enchanting, graceful, faithful, nice person who loves writing and wants to share my knowledge and understanding with you.