Oligonucleotide-based inhibitors comprising locked nucleic acid motif

Abstract
The present invention relates to chemical modification motifs for oligonucleotides. The oligonucleotides of the present invention, such as chemically modified antisense oligonucleotides, can have increased in vivo efficacy. The chemically modified oligonucleotides provide advantages in one or more of potency, efficiency of delivery, target specificity, toxicity, and/or stability. The chemically modified oligonucleotides have a specific chemical modification motif or pattern of locked nucleic acids (LNAs). The oligonucleotide (e.g. antisense oligonucleotide) can target RNA, such as miRNA or mRNA. Also provided herein are compositions comprising the chemically modified oligonucleotides and methods of using the chemically modified oligonucleotides as therapeutics for various disorders, including cardiovascular disorders.
Description
DESCRIPTION OF THE TEXT FILE SUBMITTED ELECTRONICALLY

The contents of the text file submitted electronically herewith are incorporated herein by reference in their entirety: A computer readable format copy of the Sequence Listing (filename: MIRG_036_04US_SeqList_ST25.txt, date recorded Apr. 24, 2018, file size 87 kilobytes).


FIELD OF THE INVENTION

The present invention relates to chemical modification motifs for oligonucleotides, such as antisense oligonucleotides, including mRNA and microRNA (miRNA or miR) inhibitors. The oligonucleotides of the present invention, such as chemically modified antisense oligonucleotides, for example, miRNA antisense oligonucleotides, can have advantages in potency, efficiency of delivery, target specificity, stability, and/or toxicity when administered to a subject.


BACKGROUND OF THE INVENTION

Delivery of oligonucleotides to the body, such as an antisense-based therapeutics, poses several challenges. The binding affinity and specificity to a target, efficiency of cellular uptake, and nuclease resistance are all factors in the delivery and activity of an oligonucleotide-based therapeutic. For example, when oligonucleotides are introduced into intact cells they are attacked and degraded by nucleases leading to a loss of activity. Thus, a useful oligonucleotide should have good resistance to extra- and intracellular nucleases, as well as be able to penetrate the cell membrane.


Polynucleotide analogues have been prepared in an attempt to avoid their degradation, e.g. by means of 2′ substitutions (Sproat et al., Nucleic Acids Research 17 (1989), 3373-3386). However, such modifications often affect the polynucleotide's potency for its intended biological action. Such reduced potency may be due to an inability of the modified polynucleotide to form a stable duplex with the target RNA and/or a loss of interaction with the cellular machinery. Other modifications include the use of locked nucleic acids, which has the potential to improve RNA-binding affinity (Veedu and Wengel, RNA Biology 6:3, 321-323 (2009)), however, in vivo efficacy can be low. An oligonucleotide used as an antisense therapeutic should have high affinity for its target to efficiently impair the function of its target (such as inhibiting translation of a mRNA target, or inhibiting the activity of a miRNA target). However, modification of oligonucleotides can decrease its affinity and binding specificity, as well as its ability to impair the function of its target.


Thus, despite the variety of methods described for the delivery of oligonucleotides as a therapeutic, there is a need for improved chemical modifications for stable and efficacious oligonucleotide-based inhibitors.


SUMMARY OF THE INVENTION

The present invention is based, in part, on the discovery that a specific chemical modification pattern or motif of an oligonucleotide can increase the potency, efficiency of delivery, target specificity, stability, and/or improve the toxicity profile when administered to a subject. The present inventors have discovered specific oligonucleotide chemistry modification patterns or motifs with the potential to improve the delivery, stability, potency, specificity, and/or toxicity profile of the oligonucleotide. For example, oligonucleotide chemistry modification patterns or motifs for miRNA inhibitors can improve the delivery, stability, potency, specificity, and/or toxicity profile of the miRNA inhibitor, thus, effectively targeting miRNA function in a therapeutic context.


The present invention provides oligonucleotides with a chemistry modification pattern or motif capable of inhibiting the expression (e.g., abundance) of miRNA with improved properties, such as increased in vivo efficacy. This chemistry modification pattern or motif can be applied to other oligonucleotides for targeting other therapeutic targets, such as mRNA. Thus, the present invention provides a novel therapeutic for the treatment of a variety of diseases, including cardiovascular diseases, obesity, diabetes, and other metabolic disorders.


The oligonucleotide with the specific chemical modification pattern or motif can have an increased in vivo efficacy as compared to an oligonucleotide with the same nucleotide sequence but different chemical modification pattern or motif. For example, an oligonucleotide with a specific locked nucleic acid (LNA) pattern can have an increased in vivo efficacy as compared to an oligonucleotide with the same nucleotide sequence but different LNA pattern.


In one embodiment, the oligonucleotide of the present invention comprises a sequence complementary to the seed region of a miRNA, wherein the sequence comprises at least five LNAs. The oligonucleotide can comprise at least five LNAs complementary to the seed region of a miRNA and at least one non-locked nucleotide. In some embodiments, the non-locked nucleotide is in a region that is complementary to the seed region. The oligonucleotide can have increased in vivo efficacy as compared to a second oligonucleotide comprising the same sequence and LNA composition and different LNA motif. The oligonucleotide can comprise a LNA at the 5′ end, 3′ end, or both 5′ and 3′ ends. In some embodiments, the oligonucleotide comprises three or fewer contiguous LNAs. For example, the oligonucleotide comprises no more than three contiguous LNAs. The oligonucleotide can be at least 16 nucleotides in length. In some embodiments, the oligonucleotide can be from 8 to 20 nucleotides in length, from 18 to 50 nucleotides in length, from 10 to 18 nucleotides in length, or from 11 to 16 nucleotides in length. The oligonucleotide in some embodiments is about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, or about 18 nucleotides in length.


In another embodiment, the oligonucleotide of the present invention comprises a sequence of 16 nucleotides, wherein the sequence comprises at least five LNAs, a LNA at the 5′ end, a LNA at the 3′ end, and no more than three contiguous LNAs. The oligonucleotide, from the 5′ end to the 3′ end, can comprise LNAs at positions 1, 5, 6, 8, 10, 11, 13, 15, and 16 of the sequence.


The oligonucleotide described herein can comprise one or more non-locked nucleotides. In some embodiments, at least one of the non-locked nucleotides is 2′ deoxy, 2′ O-alkyl or 2′ halo. In another embodiment, all of the non-locked nucleotides are 2′ deoxy, 2′ O-alkyl, 2′ halo, or any combination thereof.


In some embodiments, the oligonucleotide described herein comprises at least one LNA with a 2′ to 4′ methylene bridge. The oligonucleotide can have a 5′ cap structure, 3′ cap structure, or 5′ and 3′ cap structure. In some embodiments, the oligonucleotide comprises one or more phosphorothioate linkages or is fully phosphorothioate-linked. The oligonucleotide can have one to three phosphate linkages. The oligonucleotide can further comprise a pendent lipophilic or hydrophilic group.


In one embodiment, the oligonucleotide is an inhibitor of a RNA, such as an inhibitor of its expression or activity. In one embodiment, the oligonucleotide is a miRNA inhibitor. For example, the oligonucleotide can comprise a sequence that is substantially or completely complementary to a nucleotide sequence of a miRNA or fragment thereof. The miRNA can be expressed in any tissue, or selectively expressed in a tissue. In one embodiment, the tissue is cardiac tissue. For example, the miRNA is selectively expressed in cardiac tissue.


The oligonucleotide can be an inhibitor of any miRNA. In some embodiments, the oligonucleotide can be an inhibitor of any miRNA, but not miR-208a, miR-208b, or miR-499. Such inhibitors are described in, for example, International Publication No. WO 2012/083005, which is hereby incorporated by reference in its entirety. In one embodiment, the oligonucleotide is an inhibitor of a miR selected from Table 1 or Table 2. In yet another embodiment, the oligonucleotide is an inhibitor of miR-15a, miR-15b, miR-16-1, miR-16-2, miR-24, miR-25, miR-26a, miR-497, miR-195, miR-424, a let 7 family member, miR-21, miR-199a-b, miR-214, miR-10a-b, miR-16, miR-125b, miR-146a-b, miR-221, miR-222, a miR-30 family member, miR-126, miR-133, miR-1, miR-143, miR-145, miR-486, miR-92a, miR-320, miR-1-1, miR-1-2, miR-451, miR-378, miR-378*, miR-92, miR-34a, miR-34b, miR-34c, miR-29, or miR-33.


In yet another embodiment, the oligonucleotide can be an inhibitor of mRNA. For example, the sequence can be substantially or completely complementary to a nucleotide sequence of an mRNA or fragment thereof.


Also provided herein is a pharmaceutical composition comprising an effective amount of the oligonucleotide described herein, or a pharmaceutically-acceptable salt thereof, and a pharmaceutically-acceptable carrier or diluent. In some embodiments, the pharmaceutically-acceptable carrier can comprise a colloidal dispersion system, macromolecular complex, nanocapsule, nanoparticle, microsphere, bead, oil-in-water emulsion, micelle, mixed micelle, or liposome. In another embodiment, the pharmaceutically-acceptable carrier or diluent consists essentially of saline.


The present invention also provides methods of producing and using an oligonucleotide described herein. A method of reducing or inhibiting activity of a miRNA in a cell comprising contacting the cell with an oligonucleotide described herein is also provided. Also disclosed herein is a method of reducing expression of an mRNA in a cell comprising contacting the cell with an oligonucleotide disclosed herein. The cell can be any cell type, such as a heart cell. The cell can be in vivo or ex vivo. In one embodiment, the cell is a mammalian cell.


A method of preventing or treating a condition in a subject associated with or mediated by expression of an RNA is also provided. The method can comprise administering to the subject a pharmaceutical composition comprising an oligonucleotide disclosed herein. In one embodiment, a method of preventing or treating a condition in a subject associated with or mediated by the activity of a miRNA comprises administering to the subject a pharmaceutical composition comprising the oligonucleotide disclosed herein. In another embodiment, a method of preventing or treating a condition in a subject associated with or mediated by the activity of a mRNA comprises administering to the subject a pharmaceutical composition comprising the oligonucleotide disclosed herein. The condition can be a heart condition, such as pathologic cardiac hypertrophy, myocardial infarction, myocardial ischemia, ischemia-reperfusion injury, cardiomyopathy, or heart failure. The pharmaceutical composition can be administered by parenteral administration, such as by intravenous, subcutaneous, intraperitoneal, or intramuscular administration. In some embodiments, administration is by direct injection into cardiac tissue. In yet in some embodiments, the composition is administered by oral, transdermal, sustained release, controlled release, delayed release, suppository, catheter, or sublingual administration. Furthermore, the subject can be a human. In some embodiments, an oligonucleotide disclosed herein is delivered at a dose of between about 10 mg/kg to about 100 mg/kg, between about 10 mg/kg to about 50 mg/kg, between about 10 mg/kg to about 25 mg/kg. In some embodiments, an oligonucleotide disclosed herein is delivered at a dose of about 100 mg/kg or less, about 50 mg/kg or less, about 25 mg/kg or less, or about 10 mg/kg or less. In one embodiment, the oligonucleotide is formulated in saline and administered subcutaneously.





BRIEF DESCRIPTION OF THE DRAWINGS


FIG. 1A. Location of LNA and DNA bases for 16 antimiRs designed to target miR-208a (SEQ ID NOs: 76-91). LNA bases are represented by a capital letter. DNA bases are represented by a lower case letter. FIG. 1B. MiR-208a inhibition by antimiR-208a compounds. All antimiR compounds showed significant inhibition in the left ventricle. #p<0.05 vs Saline. *p<0.05 vs. control oligo, M-10591. FIG. 1C. Real-time PCR from cardiac tissue of antimiR-208a-treated rats showed differing target de-repression in vivo using Dynlt1 as a primary readout for efficacy and target de-repression. #p<0.05 vs Saline. *p<0.05 vs. control oligo, M-10591. FIG. 1D. Serum levels of toxicology parameters. Four days after injection, plasma was collected from all groups. No antimiR-208a oligonucleotide or control oligonucleotide showed increased levels of liver toxicity as assessed by ALT and AST measurements, or kidney toxicity as assessed by BUN measurements compared to saline controls. FIG. 1E. Quantification of antimiR from heart, liver, and kidney four days after a single 25 mg/kg subcutaneous dose. Distribution to the heart is much lower than liver and kidney. Efficacious compounds are not more robustly distributed to the heart.



FIG. 2A. Location of LNA and DNA bases for 9 antimiRs designed to target miR-208b (SEQ ID NOs: 92-100). LNA bases are represented by a capital letter. DNA bases are represented by a lower case letter. FIG. 2B. MiR-208b inhibition by antimiR-208b compounds. All antimiR compounds showed significant miR-208b inhibition in the left ventricle. FIG. 2C. Real-time PCR from cardiac tissue of antimiR-208b treated rats showed differing target de-repression in vivo using Dynlt1 as a primary readout for efficacy and target de-repression. *p<0.05 vs. Saline



FIG. 3A. Silencing. Location of LNA and DNA bases for 7 antimiRs designed to target miR-378 (SEQ ID NOs: 101-107). LNA bases are represented by a capital letter. DNA bases are represented by a lower case letter. FIG. 3B. MiR-378 inhibition by antimiR-378 compounds. All antimiR compounds showed significant miR-378 inhibition in the left ventricle. FIG. 3C. Real-time PCR from cardiac tissue of antimiR-378 treated rats showed differing target de-repression in vivo using Gfpt2 as a primary readout for efficacy and target de-repression. #p<0.05 vs. Saline



FIG. 4A. Location of LNA and DNA bases for 7 antimiRs designed to target miR-29 (SEQ ID NOs: 108-114). LNA bases are represented by a capital letter. DNA bases are represented by a lower case letter. FIG. 4B. MiR-29 family inhibition by antimiR-29 compounds in heart (top panel), liver (middle panel), and kidney (bottom panel). All antimiR compounds showed significant miR-29 family inhibition in heart, liver, and kidney. FIG. 4C. Real-time PCR from heart (top panel), liver (middle panel), and kidney (bottom panel) of antimiR-29 treated rats showed differing target de-repression in vivo using Dnmt3b and Mcl1 as a primary readout for efficacy and target de-repression. *p<0.05 vs. Saline; #p<0.05 vs. Control oligonucleotide M-10591. FIG. 4D. Quantification of antimiR compounds from heart and liver four days after a single 25 mg/kg subcutaneous dose. Distribution to the heart is much lower than liver. More efficacious compounds are not more robustly distributed to the heart compared to less efficacious compounds.



FIG. 5A. Location of LNA and DNA bases for 5 antimiRs designed to target miR-199a (SEQ ID NOs: 115-119). LNA bases are represented by a capital letter. DNA bases are represented by a lower case letter. FIG. 5B. MiR-199a inhibition by antimiR-199 compounds in heart, lung, liver (Li), and kidney (K). All antimiR compounds showed significant miR-199a inhibition in heart, lung, liver, and kidney. FIG. 5C. Real-time PCR from heart, lung, liver (Li), and kidney (K) of antimiR-199-treated rats showed differing target de-repression in vivo using Ddr1 as a primary readout for efficacy and target de-repression. M-10518 consistently appeared to show target de-repression across multiple tissues. *p<0.05 vs. Saline.



FIG. 6. Real-time PCR from endothelial cells isolated from heart tissue of antimiR-92a-treated rats showed differing target de-repression in vivo using Map2K4 as a primary readout for efficacy and target de-repression. *p<0.05 vs. Saline.





DETAILED DESCRIPTION OF THE INVENTION

The present invention is based, in part, on the discovery that a specific chemical modification pattern or motif of an oligonucleotide can improve the potency, efficiency of delivery, target specificity, stability, and/or toxicity when administered to a subject. The oligonucleotide with the specific chemical modification pattern or motif can have an increased in vivo efficacy as compared to an oligonucleotide with the same nucleotide sequence but different chemical modification pattern or motif. For example, an oligonucleotide with a specific LNA/DNA pattern can have an increased in vivo efficacy as compared to an oligonucleotide with the same nucleotide sequence but different LNA/DNA pattern.


The invention provides in some embodiments, oligonucleotides capable of inhibiting, in a specific fashion, the expression or abundance of an RNA species, such as a miRNA or mRNA. The invention further provides pharmaceutical compositions comprising the oligonucleotides, and methods of treating patients having conditions or disorders relating to or involving the RNA, such as miRNA or mRNA, such as a various cardiovascular conditions. In various embodiments, the oligonucleotides provide advantages in one or more of potency, efficiency of delivery, target specificity, toxicity, and/or stability.


In one aspect, the present invention provides an oligonucleotide capable of reducing the expression or abundance of an RNA, such as a mRNA or miRNA. The oligonucleotide of the present invention may have increased in vivo efficacy as compared to another oligonucleotide with the same nucleotide sequence but different chemical modification motif or pattern. For example, a first and a second oligonucleotide each have the same nucleotide sequence that targets a miRNA. The first oligonucleotide has a chemical modification motif or pattern that differs from the second oligonucleotide. Both the first and second oligonucleotides are capable of reducing the expression or abundance of a miRNA. However, the first oligonucleotide with a first chemical modification motif has a higher in vivo efficacy as compared to the second oligonucleotide with a different chemical modification motif, as measured by the amount of de-repression of one or more of the miRNA's targets.


The activity of the oligonucleotide in reducing the expression or abundance of an RNA species, such as miRNA, may be determined in vitro and/or in vivo. For example, when inhibition of a miRNA activity is determined in vitro, the activity may be determined using a dual luciferase assay, such as that described herein. The oligonucleotide significantly inhibits such activity, as determined in the dual luciferase activity, at a concentration of about 50 nM or less, or in other embodiments, 40 nM or less, 20 nM or less, or 10 nM or less. For example, the oligonucleotide may have an IC50 for inhibition of a miRNA activity of about 50 nM or less, about 40 nM or less, about 30 nM or less, or about 20 nM or less, as determined in the dual luciferase assay. The dual luciferase assay, as exemplified by the commercially available product PsiCHECK™ (Promega), involves placement of the miR recognition site in the 3′ UTR of a gene for a detectable protein (e.g., renilla luciferase). The construct is co-expressed with the target miRNA, such that inhibitor activity can be determined by change in signal. A second gene encoding a detectable protein (e.g., firefly luciferase) can be included on the same plasmid, and the ratio of signals determined as an indication of antimiR activity.


Alternatively, or in addition, the activity of the oligonucleotide in reducing the expression or abundance of an RNA species, such as miRNA, may be determined in a suitable mouse or rat model, such as those described herein, where inhibition (e.g., by at least 50%) of a miRNA is observed at an oligonucleotide dose, such as a dose of about 50 mg/kg or less, about 25 mg/kg or less, about 10 mg/kg or less or about 5 mg/kg or less. In some embodiments, the activity of the oligonucleotide is determined in an animal model, such as described in WO 2008/016924, which descriptions are hereby incorporated by reference. For example, the oligonucleotide may exhibit at least 50% target miRNA inhibition, such as a dose of about 50 mg/kg or less, about 25 mg/kg or less, such as about 10 mg/kg or less or about 5 mg/kg or less. In such embodiments, the oligonucleotide may be dosed intravenously or subcutaneously to mice, and the oligonucleotide may be formulated in saline.


The in vivo efficacy of the oligonucleotide may be determined by assessing the level or amount of de-repression of one or more of the miRNA's targets in a suitable mouse or rat model, such as those described herein. The oligonucleotide may exhibit at least 50% target de-repression at a dose of about 50 mg/kg or less, about 25 mg/kg or less, about 10 mg/kg or less or about 5 mg/kg or less. In such embodiments, the oligonucleotide may be dosed intravenously or subcutaneously to mice, and the oligonucleotide may be formulated in saline.


In these or other embodiments, the oligonucleotides of the present invention can be stable after administration, being detectable in the circulation and/or target organ for at least three weeks, at least four weeks, at least five weeks, or at least six weeks, or more, following administration. Thus, the oligonucleotides of the present invention may provide for less frequent administration, lower doses, and/or longer duration of therapeutic effect.


The nucleotide sequence of the oligonucleotide can be substantially complementary to a nucleotide sequence of a RNA, such as a mRNA or miRNA. In some embodiments, the miRNA is not miR-208a, miR-208b, or miR-499. The oligonucleotide comprises at least one LNA, such as at least five, at least seven or at least nine LNAs. In some embodiments, the oligonucleotide comprises a mix of LNA and non-locked nucleotides. For example, the oligonucleotide may contain at least five or at least seven or at least nine locked nucleotides, and at least one non-locked nucleotide.


Generally, the length of the oligonucleotide and number and position of locked nucleotides is such that the oligonucleotide reduces RNA expression or abundance, such as mRNA expression or miRNA expression, at an oligonucleotide concentration of about 50 nM or less in the in vitro luciferase assay, or at a dose of about 50 mg/kg or less, or about 25 mg/kg or less in a suitable mouse or rat model, each as described herein. In some embodiments, the oligonucleotide is a miRNA inhibitor, such that the length of the oligonucleotide and number and position of locked nucleotides is such that the oligonucleotide reduces miRNA activity as determined by target de-repression, at a dose of about 50 mg/kg or less, or about 25 mg/kg or less in a suitable mouse or rat model, such as those described herein.


The oligonucleotide of the present invention can comprise a sequence of nucleotides in which the sequence comprises at least five LNAs, a LNA at the 5′ end of the sequence, a LNA at the 3′ end of the sequence, or any combination thereof. In one embodiment, the oligonucleotide comprises a sequence of nucleotides in which the sequence comprises at least five LNAs, a LNA at the 5′ end of the sequence, a LNA at the 3′ end of the sequence, or any combination thereof, wherein three or fewer of the nucleotides are contiguous LNAs. For example, the oligonucleotide comprises no more than three contiguous LNAs. For example, the oligonucleotide may comprise a sequence with at least five LNAs, a LNA at the 5′ end, a LNA at the 3′ end, and no more than three contiguous LNAs. The oligonucleotide may comprise a sequence with at least five LNAs, a LNA at the 5′ end, a LNA at the 3′ end, and no more than three contiguous LNAs, wherein the sequence is at least 16 nucleotides in length. The sequence can be substantially or completely complementary to a RNA, such as mRNA or miRNA, wherein a substantially complementary sequence may have from 1 to 4 mismatches (e.g., 1 or 2 mismatches) with respect to its target sequence. In one embodiment, the target sequence is a miRNA, such that the oligonucleotide is a miRNA inhibitor, or antimiR. The miRNA can be any miRNA, such as, but not limited to, those listed in Table 1 or Table 2. Exemplary miRNA therapeutic utilities are disclosed in the US and PCT patent references listed in Table 2 below, each of which is hereby incorporated by reference in its entirety. The mature and pre-processed forms of miRNAs are disclosed in the patent references listed in Table 2, and such descriptions are also hereby incorporated by reference.













TABLE 1









SEQ





ID



miRNA
miRNA Sequence
NO:









  1
UGGAAUGUAAAGAAGUAUGUAU
 1







100
AACCCGUAGAUCCGAACUUGUG
 2







 10a
UACCCUGUAGAUCCGAAUUUGUG
 3







 10b
UACCCUGUAGAACCGAAUUUGUG
 4







125b
UCCCUGAGACCCUAACUUGUGA
 5







126
UCGUACCGUGAGUAAUAAUGCG
 6







128
UCACAGUGAACCGGUCUCUUU
 7







133a
UUUGGUCCCCUUCAACCAGCUG
 8







133b
UUUGGUCCCCUUCAACCAGCUA
 9







139
UCUACAGUGCACGUGUCUCCAG
10







143
UGAGAUGAAGCACUGUAGCUC
11







145
GUCCAGUUUUCCCAGGAAUCCCU
12







146a
UGAGAACUGAAUUCCAUGGGUU
13







146b
UGAGAACUGAAUUCCAUAGGCU
14







150
UCUCCCAACCCUUGUACCAGUG
15







 15a
UAGCAGCACAUAAUGGUUUGUG
16







 15b
UAGCAGCACAUCAUGGUUUACA
17







 16
UAGCAGCACGUAAAUAUUGGCG
18







181b
AACAUUCAUUGCUGUCGGUGGGU
19







195
UAGCAGCACAGAAAUAUUGGC
20







197
UUCACCACCUUCUCCACCCAGC
21







199a
CCCAGUGUUCAGACUACCUGUUC
22







199b-5p
CCCAGUGUUUAGACUAUCUGUUC
23







199b-3p
ACAGUAGUCUGCACAUUGGUUA
24







208a
AUAAGACGAGCAAAAAGCUUGU
25







208b
AUAAGACGAACAAAAGGUUUGU
26







 20a
UAAAGUGCUUAUAGUGCAGGUAG
27







 21
UAGCUUAUCAGACUGAUGUUGA
28







214
ACAGCAGGCACAGACAGGCAGU
29







 22
AAGCUGCCAGUUGAAGAACUGU
30







221
AGCUACAUUGUCUGCUGGGUUUC
31







222
AGCUACAUCUGGCUACUGGGU
32







224
CAAGUCACUAGUGGUUCCGUU
33







 23a
AUCACAUUGCCAGGGAUUUCC
34







 24
UGGCUCAGUUCAGCAGGAACAG
35







 25
CAUUGCACUUGUCUCGGUCUGA
36







 26a
UUCAAGUAAUCCAGGAUAGGCU
37







 26b
UUCAAGUAAUUCAGGAUAGGU
38







 28
AAGGAGCUCACAGUCUAUUGAG
39







 29a
UAGCACCAUCUGAAAUCGGUUA
40







 29b
UAGCACCAUUUGAAAUCAGUGUU
41







 29c
UAGCACCAUUUGAAAUCGGUUA
42







 30a
UGUAAACAUCCUCGACUGGAAG
43







 30b
UGUAAACAUCCUACACUCAGCU
44







 30c
UGUAAACAUCCUACACUCUCAGC
45







 30d
UGUAAACAUCCCCGACUGGAAG
46







 30e
UGUAAACAUCCUUGACUGGAAG
47







 33a
GUGCAUUGUAGUUGCAUUGCA
48







 33b
GUGCAUUGCUGUUGCAUUGC
49







 34a
UGGCAGUGUCUUAGCUGGUUGU
50







 34b
CAAUCACUAACUCCACUGCCAU
51







 34c
AGGCAGUGUAGUUAGCUGAUUGC
52







320
AAAAGCUGGGUUGAGAGGGCGA
53







342-3p
UCUCACACAGAAAUCGCACCCGU
54







382
GAAGUUGUUCGUGGUGGAUUCG
55







422a
ACUGGACUUAGGGUCAGAAGGC
56







378
ACUGGACUUGGAGUCAGAAGG
57







378*
CUCCUGACUCCAGGUCCUGUGU
58







424
CAGCAGCAAUUCAUGUUUUGAA
59







451
AAACCGUUACCAUUACUGAGUU
60







483-3p
UCACUCCUCUCCUCCCGUCUU
61







484
UCAGGCUCAGUCCCCUCCCGAU
62







486-5p
UCCUGUACUGAGCUGCCCCGAG
63







497
CAGCAGCACACUGUGGUUUGU
64







499
UUAAGACUUGCAGUGAUGUUU
65







542-5p
UCGGGGAUCAUCAUGUCACGAGA
66







 92a
UAUUGCACUUGUCCCGGCCUGU
67







 92b
UAUUGCACUCGUCCCGGCCUCC
68







let-7a
UGAGGUAGUAGGUUGUAUAGUU
69







let-7b
UGAGGUAGUAGGUUGUGUGGUU
70







let-7c
UGAGGUAGUAGGUUGUAUGGUU
71







let-7d
AGAGGUAGUAGGUUGCAUAGUU
72







let-7e
UGAGGUAGGAGGUUGUAUAGUU
73







let-7f
UGAGGUAGUAGAUUGUAUAGUU
74







let-7g
UGAGGUAGUAGUUUGUACAGUU
75



















TABLE 2





miRNA
Indications
Reference







miR-208a/miR-208b/miR-
Pathologic cardiac hypertrophy,
WO 2008/016924 (208a)


499
myocardial infarction, heart failure
WO 2009/018492 (208b/499)


miR-208a/miR-208b
Metabolic Disorders (obesity,
PCT/US2012/059349, filed



hyperlipidemia, diabetes, metabolic
Oct. 9, 2012



syndrome, hypercholesterolemia; hepatic



steatosis)


miR-15/miR-16/miR-195
Pathologic cardiac hypertrophy,
WO 2009/062169



myocardial infarction, heart failure


miR-29
Profibrotic agents to convert soft plaques
WO 2009/018493



(vulnerable plaques) to fibrotic tissue;



induce collagen deposition


miR-126
Pathologic vascularization
WO 2010/019574


miR-145
Muscle injury
WO 2007/070483


miR-1/miR-133
Muscle injury (antagonist/agonist of each
WO 2007/070483



miRNA applied in combination at



different times)


miR-451
Polycythemia
WO 2012/148373


miR-378/miR-378*
Metabolic disorders (obesity,
WO 2011/153542



hyperlipidemia, diabetes, metabolic



syndrome, hypercholesterolemia; hepatic



steatosis);



Pathologic cardiac hypertrophy,



myocardial infarction, heart failure


miR-92
Promotes angiogenesis and vessel repair
US 2010/0324118 A1


miR-34a
Myocardial infarction
US 2012/0238619 A1


miR-145
Pulmonary arterial hypertension
WO 2012/153135


miR-33
Statin-induced hepatotoxicity,
US 20110281933 A1



cholestasis, increasing HDL cholesterol









In some embodiments, the oligonucleotide comprises a sequence that is substantially or completely complementary to a miRNA that is selected from the group consisting of, but not limited to: miR-15a, miR-15b, miR-16-1, miR-16-2, miR-24, miR-25, miR-26a, miR-497, miR-195, miR-424, a let 7 family member, miR-21, miR-199a-b, miR-214, miR-10a-b, miR-16, miR-125b, miR-146a-b, miR-221, miR-222, a miR-30 family member, miR-126, miR-133, miR-1, miR-143, miR-145, miR-486, miR-92a, miR-320, miR-1-1, miR-1-2, miR-451, miR-378, miR-378*, miR-92, miR-34a, miR-34b, miR-34c, miR-29, or miR-33. In some embodiments, the miRNA is not miR208a, miR208b, or miR-499, such as described in International Publication No. WO 2012/083005, which is hereby incorporated by reference in its entirety. In some embodiments, the miRNA is expressed in a specific tissue, such as kidney, liver, or cardiac tissue. In yet another embodiment, the miRNA is selectively expressed in a tissue, such as kidney, liver, or cardiac tissue.


In yet another embodiment, the oligonucleotide of the present invention can comprise a sequence complementary to the seed region of a miRNA, wherein the sequence comprises at least five LNAs. The “seed region of a miRNA” is the portion spanning bases 2 to 9 at the 5′ end of the miRNA. The miRNA can be any miRNA, such as, but not limited to those listed in Table 1 or Table 2. The miRNA can be, but is not limited to: miR-15a, miR-15b, miR-16-1, miR-16-2, miR-24, miR-25, miR-26a, miR-497, miR-195, miR-424, a let 7 family member, miR-21, miR-199a-b, miR-214, miR-10a-b, miR-16, miR-125b, miR-146a-b, miR-221, miR-222, a miR-30 family member, miR-126, miR-133, miR-1, miR-143, miR-145, miR-486, miR-92a, miR-320, miR-1-1, miR-1-2, miR-451, miR-378, miR-378*, miR-92, miR-34a, miR-34b, miR-34c, miR-29, or miR-33. In some embodiments, the miRNA is not miR208a, miR208b, or miR-499. The sequence can be substantially or completely complementary to the miRNA. In some embodiments, the miRNA is expressed in a specific tissue, such as kidney, liver, or cardiac tissue. In yet another embodiment, the miRNA is selectively expressed in a tissue, such as kidney, liver, or cardiac tissue. In some embodiments, the miRNA is selectively expressed in a particular cell type, including, but not limited to, cardiomyocytes, myocytes, fibroblasts, smooth muscle cells, endothelial cells, and monocytes.


The oligonucleotide comprising a sequence complementary to the seed region of a miRNA, wherein the sequence comprises at least five LNAs, may comprise a LNA at the 5′ end or a LNA at the 3′ end, or both a LNA at the 5′ end and 3′ end. In one embodiment, the oligonucleotide comprising at least 5 LNAs, a LNA at the 5′ end and/or a LNA at the 3′ end, also has three or fewer consecutive LNAs. In some embodiments, the sequence is at least 16 nucleotides in length. The sequence complementary to the seed region of a miRNA can be substantially complementary or completely complementary.


The oligonucleotide of the present invention contains one or more locked nucleic acid (LNAs) residues, or “locked nucleotides.” LNAs are described, for example, in U.S. Pat. Nos. 6,268,490; 6,316,198; 6,403,566; 6,770,748; 6,998,484; 6,670,461; and 7,034,133, all of which are hereby incorporated by reference in their entireties. LNAs are modified nucleotides or ribonucleotides that contain an extra bridge between the 2′ and 4′ carbons of the ribose sugar moiety resulting in a “locked” conformation, and/or bicyclic structure. In one embodiment, the oligonucleotide contains one or more LNAs having the structure shown by structure A below. Alternatively or in addition, the oligonucleotide may contain one or more LNAs having the structure shown by structure B below. Alternatively or in addition, the oligonucleotide contains one or more LNAs having the structure shown by structure C below.




embedded image


Other suitable locked nucleotides that can be incorporated in the oligonucleotides of the present invention include those described in U.S. Pat. Nos. 6,403,566 and 6,833,361, both of which are hereby incorporated by reference in their entireties.


In exemplary embodiments, the locked nucleotides have a 2′ to 4′ methylene bridge, as shown in structure A, for example. In other embodiments, the bridge comprises a methylene or ethylene group, which may be substituted, and which may or may not have an ether linkage at the 2′ position.


The oligonucleotide may comprise, consist essentially of, or consist of, an antisense sequence to a mRNA or miRNA. In one embodiment, the oligonucleotide comprises an antisense sequence directed to a miRNA. For example, the oligonucleotide comprises an antisense sequence that is sufficiently complementary to a miRNA sequence to hybridize to the endogenous miRNA under physiological conditions. In such embodiments, the oligonucleotide can comprise a sequence that is at least partially complementary to a mature miRNA sequence, e.g. at least about 75%, about 80%, about 85%, about 90%, about 95%, about 96%, about 97%, about 98%, or about 99% complementary to a mature miRNA sequence, such as, but not limited to, a miRNA in Table 1, Table 2, or any one of the following miRNAs: miR-15a, miR-15b, miR-16-1, miR-16-2, miR-24, miR-25, miR-26a, miR-497, miR-195, miR-424, a let 7 family member, miR-21, miR-199a-b, miR-214, miR-10a-b, miR-16, miR-125b, miR-146a-b, miR-221, miR-222, a miR-30 family member, miR-126, miR-133, miR-1, miR-143, miR-145, miR-486, miR-92a, miR-320, miR-1-1, miR-1-2, miR-451, miR-378, miR-378*, miR-92, miR-34a, miR-34b, miR-34c, miR-29, or miR-33. In some embodiments, the miRNA is not miR208a, miR208b, or miR-499. In one embodiment, the antisense oligonucleotide comprises a sequence that is 100% complementary to a mature miRNA sequence, such as, but not limited to, a miRNA selected from the group consisting of miR-15a, miR-15b, miR-16-1, miR-16-2, miR-24, miR-25, miR-26a, miR-497, miR-195, miR-424, a let 7 family member, miR-21, miR-199a-b, miR-214, miR-10a-b, miR-16, miR-125b, miR-146a-b, miR-221, miR-222, a miR-30 family member, miR-126, miR-133, miR-1, miR-143, miR-145, miR-486, miR-92a, miR-320, miR-1-1, miR-1-2, miR-451, miR-378, miR-378*, miR-92, miR-34a, miR-34b, miR-34c, miR-29, or miR-33. In some embodiments, the miRNA is not miR208a, miR208b, or miR-499. In some embodiments, the miRNA is not miR-208a, miR-208b, or miR-499.


The oligonucleotide generally has a nucleotide sequence designed to target mature miRNA. The oligonucleotide may, in these or other embodiments, also or alternatively be designed to target the pre- or pri-miRNA forms. In certain embodiments, the oligonucleotide may be designed to have a sequence containing from 1 to 5 (e.g., 1, 2, 3, or 4) mismatches relative to the fully complementary (mature) miRNA sequence. In some embodiments, the miRNA is not miR-208a, miR-208b, or miR-499. In certain embodiments, such antisense sequences may be incorporated into shRNAs or other RNA structures containing stem and loop portions, for example.


In certain embodiments, the oligonucleotide comprises a nucleotide sequence that is completely complementary (i.e. fully complementary) to a nucleotide sequence of a miRNA. In some embodiments, the miRNA is not miR-208a, miR-208b, or miR-499. In particular embodiments, the oligonucleotide comprises, consists essentially of, or consists of a sequence completely complementary to the nucleotide sequence of a miRNA. In this context, “consists essentially of” includes the optional addition of nucleotides (e.g., one or two) on either or both of the 5′ and 3′ ends, so long as the additional nucleotide(s) do not substantially affect (as defined by an increase in IC50 of no more than 20%) the oligonucleotide's inhibition of the target miRNA activity in the dual luciferase assay or mouse model.


The oligonucleotide can be from about 8 to about 20 nucleotides in length, from about 18 to about 50 nucleotides in length, from about 10 to about 18 nucleotides in length, or from about 11 to about 16 nucleotides in length. The oligonucleotide in some embodiments is about 8, about 9, about 10, about 11, about 12, about 13, about 14, about 15, about 16, about 17, or about 18 nucleotides in length. In some embodiments, the oligonucleotide is at least 16 nucleotides in length.


The oligonucleotide generally contains at least about 5, at least about 7, or at least about 9 LNAs, but in various embodiments is not fully comprised of LNAs. Generally, the number and position of LNAs is such that the oligonucleotide reduces mRNA or miRNA activity. In one embodiment, the number and position of LNAs is such that the oligonucleotide has increased in vivo efficacy as compared to an oligonucleotide with a different number and/or position of LNAs. In certain embodiments, the oligonucleotide does not contain a stretch of nucleotides with more than four, or more than three, contiguous LNAs. For example, the oligonucleotide comprises no more than three contiguous LNAs. In these or other embodiments, the oligonucleotide can comprise a region or sequence that is substantially or completely complementary to a miRNA seed region, in which the region or sequence comprises at least three, at least four, or at least five locked nucleotides. In some embodiments, the miRNA is not miR-208a, miR-208b, or miR-499.


In various embodiments, the oligonucleotide contains at least nine locked nucleotides. For example, the oligonucleotide may contain nine locked nucleotides and seven non-locked nucleotides. The pattern of LNAs may be such that, from the 5′ end to the 3′ end of the oligonucleotide, at least positions 1, 6, 10, 13, and 15 are LNAs. In some embodiments, the pattern of LNAs may be such that, from the 5′ end to the 3′ end of the oligonucleotide, at least positions 1, 6, 10, 11, 13, and 16 are LNAs. In certain embodiments, from the 5′ end to the 3′ end of the oligonucleotide, positions 1, 5, 6, 8, 10, 11, 13, 15, and 16 are LNAs, and the remaining positions are non-locked nucleotides. In some embodiments, from the 5′ end to the 3′ end of the oligonucleotide, positions 1, 4, 5, 7, 9, 10, 12, 14, and 16 are LNAs, and remaining positions are non-locked nucleotides. For example, in one embodiment, an oligonucleotide can comprise at least 16 nucleotides, in which from the 5′ end to the 3′ end of the oligonucleotide, positions 1, 5, 6, 8, 10, 11, 13, 15, and 16 are LNAs, and the remaining positions are non-locked nucleotides, wherein the oligonucleotide is a miRNA inhibitor.


For example, the oligonucleotide can comprise at least 16 nucleotides, in which from the 5′ end to the 3′ end of the oligonucleotide, positions 1, 5, 6, 8, 10, 11, 13, 15, and 16 are LNAs, and the remaining positions are non-locked nucleotides, the oligonucleotide is at least partially complementary to a miRNA, in which the miRNA may in some embodiments, not be miR-208a, miR-208b, or miR-499. In another embodiment, the oligonucleotide can comprise at least 16 nucleotides, in which from the 5′ end to the 3′ end of the oligonucleotide, positions 1, 5, 6, 8, 10, 11, 13, 14, and 16 are LNAs, and the remaining positions are non-locked nucleotides, the oligonucleotide is at least partially complementary to a miRNA, in which the miRNA may in some embodiments, not be miR-208a, miR-208b, or miR-499. In yet another example, the oligonucleotide can comprise at least 16 nucleotides, in which from the 5′ end to the 3′ end of the oligonucleotide, positions 1, 5, 6, 8, 10, 11, 13, 15, and 16 are LNAs, and the remaining positions are non-locked nucleotides, the oligonucleotide is at least partially complementary to a seed region of a miRNA, in which the miRNA may in some embodiments, not be miR-208a, miR-208b, or miR-499. In some embodiments, the oligonucleotide is selected from Tables 3, 5, 6, 7, 8, or 9. In certain embodiments, the oligonucleotide is a compound selected from M-10101, M-10707, M-11192, M-11185, M-10518, or M-11127.


For non-locked nucleotides, the nucleotide may contain a 2′ modification with respect to a 2′ hydroxyl. For example, the 2′ modification may be 2′ deoxy. Incorporation of 2′-modified nucleotides in antisense oligonucleotides may increase both resistance of the oligonucleotides to nucleases and their thermal stability with complementary RNA. Various modifications at the 2′ positions may be independently selected from those that provide increased nuclease sensitivity, without compromising molecular interactions with the RNA target or cellular machinery. Such modifications may be selected on the basis of their increased potency in vitro or in vivo. Exemplary methods for determining increased potency (e.g., IC50) for miRNA inhibition are described herein, including the dual luciferase assay and in vivo miRNA expression or target de-repression.


In some embodiments the 2′ modification may be independently selected from O-alkyl (which may be substituted), halo, and deoxy (H). Substantially all, or all, nucleotide 2′ positions of the non-locked nucleotides may be modified in certain embodiments, e.g., as independently selected from O-alkyl (e.g., O-methyl), halo (e.g., fluoro), deoxy (H), and amino. For example, the 2′ modifications may each be independently selected from O-methyl and fluoro. In exemplary embodiments, purine nucleotides each have a 2′ OMe and pyrimidine nucleotides each have a 2′-F. In certain embodiments, from one to about five 2′ positions, or from about one to about three 2′ positions are left unmodified (e.g., as 2′ hydroxyls).


2′ modifications in accordance with the invention also include small hydrocarbon substituents. The hydrocarbon substituents include alkyl, alkenyl, alkynyl, and alkoxyalkyl, where the alkyl (including the alkyl portion of alkoxy), alkenyl and alkynyl may be substituted or unsubstituted. The alkyl, alkenyl, and alkynyl may be C1 to C10 alkyl, alkenyl or alkynyl, such as C1, C2, or C3. The hydrocarbon substituents may include one or two or three non-carbon atoms, which may be independently selected from N, O, and/or S. The 2′ modifications may further include the alkyl, alkenyl, and alkynyl as O-alkyl, O-alkenyl, and O-alkynyl.


Exemplary 2′ modifications in accordance with the invention include 2′-O-alkyl (C1-3 alkyl, such as 2′OMe or 2′OEt), 2′-O-methoxyethyl (2′-O-MOE), 2′-O-aminopropyl (2′-O-AP), 2′-O-dimethylaminoethyl (2′-O-DMAOE), 2′-O-dimethylaminopropyl (2′-O-DMAP), 2′-O-dimethylaminoethyloxyethyl (2′-O-DMAEOE), or 2′-O—N-methylacetamido (2′-O-NMA) substitutions.


In certain embodiments, the oligonucleotide contains at least one 2′-halo modification (e.g., in place of a 2′ hydroxyl), such as 2′-fluoro, 2′-chloro, 2′-bromo, and 2′-iodo. In some embodiments, the 2′ halo modification is fluoro. The oligonucleotide may contain from 1 to about 5 2′-halo modifications (e.g., fluoro), or from 1 to about 3 2′-halo modifications (e.g., fluoro). In some embodiments, the oligonucleotide contains all 2′-fluoro nucleotides at non-locked positions, or 2′-fluoro on all non-locked pyrimidine nucleotides. In certain embodiments, the 2′-fluoro groups are independently di-, tri-, or un-methylated.


The oligonucleotide may have one or more 2′-deoxy modifications (e.g., H for 2′ hydroxyl), and in some embodiments, contains from 2 to about 10 2′-deoxy modifications at non-locked positions, or contains 2′ deoxy at all non-locked positions.


In exemplary embodiments, the oligonucleotide contains 2′ positions modified as 2′OMe in non-locked positions. Alternatively, non-locked purine nucleotides are modified at the 2′ position as 2′OMe, with non-locked pyrimidine nucleotides modified at the 2′ position as 2′-fluoro.


In certain embodiments, the oligonucleotide further comprises at least one terminal modification or “cap”. The cap may be a 5′ and/or a 3′-cap structure. The terms “cap” or “end-cap” include chemical modifications at either terminus of the oligonucleotide (with respect to terminal ribonucleotides), and including modifications at the linkage between the last two nucleotides on the 5′ end and the last two nucleotides on the 3′ end. The cap structure as described herein may increase resistance of the oligonucleotide to exonucleases without compromising molecular interactions with the RNA target or cellular machinery. Such modifications may be selected on the basis of their increased potency in vitro or in vivo. The cap can be present at the 5′-terminus (5′-cap) or at the 3′-terminus (3′-cap) or can be present on both ends. In certain embodiments, the 5′- and/or 3′-cap is independently selected from phosphorothioate monophosphate, abasic residue (moiety), phosphorothioate linkage, 4′-thio nucleotide, carbocyclic nucleotide, phosphorodithioate linkage, inverted nucleotide or inverted abasic moiety (2′-3′ or 3′-3′), phosphorodithioate monophosphate, and methylphosphonate moiety. The phosphorothioate or phosphorodithioate linkage(s), when part of a cap structure, are generally positioned between the two terminal nucleotides on the 5′ end and the two terminal nucleotides on the 3′ end.


In certain embodiments, the oligonucleotide has at least one terminal phosphorothioate monophosphate. The phosphorothioate monophosphate may support a higher potency by inhibiting the action of exonucleases. The phosphorothioate monophosphate may be at the 5′ and/or 3′ end of the oligonucleotide. A phosphorothioate monophosphate is defined by the following structures, where B is base, and R is a 2′ modification as described above:




embedded image


Where the cap structure can support the chemistry of a locked nucleotide, the cap structure may incorporate a LNA as described herein.


Phosphorothioate linkages may be present in some embodiments, such as between the last two nucleotides on the 5′ and the 3′ end (e.g., as part of a cap structure), or as alternating with phosphodiester bonds. In these or other embodiments, the oligonucleotide may contain at least one terminal abasic residue at either or both the 5′ and 3′ ends. An abasic moiety does not contain a commonly recognized purine or pyrimidine nucleotide base, such as adenosine, guanine, cytosine, uracil or thymine. Thus, such abasic moieties lack a nucleotide base or have other non-nucleotide base chemical groups at the 1′ position. For example, the abasic nucleotide may be a reverse abasic nucleotide, e.g., where a reverse abasic phosphoramidite is coupled via a 5′ amidite (instead of 3′ amidite) resulting in a 5′-5′ phosphate bond. The structure of a reverse abasic nucleoside for the 5′ and the 3′ end of a polynucleotide is shown below.




embedded image


The oligonucleotide may contain one or more phosphorothioate linkages. Phosphorothioate linkages have been used to render oligonucleotides more resistant to nuclease cleavage. For example, the polynucleotide may be partially phosphorothioate-linked, for example, phosphorothioate linkages may alternate with phophodiester linkages. In certain embodiments, however, the oligonucleotide is fully phosphorothioate-linked. In other embodiments, the oligonucleotide has from one to five or one to three phosphate linkages.


In some embodiments, the nucleotide has one or more carboxamido-modified bases as described in WO 2012/061810, which is hereby incorporated by reference, including with respect to all exemplary pyrimidine carboxamido modifications disclosed therein with heterocyclic substituents.


The synthesis of oligonucleotides, including modified polynucleotides, by solid phase synthesis is well known and is reviewed in New Chemical Methods for Synthesizing Polynucleotides. Caruthers M H, Beaucage S L, Efcavitch J W, Fisher E F, Matteucci M D, Stabinsky Y. Nucleic Acids Symp. Ser. 1980; (7):215-23.


The oligonucleotide may be incorporated within a variety of macromolecular assemblies or compositions. Such complexes for delivery may include a variety of liposomes, nanoparticles, and micelles, formulated for delivery to a patient. The complexes may include one or more fusogenic or lipophilic molecules to initiate cellular membrane penetration. Such molecules are described, for example, in U.S. Pat. Nos. 7,404,969 and 7,202,227, which are hereby incorporated by reference in their entireties. Alternatively, the oligonucelotide may further comprise a pendent lipophilic group to aid cellular delivery, such as fatty acids and those described in WO 2010/129672, which is hereby incorporated by reference in its entirety. In some embodiments, the oligonucleotide may further comprise a pendent hydrophilic group to target the oligonucleotide to particular tissues. For instance, in one embodiment, the oligonucleotide may be conjugated to a sugar moiety, such as mannose-6-phosphate or an amino sugar, such as N-acetyl glucosamine.


The oligonucleotides of the invention may be formulated as a variety of pharmaceutical compositions. Pharmaceutical compositions will be prepared in a form appropriate for the intended application. Generally, this will entail preparing compositions that are essentially free of pyrogens, as well as other impurities that could be harmful to humans or animals. Exemplary delivery/formulation systems include colloidal dispersion systems, macromolecule complexes, nanocapsules, nanoparticles, microspheres, beads, and lipid-based systems including oil-in-water emulsions, micelles, mixed micelles, and liposomes. Commercially available fat emulsions that are suitable for delivering the nucleic acids of the invention to cardiac and skeletal muscle tissues include Intralipid®, Liposyn®, Liposyn® II, Liposyn® III, Nutrilipid, and other similar lipid emulsions. A preferred colloidal system for use as a delivery vehicle in vivo is a liposome (i.e., an artificial membrane vesicle). The preparation and use of such systems is well known in the art. Exemplary formulations are also disclosed in U.S. Pat. Nos. 5,981,505; 6,217,900; 6,383,512; 5,783,565; 7,202,227; 6,379,965; 6,127,170; 5,837,533; 6,747,014; and WO03/093449, all of which are hereby incorporated by reference in their entireties.


The compositions or formulations may employ a plurality of therapeutic oligonucleotides, including at least one described herein. For example, the composition or formulation may employ at least 2, 3, 4, or 5 miRNA inhibitors described herein. In another embodiment, an oligonucleotide of the present invention may be used in combination with other therapeutic modalities. Combinations may also be achieved by contacting the cell with more than one distinct compositions or formulations, at the same time. Alternatively, combinations may be administered sequentially.


In some embodiments, the oligonucleotide is formulated for conventional subcutaneous or intravenous administration, for example, by formulating with appropriate aqueous diluent, including sterile water and normal saline.


The pharmaceutical compositions and formulations may employ appropriate salts and buffers to render delivery vehicles stable and allow for uptake by target cells. Aqueous compositions of the present invention comprise an effective amount of the delivery vehicle comprising the inhibitor oligonucleotide (e.g. liposomes, nanoparticles, or other complexes), dissolved or dispersed in a pharmaceutically acceptable carrier or aqueous medium. The phrases “pharmaceutically acceptable” or “pharmacologically acceptable” refers to molecular entities and compositions that do not produce adverse, allergic, or other untoward reactions when administered to an animal or a human. As used herein, “pharmaceutically acceptable carrier” may include one or more solvents, buffers, solutions, dispersion media, coatings, antibacterial and antifungal agents, isotonic and absorption delaying agents and the like acceptable for use in formulating pharmaceuticals, such as pharmaceuticals suitable for administration to humans. The use of such media and agents for pharmaceutically active substances is well known in the art. Supplementary active ingredients also can be incorporated into the compositions.


Administration or delivery of the pharmaceutical compositions according to the present invention may be via any route so long as the target tissue is available via that route. For example, administration may be topical or by intradermal, subcutaneous, intramuscular, intraperitoneal, intraarterial, intracoronary, intrathecal, or intravenous injection, or by direct injection into target tissue (e.g., cardiac tissue). The stability and/or potency of the oligonucleotides disclosed herein allows for convenient routes of administration, including subcutaneous, intradermal, intravenous, and intramuscular. Pharmaceutical compositions comprising an oligonucleotide described herein may also be administered by catheter systems or systems that isolate coronary circulation for delivering therapeutic agents to the heart. Various catheter systems for delivering therapeutic agents to the heart and coronary vasculature are known in the art. Some non-limiting examples of catheter-based delivery methods or coronary isolation methods suitable for use in the present invention are disclosed in U.S. Pat. Nos. 6,416,510; 6,716,196; and 6,953,466; PCT Publication Nos. WO 2005/082440 and WO 2006/089340; and U.S. Patent Publication Nos. 2007/0203445, 2006/0148742, and 2007/0060907, which are all hereby incorporated by reference in their entireties.


The compositions or formulations may also be administered parenterally or intraperitoneally. By way of illustration, solutions of the conjugates as free base or pharmacologically acceptable salts can be prepared in water suitably mixed with a surfactant, such as hydroxypropylcellulose. Dispersions can also be prepared in glycerol, liquid polyethylene glycols, and mixtures thereof and in oils. Under ordinary conditions of storage and use, these preparations generally contain a preservative to prevent the growth of microorganisms.


The pharmaceutical forms suitable for injectable use or catheter delivery include, for example, sterile aqueous solutions or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. Generally, these preparations are sterile and fluid to the extent that easy injectability exists. Preparations should be stable under the conditions of manufacture and storage and should be preserved against the contaminating action of microorganisms, such as bacteria and fungi. Appropriate solvents or dispersion media may contain, for example, water, ethanol, polyol (for example, glycerol, propylene glycol, and liquid polyethylene glycol, and the like), suitable mixtures thereof, and vegetable oils. The proper fluidity can be maintained, for example, by the use of a coating, such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. The prevention of the action of microorganisms can be brought about by various antibacterial an antifungal agents, for example, parabens, chlorobutanol, phenol, sorbic acid, thimerosal, and the like. In many cases, it will be preferable to include isotonic agents, for example, sugars or sodium chloride. Prolonged absorption of the injectable compositions can be brought about by the use in the compositions of agents delaying absorption, for example, aluminum monostearate and gelatin.


Sterile injectable solutions may be prepared by incorporating the conjugates in an appropriate amount into a solvent along with any other ingredients (for example as enumerated above) as desired. Generally, dispersions are prepared by incorporating the various sterilized active ingredients into a sterile vehicle which contains the basic dispersion medium and the desired other ingredients, e.g., as enumerated above. In the case of sterile powders for the preparation of sterile injectable solutions, the preferred methods of preparation include vacuum-drying and freeze-drying techniques which yield a powder of the active ingredient(s) plus any additional desired ingredient from a previously sterile-filtered solution thereof.


Upon formulation, solutions are preferably administered in a manner compatible with the dosage formulation and in such amount as is therapeutically effective. The formulations may easily be administered in a variety of dosage forms such as injectable solutions, drug release capsules and the like. For parenteral administration in an aqueous solution, for example, the solution generally is suitably buffered and the liquid diluent first rendered isotonic for example with sufficient saline or glucose. Such aqueous solutions may be used, for example, for intravenous, intramuscular, subcutaneous and intraperitoneal administration. Preferably, sterile aqueous media are employed as is known to those of skill in the art, particularly in light of the present disclosure. By way of illustration, a single dose may be dissolved in 1 ml of isotonic NaCl solution and either added to 1000 ml of hypodermoclysis fluid or injected at the proposed site of infusion, (see for example, “Remington's Pharmaceutical Sciences” 15th Edition, pages 1035-1038 and 1570-1580). Some variation in dosage will necessarily occur depending on the condition of the subject being treated. The person responsible for administration will, in any event, determine the appropriate dose for the individual subject. Moreover, for human administration, preparations should meet sterility, pyrogenicity, general safety and purity standards as required by FDA Office of Biologics standards.


The present invention provides a method for delivering oligonucleotides to a cell (e.g., as part of a composition or formulation described herein), and methods for treating, ameliorating, or preventing the progression of a condition in a subject. As used herein, the term “subject” or “patient” refers to any vertebrate including, without limitation, humans and other primates (e.g., chimpanzees and other apes and monkey species), farm animals (e.g., cattle, sheep, pigs, goats and horses), domestic mammals (e.g., dogs and cats), laboratory animals (e.g., rodents such as mice, rats, and guinea pigs), and birds (e.g., domestic, wild and game birds such as chickens, turkeys and other gallinaceous birds, ducks, geese, and the like). In some embodiments, the subject is a mammal. In other embodiments, the subject is a human.


The oligonucleotide or pharmaceutical composition may be contacted in vitro or in vivo with a target cell (e.g., a mammalian cell). The cell may be a kidney, liver, vascular, or heart cell.


The method generally comprises administering the oligonucleotide or composition comprising the same to a subject or cell. The oligonucleotide, as described herein, can be mRNA or miRNA inhibitor. In some embodiments, the miRNA inhibitor is not a miR-208a inhibitor, miR-208b inhibitor, or miR-499 inhibitor. Thus, the patient may have a condition associated with, mediated by, or resulting from, expression or dysregulation of a mRNA or miRNA. Such conditions include, but are not limited to, cardiovascular conditions, such as cardiac hypertrophy, myocardial infarction, heart failure (e.g., congestive heart failure), myocardial ischemia, ischemia-reperfusion injury, vascular damage, coronary artery disease, peripheral artery disease, vulnerable plaque, restenosis, or pathologic cardiac fibrosis. Other conditions may include metabolic conditions, renal conditions (e.g., renal ischemia), hepatic conditions, or pulmonary conditions. Thus, the invention provides a use of the modified oligonucleotides and compositions of the present invention for treating such conditions, and for the preparation of medicaments for such treatments.


In certain embodiments, the subject (e.g., human patient) has one or more risk factors for a condition, such as, but not limited to, long standing uncontrolled hypertension, uncorrected valvular disease, chronic angina, recent myocardial infarction, congestive heart failure, congenital predisposition to heart disease and pathological hypertrophy. Alternatively or in addition, the patient may have been diagnosed as having a genetic predisposition to, for example, cardiac hypertrophy, or may have a familial history of, for example, cardiac hypertrophy.


In this aspect, the present invention may provide for an improved exercise tolerance, reduced hospitalization, better quality of life, decreased morbidity, and/or decreased mortality in a patient with heart failure or cardiac hypertrophy.


In certain embodiments, the activity of the miRNA in a tissue of interest, such as cardiac tissue, or as determined in serum, is reduced or inhibited.


In various embodiments, the pharmaceutical composition is administered by parenteral administration or by direct injection into heart tissue. The parenteral administration may be intravenous, subcutaneous, or intramuscular. In some embodiments, the composition is administered by oral, transdermal, sustained release, controlled release, delayed release, suppository, catheter, or sublingual administration. In certain embodiments, the oligonucleotide is administered at a dose of about 25 mg/kg or less, or a dose of about 10 mg/kg or less, or a dose of about 5 mg/kg or less. In these embodiments, the oligonucleotide or composition may be administered by intramuscular or subcutaneous injection, or intravenously.


In some embodiments, the methods further comprise scavenging or clearing the miRNA inhibitors following treatment. For example, a polynucleotide having a nucleotide sequence that is complementary to the inhibitor (e.g., a polynucleotide comprising a miRNA sequence) may be administered after therapy to attenuate or stop the function of the inhibitor.


The present invention is further illustrated by the following additional examples that should not be construed as limiting. Those of skill in the art should, in light of the present disclosure, appreciate that many changes can be made to the specific embodiments which are disclosed and still obtain a like or similar result without departing from the spirit and scope of the invention.


All publications and patents and patent applications mentioned in this specification are herein incorporated by reference to the same extent as if each individual publication, patent or patent application was specifically and individually indicated to be incorporated by reference.


EXAMPLES
Example 1. In Vivo Efficacy of AntimiR-208a

To determine whether the location of the LNA base affects in vivo efficacy of microRNA inhibitors (antimiRs) of identical length and LNA percentage, several antimiRs with different LNA modification patterns were designed and tested for efficacy in inhibiting miRNA function in vivo.


Sixteen antimiRs against miR-208a (FIG. 1A) with varying Tm measurements were designed, as depicted in Table 3 below:














TABLE 3





Mole-




Pre-


cule



LNA/
dicted


#
Alias
Sequence
Length
DNA
Tm







M-
208a_
1Cs; dTs; dTs; dTs;   
16
9/7
81


10101
LNA_
1Ts; 1Ts; dGs; 1Cs;   






DNA_
dTs; 1Cs; 1Gs; dTs;  






16_PS
1Cs; dTs; 1Ts; 1A







(SEQ ID NO: 76)








M-
208a 
1Cs; dTs; 1Ts; dTs;   
16
9/7
93


10679
LNA
1Ts; 1Ts; dGs; 1Cs;   






C_T_
dTs; 1Cs; dGs; 1Ts;  






DNA_
dCs; 1Ts; 1Ts; dA






16_1
(SEQ ID NO: 77)








M-
208a_
1Cs; dTs; 1Ts; 1Ts;   
16
9/7
90


10680
LNA_
1Ts, 1Ts; dGs; 1Cs;   






opt_1
dTs; 1Cs; dGs; dTs;  







1Cs; dTs; dTs; 1A







(SEQ ID NO: 78)








M-
208a_
1Cs; dTs; 1Ts; 1Ts;   
16
9/7
93


10681
LNA_
dTs; 1Ts; dGs; 1Cs;   






opt_2
1Ts; 1Cs; dGs; dTs;  







1Cs; dTs; 1Ts; dA







(SEQ ID NO: 79)








M-
208a_
1Cs; dTs; 1Ts; dTs;   
16
9/7
86


10682
LNA_
1Ts; dTs; 1Gs; dCs;   






opt_3
1Ts; dCs; 1Gs; dTs;  







1Cs; dTs; 1Ts; 1A







(SEQ ID NO: 80)








M-
208a_
1Cs; dTs; dTs; 1Ts;   
16
9/7
92


10683
LNA_
1Ts; dTs; 1Gs; dCs;   






opt_4
1Ts; 1Cs; dGs; 1Ts;  







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 81)








M-
208a_
1Cs; dTs; 1Ts; 1Ts;   
16
9/7
93


10673
LNA_
1Ts, 1Ts; dGs; 1Cs;   






opt_5
dTs; 1Cs; dGs; dTs;  







1Cs; dTs; 1Ts; dA







(SEQ ID NO: 82)








M-
208a_
1Cs; dTs; 1Ts; 1Ts;   
16
9/7
83


11184
10626
dTs; dTs; 1Gs; 1Cs;   







dTs; 1Cs; dGs; 1Ts;  







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 83)








M-
208a_
1Cs; dTs; dTs; dTs;   
16
9/7
86


11293
scr2_
1Ts; 1Ts; dGs; dCs;   






1
1Ts; 1Cs; 1Gs; dTs;  







1Cs; dTs; 1Ts; 1A







(SEQ ID NO: 84)








M-
208a_
1Cs; 1Ts; dTs; dTs;   
16
9/7
77


11294
scr2_
dTs; 1Ts; dGs; 1Cs;   






2
dTs; 1Cs; 1Gs; dTs;  







1Cs; dTs; 1Ts; 1A







(SEQ ID NO: 85)








M-
208a_
1Cs; 1Ts; dTs; dTs;   
16
9/7
76


11295
scr2_
dTs; 1Ts; 1Gs; dCs;   






3
dTs; 1Cs; 1Gs; 1Ts;  







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 86)








M-
208a_
1Cs; dTs; dTs; dTs;   
16
9/7
80


11296
scr2_
1Ts; 1Ts; dGs; 1Cs;   






4
dTs; 1Cs; 1Gs; dTs;  







1Cs; 1Ts; dTs; 1A







(SEQ ID NO: 87)








M-
208a_
1Cs; 1Ts; dTs; dTs;   
16
9/7
87


11297
scr2_
1Ts; dTs; dGs; 1Cs;   






5
1Ts; 1Cs; dGs; 1Ts;  







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 88)








M-
208a_
1Cs; dTs; 1Ts; dTs;   
16
9/7
83


11298
scr2_
1Ts; 1dTs; 1Gs; Cs;   






6
ddTs; 1Cs; Gs; 1Ts;  







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 89)








M-
208a_
1Cs; dTs; dTs; 1Ts;   
16
9/7
83


11299
scr2_
1Ts; dTs; 1Gs; 1Cs;   






7
dTs; 1Cs; dGs; 1Ts;  







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 90)








M-
208a_
1Cs; 1Ts; dTs; dTs;   
16
9/7
88


11300
scr2_
1Ts; dTs; 1Gs; dCs;   






8
1Ts; 1Cs; dGs; 1Ts;  







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 91)
















TABLE 4





Description of Notations


















deoxy A
dA



deoxy G
dG



deoxy C
dC



deoxy T
dT



lna A
lA



lnaG
lG



lna C
lC



lna T
lT



deoxy A P═S
dAs



deoxy G P═S
dGs



deoxy C P═S
dCs



deoxy T P═S
dTs



lna A P═S
lAs



lnaG P═S
lGs



lna C P═S
lCs



lna T P═S
lTs










These antimiRs were dorsally injected into 6-8 week old Sprague Dawley rats subcutaneously at a dose of 25 mg/kg (n=4 per group). Injection volume was 1.0 mL. A control oligonucleotide with similar LNA and DNA percentage (9/7) was also used as a chemistry control. This molecule number is M-10591 and was designed to target a C. elegans-specific miRNA. Four days after a single dose, these rats were sacrificed and plasma was collected for liver and kidney toxicology parameters. Additionally, heart, liver, and kidney were collected for molecular analysis including miRNA inhibition, target de-repression, and antimiR-distribution quantification. RNA was isolated from cardiac tissue and real-time PCR was performed. All antimiRs designed against miR-208a showed significant inhibition of miR-208a suggesting all antimiRs were delivered to cardiac tissue (FIG. 1B). To determine if miR-208a inhibition correlated to in vivo efficacy, miR-208a targets were assessed for de-repression by performing real-time PCR for the miR-208a target, Dynit1. Surprisingly, only four of the sixteen antimiRs tested showed significant de-repression of Dynlt1 (FIG. 1C).


To determine if treatment with any of these antimiRs resulted in elevated liver and/or kidney toxicology parameters, ELISAs for ALT, AST, and BUN were performed to assess liver and kidney function. No antimiR-treated group showed any elevation in either liver or kidney toxicology parameters (FIG. 1D).


To determine if the difference in efficacy between compounds is due to better cardiac distribution for the efficacious molecules, antimiR distribution to the heart, liver, and kidney for 2 antimiRs that showed efficacy (M-10101 and M-10683) and 2 antimiRs that did not show efficacy (M-10673 and M-10681) were assessed. ELISA-based distribution analyses showed no better cardiac presence for the efficacious compounds compared to the non-efficacious compounds. In fact, the non-efficacious compounds appeared to show better distribtuion to all tissues. (FIG. 1E).


These data suggest different LNA and DNA placement within the antimiR results in significantly different antimiR efficacy as it pertains to the heart, with the LNA/DNA “motif” of M-10101 sequnce appearing to be the best compound for cardiac efficacy.


Example 2. In Vivo Efficacy of AntimiR-208b

To test if the efficacious LNA/DNA motif of M-10101 remains efficacious for additional miRNAs, a subset of these for other miRNAs, including miR-208b, miR-29, miR-378, miR-199a, and miR-92a was tested. All experimental designs were the same as performed for miR-208a as described in Example 1.


Nine antimiRs against miR-208b with LNA and DNA placements similar to those found for the miR-208a screen were synthesized (FIG. 2A), with varying Tm measurements was designed, as depicted in Table 5 below (description of notations is as described in Table 4):














TABLE 5





Mole-




Pre-


cule



LNA/
dicted


#
Alias
Sequence
Length
DNA
Tm







M-
208b_
1Cs; dCs; dTs; dTs; 
16
9/7
82


10707
10101
1Ts; 1Ts; dGs; 1Ts; 







dTs; 1Cs; 1Gs; dTs; 







1Cs; dTs; 1Ts; 1A







(SEQ ID NO: 92)








M-
208b_
1Cs; dCs; 1Ts; dTs; 
16
9/7
91


11283
10679
1Ts; 1Ts; dGs; 1Ts; 







dTs; 1Cs; dGs; 1Ts; 







dCs; 1Ts; 1Ts; dA







(SEQ ID NO: 93)








M-
208b_
1Cs; dCs; 1Ts; 1Ts; 
16
9/7
89


11284
10680
1Ts; 1Ts; dGs; 1Ts; 







dTs; 1Cs; dGs; dTs; 







1Cs; dTs; dTs; 1A







(SEQ ID NO: 94)








M-
208b_
1Cs; dCs; 1Ts; 1Ts; 
16
9/7
94


11285
10681
dTs; 1Ts; dGs; 1Ts; 







1Ts; 1Cs; dGs; dTs; 







1Cs; dTs; 1Ts; dA







(SEQ ID NO: 95)








M-
208b_
1Cs; dCs; 1Ts; dTs; 
16
9/7
86


11286
10682
1Ts; dTs; 1Gs; dTs; 







1Ts; dCs; 1Gs; dTs; 







1Cs; dTs; 1Ts; 1A







(SEQ ID NO: 96)








M-
208b_
1Cs; dCs; dTs; 1Ts; 
16
9/7
93


11287
10683
1Ts; dTs; 1Gs; dTs; 







1Ts; 1Cs; dGs; 1Ts; 







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 97)








M-
208b_
1Cs; dCs; 1Ts; 1Ts; 
16
9/7
91


11288
10673
1Ts; 1Ts; dGs; 1Ts; 







dTs; 1Cs; dGs; dTs; 







1Cs; dTs; 1Ts; dA







(SEQ ID NO: 98)








M-
208b_
1Cs; dCs; 1Ts; 1Ts; 
16
9/7
89


11289
10626
dTs; dTs; 1Gs; 1Ts; 







dTs; 1Cs; dGs; 1Ts; 







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 99)








M-11290
208b_
1Cs; 1Cs; dTs; dTs; 
16
9/7
92



LNA_
1Ts; dTs; 1Gs; 1Ts; 






opt6
dTs; 1Cs; dGs; 1Ts; 







dCs; 1Ts; dTs; 1A







(SEQ ID NO: 100)









These antimiRs were dorsally injected into 6-8 week old Sprague Dawley rats subcutaneously at a dose of 25 mg/kg (n=4 per group). Injection volume was 1.0 mL. A control oligonucleotide with similar LNA and DNA percentage (9/7) was also used as a chemistry control. This molecule number is M-10591 and was designed to target a C. elegans-specific miRNA. Four days after a single dose, these rats were sacrificed and the heart was collected for molecular analysis including miRNA inhibition and target de-repression. RNA was isolated from cardiac tissue and real-time PCR was performed. All antimiRs designed against miR-208b showed significant inhibition of miR-208b suggesting all antimiRs were delivered to cardiac tissue (FIG. 2B). To determine if miR-208b inhibition correlated to in vivo efficacy, the miR-208b target, Dynlt1, was assessed for de-repression by performing real-time PCR. Surprisingly, only M-10707 showed significant de-repression of Dynlt1 (FIG. 2C), which is the same LNA/DNA motif that showed the best efficacy for miR-208a. (FIG. 1C).


These data suggest the LNA/DNA motif of M-10101 and M-10707 (which is the same) confers cardiac efficacy in vivo.


Example 3. In Vivo Efficacy of AntimiR-378

To determine if motif M-10101 extends beyond the miR-208 family, 7 antimiRs against miR-378 with LNA and DNA placements similar to those found for the miR-208a screen (FIG. 3A), with varying Tm measurements as depicted in Table 6 below (description of notations is as described in Table 4), were designed and synthesized:














TABLE 6





Mole-




Pre-


cule



LNA/
dicted


#
Alias
Sequence
Length
DNA
Tm







M-
378_
1Cs; dTs; dGs; dAs; 
16
9/7
86


11192
10101
1Cs; 1Ts; dCs; 1Cs; 







dAs; 1As; 1Gs; dTs; 







1Cs; dCs; 1As; 1Gs







(SEQ ID NO: 101)








M-
378_
1Cs; dTs; 1Gs; 1As; 
16
9/7
89


11193
10680
1Cs; 1Ts; dCs; 1Cs; 







dAs; 1As; dGs; dTs; 







1Cs; dCs; dAs; 1Gs







(SEQ ID NO: 102)








M-
378_
1Cs; dTs; 1Gs; 1As; 
16
9/7
89


11194
10681
dCs; 1Ts; dCs; 1Cs; 







1As; 1As; dGs; dTs; 







1Cs; dCs; 1As; dGs







(SEQ ID NO: 103)








M-
378_
1Cs; dTs; 1Gs; dAs; 
16
9/7
86


11195
10682
1Cs; dTs; 1Cs; dCs; 







1As; dAs; 1Gs; dTs; 







1Cs; dCs; 1As; 1Gs







(SEQ ID NO: 104)








M-
378_
1Cs; dTs; dGs; 1As; 
16
9/7
91


11196
10683
1Cs; dTs; 1Cs; dCs; 







1As; 1As; dGs; 1Ts; 







dCs; 1Cs; dAs; 1Gs







(SEQ ID NO: 105)








M-
378_
1Cs; dTs; 1Gs; 1As; 
16
9/7
95


11197
10673
1Cs; 1Ts; dCs; 1Cs; 







dAs; 1As; dGs; dTs; 







1Cs; dCs; 1As; dGs







(SEQ ID NO: 106)








M-
378_
1Cs; dTs; 1Gs; 1As; 
16
9/7
95


11198
10626
dCs; dTs; 1Cs; 1Cs; 







dAs; 1As; dGs; 1Ts; 







dCs; 1Cs; dAs; 1Gs







(SEQ ID NO: 107)









These antimiRs were dorsally injected into 6-8 week old Sprague Dawley rats subcutaneously at a dose of 25 mg/kg (n=4 per group). Injection volume was 1.0 mL. A control oligonucleotide with similar LNA and DNA percentage (9/7) was also used as a chemistry control. This molecule number is M-10591 and was designed to target a C. elegans-specific miRNA. Four days after a single dose, these rats were sacrificed and the heart was collected for molecular analysis including miRNA inhibition and target de-repression. RNA was isolated from cardiac tissue and real-time PCR was performed. All antimiRs designed against miR-378 showed significant inhibition of miR-378 suggesting all antimiRs were delivered to cardiac tissue (FIG. 3B). To determine if miR-378 inhibition correlated to in vivo efficacy, we assessed the miR-378 target, Gfpt2, for de-repression by performing real-time PCR. Surprisingly, only M-11192 showed significant de-repression of Gfpt2 (FIG. 3C), which is the same LNA/DNA motif that showed the best efficacy for miR-208a and miR-208b in the heart. (FIGS. 1C and 2C).


These data highly suggest the LNA/DNA motif of M-10101, M-10707, and M-11192 (which is the same) confers cardiac efficacy in vivo.


Example 4. In Vivo Efficacy of AntimiR-29

Seven antimiRs against miR-29b with LNA and DNA placements similar to those found for the miR-208a screen (FIG. 4A) were synthesized to determine if this motif confers efficacy in further miRNA families. The sequence and modification patterns of these antimiRs with their corresponding predicted Tm values are depicted in Table 7 below (description of notations is as described in Table 4):














TABLE 7





Mole-




Pre-


cule



LNA/
dicted


#
Alias
Sequence
Length
DNA
Tm







11185
29b_
1Gs; dAs; dTs; dTs; 
16
9/7
84



10101
1Ts; 1Cs; dAs; 1As; 







dAs; 1Ts; 1Gs; dGs; 







1Ts; dGs; 1Cs; 1Ts







(SEQ ID NO: 108)








11186
29b_
1Gs; dAs; 1Ts; 1Ts; 
16
9/7
91



10680
1Ts; 1Cs ;dAs; 1As; 







dAs; 1Ts; dGs; dGs; 







1Ts; dGs; dCs; 1Ts







(SEQ ID NO: 109)








11187
29b_
1Gs; dAs; 1Ts; 1Ts; 
16
9/7
87



10681
dTs; 1Cs; dAs; 1As; 







1As; 1Ts; dGs; dGs; 







1Ts; dGs; 1Cs; dTs







(SEQ ID NO: 110)








11188
29b_
1Gs; dAs; 1Ts; dTs; 
16
9/7
82



10682
1Ts; dCs; 1As; dAs; 







1As; dTs; 1Gs; dGs; 







1Ts; dGs; 1Cs; 1Ts







(SEQ ID NO: 111)








11189
29b_
1Gs; dAs; dTs; 1Ts; 
16
9/7
85



10683
1Ts; dCs; 1As; dAs; 







1As; 1Ts; dGs; 1Gs; 







dTs; 1Gs; dCs; 1Ts







(SEQ ID NO: 112)








11190
29b_
1Gs; dAs; 1Ts; 1Ts; 
16
9/7
96



10673
1Ts; 1Cs; dAs; 1As; 







dAs; 1Ts; dGs; dGs; 







1Ts; dGs; 1Cs; dTs







(SEQ ID NO: 113)








11191
29b_
1Gs; dAs; 1Ts; 1Ts; 
16
9/7
82



10626
dTs; dCs; 1As; 1As; 







dAs; 1Ts; dGs; 1Gs; 







dTs; 1Gs; dCs; 1Ts







(SEQ ID NO: 114)









These antimiRs were dorsally injected into 6-8 week old Sprague Dawley rats subcutaneously at a dose of 25 mg/kg (n=4 per group). Injection volume was 1.0 mL. A control oligonucleotide with similar LNA and DNA percentage (9/7) was also used as a chemistry control. This molecule number is M-10591 and was designed to target a C. elegans-specific miRNA. Four days after a single dose, these rats were sacrificed and the heart, liver, and kidney were collected for molecular analysis including miRNA inhibition, target de-repression, and antimiR-distribution quantification. RNA was isolated from cardiac, hepatic, and renal tissue and real-time PCR was performed. All antimiRs designed against miR-29 showed significant inhibition of miR-29 family members in all tissues suggesting all antimiRs were delivered to these three tissues (FIG. 4B).


To determine if miR-29 family inhibition correlated with in vivo efficacy, the miR-29 targets, Mcl1 and Dnmt3b, were assess for de-repression by performing real-time PCR. Surprisingly, only M-11185 showed significant de-repression of Mcl1 in the heart and trending de-repression of Dnmt3b (FIG. 4C), which is the same LNA/DNA motif that showed the best efficacy for miR-208a, miR-208b, and miR-378 in the heart. (FIGS. 1C, 2C, and 3C). Surprisingly, all antimiR-29 compounds appeared to show de-repression of Mcl1 in the liver, furthering the notion that this motif confers cardiac efficacy while the other compounds are active in other tissues.


To determine if the difference in efficacy between compounds is due to better cardiac distribution for the efficacious molecules, we quantified antimiR distribution to the heart and liver for all antimiR-29 compounds. ELISA-based distribution analyses showed no better cardiac presence for the most efficacious compound (M-11185) compared to the less efficacious compounds. For hepatic tissue where efficacy was similar among compounds, distribution was similar as well (FIG. 4D).


Example 5. In Vivo Efficacy of AntimiR-199

Five antimiRs against miR-199a with LNA and DNA placements similar to those found for the miR-208a screen (FIG. 5A) were synthesized to determine if this motif confers efficacy in further miRNA families. The sequence and modification patterns of these antimiRs with their corresponding predicted Tm values are depicted in Table 8 below (description of notations is as described in Table 4). The M-10518 compound contains the same LNA and DNA placements as M-10101 (antimiR-208a), M-10707 (antimiR-208b), M-11192 (antimiR-378), and M-11185 (antimiR-29).














TABLE 8





Mole-




Pre-


cule



LNA/
dicted


#
Alias
Sequence
Length
DNA
Tm







10518
199a_
1Gs; dTs; dAs; dGs; 
16
9/7
93



10101
1Ts; 1Cs; dTs; 1Gs; 







dAs; 1As; 1Cs; dAs; 







1Cs; dTs; 1Gs; 1Gs







(SEQ ID NO: 115)








11390
199a_
1Gs; dTs; dAs; dGs; 
16
9/7
92



10293
1Ts; 1Cs; dTs; dGs; 







1As; 1As; 1Cs; dAs; 







1Cs; dTs; 1Gs; 1Gs







(SEQ ID NO: 116)








11391
199a_
1Gs; 1Ts; dAs; dGs; 
16
9/7
86



10294
dTs; 1Cs; dTs; 1Gs; 







dAs; 1As; 1Cs; dAs; 







1Cs; dTs; 1Gs; 1Gs







(SEQ ID NO: 117)








11392
199a_
1Gs; dTs; dAs; dGs; 
16
9/7
92



10296
1Ts; 1Cs; dTs; 1Gs; 







dAs; 1As; 1Cs; dAs; 







1Cs; 1Ts; dGs; 1Gs







(SEQ ID NO: 118)








11393
199a_
1Gs; dTs; dAs; 1Gs; 
16
9/7
86



10683
1Ts; dCs; 1Ts; dGs; 







1As; 1As; dCs; 1As; 







dCs; 1Ts; dGs; 1Gs







(SEQ ID NO: 119)









These antimiRs were dorsally injected into 6-8 week old Sprague Dawley rats subcutaneously at a dose of 25 mg/kg (n=4 per group). Injection volume was 1.0 mL. A control oligo with similar LNA and DNA percentage (9/7) was also used as a chemistry control. This molecule number is M-10591 and was designed to target a C. elegans-specific miRNA. Four days after a single dose, these rats were sacrificed and plasma was collected for liver and kidney toxicology parameters. Additionally, heart, lung, liver, and kidney were collected for molecular analysis including miRNA inhibition and target de-repression. RNA was isolated from cardiac, pulmonary, hepatic, and renal tissue and real-time PCR was performed. All antimiRs designed against miR-199a showed significant inhibition of miR-199a in all tissues suggesting all antimiRs were delivered to these four tissues (FIG. 5B).


To determine if miR-199a inhibition correlated to in vivo efficacy, we assessed the miR-199 target, Ddr1 for de-repression by performing real-time PCR. Surprisingly, all antimiRs targeting miR-199a appeared to show Ddr1 target de-repression in the heart with the exception of M-11390 (FIG. 5C). For other tissues, different compounds showed varying degrees of target regulation, however, M-10518 (which is the M-10101 motif) consistently appeared to show target de-repression for all tissues, suggesting this motif confers cardiac and multi-tissue efficacy in vivo.


Example 6. In Vivo Efficacy of AntimiR-92a

Three antimiRs against miR-92a with LNA and DNA placements similar to those found for the miR-208a screen were synthesized to determine if this motif confers efficacy in further miRNA families. The sequence and modification patterns of these antimiRs with their corresponding predicted Tm values are depicted in Table 9 below (description of notations is as described in Table 4). The M-11127 compound contains the same LNA and DNA placements as M-10101 (antimiR-208a), M-10707 (antimiR-208b), M-11192 (antimiR-378), M-11185 (antimiR-29), and M-10518 (antimiR-199a).














TABLE 9





Mole-




Pre-


cule



LNA/
dicted


#
Alias
Sequence
Length
DNA
Tm







10338
92a_
1Cs; dCs; 1Gs; dGs; 
16
9/7
85



LNA_
dGs; 1As; dCs; 1As; 






16
dAs; 1Gs; 1Ts; dGs; 






_PS
1Cs; 1As; dAs; 1T







(SEQ ID NO: 120)








11127
92a_
1Cs; dCs; dGs; dGs; 
16
9/7
89



LNA_
1Gs; 1As; dCs; 1As; 






16
dAs; 1Gs; 1Ts; dGs; 






_1
1Cs; dAs; 1As; 1T







(SEQ ID NO: 121)








11130
92a_
1Cs; dCs; 1Gs; dGs; 
16
9/7
86



LNA_
dGs; 1As; dCs; dAs; 






16
1As; 1Gs; 1Ts; dGs; 






_4
1Cs; 1As; dAs; 1T







(SEQ ID NO: 122)









These antimiRs were dorsally injected into 6-8 week old Sprague Dawley rats subcutaneously at a dose of 25 mg/kg (n=4 per group). Injection volume was 1.0 mL. Two days after a single dose, these rats were sacrificed and endothelial cells from the heart were collected for molecular analysis including miRNA inhibition and target de-repression. RNA was isolated from endothelial cells and real-time PCR was performed to assess de-repression of the miR-92a target, Map2K4. Administration of antimiR M-11127 (which is the M-10101 motif) as well as antimiR M-11130 resulted in a significant increase in Map2K4 expression in endothelial cells (FIG. 6), demonstrating that these two inhibitors have in vivo efficacy.

Claims
  • 1. An oligonucleotide comprising a sequence of 16 nucleotides, wherein the sequence is complementary to miR-29a or miR-29b or miR-29c wherein the oligonucleotide comprises no more than three contiguous locked nucleic acids (LNAs), and wherein the ratio of LNAs to non-LNA nucleic acids in the oligonucleotide is 9 to 7.
  • 2. The oligonucleotide of claim 1, wherein at least one of the non-locked nucleotides is 2′ deoxy, 2′ O-alkyl, or 2′ halo.
  • 3. The oligonucleotide of claim 1, wherein all of the non-locked nucleotides are 2′ deoxy.
  • 4. The oligonucleotide of claim 1, wherein at least one LNA has a 2′ to 4′ methylene bridge.
  • 5. The oligonucleotide of claim 1, wherein the oligonucleotide has a 5′ cap structure, 3′ cap structure, or 5′ and 3′ cap structure.
  • 6. The oligonucleotide of claim 1, wherein the oligonucleotide comprises one or more phosphorothioate linkages.
  • 7. The oligonucleotide of claim 6, wherein the oligonucleotide is fully phosphorothioate-linked.
  • 8. The oligonucleotide of claim 1, further comprising a pendent lipophilic or hydrophilic group.
  • 9. The oligonucleotide of claim 1, wherein from the 5′ end to the 3′ end, positions 1, 4, 11, and 16 are LNAs.
  • 10. The oligonucleotide of claim 1, wherein the oligonucleotide comprises a sequence selected from the group consisting of M-11185, M-11186, M-11187, M-11188, M-11189, M-11190, and M-11191.
  • 11. The oligonucleotide of claim 1, the oligonucleotide is selected from the group consisting of: (a) lGs;dAs;dTs;dTs;lTs;lCs;dAs;lAs;dAs;lTs;lGs;dGs;lTs;dGs;lCs;lTs (SEQ ID NO: 108),(b) lGs;dAs;lTs;lTs;lTs;lCs;dAs;lAs;dAs;lTs;dGs;dGs;lTs;dGs;dCs;lTs (SEQ ID NO: 109),(c) lGs;dAs;lTs;lTs;dTs;lCs;dAs;lAs;lAs;lTs;dGs;dGs;lTs;dGs;lCs;dTs (SEQ ID NO: 110),(d) lGs;dAs;lTs;dTs;lTs;dCs;lAs;dAs;lAs;dTs;lGs;dGs;lTs;dGs;lCs;lTs (SEQ ID NO: 111),(e) lGs;dAs;dTs;lTs;lTs;dCs;lAs;dAs;lAs;lTs;dGs;lGs;dTs;lGs;dCs;lTs (SEQ ID NO: 112),(f) lGs;dAs;lTs;lTs;lTs;lCs;dAs;lAs;dAs;lTs;dGs;dGs;lTs;dGs;lCs;dTs (SEQ ID NO: 113), and(g) lGs;dAs;lTs;lTs;dTs;dCs;lAs;lAs;dAs;lTs;dGs;lGs;dTs;lGs;dCs;lTs (SEQ ID NO: 114).
  • 12. A pharmaceutical composition comprising an effective amount of the oligonucleotide of claim 11, or a pharmaceutically-acceptable salt thereof, and a pharmaceutically-acceptable carrier or diluent.
  • 13. The pharmaceutical composition of claim 12, wherein the pharmaceutically-acceptable carrier comprises a colloidal dispersion system, macromolecular complex, nanocapsule, nanoparticle, microsphere, bead, oil-in-water emulsion, micelle, mixed micelle, or liposome.
  • 14. The pharmaceutical composition of claim 12, wherein the pharmaceutically-acceptable carrier or diluent consists essentially of saline.
  • 15. A method of reducing or inhibiting the activity of miR-29 in a cell comprising contacting the cell with the oligonucleotide of claim 1.
  • 16. The method of claim 15, wherein the cell is a mammalian cell.
  • 17. The method of claim 15, wherein the cell is in vivo or ex vivo.
  • 18. A method of increasing MCL1 or DNMT3b expression in a subject, comprising administering to the subject a pharmaceutical composition comprising the oligonucleotide of claim 1.
  • 19. The method of claim 18, wherein the pharmaceutical composition is administered by intravenous, subcutaneous, intraperitoneal, intramuscular, oral, transdermal, sustained release, controlled release, delayed release, suppository, catheter, or sublingual administration.
  • 20. The method of claim 18, wherein the subject is a human.
  • 21. The method of claim 18, wherein the oligonucleotide comprises a sequence selected from the group consisting of M-11185, M-11186, M-11187, M-11188, M-11189, M-11190, and M-11191.
  • 22. The method of claim 18, the oligonucleotide is selected from the group consisting of: (a) lGs;dAs;dTs;dTs;lTs;lCs;dAs;lAs;dAs;lTs;lGs;dGs;lTs;dGs;lCs;lTs (SEQ ID NO: 108),(b) lGs;dAs;lTs;lTs;lTs;lCs;dAs;lAs;dAs;lTs;dGs;dGs;lTs;dGs;dCs;lTs (SEQ ID NO: 109),(c) lGs;dAs;lTs;lTs;dTs;lCs;dAs;lAs;lAs;lTs;dGs;dGs;lTs;dGs;lCs;dTs (SEQ ID NO: 110),(d) lGs;dAs;lTs;dTs;lTs;dCs;lAs;dAs;lAs;dTs;lGs;dGs;lTs;dGs;lCs;lTs (SEQ ID NO: 111),(e) lGs;dAs;dTs;lTs;lTs;dCs;lAs;dAs;lAs;lTs;dGs;lGs;dTs;lGs;dCs;lTs (SEQ ID NO: 112),(f) lGs;dAs;lTs;lTs;lTs;lCs;dAs;lAs;dAs;lTs;dGs;dGs;lTs;dGs;lCs;dTs (SEQ ID NO: 113), and(g) lGs;dAs;lTs;lTs;dTs;dCs;lAs;lAs;dAs;lTs;dGs;lGs;dTs;lGs;dCs;lTs (SEQ ID NO: 114).
  • 23. The method of claim 18; wherein the oligonucleotide is lGs;dAs;dTs;dTs;lTs;lCs;dAs;lAs;dAs;lTs;lGs;dGs;lTs;dGs;lCs;lTs (SEQ ID NO: 108).
  • 24. The method of claim 18, wherein the oligonucleotide is lGs;dAs;lTs;lTs;lTs;lCs;dAs;lAs;dAs;lTs;dGs;dGs;lTs;dGs;dCs;lTs (SEQ ID NO: 109).
CROSS-REFERENCE TO RELATED APPLICATIONS

This application is a continuation of U.S. application Ser. No. 15/154,711, filed May 13, 2016, issued as U.S. Pat. No. 9,803,202, which is a continuation of U.S. application Ser. No. 13/924,340, filed Jun. 21, 2013, issued as U.S. Pat. No. 9,388,408, which claims the benefit of U.S. Provisional Application No. 61/662,746, filed Jun. 21, 2012, and U.S. Provisional Application No. 61/801,533, filed Mar. 15, 2013, each of which is hereby incorporated by reference in its entirety.

US Referenced Citations (78)
Number Name Date Kind
7232806 Tuschl et al. Jun 2007 B2
7687617 Thrue et al. Mar 2010 B2
8222221 Corey et al. Jul 2012 B2
8258113 Dimmeler et al. Sep 2012 B2
8304397 Olson et al. Nov 2012 B2
8404659 Kauppinen et al. Mar 2013 B2
8481507 Olson et al. Jul 2013 B2
8642751 Dalby et al. Feb 2014 B2
8685946 Hutvagner et al. Apr 2014 B2
8912158 Dimmeler et al. Dec 2014 B2
8946179 Bennett et al. Feb 2015 B2
9163235 Van Rooij et al. Oct 2015 B2
9255267 Mendell et al. Feb 2016 B2
9279123 Dimmeler et al. Mar 2016 B2
9334497 Hutvagner et al. May 2016 B2
9388408 Van Rooij et al. Jul 2016 B2
9428749 Van Rooij et al. Aug 2016 B2
9803202 Van Rooij et al. Oct 2017 B2
9862949 Dimmeler et al. Jan 2018 B2
9885042 Dalby et al. Feb 2018 B2
20050059005 Tuschl et al. Mar 2005 A1
20060185027 Bartel et al. Aug 2006 A1
20070065840 Naguibneva et al. Mar 2007 A1
20070287179 Tuschl et al. Dec 2007 A1
20070292878 Raymond Dec 2007 A1
20080050744 Brown et al. Feb 2008 A1
20080176766 Brown et al. Jul 2008 A1
20080214437 Mohapatra et al. Sep 2008 A1
20090105174 Jayasena Apr 2009 A1
20090137504 Echwald et al. May 2009 A1
20090143326 Obad et al. Jun 2009 A1
20090180957 Olson et al. Jul 2009 A1
20090286753 Kauppinen et al. Nov 2009 A1
20090286969 Esau et al. Nov 2009 A1
20090291906 Esau et al. Nov 2009 A1
20090291907 Esau et al. Nov 2009 A1
20090293148 Ren et al. Nov 2009 A1
20090326049 Aristarkhov et al. Dec 2009 A1
20100004320 Elmen et al. Jan 2010 A1
20100029003 Bartel et al. Feb 2010 A1
20100173288 Zhang et al. Jul 2010 A1
20100210712 Hansen et al. Aug 2010 A1
20100269183 Olson et al. Oct 2010 A1
20100280094 Beuvink et al. Nov 2010 A1
20100286044 Litman et al. Nov 2010 A1
20100292297 Wang et al. Nov 2010 A1
20100298410 Obad et al. Nov 2010 A1
20110071211 Thum et al. Mar 2011 A1
20110098338 Hajjar et al. Apr 2011 A1
20110105593 Steel et al. May 2011 A1
20110117560 Spinale et al. May 2011 A1
20110152352 Hata et al. Jun 2011 A1
20110160285 Anderson et al. Jun 2011 A1
20110224277 Esau et al. Sep 2011 A1
20110294869 Petersen Dec 2011 A1
20110313019 Swayze et al. Dec 2011 A1
20120035243 Olson et al. Feb 2012 A1
20120041052 Beuvink et al. Feb 2012 A1
20120083596 Elmen et al. Apr 2012 A1
20120114744 Beuvink et al. May 2012 A1
20120172416 Velin et al. Jul 2012 A1
20120322851 Hardee et al. Dec 2012 A1
20130078225 Zeng et al. Mar 2013 A1
20130079505 Moeller et al. Mar 2013 A1
20130096290 Brown Apr 2013 A1
20130109738 Chang et al. May 2013 A1
20130137753 Samant et al. May 2013 A1
20130150256 Synnergren et al. Jun 2013 A1
20130156845 Manoharan et al. Jun 2013 A1
20130157883 Keller et al. Jun 2013 A1
20130171242 Lim et al. Jul 2013 A1
20140187603 Dalby et al. Jul 2014 A1
20150352055 Asin et al. Dec 2015 A1
20160208258 Dalby et al. Jul 2016 A1
20160237433 Dimmeler et al. Aug 2016 A1
20160319279 Hutvagner et al. Nov 2016 A1
20160326526 Van Rooij et al. Nov 2016 A1
20180237779 Dalby et al. Aug 2018 A1
Foreign Referenced Citations (106)
Number Date Country
101386848 Mar 2009 CN
1 959 012 Aug 2008 EP
2 113 567 Nov 2009 EP
2 194 129 Jun 2010 EP
2 208 798 Jul 2010 EP
2 388 327 Nov 2011 EP
2 388 328 Nov 2011 EP
2 447 274 May 2012 EP
2 205 737 Feb 2013 EP
2 559 442 Feb 2013 EP
2 604 690 Jun 2013 EP
2 610 342 Jul 2013 EP
2006-520760 Sep 2006 JP
2009-532044 Sep 2009 JP
2010-178741 Aug 2010 JP
2010-539959 Dec 2010 JP
2014-504857 Feb 2014 JP
WO-2001025478 Apr 2001 WO
WO-2004083430 Sep 2004 WO
WO-2004083430 Sep 2004 WO
WO-2005013901 Feb 2005 WO
WO-2005013901 Feb 2005 WO
WO-2005017145 Feb 2005 WO
WO-2005078096 Aug 2005 WO
WO-2005078096 Aug 2005 WO
WO-2005078139 Aug 2005 WO
WO-2005078139 Aug 2005 WO
WO-2005079397 Sep 2005 WO
WO-2005079397 Sep 2005 WO
WO-2005118806 Dec 2005 WO
WO-2005118806 Dec 2005 WO
WO-2006063356 Jun 2006 WO
WO-2006111512 Oct 2006 WO
WO-2006137941 Dec 2006 WO
WO-2006137941 Dec 2006 WO
WO-2007000668 Jan 2007 WO
WO-2007000668 Jan 2007 WO
WO-2007070483 Jun 2007 WO
WO-2007070483 Jun 2007 WO
WO-2007090073 Aug 2007 WO
WO-2007090073 Aug 2007 WO
WO-2007112753 Oct 2007 WO
WO-2007112753 Oct 2007 WO
WO-2007112754 Oct 2007 WO
WO-2007112754 Oct 2007 WO
WO-2008016924 Feb 2008 WO
WO-2008016924 Feb 2008 WO
WO-2008042231 Apr 2008 WO
WO-2008043521 Apr 2008 WO
WO-2008043521 Apr 2008 WO
WO-2008061537 May 2008 WO
WO-2008061537 May 2008 WO
WO-2008074328 Jun 2008 WO
WO-2008074328 Jun 2008 WO
WO-2008076324 Jun 2008 WO
WO-2008076324 Jun 2008 WO
WO-2008147839 Dec 2008 WO
WO-2009026576 Feb 2009 WO
WO-2009043353 Apr 2009 WO
WO-2009043353 Apr 2009 WO
WO-2009056116 May 2009 WO
WO-2009058818 May 2009 WO
WO-2009058818 May 2009 WO
WO-2009062169 May 2009 WO
WO-2009062169 May 2009 WO
WO-2009114681 Sep 2009 WO
WO-2009114681 Sep 2009 WO
WO-2009149182 Dec 2009 WO
WO-2010000665 Jan 2010 WO
WO-2008042231 Apr 2010 WO
WO-2010048585 Apr 2010 WO
WO-2010048585 Apr 2010 WO
WO-2010091204 Aug 2010 WO
WO-2010122538 Oct 2010 WO
WO-2010144485 Dec 2010 WO
WO-2011139911 Nov 2011 WO
WO-2011139911 Nov 2011 WO
WO-2011154553 Dec 2011 WO
WO-2011154553 Dec 2011 WO
WO-2011158191 Dec 2011 WO
WO-2012006577 Jan 2012 WO
WO-2012006577 Jan 2012 WO
WO-2012010711 Jan 2012 WO
WO-2012020307 Feb 2012 WO
WO-2012020307 Feb 2012 WO
WO-2012027206 Mar 2012 WO
WO-2012037254 Mar 2012 WO
WO-2012061810 May 2012 WO
WO-2012083005 Jun 2012 WO
WO-2012083005 Jun 2012 WO
WO-2012149646 Nov 2012 WO
WO-2013052965 Apr 2013 WO
WO-2013052965 Apr 2013 WO
WO-2013054113 Apr 2013 WO
WO-2013057527 Apr 2013 WO
WO-2013057527 Apr 2013 WO
WO-2013059496 Apr 2013 WO
WO-2013087907 Jun 2013 WO
WO-2013088338 Jun 2013 WO
WO-2013090457 Jun 2013 WO
WO-2013090457 Jun 2013 WO
WO-2013192486 Dec 2013 WO
WO-2013192576 Dec 2013 WO
WO-2013192576 Dec 2013 WO
WO-2016118612 Jul 2016 WO
WO-2016118612 Jul 2016 WO
Non-Patent Literature Citations (21)
Entry
Bellera, N. et al. (2014). “Single intracoronary injection of encapsulated antagomir-92a promotes angiogenesis and prevents adverse infarct remodeling,” J. Am. Heart Assoc. 3(5):e000946, 21 pages.
Bonauer, A. et al. (2009). “MicroRNA-92a controls angiogenesis and functional recovery of ischemic tissues in mice,” Science 324:1710-1713.
Daniel, J.M. et al. (2014). “Inhibition of miR-92a improves re-endothelialization and prevents neointima formation following vascular injury,” Cardiovasc. Res. 103:564-572.
Extended European Search Report dated on Apr. 12, 2018, for EP Application No. 18 154 472.7, filed on Jun. 21, 2013, 7 pages.
Hinkel, R. et al. (2013). “Inhibition of microRNA-92a protects against ischemia/reperfusion injury in a large-animal model,” Circulation 128:1066-1075.
Hullinger, T.G. et al. (2012). “Inhibition of miR-15 protects against cardiac ischemic injury,” Circ. Res. 110:71-81.
International Search Report for International Application No. PCT/US2011/065121, dated Jun. 5, 2012.
International Search Report for International Application No. PCT/US2013/047157, dated Feb. 10, 2014, 5 pages.
Kremp, E-M. et al. (2016). “MicroRNA target regulation is cell type specific and is influenced by stress conditions,” Circ. 134:A18709.
Lennox, K.A. et al. (2011). “Chemical modification and design of anti-miRNA oligonucleotides,” Gene Ther. 18:1111-1120.
Montgomery, R. L. et al. (2011). “Therapeutic inhibition of miR-208a improves cardiac function and survival during heart failure+ supplemental material,” Circulation 124(14):1537-1547.
Non-Final Office Action for U.S. Appl. No. 13/327,507, dated Feb. 27, 2013, 24 pages.
Non-Final Office Action for U.S. Appl. No. 14/107,784, dated Oct. 23, 2014, 26 pages.
Sedding, D. et al. (2010). “Abstract 17664: Role of miR-92a for endothelial cell function and reendothelialization following vascular injury,” Circ. 122:A17664, 3 total pages.
Sedding, D. et al. (2010). “Role of miR-92a for endothelial cell function and reendothelialization following vascular injury,” Med. Clinic I, Department of Cardiology University of Giessen, Presented at the Scientific Sessions of the American Heart Association on Resuscitation Science Symposium Conference, Nov. 13-17, 2010, 13 total pages.
Shigoka, M. et al. (2010). “Deregulation of miR-92a expression is implicated in hepatocellular carcinoma development,” Pathol. Int. 60:351-357.
Torres, A. et al. (2016). “In vitro and in vivo activity of miR-92a-Locked Nucleic Acid (LNA)-Inhibitor against endometrial cancer,” BMC Cancer 16(1):822.
Written Opinion for International Application No. PCT/US2011/065121, dated Jun. 5, 2012.
Written Opinion for International Application No. PCT/US2013/047157, dated Feb. 10, 2014, 6 pages.
Yu, S. et al. (2015). “High concentrations of uric acid inhibit angiogenesis via regulation of the Krüppel-like factor 2-vascular endothelial growth factor-A axis by miR-92a,” Circ. J. 79(11):2487-2498.
Veedu, R.N. et al. (2009). “Locked nucleic acid as a novel class of therapeutic agents,” RNA Biol. 6:321-323.
Related Publications (1)
Number Date Country
20180273944 A1 Sep 2018 US
Provisional Applications (2)
Number Date Country
61801533 Mar 2013 US
61662746 Jun 2012 US
Continuations (2)
Number Date Country
Parent 15154711 May 2016 US
Child 15703753 US
Parent 13924340 Jun 2013 US
Child 15154711 US