Compositions And Their Uses Directed To Hepcidin

MONIA; BRETT P. ;   et al.

Patent Application Summary

U.S. patent application number 13/294068 was filed with the patent office on 2012-05-10 for compositions and their uses directed to hepcidin. This patent application is currently assigned to ISIS PHARMACEUTICALS, INC.. Invention is credited to C. FRANK BENNETT, WILLIAM A. GAARDE, TRISHA LOCKHART, ROBERT MCKAY, BRETT P. MONIA.

Application Number20120115930 13/294068
Document ID /
Family ID38610239
Filed Date2012-05-10

United States Patent Application 20120115930
Kind Code A1
MONIA; BRETT P. ;   et al. May 10, 2012

COMPOSITIONS AND THEIR USES DIRECTED TO HEPCIDIN

Abstract

Disclosed herein are compounds, compositions and methods for modulating the expression of hepcidin in a cell, tissue or animal or preventing, ameliorating or treating anemia. Also provided are methods for prevention, amelioration or treatment of anemia, and for increasing red blood cell count in an animal. Also provided are methods for the prevention, amelioration and/or treatment of low serum iron levels, low red blood cell count and other clinical endpoints of anemia in an animal. These methods may be achieved by administration of compounds or compositions including antisense compounds targeted to a nucleic acid that expresses hepcidin polypeptide combined with an erythropoiesis stimulating agent.


Inventors: MONIA; BRETT P.; (ENCINITAS, CA) ; BENNETT; C. FRANK; (CARLSBAD, CA) ; GAARDE; WILLIAM A.; (CARLSBAD, CA) ; LOCKHART; TRISHA; (CARDIFF BY THE SEA, CA) ; MCKAY; ROBERT; (POWAY, CA)
Assignee: ISIS PHARMACEUTICALS, INC.
CARLSBAD
CA

Family ID: 38610239
Appl. No.: 13/294068
Filed: November 10, 2011

Related U.S. Patent Documents

Application Number Filing Date Patent Number
11734562 Apr 12, 2007 8076306
13294068
60792004 Apr 12, 2006
60821638 Aug 7, 2006
60865833 Nov 14, 2006

Current U.S. Class: 514/44A ; 536/24.5
Current CPC Class: C12N 2310/346 20130101; C12N 2310/321 20130101; C12N 2310/341 20130101; A61P 7/00 20180101; C12N 2310/3341 20130101; C12N 15/113 20130101; C12N 2310/315 20130101; C12N 2310/321 20130101; C12N 2310/11 20130101; C12N 2310/3525 20130101; A61P 7/06 20180101
Class at Publication: 514/44.A ; 536/24.5
International Class: A61K 31/711 20060101 A61K031/711; A61P 7/06 20060101 A61P007/06; A61K 31/7125 20060101 A61K031/7125; C07H 21/04 20060101 C07H021/04; A61K 31/712 20060101 A61K031/712

Claims



1. A compound comprising a modified oligonucleotide consisting of 12 to 35 linked nucleosides and having a nucleobase sequence comprising at least 8 contiguous nucleobases complementary to an equal length portion of a nucleobase sequence of a nucleic acid encoding hepcidin, wherein said modified oligonucleotide specifically hybridizes to said nucleic acid encoding hepcidin.

2. The compound of claim 1, wherein said nucleobase sequence of a nucleic acid encoding hepcidin is selected from the group consisting of SEQ ID NOs: 1, 2 and 3.

3. The compound of claim 2, wherein said nucleobase sequence of a nucleic acid encoding hepcidin is SEQ ID NO: 1.

4. The compound of claim 3, wherein said modified oligonucleotide has a nucleobase sequence comprising at least 8 contiguous nucleobases complementary to an equal length portion of nucleotides 123-566 of SEQ ID NO: 1.

5. The compound of claim 3, wherein said modified oligonucleotide has a nucleobase sequence comprising at least 8 contiguous nucleobases complementary to an equal length portion of nucleotides 442-541 of SEQ ID NO: 1.

6. The compound of claim 3, wherein said compound consists of a single-stranded modified oligonucleotide.

7. The compound of claim 1, wherein the nucleobase sequence of the modified oligonucleotide is at least 90% complementary to at least a 20 contiguous nucleobase portion of nucleotides 123-566 of SEQ ID NO: 1.

8. The compound of claim 6, wherein the nucleobase sequence of the modified oligonucleotide is 100% complementary to at least a 20 contiguous nucleobase portion of nucleotides 123-566 of SEQ ID NO: 1.

9. The compound of claim 6, wherein at least one internucleoside linkage of said modified oligonucleotide is a modified internucleoside linkage.

10. The compound of claim 9, wherein each internucleoside linkage of said modified oligonucleotide is a phosphorothioate internucleoside linkage.

11. The compound of claim 6, wherein at least one nucleoside of said modified oligonucleotide comprises a modified sugar.

12. The compound of claim 11, wherein at least one modified sugar is a bicyclic sugar.

13. The compound of claim 11, wherein at least one modified sugar is selected from the group consisting of a 2'-O-(2-methoxyethyl), and a 4'-(CH.sub.2).sub.2--O-2' bridge, wherein n is 1 or 2.

14. The compound of claim 6, wherein at least one nucleoside of said modified oligonucleotide comprises a modified nucleobase.

15. The compound of claim 14, wherein said modified nucleobase is a 5-methylcytosine.

16. The compound of claim 3, wherein said modified oligonucleotide comprises: a gap segment consisting of linked deoxynucleosides; a 5' wing segment consisting of linked nucleosides; and a 3' wing segment consisting of linked nucleosides; wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment and wherein each nucleoside of each wing segment comprises a modified sugar.

17. The compound of claim 16, wherein said modified oligonucleotide comprises: a gap segment consisting of ten linked deoxynucleosides; a 5' wing segment consisting of five linked nucleosides; and a 3' wing segment consisting of five linked nucleosides; wherein the gap segment is positioned between the 5' wing segment and the 3' wing segment, wherein each nucleoside of each wing segment comprises a 2'-O-methoxyethyl sugar, wherein each cytosine in said modified oligonucleotide is a 5-methylcytosine, and wherein each internucleoside linkage of said modified oligonucleotide is a phosphorothioate linkage.

18. The compound of claim 17, wherein said modified oligonucleotide consists of 20 linked nucleosides.

19. The compound of claim 6, wherein said modified oligonucleotide consists of 20 linked nucleosides.

20. A composition comprising the compound of claim 3 or a salt thereof and a pharmaceutically acceptable carrier or diluent.

21. The composition of claim 20, wherein said compound consists of a single-stranded modified oligonucleotide.

22. The composition of claim 20, wherein said modified oligonucleotide consists of 20 linked nucleosides.

23. The compound of claim 1, wherein said modified oligonucleotide has a nucleobase sequence comprising at least 8 contiguous nucleobases complementary to an equal length portion of a nucleotide region selected from the group consisting of nucleotides 123-566, 123-252, 295-566, 352-405, 429-566, and 456-527 of SEQ ID NO: 1, 204-341 and 231-302 of SEQ ID NO: 2, and 3249-4743, 3249-3454, 3398-3454, 4643-4703, and 4643-4743 of SEQ ID NO: 3.

24. The compound of claim 1, wherein said modified oligonucleotide has a nucleobase sequence comprising at least 8 contiguous nucleobases of a nucleobase sequence selected from the group consisting of SEQ ID NOs: 23-37.

25. The compound of claim 1, wherein said modified oligonucleotide has a nucleobase sequence consisting of a nucleobase sequence selected from the group consisting of SEQ ID NOs: 23-37.
Description



CROSS-REFERENCE TO RELATED APPLICATIONS

[0001] This application is a divisional of U.S. application Ser. No. 11/734,562, filed Apr. 12, 2007, which claims priority under 35 U.S.C. .sctn.119(e) to U.S. Provisional Application No. 60/792,004, filed Apr. 12, 2006, U.S. Provisional Application No. 60/821,638, filed Aug. 7, 2006 and U.S. Provisional Application No. 60/865,833, filed Nov. 14, 2006. Each of the above applications is herein incorporated by reference in its entirety.

SEQUENCE LISTING

[0002] The present application is being filed along with a Sequence Listing in electronic format. The Sequence Listing is provided as a file entitled BIOL0082USSEQ.txt, created on Nov. 9, 2011, which is 42 Kb in size. The information in the electronic format of the sequence listing is incorporated herein by reference in its entirety.

BACKGROUND OF THE INVENTION

[0003] Anemia is characterized by a lower than normal number of red blood cells (erythrocytes) in the blood, usually measured by a decrease in the amount of hemoglobin. The cause of anemia can include chronic inflammation, chronic kidney disease, kidney dialysis treatment, genetic disorders, chronic infection, acute infection, cancer and cancer treatments. Altered iron homeostasis and/or erythropoiesis in these conditions can result in decreased erythrocyte (red blood cell) production. Anemia can limit the use of chemotherapeutic agents during cancer treatment. Symptoms of anemia include fatigue, pallor and poor exercise tolerance. Though rarely life threatening, anemia can be severely debilitating and difficult to treat. Clinical signs of anemia include low serum iron (hypoferremia), low hemoglobin levels, low hematocrit levels, decreased red blood cells, decreased reticulocytes, increased soluble transferrin receptor and iron restricted erythropoesis.

[0004] In some cases, increasing dietary iron or intravenous iron delivery are used to treat anemia. Erythropoietin (EPO) stimulates erythroprogenitors and promotes red blood cell formation. Recombinant erythropoietin and other erythropoiesis stimulating agents (ESAs) are used for treating anemia, although certain patients respond poorly to this treatment. EPO is well known and is commercially available through Amgen (Thousand Oaks, Calif.).

[0005] Anemia of chronic disease (ACD) is a highly prevalent, inflammatory-driven disorder that is poorly treated with currently available therapies. The mechanism underlying ACD is chronic inflammation, which results in changes in iron homeostasis and utilization, resulting in a blunting of erythroid progenitor cell proliferation and red cell function.

[0006] ACD is associated with increased production of inflammatory cytokines, including, for example, tumor necrosis factor-.alpha., IL-1 .beta., IL-6, and interferon-.gamma. (Means (1995) Stem cells 13:32-37 and Means (1999) Int J Hematol 70:7-12). In several in vitro and in vivo animal model systems, inflammatory cytokines negatively affected the ability to mediate erythropoietin (EPO) production, EPO responsiveness, and the coordinate regulation of iron metabolism (Roodman et al. (1989) Adv Exp Med Biol 271:185-196; Fuchs et al. (1991) Eur J Hematol 46:65-70; Jelkmann et al. (1994) Ann NY Acad Sci 718:300-311; Vannucchi et al. (1994) Br J Hematol 87:18-23; and Oldenburg et al. (2001) Aliment Pharmacol Ther 15:429-438). Administration of EPO failed to reverse anemia in mice continuously exposed to TNF-.alpha. (Clibon et al. (1990) Exp Hematol 18:438-441). Increased levels of inflammatory cytokines, such as TNF-.alpha., IL-1 .beta., and INF-.gamma., contribute to defective EPO production and EPO resistance observed in patients with anemia of chronic disease (Jelkmann et al. (1991) Ann NY Acad Sci 718:300-311 and Macdougall and Cooper (2002) Neprol Dial Transplant 17 (11):39-43.). Therefore, various cytokines, e.g., inflammatory cytokines and cytokines associated with inflammation, are involved in many aspects of the pathogenesis of anemia of chronic disease, including inhibition of erythroid progenitors, inhibition of EPO production, and impairment of iron release and iron availability for erythropoiesis.

[0007] Hepcidin is an 8 kD polypeptide that is produced by hepatocytes in response to inflammation or to rising levels of iron in the blood. The primary role of hepcidin is to regulate blood iron levels by facilitating a decrease in these blood iron levels. Hepcidin binds with and down regulates ferroportin to reduce ferroportin mediated release of iron into the blood. Hepcidin expression is increased in conditions of acute and chronic inflammation resulting in decreased iron availability for erythropoeisis. Hepcidin is frequently measured in the urine or serum as a biomarker of anemia status. (Ganz et al. (2006) Am J Physiol Gastrointest Liver Physiol 290:G199-G203; and Ganz (2003) Blood 102 (3):783-788). Moreover, hepcidin over expression has been strongly linked to ACD mechanistically as a mediator of this disorder in animal models and in humans.

SUMMARY OF THE INVENTION

[0008] Provided herein are compounds, particularly oligomeric compounds, especially nucleic acid and nucleic acid-like oligomers, which are targeted to a nucleic acid encoding hepcidin. Preferably, the oligomeric compounds are antisense compounds targeted to a nucleic acid molecule encoding hepcidin, particularly human hepcidin (GenBank Accession Nos. BM719679.1, entered Mar. 1, 2002; NM.sub.--021175.2, entered Jul. 23, 2004; and nucleotides 7819907 to 7825131 of NT.sub.--011196.11, entered Jan. 5, 2003; all herein incorporated by reference and assigned SEQ ID NOS: 1, 2 and 3, respectively), that modulate the expression of hepcidin. In a particular embodiment, the antisense compounds are antisense oligonucleotides. In another embodiment, the antisense compounds are siRNAs. In one embodiment, the compounds comprise at least an 8 nucleobase portion, preferably at least a 12 nucleobase portion, more preferably at least a 15 nucleobase portion, of the sequences listed in Table 3. In a further embodiment, the compounds are at least 80% identical to the sequences listed in or below Table 3.

[0009] Herein, active target segments are identified for SEQ ID NOS: 1, 2 and 3. A series of antisense oligonucleotide compounds was designed to target one or more of SEQ ID NOS: 1, 2, and 3. Active target segments were then identified as being regions of the hepcidin mRNA associated with the most active antisense oligonucleotides. Compounds are provided that are targeted to an active target segment of SEQ ID NO: 1, 2, or 3 and modulate the expression of hepcidin. In one embodiment, the compounds comprise at least an 8 nucleobase portion, preferably at least a 12 nucleobase portion, more preferably at least a 15 nucleobase portion, that is complementary to an active target segment. In a further embodiment, the compounds are at least 80% complementary to an active target segments. In another embodiment, the compounds are 13 to 30 nucleobases in length. In another embodiment, the compounds are at least about 80% complementary to at least a 20 nucleobase portion of the active target segment. In yet another embodiment, the compounds are complementary to at least an 8 nucleobase portion of the active target segment.

[0010] In another aspect, the compounds comprise at least one modified internucleoside linkage, modified sugar moiety, or modified nucleobase. In another aspect, the compounds comprise a chimeric oligonucleotide. In one aspect, the compounds comprise a phosphorothioate linkage. In another aspect, the compounds comprise a 2'-MOE modification. In another aspect, the compounds comprise a 5-methylcytosine modification.

[0011] Another embodiment provides for a composition comprising erythropoietin and an antisense compound targeted to a nucleic acid encoding hepcidin. In one aspect, the compound is an antisense oligonucleotide targeted to a nucleic acid molecule encoding hepcidin

[0012] A further embodiment provides methods for prevention, amelioration or treatment of anemia in an individual comprising administering to the individual erythropoietin and an antisense compound targeted to an nucleic acid encoding hepcidin. In one aspect, the compound is an antisense oligonucleotide targeted to a nucleic acid molecule encoding hepcidin.

[0013] In one embodiment, the antisense oligonucleotide used in this method causes an increase in serum iron levels, prevention of iron restriction of erythropoiesis, or a combination thereof. In yet another embodiment, the administration causes an increase in red blood cell counts, reticulocyte counts, hemoglobin levels, hematocrit levels, or a combination thereof.

[0014] In one aspect, the cause of anemia is chronic inflammation, chronic kidney disease, kidney dialysis treatment, genetic disorders, chronic infection, acute infection, cancer, or cancer treatments. In another aspect, the anemia is associated with inflammation. In another aspect, it is anemia of chronic disease (ACD).

[0015] In another embodiment, the individual receiving the administration has received or has been continuously receiving erythropoietin prior to treatment. Another embodiment provides methods for prevention, amelioration or treatment of anemia in an individual who is being treated with erythropoietin comprising administering to the individual erythropoietin and an antisense oligonucleotide targeted to a nucleic acid molecule encoding hepcidin.

[0016] In another embodiment, the administration comprises delivery of the antisense oligonucleotide and erythropoietin in a single formation. In one aspect, the delivery of the single formulation is by injection. In another embodiment, the administration comprises delivery of the antisense oligonucleotide and erythropoietin in separate formulations. In one aspect delivery of separate formulations is by injection. In another aspect, separate formulations are delivered at distinct timepoints. In another aspect, they are delivered simultaneously.

[0017] Another embodiment provides a method for increasing red blood cell count in an animal that is being treated with erythropoietin comprising administering to the animal an oligomeric compound targeted to a nucleic acid encoding hepcidin. In one aspect, the compound is an antisense oligonucleotide targeted to a nucleic acid molecule encoding hepcidin.

[0018] Another embodiment provides a method for increasing red blood cell count in an animal comprising the steps of delivering an erythropoietin therapy and delivering an antisense oligonucleotide therapy targeted to a nucleic acid molecule encoding hepcidin.

[0019] Another embodiment provides methods for prevention, amelioration or treatment of anemia in an individual comprising administering to the individual antisense compound targeted to an nucleic acid encoding hepcidin, wherein the individual is further receiving erythropoietin therapy. In one aspect, the erythropoietin therapy had occurred or was continuously occurring at the time of administration of the antisense oligonucleotide. In another aspect the individual is therapy naive, meaning that the individual had not before received or was not receiving erythropoietin therapy at the initiation of the method. In one embodiment, the method further comprises administering to the individual erythropoietin. In one embodiment, the antisense compound and erythropoietin are administered in a single formulation. In another embodiment, they are administered in separate formulations. In one embodiment, the separate formulations are administered simultaneously. In another aspect the separate formulations are administered at distinct times. In one aspect, the formulations are administered by injection.

[0020] Another embodiment provides methods for increasing red blood cells in an individual antisense compound targeted to an nucleic acid encoding hepcidin, wherein the individual is further receiving erythropoietin therapy. In one aspect, the erythropoietin therapy had occurred or was continuously occurring at the time of administration of the antisense oligonucleotide. In another aspect the individual is therapy naive, meaning that the individual had not before received or was not receiving erythropoietin therapy at the initiation of the method. In one embodiment, the method further comprises administering to the individual erythropoietin. In one embodiment, the antisense compound and erythropoietin are administered in a single formulation. In another embodiment, they are administered in separate formulations. In one embodiment, the separate formulations are administered simultaneously. In another aspect the separate formulations are administered at distinct times. In one aspect, the formulations are administered by injection.

[0021] Also provided herein are methods for providing a combination therapy for prevention, treatment or amelioration of anemia or increasing red blood cells in an animal comprising the step of administering to the animal an antisense compound disclosed herein, wherein the animal is further receiving erythropoietin therapy. In one embodiment, the animal has been receiving erythropoietin therapy prior to the administration of the antisense compound. In another embodiment the animal begins erythropoietin therapy simultaneously with the administration of the antisense compound. In one embodiment, the combination therapy comprises receiving the antisense compound and an erythropoietin therapy compound in a single formulation. In another embodiment, the combination therapy comprises receiving the antisense compound and an erythropoietin therapy compound in separate formulations. In one embodiment, recombinant human erythropoietin is administered as the erythropoietin therapy.

[0022] Also provided herein is a use of an oligomeric compound targeted to a nucleic acid encoding hepcidin for the preparation of a medicament for the prevention, amelioration, and/or treatment of anemia. In one aspect, the compound is an antisense oligonucleotide targeted to a nucleic acid encoding hepcidin. Another embodiment provides for the use of an oligomeric composition targeted to a nucleic acid encoding hepcidin for the preparation of a medicament for the prevention, amelioration, and/or treatment of anemia. In one aspect the composition comprises an antisense oligonucleotide targeted to a nucleic acid encoding hepcidin and erythropoietin. In one aspect, the medicament is prepared for treatment of anemia in a patient being treated with erythropoietin. In one aspect, the cause of anemia is chronic inflammation, chronic kidney disease, kidney dialysis treatment, genetic disorders, chronic infection, acute infection, cancer or cancer treatments. In another embodiment, the anemia is anemia of chronic disease (ACD). In one embodiment, the medicament is formulated for delivery by injection.

BRIEF DESCRIPTION OF THE FIGURES

[0023] FIG. 1. Western blot of liver lysate following treatment with antisense oligonucleotides targeting hepcidin mRNA and showing that the hepcidin polypeptide is reduced following treatment. Hepcidin was detected with hepcidin antibody (Alpha Diagnostics, HEPC11-A, 0.3 .micro.g/ml).

[0024] FIG. 2. Graph illustrating that delivery of 10-75 mg/kg of antisense oligonucleotide targeting hepcidin mRNA reduces hepcidin expression and in turn leads to an increase in serum iron levels in normal mice.

[0025] FIG. 3. Graph illustrating that antisense oligonucleotide reduction of hepcidin increases serum iron levels in both basal and LPS states in mice (N=4, SEM).

[0026] FIGS. 4a-d. Graphs illustrating responses to treatment with a compound that reduces the expression of mRNA encoding hepcidin for a number of clinical endpoints for anemia: a) serum levels; b) red blood cell count; c) hemoglobin levels; and d) hematocrit levels.

[0027] FIG. 5 (a-c). Graphs illustrating the enhanced reticulocyte counts in turpentine-treated mice receiving erythropoietin and an antisense oligonucleotide targeted to hepcidin.

DETAILED DESCRIPTION OF THE INVENTION

[0028] Anemia is a severely debilitating condition stemming from a lower than normal number of red blood cells (erythrocytes) in the blood that characterizes a number of diseases and conditions. Therapeutic interventions for these diseases or conditions are not completely satisfactory due to lack of efficacy and/or unwanted side effects of the compounds. Provided herein are methods, uses, compositions and compounds for the prevention, amelioration, and /or treatment of anemia. As used herein, the term "prevention" means to delay or forestall onset or development of a condition or disease for a period of time from hours to days, preferably weeks to months. As used herein, the term "amelioration" means a lessening of at least one indicator of the severity of a condition or disease. By way of example, some appropriate indicators include low serum iron (hypoferremia), low hemoglobin or hematocrit levels, decreased red blood cell or reticulocyte numbers, increased soluble transferrin receptor, iron restricted erythropoiesis; symptoms include fatigue, pallor and poor exercise tolerance. The severity of indicators may be determined by subjective or objective measures which are known to those skilled in the art. As used herein, "treatment" or "therapy" means to administer a composition of antisense compounds, erythropoietin (or other ESA), or of antisense compounds combined with erythropoietin (or other ESA) to effect an alteration or improvement of the disease or condition; the antisense compounds being targeted to a nucleic acid encoding hepcidin. Prevention, amelioration, and/or treatment may require administration of multiple doses at regular intervals, or prior to exposure to an agent (e.g., a chemotherapeutic) to alter the course of the condition or disease. Moreover, a single agent may be used in a single individual for each prevention, amelioration, and treatment of a condition or disease sequentially or concurrently. Moreover, the compounds and compositions disclosed herein may be administered or delivered to a patient who is receiving a particular therapy, compound, agent or composition. In this case, the receiving can occur prior to the method of administering or delivering, or it may occur simultaneously or following the administration or delivery of the compound or composition. Additionally, regardless of when the receiving begins relative to administration or delivery, it may continue after the administration or delivery.

[0029] Disclosed herein are antisense compounds, including antisense oligonucleotides, for use in modulating the expression of nucleic acid molecules encoding hepcidin. This modulation is accomplished by providing antisense compounds that are designed to target and hybridize with one or more target nucleic acid molecules encoding hepcidin (SEQ ID NOS: 1, 2 or 3.). As used herein, the terms "target nucleic acid" and "nucleic acid molecule encoding hepcidin" have been used for convenience to encompass RNA (including pre-mRNA and mRNA or portions thereof) transcribed from DNA encoding hepcidin and also cDNA derived from such RNA. In a preferred embodiment, the target nucleic acid is an mRNA encoding hepcidin.

Target Nucleic Acids

[0030] "Targeting" refers to the design and selection of antisense compounds capable of hybridizing to or complementary to a particular target nucleic acid molecule. Antisense compounds selected by this process are said to be "targeted to" the target nucleic acid molecule. Targeting can be a multistep process that usually begins with the identification of a target nucleic acid whose expression is to be modulated. For example, the target nucleic acid can be a cellular gene (or mRNA transcribed from the gene) whose expression is associated with a particular disorder or disease state, or a nucleic acid molecule from an infectious agent. As disclosed herein, the target nucleic acid encodes hepcidin.

Variants

[0031] It is also known in the art that alternative RNA transcripts can be produced from the same genomic region of DNA. These alternative transcripts are generally known as "variants." More specifically, "pre-mRNA variants" are transcripts produced from the same genomic DNA that differ from other transcripts produced from the same genomic DNA in either their start or stop position and contain both intronic and exonic sequence. Variants can result in mRNA variants including, but not limited to, those with alternate splice junctions, or alternate initiation and termination codons. Variants in genomic and mRNA sequences can result in disease. Antisense compounds targeted to such variants are within the scope of the oligomeric compounds as described herein.

Target Names, Synonyms, Features

[0032] Accordingly, there are provided compositions and methods for modulating the expression of hepcidin. "Hepcidin" is also referred to as hepcidin antimicrobial peptide; HAMP; HAMP1; HEPC; HFE2; LEAP-1; LEAP1; and liver-expressed antimicrobial peptide. Herein the term "hepcidin" is used. Table 1 lists the GenBank accession numbers of sequences corresponding to nucleic acid molecules encoding hepcidin (nt=nucleotide), the date the version of the sequence was entered in GenBank, and the corresponding SEQ ID NO in the instant application, when assigned. Each sequence is incorporated herein by reference.

TABLE-US-00001 TABLE 1 Gene Targets SEQ ID Species Genbank # Genbank Date NO Human BM719679.1 Mar. 1, 2002 1 Human NM_021175.2 Jul. 23, 2004 2 Human nucleotides 7819907 to 7825131 of Jan. 5, 2003 3 NT_011196.11 Mouse NM_032541.1 May 28, 2001 4 Mouse the complement of nucleotides Feb. 24, 2003 5 3978217 to 3980665 of NT_039413.1

Modulation of Target Expression

[0033] Modulation of expression of a target nucleic acid can be achieved through alteration of any number of nucleic acid functions. "Modulation" means a perturbation of function, for example, either an increase (stimulation or induction) or a decrease (inhibition or reduction) in expression. As another example, modulation of expression can include perturbing splice site selection of pre-mRNA processing. "Expression" includes all the functions by which a gene's coded information is converted into structures present and operating in a cell. These structures include the products of transcription and translation. "Modulation of expression" means the perturbation of such functions. The functions of RNA to be modulated can include translocation functions, which include, but are not limited to, translocation of the RNA to a site of protein translation, translocation of the RNA to sites within the cell which are distant from the site of RNA synthesis, and translation of protein from the RNA. RNA processing functions that can be modulated include, but are not limited to, splicing of the RNA to yield one or more RNA species, capping of the RNA, 3' maturation of the RNA and catalytic activity or complex formation involving the RNA which may be engaged in or facilitated by the RNA. Modulation of expression can result in the increased level of one or more nucleic acid species or the decreased level of one or more nucleic acid species, either temporally or by net steady state level. One result of such interference with target nucleic acid function is modulation of the expression of hepcidin. Thus, in one embodiment modulation of expression can mean increase or decrease in target RNA or protein levels. In another embodiment modulation of expression can mean an increase or decrease of one or more RNA splice products, or a change in the ratio of two or more splice products.

[0034] The effect of antisense compounds on target nucleic acid expression can be tested in any of a variety of cell types provided that the target nucleic acid is present at measurable levels. The effect of antisense compounds on target nucleic acid expression can be routinely determined using, for example, PCR, Northern blot analysis or measurement of hepcidin polypeptide in the urine or serum of an animal (e.g., ELISA or Western blots). Cell lines are derived from both normal tissues and cell types and from cells associated with various disorders (e.g. hyperproliferative disorders). Cell lines derived from multiple tissues and species can be obtained from American Type Culture Collection (ATCC, Manassas, Va.) and other public sources, and are well known to those skilled in the art. Primary cells, or those cells which are isolated from an animal and not subjected to continuous culture, can be prepared according to methods known in the art, or obtained from various commercial suppliers. Additionally, primary cells include those obtained from donor human subjects in a clinical setting (i.e. blood donors, surgical patients). Primary cells are prepared by methods known in the art.

Assaying Modulation of Expression

[0035] Modulation of hepcidin expression can be assayed in a variety of ways known in the art. Hepcidin mRNA levels can be quantitated by, e.g., Northern blot analysis, competitive polymerase chain reaction (PCR), or real-time PCR. RNA analysis can be performed on total cellular RNA or poly(A)+ mRNA by methods known in the art. Methods of RNA isolation are taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.1.1-4.2.9 and 4.5.1-4.5.3, John Wiley & Sons, Inc., 1993.

[0036] Northern blot analysis is routine in the art and is taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 1, pp. 4.2.1-4.2.9, John Wiley & Sons, Inc., 1996. Real-time quantitative (PCR) can be conveniently accomplished using the commercially available ABI PRISM.TM. 7700 Sequence Detection System, available from PE-Applied Biosystems, Foster City, Calif. and used according to manufacturer's instructions. The method of analysis of modulation of RNA levels is not a limitation of the instant description.

[0037] Levels of a protein encoded by hepcidin can be quantitated in a variety of ways well known in the art, such as immunoprecipitation, Western blot analysis (immunoblotting), ELISA or fluorescence-activated cell sorting (FACS). Antibodies directed to a protein encoded by hepcidin can be identified and obtained from a variety of sources, such as the MSRS catalog of antibodies (Aerie Corporation, Birmingham, Mich.), or can be prepared via conventional antibody generation methods. Methods for preparation of polyclonal antisera are taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.12.1-11.12.9, John Wiley & Sons, Inc., 1997. Preparation of monoclonal antibodies is taught in, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 11.4.1-11.11.5, John Wiley & Sons, Inc., 1997.

[0038] Immunoprecipitation methods are standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.16.1-10.16.11, John Wiley & Sons, Inc., 1998. Western blot (immunoblot) analysis is standard in the art and can be found at, for example, Ausubel, F. M. et al., Current Protocols in Molecular Biology, Volume 2, pp. 10.8.1-10.8.21, John Wiley & Sons, Inc., 1997.

Active Target Segments

[0039] The locations on the target nucleic acid defined by having one or more active antisense compounds targeted thereto are referred to as "active target segments." When an active target segment is validated by multiple antisense compounds, the compounds are preferably separated by no more than about 30 nucleotides on the active target segment, more preferably no more than about 10 nucleotides on the target sequence, even more preferably the compounds are contiguous, and most preferably the compounds are overlapping. In a preferred embodiment, at least 50%, preferably at least 70% of the oligonucleotides targeted to the active target segment modulate expression of their target RNA at least 40%. There may be substantial variation in activity (e.g., as defined by percent reduction or activity ranking) of each antisense compounds within an active target segment. Active antisense compounds are those that modulate the expression of their target RNA. Active antisense compounds reduce expression of their target RNA at least 10%, preferably at least 20%. In other embodiments 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%, at least 95% or at least 99%. In a more preferred embodiment, the level of reduction required to define an active antisense compound is based on the results from the screen used to define the active target segments.

Hybridization

[0040] As used herein, "hybridization" means the pairing of complementary strands of antisense compounds to their target sequence. While not limited to a particular mechanism, the most common mechanism of pairing involves hydrogen bonding, which may be Watson-Crick, Hoogsteen or reversed Hoogsteen hydrogen bonding, between complementary nucleoside or nucleotide bases (nucleobases). For example, the natural base adenine is complementary to the natural nucleobases thymidine and uracil which pair through the formation of hydrogen bonds. The natural base guanine is complementary to the natural base 5-methylcytosine and the artificial base known as a G-clamp. Hybridization can occur under varying circumstances.

[0041] An antisense compound is specifically hybridizable when there is a sufficient degree of complementarity to avoid non-specific binding of the antisense compound to non-target nucleic acid sequences under conditions in which specific binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assays are performed in the case of in vitro assays.

[0042] As used herein, "stringent hybridization conditions" or "stringent conditions" refers to conditions under which an antisense compound will hybridize to its target sequence, but to a minimal number of other sequences. Stringent conditions are sequence-dependent and will be different in different circumstances, and "stringent conditions" under which antisense compounds hybridize to a target sequence are determined by the nature and composition of the antisense compounds and the assays in which they are being investigated.

Complementarity

[0043] "Complementarity," as used herein, refers to the capacity for precise pairing between two nucleobases on either two oligomeric compound strands or an antisense compound with its target nucleic acid. For example, if a nucleobase at a certain position of an antisense compound is capable of hydrogen bonding with a nucleobase at a certain position of a target nucleic acid, then the position of hydrogen bonding between the oligonucleotide and the target nucleic acid is considered to be a complementary position. The antisense compound and the further DNA or RNA are complementary to each other when a sufficient number of complementary positions in each molecule are occupied by nucleobases which can hydrogen bond with each other. Thus, "specifically hybridizable" and "complementary" are terms which are used to indicate a sufficient degree of precise pairing or complementarity over a sufficient number of nucleobases such that stable and specific binding occurs between the antisense compound and a target nucleic acid.

Identity

[0044] Antisense compounds, or a portion thereof, may have a defined percent identity to a SEQ ID NO, or a compound having a specific CMPD number. As used herein, a sequence is identical to the sequence disclosed herein if it has the same nucleobase pairing ability. For example, an RNA which contains uracil in place of thymidine in the disclosed sequences would be considered identical as they both pair with adenine. Similarly, a G-clamp modified heterocyclic base would be considered identical to a cytosine or a 5-Me cytosine in the sequences of the instant application as it pairs with a guanine. This identity may be over the entire length of the oligomeric compound, or in a portion of the antisense compound (e.g., nucleobases 1-20 of a 27-mer may be compared to a 20-mer to determine percent identity of the antisense compound to the SEQ ID NO.) It is understood by those skilled in the art that an antisense compound need not have an identical sequence to those described herein to function similarly to the antisense compound described herein. Shortened versions of antisense compound taught herein, or non-identical versions of the antisense compound taught herein fall within the scope of the current disclosure. Non-identical versions are those wherein each base does not have the same pairing activity as the antisense compounds disclosed herein. In a preferred embodiment the antisense compounds can have about 3 mismatched base pairs. Bases do not have the same pairing activity by being shorter or having at least one abasic site. Alternatively, a non-identical version can include at least one base replaced with a different base with different pairing activity (e.g., G can be replaced by C, A, or T). Percent identity is calculated according to the number of bases that have identical base pairing corresponding to the SEQ ID NO or identical bases corresponding to the antisense compound to which it is being compared. The non-identical bases may be adjacent to each other, dispersed through out the oligonucleotide, or both.

[0045] For example, a 16-mer having the same sequence as nucleobases 2-17 of a 20-mer is 80% identical to the 20-mer. Alternatively, a 20-mer containing four nucleobases not identical to another 20-mer is also 80% identical to that other 20-mer. A 14-mer having the same sequence as nucleobases 1-14 of an 18-mer is 78% identical to the 18-mer. Such calculations are well within the ability of those skilled in the art.

[0046] The percent identity is based on the percent of nucleobases in the original sequence present in a portion of the modified sequence. Therefore, a 30 nucleobase antisense compound comprising the full sequence of the complement of a 20 nucleobase active target segment would have a portion of 100% identity with the complement of the 20 nucleobase active target segment, while further comprising an additional 10 nucleobase portion. Herein, the complement of an active target segment may constitute a single portion. In a preferred embodiment, the oligomeric compounds are at least about 80%, more preferably at least about 85%, even more preferably at least about 90%, most preferably at least 95% identical to at least a portion of the complement of the active target segments presented herein.

[0047] It is well known by those skilled in the art that it is possible to increase or decrease the length of an antisense compound and/or introduce mismatch bases without eliminating activity. For example, in Woolf et al. (Proc. Natl. Acad. Sci. USA 89:7305-7309, 1992, incorporated herein by reference), a series of ASOs 13-25 nucleobases in length were tested for their ability to induce cleavage of a target RNA in an oocyte injection model. ASOs 25 nucleobases in length with 8 or 11 mismatch bases near the ends of the ASOs were able to direct specific cleavage of the target mRNA, albeit to a lesser extent than the ASOs that contained no mismatches. Similarly, target specific cleavage was achieved using a 13 nucleobase ASOs, including those with 1 or 3 mismatches. Maher and Dolnick (Nuc. Acid. Res. 16:3341-3358, 1988, incorporated herein by reference) tested a series of tandem 14 nucleobase ASOs, and a 28 and 42 nucleobase ASOs comprised of the sequence of two or three of the tandem ASOs, respectively, for their ability to arrest translation of human DHFR in a rabbit reticulocyte assay. Each of the three 14 nucleobase ASOs alone were able to reduce translation, albeit at a more modest level than the 28 or 42 nucleobase ASOs.

Therapeutics

[0048] The antisense compounds disclosed herein can be used to modulate the expression of hepcidin in an animal, such as a human. In one non-limiting embodiment, the methods comprise the step of administering to said animal in need of therapy for a disease or condition associated with hepcidin an effective amount of an antisense compound that reduces expression of hepcidin. A disease or condition associated with hepcidin includes, but is not limited to, anemia. In one embodiment, the antisense compounds effectively reduce the levels or function of hepcidin RNA. Because reduction in hepcidin mRNA levels can lead to alteration in hepcidin protein products of expression as well, such resultant alterations can also be measured. Antisense compounds that effectively reduce the level or function of hepcidin RNA or protein products of expression are considered an active antisense compounds. In one embodiment, the antisense compounds reduce the expression of hepcidin causing a reduction of target RNA by at least about 10%, by at least about 20%, by at least about 25%, by at least about 30%, by at least about 40%, by at least about 50%, by at least about 60%, by at least about 70%, by at least about 75%, by at least about 80%, by at least about 85%, by at least about 90%, by at least about 95%, by at least about 98%, by at least about 99%, or by 100%.

[0049] For example, the reduction of the expression of hepcidin can be measured in a bodily fluid, tissue or organ of the animal. Methods of obtaining samples for analysis, such as body fluids (e.g., blood, serum or urine), tissues (e.g., biopsy), or organs, and methods of preparation of the samples to allow for analysis are well known to those skilled in the art. Methods for analysis of RNA and protein levels are discussed above and are well known to those skilled in the art. The effects of treatment can be assessed by measuring biomarkers associated with the hepcidin expression in the aforementioned fluids, tissues or organs, collected from an animal contacted with one or more oligomeric compounds targeted to a nucleic acid encoding hepcidin, by routine clinical methods known in the art. These biomarkers include but are not limited to:serum iron levels, hemoglobin levels, transferrin levels, iron restricted erythropoesis, liver transaminases, bilirubin, albumin, blood urea nitrogen, creatine and other markers of kidney and liver function; interleukins, tumor necrosis factors, chemokines, cytokines and other markers of anemia.

[0050] The antisense compounds can be utilized in pharmaceutical compositions by adding an effective amount of a compound to a suitable pharmaceutically acceptable diluent or carrier. Acceptable carriers and dilutents are well known to those skilled in the art. Selection of a dilutent or carrier is based on a number of factors, including, but not limited to, the solubility of the compound and the route of administration. Such considerations are well understood by those skilled in the art. In one aspect, the antisense compounds reduce the expression of hepcidin. The compounds can also be used in the manufacture of a medicament for the treatment of diseases and conditions related to hepcidin expression.

[0051] Methods whereby bodily fluids, organs or tissues are contacted with an effective amount of one or more of the antisense compounds or compositions are also contemplated. Bodily fluids, organs or tissues can be contacted with one or more of the antisense compounds resulting in modulation of hepcidin expression in the cells of bodily fluids, organs or tissues. An effective amount can be determined by monitoring the modulatory effect of the antisense compound or compounds or compositions on target nucleic acids or their products by methods routine to the skilled artisan.

[0052] Thus, provided herein is the use of an isolated single- or double-stranded antisense compound targeted to hepcidin in the manufacture of a medicament for the treatment of a disease or disorder by means of the method described above. In a preferred embodiment, the antisense compound is a single stranded antisense compound. Also provided herein is the use of a composition comprising erythropoietin or other ESA and an antisense compound targeted to hepcidin in the manufacture of a medicament for the treatment of a disease or disorder by the means of the method described above.

Kits, Research Reagents, And Diagnostics

[0053] The antisense compounds can be utilized for diagnostics, and as research reagents and kits. Furthermore, antisense compounds, which are able to reduce gene expression with specificity, are often used by those of ordinary skill to elucidate the function of particular genes or to distinguish between functions of various members of a biological pathway.

[0054] For use in kits and diagnostics, the antisense compounds, either alone or in combination with other compounds or therapeutics, for example ESAs, can be used as tools in differential and/or combinatorial analyses to elucidate expression patterns of a portion or the entire complement of genes expressed within cells and tissues. Methods of gene expression analysis are well known to those skilled in the art.

Compounds

[0055] The term "oligomeric compound" refers to a polymeric structure capable of hybridizing to a region of a nucleic acid molecule. Generally, oligomeric compounds comprise a plurality of monomeric subunits linked together by internucleoside linking groups and/or internucleoside linkage mimetics. Each of the monomeric subunits comprises a sugar, abasic sugar, modified sugar, or a sugar mimetic, and except for the abasic sugar includes a nucleobase, modified nucleobase or a nucleobase mimetic. Preferred monomeric subunits comprise nucleosides and modified nucleosides.

[0056] An "antisense compound" or "antisense oligomeric compound" refers to an oligomeric compound that is at least partially complementary to the region of a target nucleic acid molecule to which it hybridizes and which modulates (increases or decreases) its expression. This term includes oligonucleotides, oligonucleosides, oligonucleotide analogs, oligonucleotide mimetics, antisense compounds, antisense oligomeric compounds, and chimeric combinations of these. Consequently, while all antisense compounds can be said to be oligomeric compounds, not all oligomeric compounds are antisense compounds. An "antisense oligonucleotide" is an antisense compound that is a nucleic acid-based oligomer. An antisense oligonucleotide can, in some cases, include one or more chemical modifications to the sugar, base, and/or internucleoside linkages. Nonlimiting examples of antisense compounds include antisense compounds, antisense oligonucleotides, external guide sequence (EGS) oligonucleotides, alternate splicers, and siRNAs. As such, these compounds can be introduced in the form of single-stranded, double-stranded, circular, branched or hairpins and can contain structural elements such as internal or terminal bulges or loops. Antisense double-stranded compounds can be two strands hybridized to form double-stranded compounds or a single strand with sufficient self complementarity to allow for hybridization and formation of a fully or partially double-stranded compound. The oligomeric compounds are not auto-catalytic. As used herein, "auto-catalytic" means a compound has the ability to promote cleavage of the target RNA in the absence of accessory factors, e.g. proteins.

[0057] In one embodiment, the antisense compound comprises a single stranded oligonucleotide. In some embodiments the antisense compound contains chemical modifications. In a preferred embodiment, the antisense compound is a single stranded, chimeric oligonucleotide wherein the modifications of sugars, bases, and internucleoside linkages are independently selected.

[0058] The antisense compounds may comprise an antisense compound from about 12 to about 35 nucleobases (i.e. from about 12 to about 35 linked nucleosides). In other words, a single-stranded anisense compound comprises from about 12 to about 35 nucleobases, and a double-stranded antisense compound (such as a siRNA, for example) comprises two strands, each of which is independently from about 12 to about 35 nucleobases. This includes oligonucleotides 15 to 35 and 16 to 35 nucleobases in length. Contained within the antisense compounds (whether single or double stranded and on at least one strand) are antisense portions. The "antisense portion" is that part of the antisense compound that is designed to work by one of the aforementioned antisense mechanisms. One of ordinary skill in the art will appreciate that about 12 to about 35 nucleobases includes 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, or 35 nucleobases.

[0059] Antisense compounds about 12 to 35 nucleobases in length, preferably about 15 to 35 nucleobases in length, comprising a stretch of at least eight (8), preferably at least 12, more preferably at least 15 consecutive nucleobases selected from within the active target regions are considered to be suitable antisense compounds as well.

[0060] Modifications can be made to the antisense compounds and may include conjugate groups attached to one of the termini, selected nucleobase positions, sugar positions or to one of the internucleoside linkages. Possible modifications include, but are not limited to, 2'-fluoro (2'-F), 2'-OMethyl (2'-OMe), 2'-Methoxyethoxy (2'-MOE) sugar modifications, inverted abasic caps, deoxynucleobases, and bicyclic nucleic acids such as locked nucleic acids (LNA) and ethylene nucleic acid (ENA).

[0061] In one embodiment, double-stranded antisense compounds encompass short interfering RNAs (siRNAs). As used herein, the term "siRNA" is defined as a double-stranded compound having a first and second strand, each strand having a central portion and two independent terminal portions. The central portion of the first strand is complementary to the central portion of the second strand, allowing hybridization of the strands. The terminal portions are independently, optionally complementary to the corresponding terminal portion of the complementary strand. The ends of the strands may be modified by the addition of one or more natural or modified nucleobases to form an overhang.

[0062] Each strand of the siRNA duplex may be from about 12 to about 35 nucleobases. In a preferred embodiment, each strand of the siRNA duplex is about 17 to about 25 nucleobases. The two strands may be fully complementary (i.e., form a blunt ended compound), or include a 5' or 3' overhang on one or both strands. Double-stranded compounds can be made to include chemical modifications as discussed herein.

Chemical Modifications

[0063] As is known in the art, a nucleoside is a base-sugar combination. The base portion of the nucleoside is normally a heterocyclic base (sometimes referred to as a "nucleobase" or simply a "base"). The two most common classes of such heterocyclic bases are the purines and the pyrimidines. Nucleotides are nucleosides that further include a phosphate group covalently linked to the sugar portion of the nucleoside. For those nucleosides that include a pentofuranosyl sugar, the phosphate group can be linked to the 2', 3' or 5' hydroxyl moiety of the sugar. In forming oligonucleotides, the phosphate groups covalently link adjacent nucleosides to one another to form a linear polymeric compound. Within oligonucleotides, the phosphate groups are commonly referred to as forming the internucleoside backbone of the oligonucleotide. The normal linkage or backbone of RNA and DNA is a 3' to 5' phosphodiester linkage. It is often preferable to include chemical modifications in oligonucleotides to alter their activity. Chemical modifications can alter oligonucleotide activity by, for example: increasing affinity of an antisense oligonucleotide for its target RNA, increasing nuclease resistance, and/or altering the pharmacokinetics of the oligonucleotide. The use of chemistries that increase the affinity of an oligonucleotide for its target can allow for the use of shorter oligonucleotide compounds.

[0064] The term "nucleobase" or "heterocyclic base moiety" as used herein, refers to the heterocyclic base portion of a nucleoside. In general, a nucleobase is any group that contains one or more atom or groups of atoms capable of hydrogen bonding to a base of another nucleic acid. In addition to "unmodified" or "natural" nucleobases such as the purine nucleobases adenine (A) and guanine (G), and the pyrimidine nucleobases thymine (T), cytosine (C) and uracil (U), many modified nucleobases or nucleobase mimetics known to those skilled in the art are amenable to the oligomeric compounds. The terms modified nucleobase and nucleobase mimetic can overlap but generally a modified nucleobase refers to a nucleobase that is fairly similar in structure to the parent nucleobase, such as for example a 7-deaza purine or a 5-methyl cytosine, whereas a nucleobase mimetic would include more complicated structures, such as for example a tricyclic phenoxazine nucleobase mimetic. Methods for preparation of the above noted modified nucleobases are well known to those skilled in the art.

[0065] Antisense compounds may also contain one or more nucleosides having modified sugar moieties. The furanosyl sugar ring of a nucleoside can be modified in a number of ways including, but not limited to, addition of a substituent group bridging two non-geminal ring atoms to form a bicyclic nucleic acid (BNA) and substitution of an atom or group such as --S--, -N(R)-- or --C(R.sub.1)(R.sub.2) for the ring oxygen at the 4'-position. Modified sugar moieties are well known and can be used to alter, typically increase, the affinity of the antisense compound for its target and/or increase nuclease resistance. A representative list of preferred modified sugars includes but is not limited to bicyclic modified sugars (BNA's), including LNA and ENA (4'-(CH.sub.2).sub.2--O-2' bridge); and substituted sugars, especially 2'-substituted sugars having a 2'-F, 2'-OCH.sub.2 or a 2'-O(CH.sub.2).sub.2--OCH.sub.3 substituent group. Sugars can also be replaced with sugar mimetic groups among others. Methods for the preparations of modified sugars are well known to those skilled in the art.

[0066] Included herein are internucleoside linking groups that link the nucleosides or otherwise modified monomer units together thereby forming an antisense compound. The two main classes of internucleoside linking groups are defined by the presence or absence of a phosphorus atom. Representative phosphorus containing internucleoside linkages include, but are not limited to, phosphodiesters, phosphotriesters, methylphosphonates, phosphoramidate, and phosphorothioates. Representative non-phosphorus containing internucleoside linking groups include, but are not limited to, methylenemethylimino (--CH.sub.2--N(CH.sub.3)--O--CH.sub.2--), thiodiester (--O--C(O)--S--), thionocarbamate (--O--C(O)(NH)--S--); siloxane (--O--Si(H)2--O--); and N,N'-dimethylhydrazine (--CH.sub.2--N(CH.sub.3)--N(CH.sub.3)--). Antisense compounds having non-phosphorus internucleoside linking groups are referred to as oligonucleosides. Modified internucleoside linkages, compared to natural phosphodiester linkages, can be used to alter, typically increase, nuclease resistance of the antisense compound. Internucleoside linkages having a chiral atom can be prepared racemic, chiral, or as a mixture. Representative chiral internucleoside linkages include, but are not limited to, alkylphosphonates and phosphorothioates. Methods of preparation of phosphorous-containing and non-phosphorous-containing linkages are well known to those skilled in the art.

[0067] As used herein the term "mimetic" refers to groups that are substituted for a sugar, a nucleobase, and/ or internucleoside linkage. Generally, a mimetic is used in place of the sugar or sugar-internucleoside linkage combination, and the nucleobase is maintained for hybridization to a selected target. Representative examples of a sugar mimetic include, but are not limited to, cyclohexenyl or morpholino. Representative examples of a mimetic for a sugar-internucleoside linkage combination include, but are not limited to, peptide nucleic acids (PNA) and morpholino groups linked by uncharged achiral linkages. In some instances a mimetic is used in place of the nucleobase. Representative nucleobase mimetics are well known in the art and include, but are not limited to, tricyclic phenoxazine analogs and universal bases (Berger et al., (2000) Nuc Acid Res., 28:2911-14, incorporated herein by reference). Methods of synthesis of sugar, nucleoside and nucleobase mimetics are well known to those skilled in the art.

[0068] As used herein the term "nucleoside" includes, nucleosides, abasic nucleosides, modified nucleosides, and nucleosides having mimetic bases and/or sugar groups.

[0069] As used herein, the term "oligonucleotide" refers to an oligomeric compound which is an oligomer or polymer of ribonucleic acid (RNA) or deoxyribonucleic acid (DNA). This term includes oligonucleotides composed of naturally- and non-naturally-occurring nucleobases, sugars and covalent internucleoside linkages, possibly further including non-nucleic acid conjugates.

[0070] Provided herein are compounds having reactive phosphorus groups useful for forming internucleoside linkages including for example phosphodiester and phosphorothioate internucleoside linkages. Methods of preparation and/or purification of precursors or antisense compounds are not a limitation of the compositions or methods described herein. Methods for synthesis and purification of DNA, RNA, and the antisense compounds are well known to those skilled in the art.

[0071] As used herein the term "chimeric antisense compound" refers to an antisense compound, having at least one sugar, nucleobase and/or internucleoside linkage that is differentially modified as compared to the other sugars, nucleobases and internucleoside linkages within the same oligomeric compound. The remainder of the sugars, nucleobases and internucleoside linkages can be independently modified or unmodified. In general a chimeric oligomeric compound will have modified nucleosides that can be in isolated positions or grouped together in regions that will define a particular motif. Any combination of modifications and or mimetic groups can comprise a chimeric oligomeric compound.

[0072] Chimeric oligomeric compounds typically contain at least one region modified so as to confer increased resistance to nuclease degradation, increased cellular uptake, and/or increased binding affinity for the target nucleic acid. An additional region of the oligomeric compound may serve as a substrate for enzymes capable of cleaving RNA:DNA or RNA:RNA hybrids. By way of example, RNase H is a cellular endonuclease that cleaves the RNA strand of an RNA:DNA duplex. Activation of RNase H, therefore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of reduction of gene expression. Consequently, comparable results can often be obtained with shorter oligomeric compounds when chimeras are used, compared to for example phosphorothioate deoxyoligonucleotides hybridizing to the same target region. Cleavage of the RNA target can be routinely detected by gel electrophoresis and, if necessary, associated nucleic acid hybridization techniques known in the art.

[0073] Certain chimeric as well as non-chimeric oligomeric compounds can be further described as having a particular motif. As used in herein the term "motif" refers to the orientation of modified sugar moieties and/or sugar mimetic groups in an antisense compound relative to like or differentially modified or unmodified nucleosides. As used herein, the terms "sugars", "sugar moieties" and "sugar mimetic groups` are used interchangeably. Such motifs include, but are not limited to, gapped motifs, alternating motifs, fully modified motifs, hemimer motifs, blockmer motifs, and positionally modified motifs. The sequence and the structure of the nucleobases and type of internucleoside linkage is not a factor in determining the motif of an antisense compound.

[0074] As used herein the term "gapped motif" refers to an antisense compound comprising a contiguous sequence of nucleosides that is divided into 3 regions, an internal region (gap) flanked by two external regions (wings). The regions are differentiated from each other at least by having differentially modified sugar groups that comprise the nucleosides. In some embodiments, each modified region is uniformly modified (e.g. the modified sugar groups in a given region are identical); however, other motifs can be applied to regions. For example, the wings in a gapmer could have an alternating motif. The nucleosides located in the gap of a gapped antisense compound have sugar moieties that are different than the modified sugar moieties in each of the wings.

[0075] As used herein the term "alternating motif" refers to an antisense compound comprising a contiguous sequence of nucleosides comprising two differentially sugar modified nucleosides that alternate for essentially the entire sequence of the antisense compound, or for essentially the entire sequence of a region of an antisense compound.

[0076] As used herein the term "fully modified motif" refers to an antisense compound comprising a contiguous sequence of nucleosides wherein essentially each nucleoside is a sugar modified nucleoside having uniform modification.

[0077] As used herein the term "hemimer motif" refers to a sequence of nucleosides that have uniform sugar moieties (identical sugars, modified or unmodified) and wherein one of the 5'-end or the 3'-end has a sequence of from 2 to 12 nucleosides that are sugar modified nucleosides that are different from the other nucleosides in the hemimer modified antisense compound.

[0078] As used herein the term "blockmer motif" refers to a sequence of nucleosides that have uniform sugars (identical sugars, modified or unmodified) that is internally interrupted by a block of sugar modified nucleosides that are uniformly modified and wherein the modification is different from the other nucleosides. Methods of preparation of chimeric oligonucleotide compounds are well known to those skilled in the art.

[0079] As used herein the term "positionally modified motif" comprises all other motifs. Methods of preparation of positionally modified oligonucleotide compounds are well known to those skilled in the art.

[0080] The compounds described herein contain one or more asymmetric centers and thus give rise to enantiomers, diastereomers, and other stereoisomeric configurations that may be defined, in terms of absolute stereochemistry, as (R) or (S), alpha or beta, or as (D) or (L) such as for amino acids et al. The present description of oligomeric compounds is meant to include all such possible isomers, as well as their racemic and optically pure forms.

[0081] In one aspect, antisense compounds are modified by covalent attachment of one or more conjugate groups. Conjugate groups may be attached by reversible or irreversible attachments. Conjugate groups may be attached directly to antisense compounds or by use of a linker. Linkers may be mono- or bifunctional linkers. Such attachment methods and linkers are well known to those skilled in the art. In general, conjugate groups are attached to antisense compounds to modify one or more properties. Such considerations are well known to those skilled in the art.

Oligomer Synthesis

[0082] Oligomerization of modified and unmodified nucleosides can be routinely performed according to literature procedures for DNA (Protocols for Oligonucleotides and Analogs, Ed. Agrawal (1993), Humana Press) and/or RNA (Scaringe, Methods (2001), 23, 206-217. Gait et al., Applications of Chemically synthesized RNA in RNA: Protein Interactions, Ed. Smith (1998), 1-36. Gallo et al., Tetrahedron (2001), 57, 5707-5713).

[0083] Antisense compounds can be conveniently and routinely made through the well-known technique of solid phase synthesis. Equipment for such synthesis is sold by several vendors including, for example, Applied Biosystems (Foster City, Calif.). Any other means for such synthesis known in the art may additionally or alternatively be employed. It is well known to use similar techniques to prepare oligonucleotides such as the phosphorothioates and alkylated derivatives. The current description is not limited by the method of antisense compound synthesis.

Oligomer Purification And Analysis

[0084] Methods of oligonucleotide purification and analysis are known to those skilled in the art. Analysis methods include capillary electrophoresis (CE) and electrospray-mass spectroscopy. Such synthesis and analysis methods can be performed in multi-well plates. The methods described herein are not limited by the method of oligomer purification.

Salts, Prodrugs And Bioequivalents

[0085] The antisense compounds comprise any pharmaceutically acceptable salts, esters, or salts of such esters, or any other functional chemical equivalent which, upon administration to an animal including a human, is capable of providing (directly or indirectly) the biologically active metabolite or residue thereof. Accordingly, for example, the disclosure is also drawn to prodrugs and pharmaceutically acceptable salts of the antisense compounds, pharmaceutically acceptable salts of such prodrugs, and other bioequivalents.

[0086] The term "prodrug" indicates a therapeutic agent that is prepared in an inactive or less active form that is converted to an active form (i.e., drug) within the body or cells thereof by the action of endogenous enzymes, chemicals, and/or conditions. In particular, prodrug versions of the oligonucleotides are prepared as SATE ((S-acetyl-2-thioethyl) phosphate) derivatives according to the methods disclosed in WO 93/24510 or WO 94/26764. Prodrugs can also include antisense compounds wherein one or both ends comprise nucleobases that are cleaved (e.g., phosphodiester backbone linkages) to produce the active compound.

[0087] The term "pharmaceutically acceptable salts" refers to physiologically and pharmaceutically acceptable salts of the compounds: i.e., salts that retain the desired biological activity of the parent compound and do not impart undesired toxicological effects thereto. Sodium salts of antisense oligonucleotides are useful and are well accepted for therapeutic administration to humans. In another embodiment, sodium salts of dsRNA compounds are also provided.

Combinations

[0088] Compositions can contain two or more antisense compounds. In another related embodiment, compositions can contain one or more antisense compounds, particularly oligonucleotides, targeted to a first nucleic acid and one or more additional antisense compounds targeted to a second nucleic acid target. Alternatively, compositions can contain two or more antisense compounds targeted to different regions of the same nucleic acid target. Compositions can also include other non-antisense compound therapeutic agents. For example, disclosed herein are combinations comprising an antisense oligonucleotide targeted to hepcidin combined with erythropoietin. Two or more combined compounds may be used together or sequentially. Likewise, one or more compound and one or more therapeutic agent may be used together or sequentially. For example, disclosed herein are compounds and therapeutics and methods of use wherein the compounds are administered simultaneously, or at distinct timepoints. For example, an antisense oligonucleotide targeted to a nucleic acid encoding hepcidin can be administered either simultaneously with or at a distinct timepoint from erythropoietin.

Nonlimiting Disclosure And Incorporation By Reference

[0089] While certain compounds, compositions and methods have been described with specificity in accordance with certain embodiments, the following examples serve only to illustrate the compounds and methods and are not intended to limit the same. Each of the references, GenBank accession numbers, and the like recited in the present application is incorporated herein by reference in its entirety.

EXAMPLE 1

Cell Types And Transfection Methods

[0090] The effect of oligomeric compounds on target nucleic acid expression was tested on the following cell types.

[0091] Mouse Primary Hepatocytes: Primary mouse hepatocytes were prepared from CD-1 mice purchased from Charles River Labs (Wilmington, Mass.). Primary mouse hepatocytes were routinely cultured in Hepatocyte Attachment Media supplemented with 10% fetal bovine serum, 1% penicillin/streptomycin (both from Sigma-Aldrich, St. Louis, Mo.), 1% antibiotic-antimitotic (Invitrogen Life Technologies, Carlsbad, Calif.) and 10 nM bovine insulin (Sigma-Aldrich, St. Louis, Mo.). Cells were seeded into 96-well plates (Falcon-Primaria #3872) coated with 0.1 mg/ml collagen at a density of approximately 10,000 cells/well for use in oligomeric compound transfection experiments.

[0092] HepG2: The human hepatocarcinoma cell line was obtained from the American Type Culture Collection (Manassas, Va.). HepG2 cells were routinely cultured in minimum essential medium (Eagle) with 2 mM L-glutamine and Earle's BSS (Cambrex, East Rutherford, N.J.) adjusted to contain 1.5 g/L sodium bicarbonate, 0.1 mM non-essential amino acids, and 1.0 mM sodium pyruvate, 90%; fetal bovine serum, 10% (sigma-Aldrich, St. Louis, Mo.) at a temperature of 37.degrees.C. Cells were routinely passaged by trypsinization and dilution when they reached approximately 80% confluence. Cells were seeded into 96-well plates (Falcon-Primaria #3872) at a density of approximately 5000 cells/well for use in oligomeric compound transfection experiments.

[0093] When cells reached 65-75% confluency, they were treated with oligonucleotide. Oligonucleotide was mixed with LIPOFECTIN.TM. Invitrogen Life Technologies, Carlsbad, Calif.) in OPTI-MEM.TM.-1 reduced serum medium (Invitrogen Life Technologies, Carlsbad, Calif.) to achieve the desired concentration of oligonucleotide and a LIPOFECTIN.TM. concentration of 2.5 or 3 .micro.g/mL per 100 nM oligonucleotide. This transfection mixture was incubated at room temperature for approximately 0.5 hours. For cells grown in 96-well plates, wells were washed once with 100 .micro.L OPTI-MEMT.TM.-1 and then treated with 130 .micro.L of the transfection mixture. Cells were treated and data were obtained in duplicate or triplicate. After approximately 4-7 hours of treatment at 37.degrees.C., the medium containing the transfection mixture was replaced with fresh culture medium. Cells were harvested 16-24 hours after oligonucleotide treatment.

[0094] Control oligonucleotides were used to determine the optimal oligomeric compound concentration for a particular cell line. Furthermore, when oligomeric compounds were tested in oligomeric compound screening experiments or phenotypic assays, control oligonucleotides were tested in parallel.

[0095] The concentration of oligonucleotide used varied from cell line to cell line. To determine the optimal oligonucleotide concentration for a particular cell line, the cells were treated with a positive control oligonucleotide at a range of concentrations. The concentration of positive control oligonucleotide that resulted in about an 80% reduction of the target mRNA was then utilized as the screening concentration for new oligonucleotides in subsequent experiments for that cell line. If 80% reduction was not achieved, the lowest concentration of positive control oligonucleotide that resulted in a 60% reduction of the target mRNA was then utilized as the oligonucleotide screening concentration in subsequent experiments for that cell line. If 60% reduction was not achieved, that particular cell line was deemed as unsuitable for oligonucleotide transfection experiments.

EXAMPLE 2

Real-Time Quantitative PCR Analysis of Hepcidin mRNA Levels

[0096] Quantitation of hepcidin mRNA levels was accomplished by real-time quantitative PCR using the ABI PRISM.TM. 7600, 7700, or 7900 Sequence Detection System (PE-Applied Biosystems, Foster City, Calif.) according to manufacturer's instructions.

[0097] Prior to quantitative PCR analysis, primer-probe sets specific to the hepcidin being measured were evaluated for their ability to be "multiplexed" with a GAPDH amplification reaction. After isolation the RNA was subjected to sequential reverse transcriptase (RT) reaction and real-time PCR, both of which were performed in the same well. RT and PCR reagents were obtained from Invitrogen Life Technologies (Carlsbad, Calif.). RT, real-time PCR was carried out in the same by adding 20 .micro.L PCR cocktail (2.5.times.PCR buffer minus MgCl.sub.2, 6.6 mM MgCl.sub.2, 375 .micro.M each of dATP, dCTP, dCTP and dGTP, 375 nM each of forward primer and reverse primer, 125 nM of probe, 4 Units RNAse inhibitor, 1.25 Units PLATINUM.RTM. Taq, 5 Units MuLV reverse transcriptase, and 2.5.times.ROX dye) to 96-well plates containing 30 .micro.L total RNA solution (20-200 ng). The RT reaction was carried out by incubation for 30 minutes at 48.degrees.C. Following a 10 minute incubation at 95.degrees.C. to activate the PLATINUM.RTM. Taq, 40 cycles of a two-step PCR protocol were carried out: 95.degrees.C. for 15 seconds (denaturation) followed by 60.degrees.C. for 1.5 minutes (annealing/extension).

[0098] Gene target quantities obtained by RT, real-time PCR were normalized using either the expression level of GAPDH, a gene whose expression is constant, or by quantifying total RNA using RiboGreen.TM. (Molecular Probes, Inc. Eugene, Oreg.). GAPDH expression was quantified by RT, real-time PCR, by being run simultaneously with the target, multiplexing, or separately. Total RNA was quantified using RiboGreen.TM. RNA quantification reagent (Molecular Probes, Inc. Eugene, Oreg.).

[0099] 170 .micro.L of RiboGreen.TM. working reagent (RiboGreen.TM. reagent diluted 1:350 in 10 mM Tris-HCl, 1 mM EDTA, pH 7.5) was pipetted into a 96-well plate containing 30 .micro.L purified cellular RNA. The plate was read in a CytoFluor 4000 (PE Applied Biosystems) with excitation at 485 nm and emission at 530 nm.

[0100] The GAPDH PCR probes have JOE covalently linked to the 5' end and TAMRA or MGB covalently linked to the 3' end, where JOE is the fluorescent reporter dye and TAMRA or MGB is the quencher dye. In some cell types, primers and probe designed to a GAPDH sequence from a different species are used to measure GAPDH expression. For example, a human GAPDH primer and probe set is used to measure GAPDH expression in monkey-derived cells and cell lines.

[0101] Probes and primers for use in real-time PCR were designed to hybridize to target-specific sequences. The primers and probes and the target nucleic acid sequences to which they hybridize are presented in Table 2. The target-specific PCR probes have FAM covalently linked to the 5' end and TAMRA or MGB covalently linked to the 3' end, where FAM is the fluorescent dye and TAMRA or MGB is the quencher dye.

TABLE-US-00002 TABLE 2 Hepcidin-specific primers and probes for use in real-time PCR Target SEQ SEQ ID Sequence ID Species NO Description Sequence (5' to 3') NO Human 2 Forward Primer AGGAGGCGAGACACCCACTT 6 Human 2 Reverse Primer TCCCACACTTTGATCGATGACA 7 Human 2 Probe CATCTGCATTTTCTGCTGCGGCTG 8 Mouse 4 Forward Primer TGCAGAAGAGAAGGAAGAGAGACA 9 Mouse 4 Reverse Primer CACACTGGGAATTGTTACAGCATT 10 Mouse 4 Probe CAACTTCCCCATCTGCATCTTCTGCTGT 11

EXAMPLE 3

Antisense Reduction of Human Hepcidin Expression By Oligomeric Compounds

[0102] A series of antisense oligonucleotide compounds was designed to target different regions of human hepcidin RNA, using published sequences or portions of published sequences as cited in Table 1. The compounds are shown in Table 3. Methods for designing and testing antisense oligonucleotides for reduction of mRNA target expression are discussed herein and well known to those skilled in the art. Although an antisense oligonucleotide is reported in Table 3 as being targeted to a particular sequence, one ordinarily skilled in the art will know that these sequences may also target other sequences. For example, in Table 3 SEQ ID NO: 50 is reportedly designed to target nucleotides 261 to 280 of SEQ ID NO: 2. However, SEQ ID NO: 50 is also fully complementary to nucleotides 486 to 505 of SEQ ID NO: 1 and nucleotides 4662 to 4681 of SEQ ID NO: 3.

[0103] The compounds were analyzed for their effect on target mRNA levels by using quantitative real time RT-PCR as described above. The screen identified active target segments within these regions of the human hepcidin mRNA sequence and the genomic sequence encoding hepcidin mRNA, specifically GenBank numbers BM719679.1; NM.sub.--021175.2; and nucleotides 7819907 to 7825131 of NT.sub.--011196.11 (SEQ ID NOS: 1; 2; and 3, respectively). The activity of these antisense oligonucleotides to reduce hepcidin mRNA expression is ranked as follows: +++++ (reduction of target by greater than about 70%), ++++ (reduction of target by greater than about 70% to about 74%), +++ (reduction of target by greater than about 60% to about 69%), ++ (reduction of target by greater than about 55% to about 60%), + (reduction of target by greater than about 50% to about 55%)or bc (below cut-off or lesser than about 50%). Antisense compounds having an activity ranking of bc may have shown some reduction of hepcidin expression but fell below the cut-off level for this screening assay.

TABLE-US-00003 TABLE 3 Reduction of Human Hepcidin mRNA Levels by Antisense Oligonucleotides Target Target SEQ CMPD SEQ ID Start Activity ID # NO Site Sequence (5' to 3') Ranking NO 392287 1 32 AATGCACAGGCCCTGCCATC + 12 392288 1 38 CACAGAAATGCACAGGCCCT bc 13 392289 1 43 TCAAGCACAGAAATGCACAG bc 14 392290 1 48 CCCACTCAAGCACAGAAATG bc 15 392291 1 53 CAAGGCCCACTCAAGCACAG +++ 16 392292 1 58 ACTTTCAAGGCCCACTCAAG ++++ 17 392280 1 66 GCTGAACCACTTTCAAGGCC bc 18 392281 1 71 TGGTTGCTGAACCACTTTCA bc 19 392282 1 91 GAGGAATGAACACTTCTTCC bc 20 392283 1 96 TTGTCGAGGAATGAACACTT ++ 21 392284 1 101 TGTTGTTGTCGAGGAATGAA bc 22 392285 1 123 CCAAGTCACCAGAGCCCGGG +++++ 23 392286 1 128 GTCAGCCAAGTCACCAGAGC ++++ 24 392304 1 133 CCAGTGTCAGCCAAGTCACC bc 25 392305 1 138 GCCATCCAGTGTCAGCCAAG + 26 392306 1 143 CCAGGGCCATCCAGTGTCAG +++ 27 392307 1 148 TCATTCCAGGGCCATCCAGT ++++ 28 392308 1 153 CTTTTTCATTCCAGGGCCAT ++ 29 392309 1 179 GGCCCTTGCACATTTTGCCT + 30 392293 1 186 CCAGATGGGCCCTTGCACAT ++++ 31 392294 1 191 TGGTTCCAGATGGGCCCTTG bc 32 392295 1 196 GGCCTTGGTTCCAGATGGGC +++ 33 392296 1 215 GTGACAGTCGCTTTTATGGG bc 34 392297 1 220 ACCGAGTGACAGTCGCTTTT bc 35 392298 1 225 CTGGGACCGAGTGACAGTCG + 36 392299 1 547 CAGGGCAGGTAGGTTCTACG +++ 37 392316 2 3 TGTCTGGGACCGAGTGACAG bc 38 392323 2 8 TCTGGTGTCTGGGACCGAGT + 39 392318 2 70 CTGGGAGCTCAGTGCCATCG +++++ 40 392319 2 75 CAGATCTGGGAGCTCAGTGC bc 41 392300 2 127 AGAGCCACTGGTCAGGCTGG +++++ 42 392320 2 132 AAAACAGAGCCACTGGTCAG bc 43 392302 2 137 GTGGGAAAACAGAGCCACTG +++ 44 392310 2 161 GCTCTGCAAGTTGTCCCGTC ++++ 45 392311 2 166 TTGCAGCTCTGCAAGTTGTC bc 46 392303 2 212 GGAACATGGGCATCCAGCTG bc 47 392313 2 217 CCTCTGGAACATGGGCATCC ++++ 48 392314 2 242 GGAAGTGGGTGTCTCGCCTC +++++ 49 392315 2 261 CAGCAGAAAATGCAGATGGG +++++ 50 392326 2 268 GCAGCCGCAGCAGAAAATGC +++++ 51 392327 2 273 TGACAGCAGCCGCAGCAGAA +++ 52 392328 2 278 ATCGATGACAGCAGCCGCAG +++++ 53 392330 2 287 CACACTTTGATCGATGACAG ++ 54 392329 2 292 CATCCCACACTTTGATCGAT ++++ 55 392321 2 297 CAGCACATCCCACACTTTGA ++ 56 392322 2 302 TCTTGCAGCACATCCCACAC bc 57 392301 2 382 TTATTCCAAGACCTATGTTC ++ 58 392317 2 390 CAGCCATTTTATTCCAAGAC +++ 59 392324 2 395 AGAACCAGCCATTTTATTCC bc 60 392325 2 400 ACAAAAGAACCAGCCATTTT bc 61 392340 3 2917 CCTGACCAGGATGGAGAAAT +++ 62 392341 3 2924 CAAATTCCCTGACCAGGATG + 63 392339 3 2929 TTTAACAAATTCCCTGACCA bc 64 392338 3 2934 CAGTCTTTAACAAATTCCCT bc 65 392344 3 2942 GTTTTCATCAGTCTTTAACA +++ 66 392343 3 2963 TAGACACAATTATTTATTCA bc 67 392342 3 2968 TGTACTAGACACAATTATTT ++ 68 392332 3 2973 TAGAATGTACTAGACACAAT + 69 392333 3 2982 ATTCACGAATAGAATGTACT +++ 70 392331 3 2987 ATGAGATTCACGAATAGAAT bc 71 392337 3 3018 ATGGGTCACGGTCACTCTAC ++ 72 392336 3 3023 GGCGAATGGGTCACGGTCAC +++ 73 392335 3 3028 TGTGTGGCGAATGGGTCACG bc 74 392334 3 3227 CAGCCCCTTGACCATGCCTG ++++ 75 392357 3 3260 TGTCACTTTCTCATGAAACA +++ 76 392356 3 3278 TCCAAGACGAAGGTCAACTG bc 77 392355 3 3349 TTGTTCAGCCTGACCCTGGG +++++ 78 392354 3 3375 TTATCCCCAAGTCCCACCAT bc 79 392353 3 3383 CCTCAGCCTTATCCCCAAGT bc 80 392352 3 3398 TAGGGACTGCCCACCCCTCA +++++ 81 392348 3 3420 CTGCATGGTTGCCCACAAGA ++++ 82 392347 3 3425 AGTGTCTGCATGGTTGCCCA +++++ 83 392346 3 3430 AAATCAGTGTCTGCATGGTT +++++ 84 392345 3 3435 AGGAAAAATCAGTGTCTGCA ++++ 85 392351 3 3782 TCCATTTTAAAAACCTACAG bc 86 392350 3 3803 GCCAGTGAGATTGTGGTTTT bc 87 392349 3 3808 ACATGGCCAGTGAGATTGTG +++ 88 392312 3 4516 GGCATCCAGCTGGCCCTGGC ++++ 89

[0104] Each active target segment was targeted by multiple, active antisense oligonucleotides. This current example includes identification of active target segments identified on SEQ ID NO: 1, which active target segments are referred to as Regions A-E. The active target segments include nucleotides 456 to 527 for Region A, nucleotides 123 to 252 for Region B, nucleotides 352 to 405 for Region C, nucleotides 429 to 566 for Region D, and nucleotides 295 to 566 for Region E. Each of the oligonucleotides tested within Region A reduced expression of human hepcidin at least 55% (activity ranking ++). Four-fifths of the oligonucleotides tested in this region reduced expression by at least 75% (activity ranking +++++). Moreover, in Region D more than four-fifths of the oligonucleotides tested reduced expression by at least 55% (activity ranking ++) and more than one-half reduced expression by at least 70% (activity ranking ++++). In Region E, three-fourths of the oligonucleotides tested reduced expression by at least 50% (activity ranking +) and nearly one-half reduced expression by at least 70% (activity ranking ++++). Other active target segments on SEQ ID NO: 1 include Region C where three-fourths of the oligonucleotides tested reduced expression by at least 60% (activity ranking +++), and Region B where two-thirds of the oligonucleotides tested reduce expression by at least 50% (activity ranking +).

[0105] Active target segments were also identified on SEQ ID NO: 2 and SEQ ID NO: 3. The active target segments of SEQ ID NO: 2 are referred to as Regions F and G. Region F includes nucleotides 231 to 302. All of the nucleotides tested in Region F reduce expression by at least 55% (activity ranking ++), and two-thirds reduce at least 70% (activity ranking ++++). Region G includes nucleotides 204 to 341. More than four-fifths of the nucleotides tested in Region G reduce expression by at least 55% (activity ranking ++) and more than one-half reduce expression by at least 70% (activity ranking ++++).

[0106] The active target segments of SEQ ID NO: 3 are referred to as Regions H-K. Region H includes nucleotides 3398 to 3454 and all of the oligonucleotides tested in this region reduce expression by at least 70% (activity ranking ++++). Region I includes nucleotides 3249 to 3454. In Region I, three-fourths of the oligonucleotides tested reduced expression by at least 70% (activity ranking ++++) and one-half reduce by at least 75% (activity ranking +++++). Region J includes nucleotides 4643 to 4703. All of the oligonucleotides tested in this region reduce expression by at least 60% (activity ranking +++) and four-fifths reduce expression by at least 75% (activity ranking +++++). Region K includes nucleotides 4643 to 4743 and at least one-half of the oligonucleotides tested in this region reduce expression by at least 70% (activity ranking ++++).

[0107] Thus, Applicant has discovered regions of the human hepcidin mRNA that are highly active in response to antisense reduction. Identification of these regions allows for the design of antisense oligonucleotides that will modulate the expression of hepcidin. Antisense oligonucleotides can be designed the active target segments with at least a + activity ranking, more preferably with at least a ++ activity ranking and most preferably with a +++ activity ranking. One skilled in the art will readily recognize that the percent reduction is referred to herein as a relative amount of reduction compared to other oligonucleotides tested against SEQ ID NOS: 1, 2 or 3. Assay to assay variations in conditions results in changes from the absolute data received, however, the general trends and relative reduction will be substantially the same.

EXAMPLE 4

Antisense Reduction of Mouse Hepcidin Expression By Oligomeric Compounds

[0108] A series of antisense compounds was designed to target different regions of mouse hepcidin RNA, using published sequences or portions of published sequences as cited in Table 1. The compounds are shown in Table 4. The screen identified active target segments within these regions of the human hepcidin mRNA sequence and the genomic sequence encoding hepcidin mRNA, specifically GenBank numbers NM.sub.--032541.1 and the complement of nucleotides 3978217 to 3980665 of NT.sub.--039413.1 (SEQ ID NOS: 4 and 5, respectively). The activity of these antisense oligonucleotides are ranked based on measured percent reduction of the target from this study. The activity of these antisense oligonucleotides to reduce hepcidin mRNA expression is ranked as follows: +++++ (reduction of target by greater than about 70%), ++++ (reduction of target by greater than about 70% to about 74%), +++ (reduction of target by greater than about 60% to about 69%), ++ (reduction of target by greater than about 55% to about 60%), + (reduction of target by greater than about 50% to about 55%) or bc (below cut-off or lesser than about 50%). Antisense compounds having an activity ranking of bc may have shown some reduction of hepcidin expression but fell below the cut-off level for this screening assay.

TABLE-US-00004 TABLE 4 Reduction of Mouse Hepcidin mRNA Levels by Antisense Oligonucleotides Target Target SEQ CMPD SEQ ID Start Activity ID # NO Site Sequence (5' to 3') Ranking NO 326810 4 1 TGCTGTGCAGTCTAAGGACT +++ 90 326811 4 6 TGTTCTGCTGTGCAGTCTAA bc 91 326812 4 10 CTTCTGTTCTGCTGTGCAGT ++ 92 326813 4 15 CATGCCTTCTGTTCTGCTGT ++++ 93 326814 4 17 ATCATGCCTTCTGTTCTGCT +++ 94 326815 4 22 GTGCCATCATGCCTTCTGTT +++++ 95 326816 4 24 GAGTGCCATCATGCCTTCTG ++++ 96 326817 4 27 GCTGAGTGCCATCATGCCTT +++++ 97 326818 4 29 GTGCTGAGTGCCATCATGCC +++ 98 326819 4 32 CGAGTGCTGAGTGCCATCAT bc 99 326820 4 37 GGGTCCGAGTGCTGAGTGCC +++ 100 326821 4 41 GCCTGGGTCCGAGTGCTGAG +++ 101 326822 4 45 GGCAGCCTGGGTCCGAGTGC +++++ 102 326823 4 50 AGACAGGCAGCCTGGGTCCG bc 103 326824 4 55 GCAGGAGACAGGCAGCCTGG +++++ 104 326825 4 71 CTGGCAAGGAGGAGAAGCAG bc 105 326826 4 80 CTGCTCAGGCTGGCAAGGAG bc 106 326827 4 84 GGTGCTGCTCAGGCTGGCAA bc 107 326828 4 86 GTGGTGCTGCTCAGGCTGGC bc 108 326829 4 88 AGGTGGTGCTGCTCAGGCTG ++++ 109 326830 4 89 TAGGTGGTGCTGCTCAGGCT +++ 110 326831 4 90 ATAGGTGGTGCTGCTCAGGC + 111 326832 4 91 GATAGGTGGTGCTGCTCAGG bc 112 326833 4 98 TGATGGAGATAGGTGGTGCT bc 113 326834 4 99 TTGATGGAGATAGGTGGTGC bc 114 326835 4 104 ATCTGTTGATGGAGATAGGT bc 115 326836 4 105 CATCTGTTGATGGAGATAGG bc 116 326837 4 112 TCTGTCTCATCTGTTGATGG bc 117 326838 4 115 TAGTCTGTCTCATCTGTTGA bc 118 326839 4 116 GTAGTCTGTCTCATCTGTTG bc 119 326840 4 121 GCTCTGTAGTCTGTCTCATC + 120 326841 4 124 GCAGCTCTGTAGTCTGTCTC +++++ 121 326842 4 127 GCTGCAGCTCTGTAGTCTGT bc 122 326843 4 130 AAGGCTGCAGCTCTGTAGTC +++ 123 326844 4 135 GTGCAAAGGCTGCAGCTCTG +++ 124 326845 4 141 TTCCCCGTGCAAAGGCTGCA bc 125 326846 4 145 TTTCTTCCCCGTGCAAAGGC ++ 126 326847 4 148 TGCTTTCTTCCCCGTGCAAA bc 127 326848 4 158 ATGTCTGCCCTGCTTTCTTC bc 128 326849 4 172 GCATTGGTATCGCAATGTCT + 129 326850 4 176 TTCTGCATTGGTATCGCAAT bc 130 326851 4 180 TCTCTTCTGCATTGGTATCG bc 131 326852 4 200 AAGTTGGTGTCTCTCTTCCT ++ 132 326853 4 203 GGGAAGTTGGTGTCTCTCTT +++++ 133 326854 4 225 TTTACAGCAGAAGATGCAGA bc 134 326855 4 229 AGCATTTACAGCAGAAGATG ++ 135 326856 4 231 ACAGCATTTACAGCAGAAGA + 136 326857 4 233 TTACAGCATTTACAGCAGAA +++ 137 326858 4 238 AATTGTTACAGCATTTACAG bc 138 326859 4 242 TGGGAATTGTTACAGCATTT bc 139 326860 4 245 CACTGGGAATTGTTACAGCA bc 140 326861 4 252 GATACCACACTGGGAATTGT bc 141 326862 4 256 AACAGATACCACACTGGGAA bc 142 326863 4 258 GCAACAGATACCACACTGGG +++++ 143 326864 4 261 TTTGCAACAGATACCACACT +++ 144 326865 4 263 GTTTTGCAACAGATACCACA + 145 326866 4 266 TATGTTTTGCAACAGATACC bc 146 326867 4 275 GCTCTAGGCTATGTTTTGCA +++++ 147 326868 4 287 GGTCAGGATGTGGCTCTAGG +++++ 148 326869 4 291 GAGAGGTCAGGATGTGGCTC +++++ 149 326870 4 293 TAGAGAGGTCAGGATGTGGC ++++ 150 326871 4 296 GTGTAGAGAGGTCAGGATGT +++++ 151 326872 4 334 AGGGCAGGAATAAATAATGG bc 152 326873 4 336 GGAGGGCAGGAATAAATAAT bc 153 326874 4 355 ATTTCAAGGTCATTGGTGGG bc 154 326875 4 357 TTATTTCAAGGTCATTGGTG +++ 155 326876 4 362 CGTCTTTATTTCAAGGTCAT +++++ 156 326877 4 367 AAAATCGTCTTTATTTCAAG bc 157 326878 4 369 ATAAAATCGTCTTTATTTCA bc 158 326879 4 371 AAATAAAATCGTCTTTATTT bc 159 326880 5 512 GGGTGCTCACCTGTTGATGG bc 160 326881 5 1239 TGCTTACTGCTGACTGCCAT + 161 326882 5 1244 ATGCATGCTTACTGCTGACT + 162 326883 5 1259 GCCTATGTGTATCATATGCA ++ 163 326884 5 1347 CAGCAGAACCTCTACAGCCC bc 164 326885 5 1686 TCTGTCTCATCTGTGAAAGC bc 165 326886 5 1746 ATGCTCTTACCGCAATGTCT bc 166 326887 5 1829 GCATTGGTATCTGTGTAAAG + 167

[0109] Each active target segment was targeted by multiple, active antisense oligonucleotides. This current example includes identification of active target segments identified on SEQ ID NO: 4, which active target segments are referred to as Regions L-0. The active target segments include nucleotides 287 to 315 for Region L, nucleotides 258 to 315 for Region M, nucleotides 1 to 74 for Region N, and nucleotides 80 to 199 for Region O. Each of the oligonucleotides tested within Region L reduced expression by at least 70% (++++) and three-fourths of them by at least 75% (+++++). Two-thirds of the oligonucleotides tested within Region M reduced expression by at least 70% (++++). About one-half of the oligonucleotides tested in Region N reduced expression by at least 70% (++++). This screen also identified regions with low activity when targeted with an antisense oligonucleotide. In Region O more than two-thirds of the oligonucleotides tested reduced expression by less than 50% (bc) and more than three-fifths by less than 35% (bc).

[0110] The active target segments for SEQ ID NO: 5 are referred to as Regions P-R. Region P includes nucleotides 1942 to 1972 and all of the oligonucleotides tested in this region reduce expression by at least 70% (+++). Region Q includes nucleotides 1991 to 2047 and three-fourths of the oligonucleotides tested in this region reduced expression by less than 50% (bc), and thus is considered a low-activity region. Similarly, for Region R, which includes nucleotides 474 to 531, more than two-thirds of the oligonucleotides tested in this region reduce expression by less than 50% (bc).

[0111] Thus, Applicant has discovered regions of the hepcidin mRNA that are highly responsive to antisense reduction and regions of the hepcidin mRNA that are less responsive to antisense reduction. Identification of these regions allows for the design of antisense oligonucleotides that will modulate the expression of hepcidin. One skilled in the art will readily recognize that the percent reduction is referred to herein as a relative amount of reduction compared to other oligonucleotides tested against SEQ ID NOS: 4 or 5. Assay to assay variations in conditions will results in changes from the absolute data received, however, the general trends and relative reduction will be substantially the same.

EXAMPLE 5

Antisense Oligonucleotides From About 12 To About 35 Nucleobases

[0112] As stated above, antisense oligonucleotides directed to a target or more preferably to an active target segment can be from about 12 to about 35 linked nucleobases. The following Table 5a provides a non-limiting example of such antisense oligonucleotides targeting SEQ ID NO 4.

TABLE-US-00005 TABLE 5a Antisense Oligonucleotides from about 12 to about 35 Nucleobases Sequence Length GGTCAGGATGTGGCTCTAGG 20 nucleobases (SEQ ID NO: 148) GGTCAGGATGTGGCT 15 nucleobases (SEQ ID NO: 170) CAGGATGTGGCTCTA 15 nucleobases (SEQ ID NO: 171) GTCAGGATGTGG 12 nucleobases (SEQ ID NO: 172) GAGAGGTCAGGATGTGGCTCTAGGCTATGTTTTGCA 35 nucleobases (SEQ ID NO: 173) GTCAGGATGTGGCTCTAGGCTATGTTTT 27 nucleobases (SEQ ID NO: 174) TGTGGCTCTAGGCTATGTTTTGC 22 nucleobases (SEQ ID NO: 175)

[0113] Antisense oligonucleotides directed to a target or more preferably to an active target segment can also contain mismatched nucleobases when compared to the target sequence. The following Table 5b provides a non-limiting example of such antisense oligonucleotides targeting nucleobases 287 to 306 of SEQ ID NO 4. Mismatched nucleobases are underlined.

TABLE-US-00006 TABLE 5b Antisense Oligonucleotides from about 1-3 Nucleobases Mismatched to the Target Sequence Number of mismatches to Sequence SEQ ID NO: 4 GGTCAGGATGTGGCTCTAGG None (SEQ ID NO: 148) GGTCAGGATGTGGCTCAAGG One mismatch (SEQ ID NO: 176) GGTCAGGAAGTGGCTCTAGG One mismatch (SEQ ID NO: 177) GGTCAGTTTGTGGCTCTAGG Two mismatches (SEQ ID NO: 178) GGTGAGGATGTGGCTCCAGG Two mismatches (SEQ ID NO: 179) GGTCTGGATGTGCCTCTCGG Three mismatches (SEQ ID NO: 180)

EXAMPLE 6

Effects of Antisense Oligonucleotides Targeted To Mouse Hepcidin mRNA: In Vivo Evaluation In Normal Mice

[0114] C57/131/6 mice were divided into treatment groups for injection with one of the following: antisense oligonucleotides targeting hepcidin mRNA (SEQ ID NOS: 4 and 5) at 50 mg/kg; antisense oligonucleotides that do not target hepcidin mRNA at 50 mg/kg; or saline. The animals received an initial dose and then were dosed twice per week for two weeks (5 doses total). The antisense oligonucleotides used in the study were all chimeric oligonucleotides ("gapmers") 20 nucleobases in length, composed of a central "gap" region consisting of 10 2'-deoxynucleotides, flanked on both the 5' and 3' ends by 5-nucleotide "wings." The wings are composed of 2'-methoxyethoxy nucleotides (2'-MOE). The internucleoside linkages are phosphorothioate (P=S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines. The sequences of the oligonucleotide are presented in Table 6.

TABLE-US-00007 TABLE 6 Antisense oligonucleotides used in normal mouse in vivo screen assay SEQ ID Sequence NO: Targeted to hepcidin mRNA GGTCAGGATGTGGCTCTAGG 148 CGTCTTTATTTCAAGGTCAT 156 GTGCCATCATGCCTTCTGTT 95 Not targeted to hepcidin mRNA CCTTCCCTGAAGGTTCCTCC 168 GCTCATAGAACTGTCAGGCT 169

[0115] At the end of the treatment period animals were sacrificed and tissues were harvested for assessment of levels of hepcidin mRNA in the liver, levels of hepcidin polypeptide in the liver and organ weight (specifically spleen and liver. Over the course of the study, data collected to evaluate overt toxicity, changes in body weight, changes in organ weight (specifically spleen and liver), changes in food intake, blood levels of aspartate aminotransferase (AST) and blood levels of alanine aminotransferase (ALT). There were no signs of overt toxicity or in changes to organ or body weight. There was an increase in levels of AST and ALT in the mice receiving one of the antisense oligonucleotides (SEQ ID NO: 95); however, levels for the other hepcidin targeting antisense oligonucleotides remained comperable to the saline control. Liver hepcidin levels were reduced by 60-70 percent for the hepcidin targeting antisense oligonucleotides compared to controls. A similar reduction was observed for liver hepcidin polypeptides. Hepcidin polypeptide levels were determined by Western blot of whole-liver lysates. Briefly, 50 .micro.g of whole-liver lysate was electrophoresed on a 16% TG-gel. Hepcidin was detected with hepcidin antibody (Alpha Diagnostics, HEPC11-A, 0.3 .micro.g/ml). FIG. 1. Thus, antisense oligonucleotides targeting hepcidin mRNA can result in a significant reduction in the levels of that mRNA and in turn the expression thereof.

Example 7

Dose-Response of Antisense Oligonucleotide Targeting Hepcidin mRNA In Normal Mice

[0116] C57/B1/6 mice were dosed with 10, 25, 50 or 75 mg/kg of antisense oligonucleotide targeting hepcidin mRNA (SEQ ID NO: 145). C57/B1/6 control mice received either saline or 10, 25, 50 or 75 mg/kg of an antisense oligonucleotide not targeted to hepcidin mRNA (SEQ ID NO: 168) The groups were given an initial dose and then were dosed twice per week for 2 weeks. Liver hepcidin mRNA and serum iron levels were measured at the end of the study. All antisense oligonucleotides used in this example were chimeric oligonucleotides ("gapmers") 20 nucleobases in length, composed of a central "gap" region consisting of 10 2'-deoxynucleotides, flanked on both the 5' and 3' ends by 5-nucleotide "wings." The wings are composed of 2'-methoxyethoxy nucleotides (2'-MOE). The internucleoside linkages are phosphorothioate (P=S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines.

[0117] There was a reduction in liver hepcidin mRNA for the group receiving the antisense oligonucleotide targeting hepcidin as compared to saline control. A reduction was not observed for the control antisense oligonucleotide group as shown in FIG. 2. Also observed was a corresponding increase in serum iron levels for the treatment groups compared to saline. This same increase was not seen in the control oligonucleotide group. These data demonstrate that antisense oligonucleotides targeting hepcidin mRNA will cause a decrease in liver hepcidin mRNA and a corresponding increase in serum iron levels.

EXAMPLE 8

Effects of Antisense Reduction of Hepcidin In Normal And LPS Stimulated Mice: Dose-Response

[0118] An ACD mouse model was prepared prior to treatment by injecting C57B1/6 mice (Charles River Labs, Wilmington, Mass.) with lipopolysaccaride (LPS, Sigma-Aldrich, St. Louis, Mo.) for 6 hours (0.1 to 1 mg/kg intraperitoneal injection). (See, e.g. Ganz (2003) Blood, 102(3):783-785, and references therein). Injection of LPS caused an increase in cytokine production in these mice, which in turn resulted in a three fold increase in hepcidin production and an 80% reduction in serum iron levels.

[0119] A normal unstimulated mouse population and an LPS stimulated mouse population were each separated into treatment groups, antisense control groups and saline control groups. Mice in the LPS stimulated population were prepared using a 1 mg/kg intraperitoneal LPS injection for 6 hours. The treatment groups were treated with an antisense oligonucleotide targeted to mouse hepcidin mRNA (SEQ ID NOS: 4 and 5). The antisense control groups were treated with antisense oligonucleotide not targeted to mouse hepcidin. The saline control groups were injected with saline alone. The antisense oligonucleotide targeted to mouse hepcidin mRNA was (SEQ ID NO: 145). The control antisense oligonucleotide was SEQ ID NO 168. All mice were dosed at time point zero and then twice a week for two weeks using either 10, 25, 50 or 75 mg/kg of antisense oligonucleotide; 50 mg/kg of control oligonucleotide or using saline. All antisense oligonucleotides used in this example were chimeric oligonucleotides ("gapmers") 20 nucleobases in length, composed of a central "gap" region consisting of 10 2'-deoxynucleotides, flanked on both the 5' and 3' ends by 5-nucleotide "wings." The wings are composed of 2'-methoxyethoxy nucleotides (2'-MOE). The internucleoside linkages are phosphorothioate (P=S) throughout the oligonucleotide. All cytidine residues are 5-methylcytidines.

[0120] In both the normal mice and the LPS stimulated mice there was a reduction in levels of liver hepcidin mRNA for the 25, 50 and 75 mg/kg doses as compared to saline control. This same reduction was not seen in the antisense oligonucleotide control group. Additionally, there was a corresponding increase in serum iron levels for both the normal and LPS stimulated groups receiving antisense oligonucleotide targeting hepcidin as compared to saline. This same increase was not seen in the antisense oligonucleotide control group. This study shows that hepcidin increases serum iron levels in both normal and LPS stimulated mice. FIG. 3. Antisense oligonucleotides targeting hepcidin mRNA will reduce the levels of hepcidin mRNA in the liver, correlating with an increase in serum iron levels. This effect of the antisense oligonucleotide is observed in normal and LPS stimulated mice (ACD models). Thus, the antisense oligonucleotides are useful for treating subjects having low serum iron levels and disorders associated therewith.

EXAMPLE 9

Effects of Antisense Reduction of Hepcidin In Normal And Chronic Anemia Mice: Turpentine Mouse Model

[0121] The effect of antisense compound treatment on acute and chronic anemia was determined using a turpentine induced anemia mouse model. Male C57B1/6 mice (Charles River Laboratories, Wilmington, Mass.) weighing approximately 25 g were separated into one of a control group, an oligonucleotide control group or a treatment group. The control group mice received either no treatment (n=5) or treatment with 100 microliters turpentine only (n=5). The oligonucleotide control group mice received 100 microliters turpentine and a dosing regimen comprising either 50 mg/kg/dose of control compound (n=5) or 75 mg/kg/dose of control compound (n=5). The control compound was an antisense compound that is not targeted to a nucleic acid encoding hepcidin (CCTTCCCTGAAGGTTCCTCC, SEQ ID NO: 168). The treatment group mice received 100 microliters of turpentine and a dosing regimen comprising either 10 mg/kg/dose of treatment compound (n=5), 25 mg/kg/dose of treatment compound (n=5), 50 mg/kg/dose of treatment compound (n=5), or 75 mg/kg/dose of treatment compound (n=4). The treatment oligonucleotide is Cmpd 326868 (SEQ ID NO: 148). The turpentine injection stimulates an inflammatory response in the mice, and subsequently produces anemia. Turpentine mouse models of anemia are discussed in the literature. (Liuzzi, J P, Lichten, L A, Rivera, S, Blanchard, R K, Aydemir, T B, Knutson, M D, Ganz, T, and Cousins, R J, PNAS, v. 102, n. 19, 6843-6848 (2005); Venihaki, M, Dikkes, P, Carrigan, A, and Karalis, K P, J. Clin. Invest., 108:1159-1166 (2001)).

[0122] Mice in all groups were placed on a no-iron diet for two weeks, followed by a low-iron diet. At the same time the low-iron diet was started, the mice were subjected to the appropriate dosing regimen as described above. Briefly, the control group received no oligonucleotide, the oligonucleotide control group received 50 or 75 mg/kg/dose of SEQ ID NO: 168 twice a week for two weeks, and the treatment group received 10, 25, 50 or 75 mg/kg/dose of SEQ ID NO: 148 twice a week for two weeks. After two weeks of the dosing regimen, all mice in the oligonucleotide group and in the treatment group received an injection comprising 100 microliters of turpentine (Sigma-Aldrich, St. Louis, Mo.). In the control group, only half of the mice population received an injection comprising 100 microliters of turpentine and the other half of the population received a saline injection only. The injections were delivered subcutaneously to the left hind limb of each mouse. To obtain acute anemia endpoints, blood was drawn at 16 hours post turpentide injection. For chronic anemia endpoints, the dosing regimen described above continued for another two weeks after turpentine injection. Blood was drawn at two weeks post-turpentine injection and at 24 hours after the final oligonucleotide injection was delivered. Mice were then sacrificed.

[0123] Analysis of the serum iron from acute blood draw showed that the turpentine injection produced a significant reduction in the serum iron levels at 16 hours post turpentine injection. The serum iron level reduction was reversed in a dose responsive manner for the treatment group. At two weeks post injection there was a dose dependent improvement in the clinical endpoints of anemia compared to the control group and the oligonucleotide control group. Table 7 and FIGS. 4a-d shows the dose dependent improvement in serum iron levels, red blood cell count, hematocrit levels and hemoglobin levels.

TABLE-US-00008 TABLE 7 The Effect of Hepcidin ASO in a Chronic Turpentine Model: Serum Iron, Red Blood Cell (RBC) Count, Hemoglobin Levels and Hematocrit Levels Oligonucleotide Turpentine Control Group - Control Group Treatment Group (.micro.l) 0 100 100 100 100 100 100 100 CMPD none none 50 mg/kg 75 mg/kg 10 mg/kg 25 mg/kg 50 mg/kg 75 mg/kg 141923 141923 326868 326868 326868 326868 Serum Iron 190.6 170.6 186.0 208.2 182.8 260.8 266.6 325.5 (.micro.g/ (.+-.24.1) (.+-.28.1) (.+-.44.4) (.+-.40.0) (.+-.44.3) (.+-.22.9) (.+-.46.0) (.+-.55.7) dL) (SD) RBC (10.sup.6/ 10.4 9.4 9.9 9.8 9.4 10.2 10.2 10.6 .micro.l) (SD) (.+-.0.4) (.+-.1.0) (.+-.0.2) (.+-.0.7) (.+-.0.5) (.+-.0.3) (.+-.0.3) (.+-.0.8) Hemoglobin 14.8 13.0 13.3 13.2 13.1 14.0 14.0 14.3 (g/dL) (SD) (.+-.0.8) (.+-.1.2) (.+-.0.4) (.+-.0.8) (.+-.0.6) (.+-.0.5) (.+-.0.3) (.+-.0.9) Hematocrit 53.4 47.6 49.8 47.8 47.6 51.2 51.0 51.5 (%) (SD) (.+-.2.1) (.+-.2.6) (.+-.1.1) (.+-.3.3) (.+-.1.9) (.+-.1.9) (.+-.1.7) (.+-.3.5)

[0124] The turpentine mouse model of anemia provides data for endpoints of chronic anemia, such as RBC count, hemoglobin levels and hematocrit levels, as well as serum iron levels. Treating these anemic mice with an antisense compound that targets and inhibits expression of nucleic acids encoding hepcidin polypeptide results in a dose dependent improvement in these endpoints.

EXAMPLE 10

Effects of Antisense Reduction of Hepcidin In Chronic Anemia (Turpentine Mouse Model) Treated With Erythropoietin

[0125] The effect of antisense compound treatment on chronic anemia, in combination with erythropoietin (EPO) treatment was determined using the chronic turpentine induced anemia mouse model. Male C57B1/6 mice (Charles River Laboratories, Wilmington, Mass.) weighing approximately 25 g were fed a "no iron" diet (2.7 ppm iron) for 2 weeks and subsequently switched to a "low iron" diet (47 ppm iron) for the remainder of the study. Upon switch to the low iron diet, two weeks prior to sacrifice, mice were treated via subcutaneous injection with saline (vehicle) or a single 100 .micro. L dose of turpentine. Hepcidin antisense oligonucleotide (CMPD # 326868, 50 mg/kg, which is SEQ ID NO: 148) was administered via subcutaneous injection to the appropriate groups of animals (as indicated in FIG. 5a-5c), two times per week, beginning 24 hours following turpentine treatment. EPO was administered daily, for the last four days of the study, by intraperitoneal injection to indicated animals at indicated doses (0-25 units/day). Thus, in this study, in indicated mice, regular treatment with antisense oligonucleotide targeted to hepcidin began prior to regular treatment with erythropoietin. After the first administration, each treatment was continued at regular intervals for the remainder of the study. At the end of the treatment period, animals were sacrificed and tissues and blood taken for assessment of levels hepcidin mRNA and phenotypic endpoints. Results from this study showed that hepcidin ASO in combination with EPO was well tolerated. Hepcidin mRNA levels in the liver, serum iron (.micro.g/mL) and % reticulocytes (immature red blood cells) were measured as endpoints. The results are shown in FIG. 5a-5c.

[0126] As shown in FIG. 5, in this study using the turpentine mouse model of chronic anemia, erythropoietin treatment decreased liver hepcidin mRNA expression and serum iron levels and increased reticulocyte counts. Administration of the antisense oligonucleotide targeted to hepcidin caused decreased liver hepcidin mRNA, but an increase in serum iron. Administration of an antisense oligonucleotide targeted to hepcidin in addition to erythropoietin resulted in a further increase in reticulocyte counts compared to erythropoietin treatment alone. Thus the effects of erythropoietin and the antisense compound targeted to hepcidin were additive. These results show that treatment with both erythropoietin and an antisense compound targeted to hepcidin is effective in improving endpoints of chronic anemia in a mouse model for hypoferremia and anemia associated with inflammation.

[0127] It is contemplated herein that a further experiment, evalutating the effectiveness of combination therapy for treating anemia or increasing red blood cells could be performed, according to the methods and using the mouse model described above, by evaluating multiple experimental mice groups.

[0128] In such an experiment, a control group would receive only saline. Experimental groups would all receive turpentine. A first experimental group would receive only turpentine (no treatment or therapy). A second experimental group would receive erythropoietin treatment continuously, meaning regularly, for over a week, prior to receiving antisense compound targeted to hepcidin, and would continue to receive erythropoietin treatment throughout the treatment period with antisense compound. Thus, this second experimental group will have been receiving erythropoietin therapy prior to administration of the antisense compound. Erythropoietin therapy treatment regimens are well known in the art. Thus, in the second experimental group, the antisense compound and the erythropoietin would be administered in separate formulations, at distinct timepoints. A third experimental group would have received erythropoietin therapy at some time prior to receiving antisense compound. A fourth experimental group would receive erythropoietin therapy and antisense compound treatment simultaneously. Thus, in this group, the group would begin erythropoietin therapy simultaneously with the administration of the antisense compound. In this case, the two treatments could be administered in separate formulations or in single formulations. A fifth experimental group would receive antisense compound prior to erythropoietin therapy. Thus, the second through fifth experimental groups would be receiving a combination therapy, wherein they would receive antisense compound and be further receiving erythropoietin. A sixth experimental group would receive only erythropoietin. A seventh experimental group would receive only antisense compound.

[0129] It is further contemplated that a combination therapy method could be used for preventing, treating or ameliorating anemia or for increasing red blood cell counts in a patient. It is contemplated that this combination method could include administration of an antisense compound targeted to hepcidin and an ESA. It is contemplated that the ESA used in such a therapy could be erythropoietin, recombinant human erythropoietin, EPOGEN or PROCRIT. It is further contemplated that such a combination therapy method could comprise administration of the antisense compound and erythropoietin simultaneously. Alternatively, administration of the erythropoietin and antisense compound could be done at distinct timepoints.

[0130] If administered simultaneously, the antisense compound and erythropoietin could be given in a single formulation, or in separate formulations. Also, this combination therapy could be used to treat a patient who is receiving erythropoietin. The patient could be receiving erythropoietin prior to initiation of the combination therapy, or the patient could begin receiving the erythropoietin at the same time the combination therapy is initiated.

[0131] It is contemplated that an antisense compound used in this therapy could be an antisense oligonucleotide targeted to a nucleic acid molecule encoding hepcidin. Alternatively, the antisense compound could be double stranded. It is contemplated that the double stranded antisense compound could be an siRNA.

Sequence CWU 1

1

1801588DNAHomo sapiens 1gagcctccca cgtggtgtgg atgaggaggc agatggcagg gcctgtgcat ttctgtgctt 60gagtgggcct tgaaagtggt tcagcaacca ggaagaagtg ttcattcctc gacaacaaca 120tccccgggct ctggtgactt ggctgacact ggatggccct ggaatgaaaa aggcaaagag 180gcaaaatgtg caagggccca tctggaacca aggccccata aaagcgactg tcactcggtc 240ccagacacca gagcaagctc aagacccagc agtgggacag ccagacagac ggcacgatgg 300cactgagctc ccagatctgg gccgcttgcc tcctgctcct cctcctcctc gccagcctga 360ccagtggctc tgttttccca caacagacgg gacaacttgc agagctgcaa ccccaggaca 420gagctggagc cagggccagc tggatgccca tgttccagag gcgaaggagg cgagacaccc 480acttccccat ctgcattttc tgctgcggct gctgtcatcg atcaaagtgt gggatgtgct 540gcaagacgta gaacctacct gccctgcccc cgtcccctcc cttcctta 5882430DNAHomo sapiens 2gactgtcact cggtcccaga caccagagca agctcaagac ccagcagtgg gacagccaga 60cagacggcac gatggcactg agctcccaga tctgggccgc ttgcctcctg ctcctcctcc 120tcctcgccag cctgaccagt ggctctgttt tcccacaaca gacgggacaa cttgcagagc 180tgcaacccca ggacagagct ggagccaggg ccagctggat gcccatgttc cagaggcgaa 240ggaggcgaga cacccacttc cccatctgca ttttctgctg cggctgctgt catcgatcaa 300agtgtgggat gtgctgcaag acgtagaacc tacctgccct gcccccgtcc cctcccttcc 360ttatttattc ctgctgcccc agaacatagg tcttggaata aaatggctgg ttcttttgtt 420ttccaaaaaa 43035225DNAHomo sapiens 3atgcctggta gggctggggg ctgctcctgt gtctccccag gtgagcacac ccctattcac 60tgggccctgc ttcagcctgc agcacccttc aactcccagg agctgggctt gccactctgc 120tcaccttgtg gagctccatc tgcctttcct ccccaattcc cccactccct gcactcgtct 180cttcccacaa gagccctgtc tccttttcct agctattccc atctgaggcc atctttattc 240atttagtttt tagagacagg gtttcactct cacccaggct ggggtgcagt ggcacacaat 300cacggctcac tgcagccttg accaactaca ggtgcgtagc accacagcca agtttttgta 360tagatggggt ctcgctttgt tacccaggct gtgacaagag gagcctccca cgtggtgtgg 420atgaggaggc agatggcagg gcctgtgcat ttctgtgctt gagtgggcct tgaaagtggt 480tcagcaacca ggaagaagtg ttcattcctc gacaacaaca tccccgggct ctggtgactt 540ggctgacact ggatggccct ggaatgaaaa aggcaaagag gcaaaatgtg caagggccca 600tctggaacca aggtttgttg atcccctggg ccgtgtgcac cctgagctgg gcctggtagt 660ggaaaggaat gaaggcactg cagtcaggca gcctgggttc atcccccagc tagtggtgtc 720ctaaggaacc ggctccccaa aaacatccct ggcttgtagt gcttgccaat ttctgggtgt 780caagactccc actgctgctg atttcaggat accagcatga tgccactgaa tgcagagttt 840cgagatgtgc atggtctgct atgttgagcc aggtctagca taccgctgtg ccctgctgtg 900ttttagggga gatggggaaa cctggtgggt aagagcaaaa gccctggagt caggctgtcc 960aggctagaat ctcagctctg cctctggctg agcaagcttg ggccatgccc tgatctctgc 1020cttcagtgcc ttttctgtaa agtgaaggaa atgagtgtcc gacggggagg aggttcctaa 1080aagggagcag ggtctgggga gcccaggcct ctggggttgg gtgactgaga aggcagcccc 1140tgaatacaga gcagagctga aggtggggca gtaagtgctg ctgggagaac aggcagcaca 1200ggctgagttg gtgcagaagt gagtcaacat atgtgccatc gtataaaatg tactcatcgg 1260actgtagatg ttagctatta ctattactgc tattttatgt tttatagaca gggtctcact 1320ctgtcaccca ggctggagtg cagtcacaca atcatagctc actgcaacct cagcctcctg 1380ggcttaagcg atctgcctca gcctcccaag tagctgggac tacagatgtg tgccaccacg 1440cctggctaaa tttgtttaaa attttttttg tagagatggg gtctccctat gttgcccagg 1500ctagtcttga acttctgggc tcaagcgacc ctcctgcctt ggcctcccaa attgctggga 1560ttacaggcat aagccactgt gctgggccat attactgctg tcatttatgg ccaaaagttt 1620gctcaaacat tttccagtta ccagagccac atctcaaggg tctgacactg ggaaaacacc 1680acgtgcggat cgggcacacg ctgatgcttg ccctgctcag ggctatctag tgttccctgc 1740cagaacctat gcacgtgtgg tgagagctta aagcaatgga tgcttccccc aacatgccag 1800acactcctga ggagcctggc ggctgctggc catgccccgt gtgcatgtag gcgatgggga 1860agtgagtgga ggagagcgga accttgattc tgctcatcaa actgcttaac cgctgaagca 1920aaagggggaa cttttttccc gatcagcaga atgacatcgt gatggggaaa gggctcccca 1980gatggctggt gagcagtgtg tgtctgtgac cccgtctgcc ccaccccctg aacacacctc 2040tgccggctga gggtgacaca accctgttcc ctgtcgctct gttcccgctt atctctcccg 2100ccttttcggc gccaccacct tcttggaaat gagacagagc aaaggggagg gggctcagac 2160caccgcctcc cctggcaggc cccataaaag cgactgtcac tcggtcccag acaccagagc 2220aagctcaaga cccagcagtg ggacagccag acagacggca cgatggcact gagctcccag 2280atctgggccg cttgcctcct gctcctcctc ctcctcgcca gcctgaccag tggctctgtt 2340ttcccacaac aggtgagagc ccagtggcct gggtccttag cagggcagca gggatgggag 2400agccaggcct cagcctaggg cactggagac acccgagcac tgagcagagc tcaggacgtc 2460tcaggagtac tggcagctga acaggaacca ggacaggcac ggtggctcat gcctgtaatc 2520ccagcacttt gggaggttga ggcaggcagc ccacttgagg tcagtttgag accagcctgg 2580ccaacatggt aaaaccccgt ctctactaaa aatacaaaag ttagccaggc ttggtggcag 2640gtgcctgtaa tcccagctac tcgggagact gaggcaggag aattgcttga acccgcaagg 2700tggaggttgc acagtgagct gagattgcac cactgcactc cagcctggca acagagcaag 2760actccatctc caaaaaagaa cagaaatcaa tgaagcaccg agtgacaggg actggaaggt 2820cctaattcca tgggtattta cggaacccct acgccgtgtg gagtcttatt ctagacagtg 2880gggacgaggc catgaacaag gtagatgaga gaggagattt ctccatcctg gtcagggaat 2940ttgttaaaga ctgatgaaaa catgaataaa taattgtgtc tagtacattc tattcgtgaa 3000tctcataaca gacagtggta gagtgaccgt gacccattcg ccacacagta gagtcacttt 3060tttggtttgt tttttagaga cagggtcttc ctctgttgct gaggctggag tgcagtggtg 3120cagtcatagt tcactgcagc ctcaacctcc tgtgctcaag caatcctccc acctcagcgt 3180cccaagtagc tgggacagca ggcacatgcc acgggttggg ggaccacagg catggtcaag 3240gggctggcag tcaagcaagt gtttcatgag aaagtgacag ttgaccttcg tcttggaggg 3300tgagagatgg aggcagcaaa gacctaagga gaggacaagc cagcatagcc cagggtcagg 3360ctgaacaaga ggagatggtg ggacttgggg ataaggctga ggggtgggca gtccctaagt 3420cttgtgggca accatgcaga cactgatttt tccttggaat aaagaggaag cccccataag 3480cttttttttt tttttctgag atagggtctc gctctgtcgt tcaggctggt gtgcagtggc 3540atcatctggg ctcactgcaa cctccgcctc ccgggttcaa gcaattctcc tgcctcagct 3600tcccgagcag ctgggattac aggcggctgc caccacgccc ggctaatttt tgttttttta 3660gtagagacag ggtttcacca tgttggccag actggtcttg aactcctgac ctcaggtgat 3720tctcccacct cggcttccca aagtgctggg attacaggcg tgagccactg cgcccagcct 3780cctgtaggtt tttaaaatgg agaaaaccac aatctcactg gccatgtttt aaaaaactta 3840atctgccagt caggcaccat ggctcacacc tgtaatccca gagttttggg aggccaaggt 3900aggaagatca gttgagccca ggagttcaag accagcttgg gcaacacaac cagaccccac 3960ctctacaaaa aattaaaaaa ttagccgggt gtggtggcgt gcacctgctg tcccagctac 4020tcgggaagct gaggcgggag catcgcttga gcacaggagg tcaaggctgc agggagctat 4080gactgtgcca ctgcactctg gcctgggcaa cagaggaaga ctctgtctaa aaaacaaaca 4140aaaaaagtga ctctgctgtg tggcaaatgg attgaggggc aagaatgcag ggaggtgtgt 4200taggaggctg gcactggcat ccaggcaggg gaaggtgata tcccaaagaa gagtagcagc 4260tgtggaaaga ggaggaggcg gatctgggag gttttttttt ttaggaaaag ccgcccatgg 4320gaaggtgagc agaagcaaga aagcaaggcc cctcctaaga gtccatttga gctctgggtt 4380taaaccactt ggagaggagc aggttgccgg gagccagtct cagaggtcca ctgggccccc 4440tgccatcctc tgcaccccct tctgctttca cagacgggac aacttgcaga gctgcaaccc 4500caggacagag ctggagccag ggccagctgg atggtgagcg caacagtgat gcctttccta 4560gccccctgct ccctccccat gctaaggccg gttccctgct cacattccct tccttcccac 4620agcccatgtt ccagaggcga aggaggcgag acacccactt ccccatctgc attttctgct 4680gcggctgctg tcatcgatca aagtgtggga tgtgctgcaa gacgtagaac ctacctgccc 4740tgcccccgtc ccctcccttc cttatttatt cctgctgccc cagaacatag gtcttggaat 4800aaaatggctg gttcttttgt tttccaaacc agagtgtctg ttgtcctttc tctctgccga 4860gtgtctgtgc taagagcttg tcctgaccct gccttgcaag caccagtgct tggtgggtca 4920tgtggggctg gtgtgtcctg gaggttgcca ggaaagttgg tgaagaaaat ttgtttctgt 4980tctccccctt catgttgcaa taatagggga tgaaagttaa tgtttcctct ccttgagatc 5040ttcctaaaac agctgtagaa atcagtgcct gtaaggcaag cttgtccaac ctggaggcca 5100catgcagccc tggatggctt tgaatgcacc caacacaaat ttgtagtttc ttaaggcatt 5160atgagatttt tccgcaattt ttttttttct catcagctgt cattagtgtt agtgtgtttt 5220atgtg 52254410DNAMus musculus 4agtccttaga ctgcacagca gaacagaagg catgatggca ctcagcactc ggacccaggc 60tgcctgtctc ctgcttctcc tccttgccag cctgagcagc accacctatc tccatcaaca 120gatgagacag actacagagc tgcagccttt gcacggggaa gaaagcaggg cagacattgc 180gataccaatg cagaagagaa ggaagagaga caccaacttc cccatctgca tcttctgctg 240taaatgctgt aacaattccc agtgtggtat ctgttgcaaa acatagccta gagccacatc 300ctgacctctc tacacccctg cagcccctca accccattat ttattcctgc cctccccacc 360aatgaccttg aaataaagac gattttattt tcaaaaaaaa aaaaaaaaaa 41052449DNAMus musculus 5gacattgctg ggtccttctg agaaacccag tgagtaacag ccatactgaa ggcactgata 60ggggtaaagg tacagttctt ccactcacca atccaatcac tgtttagggg aaagaagggg 120aatttttctg agagccacag tgtgacatca caggtggctg gctgcaggct tgtgtccctg 180gttctgtctg ccccaccctc tggatgcacc tctgctggct gtaggtgaca caaccctgtc 240ccctgtcact gttcccgctt atctctcccg cctgtttggc gccactattt tcttggaaat 300gagtcagagc aaaatggggg tgggtgaggc gcaggtgacc ctcccctacc actagtccca 360taaaaaggac tgggactggc tcctagacag ccaccacaca agtccttaga ctgcacagca 420gaacagaagg catgatggca ctcagcactc ggacccaggc tgcctgtctc ctgcttctcc 480tccttgccag cctgagcagc accacctatc tccatcaaca ggtgagcacc ccaggcccat 540tgtggtggga gagccaggtc ccaggcaggc aggagctgct caccactgag tagttagaat 600ggctcaggag tgatggcagc tgctgacaag gaagagggtg gtccttagtg ggagctggga 660agctgcacag gtgtccttga atagctactc tgttgtccta ctgtggaaaa tgaagcatgg 720tgggagccaa acaaaagtgt tccttggctg tcccaccccg tcagggcatt ctttaagcag 780cctttacatg agtattttat aaagaattac tgtggatagt acaaaagaca atgggcagaa 840aaactctaat gaggaaggac cagaggtggg gctaagaggc tgacagccag gcaaagtatt 900ctatgagaaa atgatacaga agtcgggcag tggtggcaca tgcctttaat cccagcattt 960gggaggcaga ggcaggtgga tttctgagtt tgaatccagc ctggtctaca aagtgagttt 1020caagacagcc agggctacac agagaaatcc tgtctgaaaa aaaaaaaaaa acaaaaaaag 1080aaaaaaaaaa tgatacagaa gggtctggag agatggctta gctgttagga acatttgatg 1140cttgtgcata ggacctagag tcagttccca gcacccatgt ggtggatcac aaccatcctg 1200aactctactt ccagggtacc tgatgccttc tgccctagat ggcagtcagc agtaagcatg 1260catatgatac acataggcac tcaaggcaat cacaagaccc ttggggactg tagggtctga 1320taagtgaagc cagtgttggc aataaagggc tgtagaggtt ctgctgtgcc gagctttgtg 1380gacagctgtg cagatgatga tctgtcctgg aaagccacaa tccagatgaa tgtgctataa 1440gcctttgtgc tatggggtga cctggttata agagataaga tgcagggaaa actgtccgga 1500gtgtgcaaaa gcaaagaaaa gtgggtgctt ttaggagcat ccaaggaatg gtgaggggac 1560acagggcagt aggagcccct tctagaaatt ctgtctaagc acagtcccta aatctctggg 1620gagaagctgg cagagaaaag tcaggaagct atgccgggta ctccacaaga ttcaatacct 1680cttctgcttt cacagatgag acagactaca gagctgcagc ctttgcacgg ggaagaaagc 1740agggcagaca ttgcggtaag agcatctggg actccctccc tgatccccag cctctcccat 1800gcccaagcta ggctgcttac ctctctttct ttacacagat accaatgcag aagagaagga 1860agagagacac caacttcccc atctgcatct tctgctgtaa atgctgtaac aattcccagt 1920gtggtatctg ttgcaaaaca tagcctagag ccacatcctg acctctctac acccctgcag 1980cccctcaacc ccattattta ttcctgccct ccccaccaat gaccttgaaa taaagacgat 2040tttattttca aacctaggtg tctaggtttt ttaccctttt tttttcttgc caaagaaatg 2100gactttttgc aagccctgct gaggacaggc actgagctgg gtttacaggt ctatgtacaa 2160tgaggctgct ggaacgctgg ggaacagcat ttcttgtctg ttcttcactt tgcttcttat 2220ttcaaaagat ctggagttta tgtagcagcc tggaaggaac ctgaaaagac tttccttagt 2280ctttttctgt ctttcattct tcttaaaagt tttttacaaa tgtgtgtatg catgtgtgtg 2340tgtgtgtgtg tgtgtgtgtg agagagagag cacatggaag agccagagta gccccagctg 2400atatgaaact tgctttgtca aactcagaga tcatttgcct ttgccttct 2449620DNAArtificial SequenceOligomeric Compound 6aggaggcgag acacccactt 20722DNAArtificial SequenceOligomeric Compound 7tcccacactt tgatcgatga ca 22824DNAArtificial SequenceOligomeric Compound 8catctgcatt ttctgctgcg gctg 24924DNAArtificial SequenceOligomeric Compound 9tgcagaagag aaggaagaga gaca 241024DNAArtificial SequenceOligomeric Compound 10cacactggga attgttacag catt 241128DNAArtificial SequenceOligomeric Compound 11caacttcccc atctgcatct tctgctgt 281220DNAArtificial SequenceOligomeric Compound 12aatgcacagg ccctgccatc 201320DNAArtificial SequenceOligomeric Compound 13cacagaaatg cacaggccct 201420DNAArtificial SequenceOligomeric Compound 14tcaagcacag aaatgcacag 201520DNAArtificial SequenceOligomeric Compound 15cccactcaag cacagaaatg 201620DNAArtificial SequenceOligomeric Compound 16caaggcccac tcaagcacag 201720DNAArtificial SequenceOligomeric Compound 17actttcaagg cccactcaag 201820DNAArtificial SequenceOligomeric Compound 18gctgaaccac tttcaaggcc 201920DNAArtificial SequenceOligomeric Compound 19tggttgctga accactttca 202020DNAArtificial SequenceOligomeric Compound 20gaggaatgaa cacttcttcc 202120DNAArtificial SequenceOligomeric Compound 21ttgtcgagga atgaacactt 202220DNAArtificial SequenceOligomeric Compound 22tgttgttgtc gaggaatgaa 202320DNAArtificial SequenceOligomeric Compound 23ccaagtcacc agagcccggg 202420DNAArtificial SequenceOligomeric Compound 24gtcagccaag tcaccagagc 202520DNAArtificial SequenceOligomeric Compound 25ccagtgtcag ccaagtcacc 202620DNAArtificial SequenceOligomeric Compound 26gccatccagt gtcagccaag 202720DNAArtificial SequenceOligomeric Compound 27ccagggccat ccagtgtcag 202820DNAArtificial SequenceOligomeric Compound 28tcattccagg gccatccagt 202920DNAArtificial SequenceOligomeric Compound 29ctttttcatt ccagggccat 203020DNAArtificial SequenceOligomeric Compound 30ggcccttgca cattttgcct 203120DNAArtificial SequenceOligomeric Compound 31ccagatgggc ccttgcacat 203220DNAArtificial SequenceOligomeric Compound 32tggttccaga tgggcccttg 203320DNAArtificial SequenceOligomeric Compound 33ggccttggtt ccagatgggc 203420DNAArtificial SequenceOligomeric Compound 34gtgacagtcg cttttatggg 203520DNAArtificial SequenceOligomeric Compound 35accgagtgac agtcgctttt 203620DNAArtificial SequenceOligomeric Compound 36ctgggaccga gtgacagtcg 203720DNAArtificial SequenceOligomeric Compound 37cagggcaggt aggttctacg 203820DNAArtificial SequenceOligomeric Compound 38tgtctgggac cgagtgacag 203920DNAArtificial SequenceOligomeric Compound 39tctggtgtct gggaccgagt 204020DNAArtificial SequenceOligomeric Compound 40ctgggagctc agtgccatcg 204120DNAArtificial SequenceOligomeric Compound 41cagatctggg agctcagtgc 204220DNAArtificial SequenceOligomeric Compound 42agagccactg gtcaggctgg 204320DNAArtificial SequenceOligomeric Compound 43aaaacagagc cactggtcag 204420DNAArtificial SequenceOligomeric Compound 44gtgggaaaac agagccactg 204520DNAArtificial SequenceOligomeric Compound 45gctctgcaag ttgtcccgtc 204620DNAArtificial SequenceOligomeric Compound 46ttgcagctct gcaagttgtc 204720DNAArtificial SequenceOligomeric Compound 47ggaacatggg catccagctg 204820DNAArtificial SequenceOligomeric Compound 48cctctggaac atgggcatcc 204920DNAArtificial SequenceOligomeric Compound 49ggaagtgggt gtctcgcctc 205020DNAArtificial SequenceOligomeric Compound 50cagcagaaaa tgcagatggg 205120DNAArtificial SequenceOligomeric Compound 51gcagccgcag cagaaaatgc 205220DNAArtificial SequenceOligomeric Compound 52tgacagcagc cgcagcagaa 205320DNAArtificial SequenceOligomeric Compound 53atcgatgaca gcagccgcag 205420DNAArtificial SequenceOligomeric Compound 54cacactttga tcgatgacag 205520DNAArtificial SequenceOligomeric Compound 55catcccacac tttgatcgat 205620DNAArtificial SequenceOligomeric Compound 56cagcacatcc cacactttga 205720DNAArtificial SequenceOligomeric Compound 57tcttgcagca catcccacac 205820DNAArtificial SequenceOligomeric Compound 58ttattccaag acctatgttc 205920DNAArtificial SequenceOligomeric Compound 59cagccatttt attccaagac 206020DNAArtificial SequenceOligomeric Compound 60agaaccagcc attttattcc 206120DNAArtificial SequenceOligomeric Compound 61acaaaagaac cagccatttt 206220DNAArtificial SequenceOligomeric Compound 62cctgaccagg atggagaaat 206320DNAArtificial

SequenceOligomeric Compound 63caaattccct gaccaggatg 206420DNAArtificial SequenceOligomeric Compound 64tttaacaaat tccctgacca 206520DNAArtificial SequenceOligomeric Compound 65cagtctttaa caaattccct 206620DNAArtificial SequenceOligomeric Compound 66gttttcatca gtctttaaca 206720DNAArtificial SequenceOligomeric Compound 67tagacacaat tatttattca 206820DNAArtificial SequenceOligomeric Compound 68tgtactagac acaattattt 206920DNAArtificial SequenceOligomeric Compound 69tagaatgtac tagacacaat 207020DNAArtificial SequenceOligomeric Compound 70attcacgaat agaatgtact 207120DNAArtificial SequenceOligomeric Compound 71atgagattca cgaatagaat 207220DNAArtificial SequenceOligomeric Compound 72atgggtcacg gtcactctac 207320DNAArtificial SequenceOligomeric Compound 73ggcgaatggg tcacggtcac 207420DNAArtificial SequenceOligomeric Compound 74tgtgtggcga atgggtcacg 207520DNAArtificial SequenceOligomeric Compound 75cagccccttg accatgcctg 207620DNAArtificial SequenceOligomeric Compound 76tgtcactttc tcatgaaaca 207720DNAArtificial SequenceOligomeric Compound 77tccaagacga aggtcaactg 207820DNAArtificial SequenceOligomeric Compound 78ttgttcagcc tgaccctggg 207920DNAArtificial SequenceOligomeric Compound 79ttatccccaa gtcccaccat 208020DNAArtificial SequenceOligomeric Compound 80cctcagcctt atccccaagt 208120DNAArtificial SequenceOligomeric Compound 81tagggactgc ccacccctca 208220DNAArtificial SequenceOligomeric Compound 82ctgcatggtt gcccacaaga 208320DNAArtificial SequenceOligomeric Compound 83agtgtctgca tggttgccca 208420DNAArtificial SequenceOligomeric Compound 84aaatcagtgt ctgcatggtt 208520DNAArtificial SequenceOligomeric Compound 85aggaaaaatc agtgtctgca 208620DNAArtificial SequenceOligomeric Compound 86tccattttaa aaacctacag 208720DNAArtificial SequenceOligomeric Compound 87gccagtgaga ttgtggtttt 208820DNAArtificial SequenceOligomeric Compound 88acatggccag tgagattgtg 208920DNAArtificial SequenceOligomeric Compound 89ggcatccagc tggccctggc 209020DNAArtificial SequenceOligomeric Compound 90tgctgtgcag tctaaggact 209120DNAArtificial SequenceOligomeric Compound 91tgttctgctg tgcagtctaa 209220DNAArtificial SequenceOligomeric Compound 92cttctgttct gctgtgcagt 209320DNAArtificial SequenceOligomeric Compound 93catgccttct gttctgctgt 209420DNAArtificial SequenceOligomeric Compound 94atcatgcctt ctgttctgct 209520DNAArtificial SequenceOligomeric Compound 95gtgccatcat gccttctgtt 209620DNAArtificial SequenceOligomeric Compound 96gagtgccatc atgccttctg 209720DNAArtificial SequenceOligomeric Compound 97gctgagtgcc atcatgcctt 209820DNAArtificial SequenceOligomeric Compound 98gtgctgagtg ccatcatgcc 209920DNAArtificial SequenceOligomeric Compound 99cgagtgctga gtgccatcat 2010020DNAArtificial SequenceOligomeric Compound 100gggtccgagt gctgagtgcc 2010120DNAArtificial SequenceOligomeric Compound 101gcctgggtcc gagtgctgag 2010220DNAArtificial SequenceOligomeric Compound 102ggcagcctgg gtccgagtgc 2010320DNAArtificial SequenceOligomeric Compound 103agacaggcag cctgggtccg 2010420DNAArtificial SequenceOligomeric Compound 104gcaggagaca ggcagcctgg 2010520DNAArtificial SequenceOligomeric Compound 105ctggcaagga ggagaagcag 2010620DNAArtificial SequenceOligomeric Compound 106ctgctcaggc tggcaaggag 2010720DNAArtificial SequenceOligomeric Compound 107ggtgctgctc aggctggcaa 2010820DNAArtificial SequenceOligomeric Compound 108gtggtgctgc tcaggctggc 2010920DNAArtificial SequenceOligomeric Compound 109aggtggtgct gctcaggctg 2011020DNAArtificial SequenceOligomeric Compound 110taggtggtgc tgctcaggct 2011120DNAArtificial SequenceOligomeric Compound 111ataggtggtg ctgctcaggc 2011220DNAArtificial SequenceOligomeric Compound 112gataggtggt gctgctcagg 2011320DNAArtificial SequenceOligomeric Compound 113tgatggagat aggtggtgct 2011420DNAArtificial SequenceOligomeric Compound 114ttgatggaga taggtggtgc 2011520DNAArtificial SequenceOligomeric Compound 115atctgttgat ggagataggt 2011620DNAArtificial SequenceOligomeric Compound 116catctgttga tggagatagg 2011720DNAArtificial SequenceOligomeric Compound 117tctgtctcat ctgttgatgg 2011820DNAArtificial SequenceOligomeric Compound 118tagtctgtct catctgttga 2011920DNAArtificial SequenceOligomeric Compound 119gtagtctgtc tcatctgttg 2012020DNAArtificial SequenceOligomeric Compound 120gctctgtagt ctgtctcatc 2012120DNAArtificial SequenceOligomeric Compound 121gcagctctgt agtctgtctc 2012220DNAArtificial SequenceOligomeric Compound 122gctgcagctc tgtagtctgt 2012320DNAArtificial SequenceOligomeric Compound 123aaggctgcag ctctgtagtc 2012420DNAArtificial SequenceOligomeric Compound 124gtgcaaaggc tgcagctctg 2012520DNAArtificial SequenceOligomeric Compound 125ttccccgtgc aaaggctgca 2012620DNAArtificial SequenceOligomeric Compound 126tttcttcccc gtgcaaaggc 2012720DNAArtificial SequenceOligomeric Compound 127tgctttcttc cccgtgcaaa 2012820DNAArtificial SequenceOligomeric Compound 128atgtctgccc tgctttcttc 2012920DNAArtificial SequenceOligomeric Compound 129gcattggtat cgcaatgtct 2013020DNAArtificial SequenceOligomeric Compound 130ttctgcattg gtatcgcaat 2013120DNAArtificial SequenceOligomeric Compound 131tctcttctgc attggtatcg 2013220DNAArtificial SequenceOligomeric Compound 132aagttggtgt ctctcttcct 2013320DNAArtificial SequenceOligomeric Compound 133gggaagttgg tgtctctctt 2013420DNAArtificial SequenceOligomeric Compound 134tttacagcag aagatgcaga 2013520DNAArtificial SequenceOligomeric Compound 135agcatttaca gcagaagatg 2013620DNAArtificial SequenceOligomeric Compound 136acagcattta cagcagaaga 2013720DNAArtificial SequenceOligomeric Compound 137ttacagcatt tacagcagaa 2013820DNAArtificial SequenceOligomeric Compound 138aattgttaca gcatttacag 2013920DNAArtificial SequenceOligomeric Compound 139tgggaattgt tacagcattt 2014020DNAArtificial SequenceOligomeric Compound 140cactgggaat tgttacagca 2014120DNAArtificial SequenceOligomeric Compound 141gataccacac tgggaattgt 2014220DNAArtificial SequenceOligomeric Compound 142aacagatacc acactgggaa 2014320DNAArtificial SequenceOligomeric Compound 143gcaacagata ccacactggg 2014420DNAArtificial SequenceOligomeric Compound 144tttgcaacag ataccacact 2014520DNAArtificial SequenceOligomeric Compound 145gttttgcaac agataccaca 2014620DNAArtificial SequenceOligomeric Compound 146tatgttttgc aacagatacc 2014720DNAArtificial SequenceOligomeric Compound 147gctctaggct atgttttgca 2014820DNAArtificial SequenceOligomeric Compound 148ggtcaggatg tggctctagg 2014920DNAArtificial SequenceOligomeric Compound 149gagaggtcag gatgtggctc 2015020DNAArtificial SequenceOligomeric Compound 150tagagaggtc aggatgtggc 2015120DNAArtificial SequenceOligomeric Compound 151gtgtagagag gtcaggatgt 2015220DNAArtificial SequenceOligomeric Compound 152agggcaggaa taaataatgg 2015320DNAArtificial SequenceOligomeric Compound 153ggagggcagg aataaataat 2015420DNAArtificial SequenceOligomeric Compound 154atttcaaggt cattggtggg 2015520DNAArtificial SequenceOligomeric Compound 155ttatttcaag gtcattggtg 2015620DNAArtificial SequenceOligomeric Compound 156cgtctttatt tcaaggtcat 2015720DNAArtificial SequenceOligomeric Compound 157aaaatcgtct ttatttcaag 2015820DNAArtificial SequenceOligomeric Compound 158ataaaatcgt ctttatttca 2015920DNAArtificial SequenceOligomeric Compound 159aaataaaatc gtctttattt 2016020DNAArtificial SequenceOligomeric Compound 160gggtgctcac ctgttgatgg 2016120DNAArtificial SequenceOligomeric Compound 161tgcttactgc tgactgccat 2016220DNAArtificial SequenceOligomeric Compound 162atgcatgctt actgctgact 2016320DNAArtificial SequenceOligomeric Compound 163gcctatgtgt atcatatgca 2016420DNAArtificial SequenceOligomeric Compound 164cagcagaacc tctacagccc 2016520DNAArtificial SequenceOligomeric Compound 165tctgtctcat ctgtgaaagc 2016620DNAArtificial SequenceOligomeric Compound 166atgctcttac cgcaatgtct 2016720DNAArtificial SequenceOligomeric Compound 167gcattggtat ctgtgtaaag 2016820DNAArtificial SequenceOligomeric Compound 168ccttccctga aggttcctcc 2016920DNAArtificial SequenceOligomeric Compound 169gctcatagaa ctgtcaggct 2017015DNAArtificial SequenceOligomeric Compound 170ggtcaggatg tggct 1517115DNAArtificial SequenceOligomeric Compound 171caggatgtgg ctcta 1517212DNAArtificial SequenceOligomeric Compound 172gtcaggatgt gg 1217336DNAArtificial SequenceOligomeric Compound 173gagaggtcag gatgtggctc taggctatgt tttgca 3617428DNAArtificial SequenceOligomeric Compound 174gtcaggatgt ggctctaggc tatgtttt 2817523DNAArtificial SequenceOligomeric Compound 175tgtggctcta ggctatgttt tgc 2317620DNAArtificial SequenceOligomeric Compound 176ggtcaggatg tggctcaagg 2017720DNAArtificial SequenceOligomeric Compound 177ggtcaggaag tggctctagg 2017820DNAArtificial SequenceOligomeric Compound 178ggtcagtttg tggctctagg 2017920DNAArtificial SequenceOligomeric Compound 179ggtgaggatg tggctccagg 2018020DNAArtificial SequenceOligomeric Compound 180ggtctggatg tgcctctcgg 20

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed