<<

US009078919B2 a2) United States Patent (0) Patent No.: US 9,078,919 B2 Olsonet al. (45) Date of Patent: *Jul. 14, 2015

(54) MICRO-RNAS OF THE MIR-15 FAMILY 2005/0182011 Al 8/2005 Olson etal. MODULATE CARDIOMYOCYTE SURVIVAL 2006/0058266 Al 3/2006 Manoharan etal. AND CARDIAC REPAIR 2006/0105360 Al 5/2006 Croce etal. 2006/0165659 Al 7/2006 Croceet al. 2006/0185027 Al 8/2006 Bartelet al. (71) Applicant: The Board of Regents, The University 2006/0189557 Al 8/2006 Slack et al. of Texas System, Austin, TX (US) 2006/0247193 Al 11/2006 Taira et al. 2006/0252722 Al 11/2006 Lollo et al. (72) Inventors: Eric N. Olson, Dallas, TX (US); Eva 2007/0026403 Al 2/2007 Hatzigeorgiouetal. 2007/0054287 Al 3/2007 Bloch van Rooij, Utrecht (NL) 2007/0213292 Al 9/2007 Stoffel et al. 2007/0259827 Al 11/2007 Aronin et al. (73) Assignee: The Board of Regents, the University 2007/0287179 Al 12/2007 Tuschletal. of Texas System, Austin, TX (US) 2007/0292878 Al 12/2007 Raymond. 2008/0050744 Al 2/2008 Brownetal. (*) Notice: Subject to any disclaimer, the term of this 2008/0176766 Al 7/2008 Brownetal. 2008/0220423 Al 9/2008 Molleret al. patent is extended or adjusted under 35 2009/0053718 Al 2/2009 Naguibnevaetal. U.S.C. 154(b) by 0 days. 2009/0092980 Al 4/2009 Arenzet al. 2009/0131356 Al 5/2009 Baderet al. This patent is subject to a terminal dis- 2009/0176723 Al 7/2009 Brownetal. claimer. 2009/0214477 Al 8/2009 Betzet al. 2009/0221685 Al 9/2009 Esauetal. (21) Appl. No.: 13/970,379 2009/0226528 Al 9/2009 Czechet al. 2009/0281167 Al 11/2009 Shenetal. (22) Filed: Aug. 19, 2013 2009/0286969 Al 11/2009 Esauet al. 2009/0286973 Al 11/2009 Manoharan etal. 2009/0291906 Al 11/2009 Esauet al. (65) Prior Publication Data 2009/0291907 Al 11/2009 Esauet al. US 2014/0051745 Al Feb. 20, 2014 2009/0293148 Al 11/2009 Renetal. 2009/0298174 Al 12/2009 Esauet al. 2009/0306181 Al 12/2009 Ikedaet al. 2009/0326049 Al 12/2009 Aristarkhovet al. Related U.S. Application Data 2010/0029003 Al 2/2010 Bartel et al. 2010/0069471 Al 3/2010 Manoharan etal. (63) Continuation of application No. 12/742,233, filed as 2010/0087512 Al 4/2010 Tuschletal. application No. PCT/US2008/083020 on Nov. 10, 2010/0087513 Al 4/2010 Tuschletal. 2008, now Pat. No. 8,513,209. (Continued) (60) Provisional application No. 60/986,798,filed on Nov. 9, 2007. FOREIGN PATENT DOCUMENTS (51) Int. Cl. EP 1627925 Al 2/2006 AG6IK 48/00 (2006.01) EP 1676914 Al 7/2006 AGIK 45/06 (2006.01) (Continued) AOIK 67/027 (2006.01) CIIN 15/113 (2010.01) OTHER PUBLICATIONS AGIK 31/7105 (2006.01) (52) U.S.C Young, “International Search Report,” 4 pages, from International CPC veeceseseeee A61K 45/06 (2013.01); AOLK 67/0275 Patent Application No. PCT/US08/83020, USPTO, Alexandria, Vir- (2013.01); A6LK 31/7105 (2013.01); C12N ginia, US (mailed May 13, 2009). 15/113 (2013.01); AOLK 2207/05 (2013.01); (Continued) AOLK 2217/058 (2013.01); AOLK 2227/105 (2013.01); AOLK 2267/0375 (2013.01); C12N 2310/113 (2013.01); CI2N 2310/14 (2013.01); Primary Examiner — Kimberly Chong C12N 2330/10 (2013.01); C12N 2830/008 (2013.01) (74) Attorney, Agent, or Firm — Cooley LLP (58) Field of Classification Search None (57) ABSTRACT See application file for complete search history. A family of microRNAs, called the miR-15 family, which (56) References Cited includes miR-195, are shown to be up-regulated during U.S. PATENT DOCUMENTS pathological cardiac remodeling and repress the expression of mRNAsrequired for cell proliferation and survival, with 7,232,806 B2 6/2007 Tuschletal. consequent loss of cardiomyocytes. Strategies to block 7,482,117 B2 1/2009 Cargill et al. expression ofthe miR-15 family in the heart as a treatment for 7,582,744 B2 9/2009 Manoharan et al. diverse cardiac disease are provided. 7,759,319 B2 7/2010 Lollo et al. 2005/0026169 Al 2/2005 Cargill et al. 2005/0059005 Al 3/2005 Tuschlet al. 2005/0075492 Al 4/2005 Chenetal. 18 Claims, 9 Drawing Sheets US 9,078,919 B2 Page 2

(56) References Cited WO WO2008/061537 A2 5/2008 WO _-WO 2008/073920 A2 —-6/2008 — U.S. PATENT DOCUMENTS WO WO 2008/073921 A2 6/2008 WO WO 2008/073922 A2 6/2008 2010/0093837 Al 4/2010 Tuschlet al. WO WO 2008/073923 A2 6/2008 —_ 2010/0099748 Al 4/2010 Tuschlet al. WO WO 2008/074328 A2 6/2008 2010/0113284 Al 5/2010 Aristarkhovet al. WO WO 2008/085797 A2 7/2008 2010/0113561 Al 5/2010 Tuschlet al. WO WO 2008/112226 A2 —_-9/2008 WO WO 2008/116267 Al 10/2008 — WO WO 2008/147430 A2 12/2008 — FOREIGN PATENT DOCUMENTS WO WO 2008/147839 Al 12/2008 — WO WO 2009/012263 A2 1/2009 EP 1777301 A2 4/2007 WO WO 2009/043353 A2 4/2009 EP 1959012 A2 8/2008 WO WO 2009/044895 Al 4/2009 EP 2105145 Al 9/2009 WO WO 2009/058818 A2 5/2009 EP 2113567 Al 11/2009 WO WO 2009/062169 A2 5/2009 JP 2006-506469 2/2006 WO WO 2009/121031 Al 10/2009 JP 2006-5 19008 8/2006 WO WO 2009/149182 Al 12/2009 JP 2006-292367 10/2006 WO WO 2010/036939 A2 4/2010 WO WO 03/029459 A2 4/2003 WO WO 2010/048585 A2 4/2010 WO WO 03/093441 A2 11/2003 WO WO 2004/043387 A2 5/2004 OTHER PUBLICATIONS WO WO 2004/076622 A2 9/2004 WO WO 2005/013901 A2 2/2005 Lagos-Quintanaet al., “New microRNAsfrom mouse and. human,” Wo WO 2005/017145 Al 2/2005 WO WO 2005/040419 Al 5/2005 RNA,vol. 9:175-179, 2003. — Wo WO 2005/056797 Al 6/2005 Lagos-Quintanaet al., “Identification of tissue-specific microRNAs WO WO 2005/078096 A2 8/2005 from mouse,” Current Biology, vol. 12:735-739, 2002. WO WO2005/078139 A2 8/2005 Sempere et al., “Expression profiling of mammalian microRNAs —- WoO WO 2005/097205 A2 10/2005 uncoversa subset ofbrain-expressed microRNAswith possible roles — WO WO 2005/098029 A2 10/2005 in murine and human neuronal differentiation”’ Genome Biology, — WO WO 2005/118806 A2 12/2005 = vol. 5:R13, 2004. WO WO 2006/020768 A2 2/2006 Wo WO 2006/048553 Al 5/2006 Van Rooij et al. “A signature pattern of stress-responsive Wo WO 2006/063356 Al 6/2006 microRNAsthat can evoke cardiac hypertrophy and heart failure,” Wo WO 2006/071884 A2 7/2006 Proc. Natl. Acad. Sci. USA, vol. 103: 18255-18260, 2006. WO WO 2006/108473 Al 10/2006 Landgrafet al., “A mammalian microRNAexpression atlas based on — WO WO 2006/111512 Al 10/2006 small RNAlibrary sequencing.” Cell, vol. 129: 1401-1414, 2007. WO WO 2006/133022 A2 12/2006 Landgrafet al., “A mammalian microRNAexpressionatlas based on — wo wo soooOooce& “ pos sma RNA library sequencing,” Cell, Supplementary Table S12, we we Rees ‘ seen Van Rooij et al., “Control of stress-dependent cardiac growth and WO WO 2007/044362 A2 4/2007 gene expression by a microRNA,”Science, vol. 316: 575-579, 2007. Wo WO 2007/053184 A2 5/2007 Van Roojj et al., “MicroRNA function during cardiac hypertrophy,” WO WO2007/056826 Al 5/2007 Abstract and Poster from 3rd Annual Symposium of the American Wo WO 2007/087451 A2 8/2007 Heart Association Council on Basic Cardiovascular Sciences: Trans- WO WO 2007/089607 A2 8/2007 lation of Basic Insights Into Clinical Practice: Jul. 31-Aug. 3, 2006 WO WO2007/090073 A2 8/2007 Keystone Conference Center Keystone, CO. — WO WO2007/092181 A2 8/2007 Ping et al., “The Study and Use of Antisense Oligonucleotide Tech- WO WO 2007/092182 A2 8/2007 nique in Cardiovascular Diseases,” Chinese Journal of Clinical Phar- WO WO 2007/095387 A2 8/2007 macology and Therapeutics, vol. 11: 241-245, 2006. WO WO 2007/103808 A2 9/2007 English Translation of Examination Report for Chinese Application WO WO 2007/112754 A2 10/2007 — WO WO 2007/147067 A2 12/2007 No. 200880 124495.1 issued Nov. 14, 2011. - wo WO 2007/147409 A2 12/2007 Seranski, Supplementary European Search Report and Opinion for wo WO 2008/024499 A? 2/2008 European Application No. 08847645.2, 9 pages, European Patent wo WO 2008/036776 A2 3/2008 Office, Munich, mailed Jan. 27, 2012. WO WO 2008/042231 A2 4/2008 Young, Written Opinion of the International Search Authority for WO WO 2008/043521 A2 4/2008 PCT/US2008/083020, 7 pages, mailed May 13, 2009. U.S. Patent Jul. 14, 2015 Sheet 1 of 9 US 9,078,919 B2

mF ei 3 g i nie,umoy

Beh

Pole)

CHE

nhesdr

OBIOY Rey L

avi ULTOUWORS Aq

peyninBey Lue L eanci | U.S. Patent Jul. 14, 2015 Sheet 2 of 9 US 9,078,919 B2

LE wah

BY

OSL

ALCUOUE

wwe

=

POPU

GR

ya

Sy WG2L

MMCo wel

pageiianir: PE

aoe YEG wie LAA YEO LE RU LAS

| U.S. Patent Jul. 14, 2015 Sheet 3 of 9 US 9,078,919 B2

A microRNAsignature of the hypertrophic heart

Normal | Failing Fold

miR-23 _

miR-24 1.5

miR-125b 1.3

miR-195 3.0

miR-199a 2.1

miR-214 1.6 Figure 2 U.S. Patent Jul. 14, 2015 Sheet 4 of 9 US 9,078,919 B2

€ &&. &.8 we OS

neo 8‘ x*

SyYBOMA FZ SHAM O

3

PiQure

B2 pyaunbi4

Awa ‘Socae mean 2 smwom@ aL pus E | [ ann ot [ ue o ceo

9,078,919 | 1 L,

US +3

‘et ret

ca PT

9 dNd S6l VNU q of 5 Sheet OL Be

n 2se)ame 2015 FRB 14, RR Jul. 8k aday-DaAy

&# st

5 8

Surasy204s JAPWEIPAT SSOOyONFEM, ee eee V jeuonpesy

uoljeinBaidn gGp-ylw 0} esuodsau ulAudosedAy pue uolesouajep jeuonouny Patent U.S.

U.S. Patent Jul. 14, 2015 Sheet 6 of 9 US 9,078,919 B2 Tg miR-214

Wild-byoe

age oe ben

ya Ras

U.S. Patent Jul. 14, 2015 Sheet 7 of 9 US 9,078,919 B2

(ITT

(wT

(9T

{IT

(4

(6

-ON

=ON

?ON

?ON

‘ON

‘ON

aI

dI

I

dI

dI

dr

528)-ODD9NNVNVVVNDIVODVODVN

O28)

525)

O48)

Sas)

Sas)

-VOVANNDONVINVOVOOVOOWN

ONONANNDONVVAVIVOSVO9"N

DODINAVAVVVNDIVODVOO"NN

~DOONNVNVVYVOVOVODVOOVN

NONNNDONSNOVIVIDVINVO

oluaHsuely) BY NRL

# ee

ed dk pS

TLRS

VSZ Z

sjoBue} J0}Oe}

oLUL-

[eAIAINS

7 8/9A9

an

.

‘La peonpul-q45L

eth

Ajiuues UOISIAIP

eT

|]99 .

sarees

‘ba

sunoAg

GL

12194

161948 7494

-

LLU /-— yi

[12D se

he To

ett

ae

be)

=

|

L G6L-ylw

9

ainBi4 qd V U.S. Patent Jul. 14, 2015 Sheet 8 of 9 US 9,078,919 B2

“tn aE : oarfq ut ay‘ che

m4 rq fh, GPC.

Z C

Tes Pa

why BPS

Bs.

3

us &

ott ! me wh ie9 ad fy da te i] os ge Sn ad & Te

Piers Bs & Rew =

"ITE oo ek ie

U.S. Patent Jul. 14, 2015 Sheet 9 of 9 US 9,078,919 B2

S60

ohn

bea

ee ghe

eu

BAIEL

LIBSE

POULICN US 9,078,919 B2 1 2 MICRO-RNAS OF THE MIR-15 FAMILY all forms of cardiac disease, including those arising from MODULATE CARDIOMYOCYTE SURVIVAL hypertension, mechanical load, myocardial infarction, car- AND CARDIAC REPAIR diac arrhythmias, endocrine disorders, and genetic mutations in cardiac contractile protein genes. While the hypertrophic CROSS-REFERENCE TO RELATED response is initially a compensatory mechanism that aug- APPLICATIONS ments cardiac output, sustained hypertrophy can lead to dilated cardiomyopathy (DCM), heart failure, and sudden This application is a continuation of U.S. application Ser. death. In the United States, approximately half a million individuals are diagnosed with heart failure each year, with a No. 12/742,233, filedAug. 24, 2010, now U.S. Pat. No. 8,513, 10 mortality rate approaching 50%. The causes andeffects of 209, which is a national stage application of International Application No. PCT/US2008/083020,filed Nov. 10, 2008, cardiac hypertrophy have been extensively documented, but the underlying molecular mechanisms have not been eluci- which claimsthe benefit ofU.S. Provisional Application No. 60/986,798, filed Nov. 9, 2007, each ofwhich is herein incor- dated. Understanding these mechanismsis a major concern in the prevention and treatment of cardiac disease and will be porated by reference in its entirety. 15 crucialas a therapeutic modality in designing new drugsthat STATEMENT OF GOVERNMENT SUPPORT specifically target cardiac hypertrophy andcardiac heart fail- ure. This invention was made with grant support undergrantno. Treatment with pharmacological agents still represents the HL53351-06 from the NationalInstitutes of Health. The gov- primary mechanism for reducing or eliminating the manifes- ernmenthas certain rights in the invention. 20 tations of heart failure. Diuretics constitute the first line of treatment for mild-to-moderate heart failure. If diuretics are DESCRIPTION OF THE TEXT FILE SUBMITTED ineffective, vasodilatory agents, such as convert- ELECTRONICALLY ing enzyme (ACE) inhibitors(e.g., enalopril and ) or inotropic agent therapy (i.e., a drug that improves cardiac The contents of the text file submitted electronically here- 25 output by increasing the force ofmyocardial muscle contrac- with are incorporated herein by reference in their entirety: A tion) may be used. Unfortunately, many of these standard computer readable format copy ofthe Sequence Listing(file- therapies have numerous adverse effects and are contraindi- name: MIRG_002__02US_SeqList_ST25 txt, date cated in some patients. Thus, the currently used pharmaco- recorded: Aug. 19, 2013, file size 4 kilobytes). logical agents have severe shortcomingsin particular patient 30 populations. The availability ofnew,safe and effective agents FIELD OF THE INVENTION would undoubtedly benefit patients who either cannot use the pharmacological modalities presently available, or who do The present invention relates generally to the fields of not receive adequate relief from those modalities. developmental biology and molecular biology. More particu- The adult heart is a dynamic organ capable of significant larly, it concerns gene regulation and cellular physiology in 35 remodeling and hypertrophic growth as a meansof adapting cardiomyocytes. Specifically, the inventionrelates to a family function to altered workloads or injury. Hemodynamicstress of miRNAs, designated as the miR-15 family, that regulate or neuroendocrine signaling associated with myocardial inf- cardiomyocyte survival and cardiac repair. Inhibition ofthese arction, hypertension, aortic stenosis, and valvular dysfunc- miRNAsprovides for reduced apoptosis in cardiac cells and tion evoke a pathologic remodeling response through the thus inhibits cardiac hypertrophy andheart failure. 40 activation of intracellular signaling pathways and transcrip- tional mediators in cardiac myocytes. Activation of these BACKGROUND OF THE INVENTION molecular pathways enhances cardiomyocyte size and pro- tein synthesis, induces the assembly of sarcomeres, and Heart disease and its manifestations, including coronary causes reexpression of fetal cardiac genes. Although aspects artery disease, myocardial infarction, congestive heart failure 45 of the hypertrophic response after acute and chronic stress and cardiac hypertrophy, clearly present a major health risk in mayinitially augmentcardiac output, prolonged hypertrophy the United States today. The cost to diagnose, treat and sup- is a major predictor of heart failure and sudden death. There port patients suffering from these diseases is well into the have been major advancesin the identification of genes and billions of dollars. Two particularly severe manifestations of signaling pathways involvedin this disease process, but the heart disease are myocardial infarction and cardiac hypertro- 50 overall complexity of hypertrophic remodeling suggests that phy. With respect to myocardial infarction, typically an acute additional regulatory mechanisms remain to be identified. thrombocytic coronary occlusion occurs in a coronary artery MicroRNAs (miRNAsor miRs) have recently been impli- as a result of atherosclerosis and causes myocardial cell cated in a numberofbiological processes including regula- death. Because cardiomyocytes, the heart muscle cells, are tion of developmental timing, apoptosis, fat metabolism, and terminally differentiated and generally incapableof cell divi- 55 hematopoietic cell differentiation among others. MiRNAsare sion, they are generally replaced by scar tissue when they die small, non-protein coding RNAs of about 18 to about 25 during the course of an acute myocardial infarction. Scar in length that regulate gene expression in a tissue is not contractile, fails to contribute to cardiac function, sequence-specific manner. MiRNAsact as repressors of tar- and often plays a detrimentalrole in heart function by expand- get mRNAs by promoting their degradation, when their ing during cardiac contraction, or by increasing the size and 60 sequences are perfectly complementary, or by inhibiting effective radius of the ventricle, for example, becoming translation, when their sequences contain mismatches. hypertrophic. MiRNAsare transcribed by RNA polymeraseII (pol II) or With respect to cardiac hypertrophy, one theory regards RNA polymeraseIII (pol IIT; see Qi et al. (2006) Cellular & this as a disease that resembles aberrant developmentand,as Molecular Immunology Vol. 3:411-419) andarise from initial such,raises the question ofwhether developmental signals in 65 transcripts, termed primary miRNA transcripts (pri-miR- the heart can contribute to hypertrophic disease. Cardiac NAs), that are generally several thousand bases long and are hypertrophy is an adaptive response of the heart to virtually derived from individual miRNA genes, from introns of pro- US 9,078,919 B2 3 4 tein coding genes, or from poly-cistronictranscripts that often ribozyme, siRNA or shRNA molecule. In still another encode multiple, closely related miRNAs. See review of Car- embodiment, expression or activity of one or more miR-15 rington et al. (2003). Pri-miRNAs are processed in the family membersis inhibited by administering a nucleic acid nucleus by the RNase Drosha into about 70- to about 100- comprising one or more miR-15 binding sites. A miR-15 hairpin-shaped precursors (pre-miRNAs). Fol- binding site may comprise a sequencethat is complementary lowing transport to the cytoplasm,the hairpin pre-miRNA is to aseed sequence ofmiR-15. The one or more miR-15 family further processed by Dicer to produce a double-stranded members may be miR-15a, miR-15b, miR-16-1, miR-16-2, miRNA (Leeet al., 1993). The mature miRNA strandis then miR-195, miR-424 and miR-497. incorporated into the RNA-induced silencing complex The present invention also provides a methodofpreventing (RISC), where it associates with its target mRNAs by base- pathologic hypertrophy or heart failure in a subject in need pair complementarity. In the relatively rare cases in which a thereof. In one embodiment, the method comprises identify- miRNA base pairs perfectly with an mRNAtarget, it pro- ing a subject at risk of developing pathologic cardiac hyper- motes mRNA degradation. More commonly, miRNAs form trophyor heart failure; and inhibiting expression or activity of imperfect heteroduplexes with target mRNAs, affecting one or more miR-15 family membersin heart cells of said either mRNAstability or inhibiting mRNAtranslation. subject. In one embodiment, inhibiting comprises delivering The 5' portion of a miRNAspanning bases 2-8, termed the ‘seed’ region, is especially important for target recognition to the heart cells an inhibitor of one or more miR-15 family (Krenz and Robbins, 2004; Kiriazis and Kranias, 2000). The members. In another embodiment, the subject at risk may sequence of the seed, together with phylogenetic conserva- exhibit one or more risk factors selected from the group tion of the target sequence, forms the basis for many current 20 consisting of uncontrolled hypertension, uncorrected valvu- target prediction models. Although increasingly sophisti- lar disease, chronic angina, recent myocardial infarction, con- cated computational approachesto predict miRNAsandtheir genital predisposition to heart disease, and pathological targets are becoming available, target prediction remains a hypertrophy. major challenge and requires experimental validation. Antagomirs, antisense oligonucleotides, inhibitory RNA Ascribing the functions of miRNAsto the regulation of spe- 25 molecules, nucleic acids comprising miR-15 binding sites or cific mRNA targets is further complicated by the ability of other modulators of the expressionor activity of one or more individual miRNAsto base pair with hundreds of potential miR-15 family members may be administered by any method high and low affinity mRNA targets and by the targeting of knownto those in the art suitable for delivery to the targeted multiple miRNAsto individual mRNAs. organ, tissue, or cell type. For example, in certain embodi- The high sequence conservation of many miRNAsacross 30 mentsof the invention, the modulator of one or more miR-15 metazoan species suggests strong evolutionary pressure and family members may be administered by parenteral admin- participation in essential biologic processes (Reinhart et al., istration, such as intravenousinjection,intraarterial injection, 2000; Stark et al., 2005). Indeed, miRNAs have been shown intrapericardial injection, or subcutaneous injection, or by to play fundamentalroles in diverse biological and pathologi- direct injection into the tissue (e.g., cardiac tissue). In some cal processes, including cell proliferation, differentiation, embodiments, the modulator of one or more miR-15 family apoptosis, and carcinogenesis in species ranging from Cae- members may be administered by oral, transdermal, intrap- norhabditis elegans and Drosophila melanogaster to eritoneal, subcutaneous, sustained release, controlled release, humans. However, there remains limited information on the delayed release, suppository, or sublingual routes of admin- role that miRNAsplay in cardiogenesis and molecular events istration. In other embodiments, the modulator ofone or more that can contribute to heart disease. 40 miR-15 family members may be administered by a catheter system. SUMMARYOF THE INVENTION Thepresent invention also encompassesa transgenic, non- human mammal, the cells ofwhichfail to express a functional Thepresent invention provides a method oftreating patho- form of one or more miR-15 family members(e.g. miR-15a, logic cardiac hypertrophy, heart failure, or myocardial infare- 45 miR-15b, miR-16-1, miR-16-2, miR-195, miR-424 and miR- tion in a subject in need thereof. In one embodiment, the 497). In another embodiment, the present invention includes method comprises identifying a subject having cardiac hyper- a transgenic, non-human mammal, the cells of which com- trophy, heart failure, or myocardial infarction; and inhibiting prise a coding region of a miR-15 family member under the expression or activity of one or more milk-15 family mem- control of a heterologous promoter active in the cells of said bersin heart cells of said subject. In another embodiment, the 50 non-human mammal. In some embodiments, the mammalis method further comprises administering to the subject a sec- a mouse. ond therapy. The second therapy maybe, for example, a beta The present invention provides a methodfor identifying a blocker, an ionotrope, a diuretic, ACE inhibitor, All antago- modulator of a miR-15 family member comprising (a) con- nist, BNP, a Ca++-blocker, and ERA, or an Hum inhibitor. tacting a cell with a candidate compound; (b) assessing activ- In some embodiments of the invention, inhibiting the ity or expression of an miR-15 family member; and (c) com- expression oractivity ofone or more miR-15 family members paring the activity or expressionin step (b) with the activity or comprises administering an antagomir ofone or more miR-15 expression in the absence of the candidate compound, family members. In one embodiment, the expressionor activ- wherein a difference between the measured activities or ity of one or more miR-15 family members is inhibited by expression indicates that the candidate compoundis a modu- administering an antisense oligonucleotide that targets the lator of said miR-15 family member. The cell may be con- mature sequence of a milk-15 family member. In yet another tacted with the candidate compoundin vitro or in vivo. The embodiment, expression or activity of one or more miR-15 candidate compound maybea protein, a peptide, a polypep- family members is inhibited by administering an inhibitory tide, a polynucleotide, an oligonucleotide, or small molecule. RNA molecule, wherein the inhibitory RNA molecule com- The modulator ofa miR-15 family member may be an agonist prises a double stranded region that is at least partially iden- or inhibitor of the miR-15 family member. The modulator of tical and complementary to a mature sequence of a miR-15 amiR-15 family member maybe an agonistor inhibitor of an family member. The inhibitory RNA molecule may be a upstream regulator of the miR-1.5 family member. US 9,078,919 B2 5 6 The present invention also provides a pharmaceutical com- FIG. 8. Enhanced expression ofmiR-15 family membersin position comprising an inhibitor of one or more miR-15 fam- samples from failing human hearts. A. Left panel shows RNA ily members. In one embodiment, the composition is formu- blots from normal humanhearts and right panel shows RNA lated for injection. In another embodiment, the blots from failing human hearts. B. The conserved seed pharmaceutical composition is combined with a kit for region among milk-15 family members allows for an addi- administration, such as parenteral or catheter administration. tional approach to knockdownthe whole miR-15 family. This approach entails overexpression of multiple miR-15 binding BRIEF DESCRIPTION OF THE DRAWINGS sites underthe control of a cardiac specific promoter, such as the alpha myosin heavy chain promoter (@MHC). Each ofthe The following drawings form part of the present specifica- miR-15 binding sites contains a sequence complementary to tion and are includedto further demonstrate certain aspects of the sequenceof the conserved seed region and allows for the the present invention. The invention maybebetter understood scavenging of all family members, thereby preventing them by reference to one or more of these drawings in combination from binding to their endogenoustargets. with the detailed description of specific embodiments pre- 15 sented herein. DETAILED DESCRIPTION OF ILLUSTRATIVE FIG. 1. MiRNA expression during cardiac hypertrophy. A. EMBODIMENTS H&Estained sections of representative hearts from mice fol- lowing sham andthoracic aortic banding (TAB)for 21 days and from wild-type (WT) and activated calcineurin trans- The present invention is based, in part, on the discovery genic (CnA Tg) mice. Scale bar equals 2 mm. B. Numbers of that members of the miR-15 family of microRNAs, such as miRNAsthat were regulated in response to CnA or TABare miR-195, are upregulated in myocardial tissue from human indicated. Although some changes were unique for either failing hearts as well as in animal models of pathologic car- TAB or CnA-induced hypertrophy, most miRNAsthat were diac hypertrophy. Cardiac overexpression ofmiR-195 is suf- induced or repressed overlapped for the different hyper- ficient to induce cardiac hypertrophy andcanleadto a dilated trophic stimuli. C. Northern blot analysis of particular miR- phenotype. Accordingly, the present invention provides NAs in WT and CnA Tg hearts. U6 RNA wasdetected as a methods of treating or preventing various forms of heart loading control. disease in a subject by inhibiting the expression oractivity of FIG. 2. MiRNA expression in humanheart failure. North- one or more members of the miR-15 family in heart cells of ern blot analysis ofmiRNAsin 4 normaland6 failing human 30 the subject. hearts. The average fold-change ofeach miRNAinthe failing The miR-15 family is a small family of microRNAsthat samples is shownatthe right. includes miR-195, miR-16-1, miR-15a, miR-15b, miR-16-2, FIG. 3. Cardiac specific over-expression ofmiRNA 195 is miR-424, and miR-497. Four of the miR-15 family members sufficient to drive cardiomyopathy. H&E stained sections of are expressedas three clustered transcripts (FIG. 6A). MiR- hearts from wild-type (WT) and twodifferent lines of miR- 35 195 transgenic (Tg) animals. MiRNA-195 Tgline 3 animals 195 and miR-497 are expressedas a duster from the intron of died two weeks after birth due to cardiac dilation. Northern a gene located on chromosome 17. There are two copies of blot analysis on hearts from WTand miR-195 transgenic lines miR-16 located on different chromosomes. One copy (miR- 1 and 3 confirming a 26.5-fold and 29.2-fold cardiac specific 16-1)is expressed as a cluster with miR-15a from an intron of miRNAoverexpression, respectively. 40 a gene encoded on chromosome 13. The second copy (miR- FIG. 4. Overexpression of milt-195 induces cardiac dys- 16-2) is expressed as a cluster with 15b from the intron of the function due to cardiac growth. A. Echocardiographic analy- SMC4gene located on chromosome 3. MiR-424is expressed ses indicate miR-195 transgenic (Tg) mice showleft ventricu- from the X chromosome. The pre-miRNA sequences(e.g. lar (LV) dilation and wall thinning, resulting in a decreased stem loop sequences) for each ofthe miR-15 family members fractional shortening compared to wild-type (WT)litter- 45 are listed below: mates. B. Heart weight to body weight ratio increases in response to cardiac specific overexpression of milk-195. C. Realtime PCR analysis shows an upregulation of hyper- Human pre-miR-195 trophic genes in miR-195 Tg animals compared to WT ani- (SEQ ID NO: 1) AGCUUCCCUG GCUCUAGCAG CACAGAAAUA UUGGCACAGG mals (n=3). *P<0.05 compared to wild-type. 50 FIG. 5. Cardiac remodeling specific for miR-195. Over- GAAGCGAGUC UGCCAAUAUU GGCUGUGCUG CUCCAGGCAG expression of miR-195 induces cardiac growth at 2 weeks of age, which within 6 weeksprogressesto a dilated phenotype. GGUGGUG

Cardiac over-expression of miR-214 has no phenotypic Human pre-miR-497 effect, indicating the specific effect of miR-195 on cardiac 55 (SEQ ID NO: 2) pathology. Scale bar equals 2 mm. CCACCCCGGU CCUGCUCCCG CCCCAGCAGC ACACUGUGGU FIG. 6. MiR-195is part of the miR-15 family that targets UUGUACGGCA CUGUGGCCAC GUCCAAACCA CACUGUGGUG pro-survival proteins. A. MiR-195 is part of the milk-15 fam- ily that consists of five different miRs: miR-15, miR-16, UUAGAGCGAG GGUGGGGGAG GCACCGCCGA GG miR-195, miR-424, and miR-497. Four ofthe miR-15 family 60 membersare expressed as three dusters of two miRNAs. B. Human pre-miR-16-1 MiR-15 family members target proteins involved in prolif- (SEQ ID NO: 3) eration, survival and anti-apoptosis. Thus, up-regulation of GUCAGCAGUG CCUUAGCAGC ACGUAAAUAU UGGCGUUAAG miR-195 results in down-regulation ofthese mRNAsandcell AUUCUAAAAU UAUCUCCAGU AUUAACUGUG CUGCUGAAGU death. 65 FIG. 7. MiR-195 target sequence in the 3' UTR of FGF2 AAGGUUGAC mRNA. US 9,078,919 B2 7 8 -continued -continued

Human pre-miR-16-2 Human miR-15b* (SEQ ID No: (SEQ ID NO: 17) GUUCCACUCU AGCAGCACGU AAAUAUUGGC GUAGUGAAAU CGAAUCAUUAUUUGCUGCUCUA

Human mature miR-424 AUAUAUUAAA CACCAAUAUU ACUGUGCUGC UUUAGUGUGA (SEQ ID NO: 20) CAGCAGCAAUUCAUGUUUUGAA Human pre-miR-15a (SEQ ID No: Human miR-424* CCUUGGAGUA AAGUAGCAGC ACAUAAUGGU UUGUGGAUUU 10 (SEQ ID NO: 21) CAAAACGUGAGGCGCUGCUAU UGAAAAGGUG CAGGCCAUAU UGUGCUGCCU CAAAAAUACA AGG Althoughthe seed region (e.g. bases spanning 2 to 8 nucle- Human pre-miR-15b otides ofmature miRNA sequence)for all family membersis (SEQ ID No: highly conserved (AGCAGCAC; SEQ ID NO:18), the 3' end UUGAGGCCUU AAAGUACUGU AGCAGCACAU CAUGGUUUAC 15 of the mature miRNA differs among the different family AUGCUACAGU CAAGAUGCGA AUCAUUAUUU GCUGCUCUAG members(FIG. 6A). The present invention provides a methodoftreating patho- AAAUUUAAGG AAAUUCAU logic cardiac hypertrophy, heart failure, or myocardialinfare- tion in a subject in need thereof by inhibiting expression or Human pre-miR-424 20 activity of one or more miR-15 family members. In one (SEQ ID NO: 19) CGAGGGGAUA CAGCAGCAAU UCAUGUUUUG AAGUGUUCUA embodiment, the method comprises identifying a subject hav- ing cardiac hypertrophy, heart failure, or myocardial infarc- AAUGGUUCAA AACGUGAGGC GCUGCUAUAC CCCCUCGUGG tion; and inhibiting expression or activity of one or more 25 milk-15 family members in heart cells of said subject. “Heart GGAAGGUAGA AGGUGGGG cells” as used herein include cardiomyocytes, cardiac fibro- blasts, and cardiac endothelial cells. In another embodiment, Eachof the pre-miRNA sequences for each miR-15 family the method comprises administering to the subject an inhibi- member is processed into a mature sequence and star a tor of one or more miR-15 family members. Instill another sequence. The star sequence is processed from the other 30 embodiment, the method comprises identifying a subject at strand of the stem loop structure. The mature and star risk of developing pathologic cardiac hypertrophy or heart sequences for each of the miR-15 family membersis given failure and inhibiting expression or activity of one or more below: miR-15 family members in heart cells of the subject. The subject at risk of developing pathologic cardiac hypertrophy 35 or heart failure may exhibit one or moreriskfactors including, Human mature miR-195 for example, uncontrolled hypertension, uncorrected valvular (SEQ ID NO: 7) UAGCAGCACAGAAAUAUUGGC disease, chronic angina, recent myocardial infarction, con- genital predispositionto heart disease or pathological hyper- Human miR-195* trophy. In certain embodiments, the subject at risk may be (SEQ ID NO: 8) 40 diagnosed as having a genetic predisposition to cardiac CCAAUAUUGGCUGUGCUGCUCC hypertrophy. In some embodimentsofthe invention, the sub- Human mature miR-497 ject at risk may have a familialhistory ofcardiac hypertrophy. (SEQ ID NO: 9) In another embodiment, the present invention provides a CAGCAGCACACUGUGGUUGU methodof preventing cardiac hypertrophy anddilated cardi- Human miR-497* 45 omyopathy in a subject in need thereof comprising inhibiting (SEQ ID NO: 10) expression oractivity ofone or more miR-15 family members CAAACCACACUGUGGUGUUAGA in heart cells of the subject. In yet a further embodiment, the present invention provides a methodof inhibiting progression Human mature miR-16-1/miR-16-2 (SEQ ID NO: 11) ofcardiac hypertrophy ina subject in need thereofcomprising UAGCAGCACGUAAAUAUUGGCG 50 inhibiting expression or activity of one or more milk-15 fam- ily membersin heart cells of the subject. In further embodi- Human miR-16-1* ments, the present invention provides a method of increasing (SEQ ID NO: 12) CCAGUAUUAACUGUGCUGCUGA exercise tolerance, reducing hospitalization, improving qual- ity of life, decreasing morbidity, and/or decreasing mortality Human miR-16-2* 55 in a subject with heart failure or cardiac hypertrophy com- (SEQ ID NO: 13) prising inhibiting expression oractivity of one or more milk- CCAAUAUUACUGUGCUGCUUA 115 family membersin heart cells of the subject. Human mature miR-15a Thus, the present invention provides methodsfor the treat- (SEQ ID NO: 14) ment of cardiac hypertrophy, heart failure, or myocardial UAGCAGCACAUAAUGGUUUGUG 60 infarction utilizing inhibitors of miR-15 members, such as Human miR-15a* miR-195, miR-15a, miR-15b, miR-16-1, miR-424, and miR- (SEQ ID NO: 15) 497. Preferably, administration of an inhibitor of a miR-15 CAGGCCAUAUUGUGCUGCCUCA family memberresults in the improvement of one or more symptomsof cardiac hypertrophy, heart failure, or myocar- Human mature miR-15b (SEQ ID NO: 16) 65 dial infarction in the subject, or in the delay in the transition UAGCAGCACAUCAUGGUUUACA from cardiac hypertrophy to heart failure. The one or more improved symptoms maybe, for example, increased exercise US 9,078,919 B2 9 10 capacity, increased cardiac ejection volume, decreased left modifications. In some embodiments, antagomirs comprise ventricular end diastolic pressure, decreased pulmonary cap- only modified nucleotides. Antagomirs may also comprise illary wedge pressure, increased cardiac output, increased one or more phosphorothioate linkages resulting in a partial cardiac index, lowered pulmonary artery pressures, or full phosphorothioate backbone.To facilitate in vivo deliv- decreased left ventricular end systolic and diastolic dimen- ery andstability, the antagomir maybe linked to a cholesterol sions, decreased left and right ventricular wall stress, or other moiety at its 3" end. Antagomirs suitable for inhibit- decreased wall tension, increased quality of life, and ing miRNAs may be about 15 to about 50 nucleotides in decreased disease related morbidity or mortality. In addition, length, more preferably about 18 to about 30 nucleotides in use of inhibitors of milk-15 family members may prevent length, and most preferably about 20 to about 25 nucleotides cardiac hypertrophy and its associated symptoms from aris- 10 in length,“Partially complementary”refers to a sequencethat ing. In one embodiment, administration ofan inhibitor ofone is at least about 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or more miR-15 family members to a subject suffering from or 99% complementary to a target polynucleotide sequence. myocardial infarction may reduce infarct size by decreasing The antagomirs maybeat least about 75%, 80%, 85%, 90%, the loss of heart cells. In another embodiment, cardiac func- 95%, 96%, 97%, 98%, or 99% complementary to a mature tion is stabilized in a subject suffering from myocardial inf- 15 miRNAsequence.In some embodiments, the antagomir may arction following administration of an inhibitor of one or be substantially complementary to a mature miRNA more miR-15 family members. sequence, that is at least about 95%, 96%, 97%, 98%, or 99% Inhibition of microRNA function may be achieved by complementary to a target polynucleotide sequence. In other administering antisense oligonucleotides targeting a mature embodiments, the antagomirs are 100% complementary to sequence of a miR-15 family member. The antisense oligo- 20 the mature miRNA sequence. nucleotides may be ribonucleotides or deoxyribonucleotides. Another approach for inhibiting the function of a miR-15 Preferably, the antisense oligonucleotides have at least one family memberis administering an inhibitory RNA molecule chemical modification. Antisense oligonucleotides may be having a double strandedregionthatis at least partially iden- comprised of one or more “locked nucleic acids”, “Locked tical and partially complementary to a mature sequenceofthe nucleic acids” (LNAs) are modified ribonucleotides that con- 25 miR-15 family member. The inhibitory RNA molecule may tain an extra bridge betweenthe 2' and 4' carbonsofthe ribose be a double-stranded, small interfering RNA (siRNA)or a sugar moiety resulting in a “locked” conformation that con- short hairpin RNA molecule (shRNA) comprising a stem- fers enhanced thermalstability to oligonucleotides contain- loop structure. The double-stranded regions of the inhibitory ing the LNAs. Alternatively, the antisense oligonucleotides RNA molecule may comprise a sequence that is at least may comprise peptide nucleic acids (PNAs), which contain a 30 partially identical and partially complementary, e.g. about peptide-based backbonerather than a sugar-phosphate back- 75%, 80%, 85%, 90%, 95%, 96%, 97%, 98%, or 99% iden- bone. Other chemical modifications that the antisense oligo- tical and complementary, to the mature miRNA sequence. In nucleotides may contain include, but are not limited to, sugar some embodiments, the double-stranded regions of the modifications, such as 2'-O-alkyl (e.g. 2'-O-methyl, 2'-O- inhibitory RNA comprise a sequencethat is at least substan- methoxyethyl), 2'-fluoro, and 4' thio modifications, and back- 35 tially identical and substantially complementary to the bone modifications, such as one or more phosphorothioate, mature miRNA sequence. “Substantially identical and sub- morpholino, or phosphonocarboxylate linkages (see, for stantially complementary”refers to a sequencethatis at least example, U.S. Pat. Nos. 6,693,187 and 7,067,641, which are about 95%, 96%, 97%, 98%, or 99% identical and comple- herein incorporated by reference in their entireties). In some mentary to a target polynucleotide sequence. In other embodiments, suitable antisense oligonucleotides are 2'-O- 40 embodiments, the double-stranded regions of the inhibitory methoxyethyl “gapmers” which contain 2'-O-methoxyethyl- RNA molecule may contain 100% identity and complemen- modified ribonucleotides on both 5' and 3' ends with at least tarity to the target miRNA sequence. ten deoxyribonucleotides in the center. These “gapmers”are The inhibitory nucleotide molecules described hereinpref- capable of triggering RNase H-dependent degradation erably target a mature sequence ofone or more miR-15 family mechanisms of RNA targets. Other modifications of anti- 45 members(e.g. SEQ ID NOs: 7, 9, 11, 14, 16, and 20) or a star sense oligonucleotides to enhancestability and improveeffi- sequence of one or more miR-15 family members (e.g. SEQ cacy, such as those described in U.S. Pat. No. 6,838,283, ID NOs: 8, 10, 12, 13, 15, 17, and 21). Insome embodiments, whichis herein incorporated by reference in its entirety, are inhibitors of miR-15 family members are antagomirs com- knownintheart andare suitable for use in the methods ofthe prising a sequence that is perfectly complementary to a invention. Preferable antisense oligonucleotides useful for 50 mature sequence ofa miR-15 family member. In one embodi- inhibiting the activity ofmicroRNAsare about 19 to about 25 ment, an inhibitor of a miR-15 family memberis an antago- nucleotides in length. Antisense oligonucleotides may com- mir having a sequencethat is partially or perfectly comple- prise a sequencethatis at least partially complementary to a mentary to SEQ ID NO: 7, SEQ ID NO: 9, SEQ ID NO: 11, mature miRNA sequence,e.g. at least about 75%, 80%, 85%, SEQ ID NO: 14, SEQ ID NO: 16, or SEQ ID NO:20. In 90%, 95%, 96%, 97%, 98%, or 99% complementary to a 55 another embodiment, an inhibitor of a miR-15 family mem- mature miRNA sequence. In some embodiments, the anti- ber is an antagomir having a sequence that is partially or sense oligonucleotide may be substantially complementary perfectly complementary to SEQ ID NO: 8, SEQ ID NO:10, to amature miRNAsequence,thatis at least about 95%, 96%, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID NO: 15, SEQ ID 97%, 98%, or 99% complementary to a target polynucleotide NO: 17, or SEQ ID NO:21. sequence. In one embodiment, the antisense oligonucleotide 60 In some embodiments, inhibitors of one or more miR-15 comprises a sequence that is 100% complementary to a family members are chemically-modified antisense oligo- mature miRNA sequence. nucleotides, in one embodiment, an inhibitor of a miR-15 In some embodiments, the antisense oligonucleotides are family member is a chemically-modified antisense oligo- antagomirs. “Antagomirs” are single-stranded, chemically- nucleotide comprising a sequence substantially complemen- modified ribonucleotides that are at least partially comple- 65 tary to SEQ ID NO:7, SEQ ID NO:9, SEQ ID NO: 11, SEQ mentary to the miRNA sequence. Antagomirs may comprise ID NO: 14, SEQ ID NO: 16, or SEQ ID NO:20. In another one or more modified nucleotides, such as 2'-O-methy]-sugar embodiment, an inhibitor of a miR-15 family memberis a US 9,078,919 B2 11 12 chemically-modified antisense oligonucleotide comprising a all pool ofmiRNAsthat could impacta given target sequence. sequence substantially complementary to SEQ ID NO:8, The term “miR-15 binding site” as used herein refers to a SEQ ID NO: 10, SEQ ID NO: 12, SEQ ID NO: 13, SEQ ID nucleotide sequence that is capable of binding a mature NO: 15, SEQ ID NO: 17, or SEQ ID NO:21. As used herein sequence of miR-15a, miR-15b, miR-16-1, miR-16-2, miR- “substantially complementary”refers to a sequencethatis at 195, miR-424, miR-497, or combinationsthereof. Preferably, least about 95%, 96%, 97%, 98%, 99%, or 100% comple- a miR-15 binding site comprises a sequence that is substan- mentary to a target polynucleotide sequence (e.g. mature or tially complementary to the miR-15 seed sequence. The seed precursor miRNA sequence). sequence or seed region refers to nucleotides 2-8 of the 5' Antisense oligonucleotides may comprise a sequence that portion of the mature miRNA sequence. In one embodiment, is substantially complementary to a precursor miRNA 10 the miR-15 binding site comprises a sequencethat is substan- sequence(pre-miRNA)for one or more miR-15 family mem- tially complementary to SEQ ID NO: 18. The inhibitory bers (e.g. pre-miR-195, pre-miR-497, pre-miR-424, pre- nucleic acid comprising one or more miR-15 bindingsites miR-15a, pre-miR-15b, pre-miR-16-1, or pre-miR-16-2). In may be from about 20 to about 500 nucleotides in length, some embodiments, the antisense oligonucleotide comprises about 25 to about 400 nucleotides in length, about 30 to about a sequencethat is substantially complementary to a sequence 15 300 nucleotides in length, about 40 to about 200 nucleotides located outside the stem-loop region of the pre-miRNA in length, or about 50 to about 100 nucleotides in length. For sequence. In one embodiment, an inhibitor of a miR-15 fam- example, the nucleic acid may be 20, 21, 22, 23, 24, 25, 26, ily memberis an antisense oligonucleotide having a sequence 27, 28, 29, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, that is substantially complementary to a pre-miRNA 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, 200, 225, sequenceselected from the group consisting of SEQ ID NO: 20 250, 275, 300, 350, 400, 500 nucleotides in length. The 1, SEQ ID NO:2, SEQID NO: 3, SEQID NO: 4, SEQID NO: nucleic acid may contain 1, 2,3, 4, 5, 6, 7, 8, 9, 10, 12, 15, or 5, SEQ ID NO:6, and SEQ ID NO:19. 20 miR-15 binding sites. The multiple ma-15 bindingsites In other embodiments of the invention, inhibitors of one or may be adjacent or may be separated by spacers of 1, 2, 3, 4, more miR-15 family members may be inhibitory RNA mol- 5, 6, 7, 8, 9, 10 or more nucleotides. ecules, such as ribozymes, siRNAs, or shRNAs. In one 25 In another embodiment, an expression vector may be used embodiment, an inhibitor of a miR-15 family memberis an to deliveran inhibitor ofone or more miR-15 family members inhibitory RNA molecule comprising a double-stranded to a cell or subject. A “vector” is a composition of matter region, wherein the double-stranded region comprises a which can be used to deliver a nucleic acid of interest to the sequence having 100% identity and complementarity to a interior of a cell. Numerous vectors are known in the art mature sequence of a miR-15 family member (e.g. SEQ ID 30 including, but not limited to, linear polynucleotides, poly- NOs: 7, 9, 11, 14, 16, and 20). In another embodiment, an nucleotides associated with ionic or amphiphilic compounds, inhibitor of a miR-115 family memberis an inhibitor RNA plasmids, and viruses. Thus, the term “vector” includes an molecule comprising a double-stranded region, wherein the autonomously replicating plasmid or a virus. Examples of double-stranded region comprises a sequence having 100% viral vectors include, but are not limited to, adenoviral vec- identity and complementarity to a star sequence of a miR-15 35 tors, adeno-associated virus vectors, retroviral vectors, and family member(e.g. SEQ ID NOs: 8, 10, 12, 13, 15, 17, and the like. An expression construct can be replicated in a living 21). In some embodiments, inhibitors of one or more miR-15 cell, or it can be made synthetically. For purposes of this family members are inhibitory RNA molecules which com- application, the terms “expression construct,” “expression prise a double-stranded region, wherein said double-stranded vector,” and “vector,” are used interchangeably to demon- region comprises a sequence of at least about 75%, 80%, 40 strate the application of the invention in a general, illustrative 85%, 90%, 95%, 96%, 97%, 98%, or 99% identity and sense, and are not intended to limit the invention. complementarity to a mature sequence of one or more miR- In one embodiment, an expression vector for expressing an 15 family members. inhibitor ofone or more miR-15 family members comprises a Multiple members of the miR-15 family (e.g. miR-15a, promoter operably linked to a polynucleotide encoding an miR-15b, miR-16-1, miR-16-2, miR-195, miR-424, and 45 antisense oligonucleotide, wherein the sequence of the miR-497) maybe inhibited simultaneously by administering expressed antisense oligonucleotide is partially or perfectly multiple inhibitors, wherein each inhibitor targets a separate complementary to a mature sequence of one or more miR-15 miR-15 family member. For example, in some embodiments, family members. The phrase “operably linked” or “under at least two membersof the miR-15 family are inhibited by transcriptional control” as used herein meansthat the pro- administering two separate inhibitors. In other embodiments, 50 moteris in the correct location andorientation in relation to a at least three members of the miR-15 family are inhibited by polynucleotide to control the initiation of transcription by administering three separate inhibitors. In still other embodi- RNA polymerase and expression of the polynucleotide. In ments, at least four members of the miR-15 family are inhib- another embodiment, an expression vector for expressing an ited by administering four separate inhibitors. In further inhibitor of one or more miR-15 family members comprises embodiments,at least five members of the miR-15 family are 55 one or more promoters operably linked to a polynucleotide inhibited by administering five separate inhibitors. In one encoding a shRNAor siRNA, wherein the expressed shRNA embodiment,all six members ofthe miR-15 family are inhib- or siRNA comprises a double stranded regionthatis identical ited by administering six separate inhibitors. and complementary or partially identical and partially In another embodiment, an inhibitor of miR-15 family complementary to a mature sequence of one or more miR-15 function is a nucleic acid comprising one or more miR-15 60 family members. “Partially identical and partially comple- binding sites. The seed region (AGCAGCAC; SEQ ID NO: mentary”refers to a sequencethatis at least about 75%, 80%, 18) of all the miR-15 family membersis highly conserved. 85%, 90%, 95%, 96%, 97%, 98%, or 99% identical and Therefore, a nucleic acid comprising a binding site having complementary to a target polynucleotide sequence. substantial complementarity to the miR-15 seed sequence In certain embodiments, the nucleic acid encoding a poly- would bind all membersof the miR-15 family. This approach nucleotide of interest is under transcriptional control of a has been analogized to the use of a sponge to “soak up” the promoter. A “promoter”refers toa DNA sequence recognized effective miR-15 family members, thereby reducing the over- by the synthetic machinery ofthecell, or introduced synthetic US 9,078,919 B2 13 14 machinery, required to initiate the specific transcription of a responseto specific physiologic signals can permit inducible gene. The term promoterwill be used hereto refer to a group expression of the gene product. Tables 1 and 2 list several of transcriptional control modules that are clustered around regulatory elements that may be employed,in the context of the initiation site for RNA polymeraseI, IT, or IT. the present invention, to regulate the expression of the poly- nucleotide of interest (e.g. inhibitor of miR-15 members). . In some embodiments, the human cytomegalovirus (CMV) This list is not intended to be exhaustive of all the possible immediate early gene promoter, the SV40 early promoter, the : : . : - : :. elements involved in the promotion of gene expression but, Rous sarcoma virus long terminal repeat, rat insulin pro- merely, to be exemplary thereof. moter, and glyceraldehyde-3-phosphate dehydrogenase pro- Below is a list of viral promoters, cellular promoters/en- moter can be used to obtain high-level expression of the io hancers and inducible promoters/enhancers that could be coding sequenceofinterest. The use of other viral or mam- used in combination with the polynucleotide of interest in an malian cellular or bacterial phage promoters which are well- expression construct (Table 1 and Table 2). Additionally, any knownin the art to achieve expression ofa coding sequence of promoter/enhancer combination as per the Eukaryotic Pro- interest is contemplated as well, provided that the levels of moter Data Base EPDB)couldalso be used to drive expres- expression are sufficient for a given purpose. 15 sion of the gene. Eukaryotic cells can support cytoplasmic By employing a promoter with well-knownproperties, the transcription from certain bacterial promotersifthe appropri- level and pattern ofexpression of the polynucleotideof inter- ate bacterial polymerase is provided, either as part of the est following transfection or transformation can be opti- delivery complex or as an additional genetic expression con- mized. Further, selection of a promoter that is regulated in struct.

TABLE 1

Promoter and/or Enhancer

Promoter/Enhancer References

Immunoglobulin Heavy Chain Banerji et al., 1983; Gilles et al., 1983; Grossched| et al., 1985; Atchinsonet al., 1986, 1987; Imler et al., 1987; Weinbergeret al., 1984; Kiledjian et al., 1988; Porton et al.; 1990 Immunoglobulin Light Chain Queenet al., 1983; Picard et al., 1984 T-Cell Receptor Luriaet al., 1987; Winoto et al., 1989; Redondo et al.; 1990 HLA DQ aand/or DQ 6 Sullivan et al., 1987 §-Interferon Goodbournet al., 1986; Fujita et al., 1987; Goodbourn etal., 1988 Interleukin-2 Greeneet al., 1989 Interleukin-2 Receptor Greeneet al., 1989; Lin et al., 1990 MHCClass II 5 Kochet al., 1989 MHCClass III HLA-DRa Sherman etal., 1989 f-Actin Kawamotoet al., 1988; Ng etal.; 1989 Muscle Creatine Kinase (MCK) Jaynes et al., 1988; Horlick et al., 1989; Johnson et al., 1989 Prealbumin (Transthyretin) Costaet al., 1988 Elastase I Ornitz et al., 1987 Metallothionein (MTII) Karin et al., 1987; Culottaet al., 1989 Collagenase Pinkert et al., 1987; Angel et al., 1987a Albumin Pinkert et al., 1987; Troncheet al., 1989, 1990 a-Fetoprotein Godboutet al., 1988; Campereet al., 1989 t-Globin Bodineet al., 1987; Perez-Stableet al., 1990 6-Globin Trude et al., 1987 c-fos Cohenet al., 1987 c-HA-ras Triesman, 1986; Deschampset al., 1985 Insulin Edlundet al., 1985 Neural Cell Adhesion Molecule Hirshet al., 1990 (NCAM) q,-Antitrypain Latimeret al., 1990 H2B (TH2B) Histone Hwanget al., 1990 Mouseand/or Type I Collagen Ripe et al., 1989 Glucose-Regulated Proteins Changet al., 1989 (GRP94 and GRP78) Rat Growth Hormone Larsenet al., 1986 Human Serum Amyloid A (SAA) Edbrookeetal., 1989 Troponin I (TN I) Yutzey et al., 1989 Platelet-Derived Growth Factor Pechet al., 1989 (PDGF) Duchenne Muscular Dystrophy Klamutet al., 1990 SV40 Banerji et al., 1981; Moreauet al., 1981; Sleigh et al., 1985; Firak et al., 1986; Herr et al., 1986; Imbraet al., 1986; Kadeschet al., 1986; Wang et al., 1986; Ondeket al., 1987; Kuhlet al., 1987; Schaffner et al., 1988 Polyoma Swartzendruberet al., 1975; Vasseuret al., 1980; Katinkaet al., 1980, 1981; Tyndell et al., 1981; Dandolo et al., 1983; de Villiers et al., 1984; Hen et al., 1986; Satake et al., 1988; Campbell and/or Villarreal, 1988 US 9,078,919 B2 15 16 TABLE1-continued

Promoter and/or Enhancer

Promotetr/Enhancer References

Retroviruses Kriegler et al., 1982, 1983; Levinson et al., 1982; Kriegler et al., 1983, 1984a, b, 1988; Boszeet al., 1986; Miksicek et al., 1986; Celanderet al., 1987; Thiesen et al., 1988; Celanderet al., 1988; Choi et al., 1988; Reisman et al., 1989 Papilloma Virus Campoet al., 1983; Lusky et al., 1983; Spandidos and/or Wilkie, 1983; Spalholz et al., 1985; Lusky et al., 1986; Cripe et al., 1987; Gloss et al., 1987; Hirochika et al., 1987; Stephenset al., 1987 Hepatitis B Virus Bulla et al., 1986; Jameelet al., 1986; Shauletal., 1987; Spandauet al., 1988; Vannice et al., 1988 Human Immunodeficiency Virus Muesinget al., 1987; Hauberet al., 1988; Jakobovits et al., 1988; Feng et al., 1988; Takebe et al., 1988; Rosen etal., 1988; Berkhoutet al., 1989; Laspia et al., 1989; Sharp etal., 1989; Braddocketal., 1989 Cytomegalovirus (CMV) Weberet al., 1984; Boshart et al., 1985; Foecking et al., 1986 Gibbon Ape Leukemia Virus Holbrooketal., 1987; Quinn et al., 1989

TABLE 2 A polyadenylation signal may be includedto effect proper

polyadenylation of the gene transcript where desired. The 25 Inducible Elements nature of the polyadenylation signal is not believed to be

Element Inducer References crucial to the successful practice of the invention, and any

such sequence may be employed such as human growth hor- MT II Phorbol Ester (TFA) Palmiter et al., 1982; mone and SV40polyadenylation signals. Also contemplated Heavy metals Haslinger et al., 1985; Searle et al., 1985; Stuart 30 as an elementofthe expression cassette is a terminator. These et al., 1985; Imagawa elements can serve to enhance message levels and to mini- et al., 1987, Karin et al., mize read through from the cassette into other sequences. 1987; Angelet al., 1987b; McNeall et al., 1989 In certain embodimentsof the invention, the cells contain- MMTV (mouse Glucocorticoids Huanget al., 1981; Lee ing nucleic acid constructs ofthe present invention,a cell may mammary tumorvirus) et al., 1981; Majors etal., 35 be identified in vitro or in vivo by including a marker in the 1983; Chandleret al., expression construct. Such markers would confer an identi- 1983; Ponta et al., 1985; Sakaiet al., 1988 fiable changeto the cell permitting easy identificationof cells §-Interferon poly(@)x Tavernieret al., 1983 containing the expression construct. Usually the inclusion of poly(re) a drug selection markeraids in cloning andin the selection of Adenovirus 5 E2 EIA Imperiale et al., 1984 40 transformants, for example, genes that confer resistance to Collagenase Phorbol Ester (TPA) Angelet al., 1987a Stromelysin Phorbol Ester (TPA) Angel et al, 1987b neomycin, puromycin, hygromycin, DHFR, GPT, zeocin and SV40 Phorbol Ester (TPA) Angelet al., 1987b histidinol are useful selectable markers. Alternatively, Murine MX Gene Interfron, Newcastle Huget al., 1988 enzymes such as herpes simplex virus thymidine kinase (tk) Disease Virus or chloramphenicol acetyltransferase (CAT) may be GRP78 Gene A23187 Resendezet al., 1988 a-2-Macroglobulin IL-6 Kunzet al., 1989 45 employed. Immunologic markers also can be employed. The Vimentin Serum Rittling et al., 1989 selectable marker employedis not believed to be important, MHCClass I Gene Interferon Blanar et al., 1989 so long as it is capable of being expressed simultaneously H-2kb with the nucleic acid encoding a gene product. Further HSP70 EIA, SV40 Large T Tayloret al., 1989, 1990a, Antigen 1990b examples of selectable markers are well known to one of skill Proliferin Phorbol Ester-TPA Mordacq et al., 1989 50 in the art. Tumor Necrosis Factor PMA Henselet al., 1989 There are a numberof ways in which expression vectors Thyroid Stimulating Thyroid Hormone Chatterjee et al., 1989 may be introducedinto cells. In certain embodiments of the Hormone a@ Gene invention, the expression construct comprises a virus or engi- neered construct derived from a viral genome. Theability of 55 certain viruses to enter cells via receptor-mediated endocyto- Ofparticular interest are muscle specific promoters, and sis, to integrate into host cell genomeandexpressviral genes moreparticularly, cardiac specific promoters. These include stably andefficiently have made them attractive candidates the myosin light chain-2 promoter (Franz etal., 1994; Kelly et for the transfer offoreign genes into mammaliancells (Ridge- al., 1995), the alpha actin promoter (Mossetal., 1996), the way, 1988; Nicolas and Rubenstein, 1988; Baichwal and troponin 1 promoter (Bhaysar et al., 1996); the Na*/Ca?* 60 Sugden, 1986; Temin, 1986). exchanger promoter (Barnes et al., 1997), the dystrophin Oneofthe preferred methodsfor in vivo delivery involves promoter (Kimuraet al., 1997), the alpha7 integrin promoter the use of an adenovirus expression vector. “Adenovirus (Ziober and Kramer, 1996), the brain natriuretic peptide pro- expression vector” is meant to include those constructs con- moter (LaPointe etal., 1996) and the alpha B-crystallin/small taining adenovirus sequencessufficient to (a) support pack- heat shock protein promoter (Gopal-Srivastava, 1995), alpha 65 aging of the construct and (b) to express an antisense poly- myosin heavy chain promoter (Yamauchi-Takihara et al., nucleotide (or other inhibitory polynucleotide) that has been 1989) and the ANF promoter (LaPointe etal., 1988). clonedtherein. The expression vector comprises a genetically US 9,078,919 B2 17 18 engineered for ofadenovirus, Knowledgeofthe genetic orga- presentat the 5' and 3' ends ofthe viral genome.These contain nization ofadenovirus, a 36 kB, linear, double-stranded DNA strong promoter and enhancer sequences and are also virus, allows substitution of large pieces of adenoviral DNA required for integration in the host cell genome (Coffin, with foreign sequences up to 7 kB (Grunhaus and Horwitz, 1990). 1992). In contrast to retrovirus, the adenoviral infection of In order to construct a retroviral vector, a nucleic acid host cells does not result in chromosomalintegration because encoding a geneofinterestis insertedinto the viral genome in adenoviral DNA canreplicate in an episomal manner without the place of certain viral sequences to produce a virusthatis potential genotoxicity. Also, adenoviruses are structurally replication-defective. In order to produce virions, a packag- stable, and no genomerearrangementhas been detectedafter ing cell line containing the gag,pol, and env genes but without extensive amplification. Adenovirus can infect virtually all the LTR and packaging components is constructed (Mann et epithelial cells regardless of their cell cycle stage. al., 1983). When a recombinant plasmid containing a cDNA, Adenovirusis particularly suitable for use as a gene trans- together with the retroviral LTR and packaging sequencesis fer vector because of its mid-sized genome, ease of manipu- lation, high titer, wide target cell range and high infectivity. introduced into this cell line (by calctum phosphate precipi- Both ends of the viral genome contain 100-200 base pair tation for example), the packaging sequence allows the RNA inverted repeats (ITRs), which are cis elements necessary for transcript of the recombinant plasmid to be packaged into viral DNA replication and packaging. viral particles, which are then secreted into the culture media Other than the requirement that the adenovirus vector be (Nicolas and Rtibenstein, 1988; Temin, 1986; Mann etal., replication defective, or at least conditionally defective, the 1983). The media containing the recombinantretrovirusesis nature of the adenovirus vector1s not believed to be crucial to then collected, optionally concentrated, and used for gene the successful practice of the invention. The adenovirus may transfer. Retroviral vectors are able to infect a broad variety of be of any of the 42 different known serotypes or subgroups cell types. However, integration and stable expression require A-F. Adenovirus type 5 of subgroup C is the preferred starting the division of host cells (Paskindet al., 1975). material in order to obtain the conditional replication-defec- Other viral vectors may be employed as expression con- tive adenovirus vector for use in the present invention. This is 25 structs in the present invention. Vectors derived from viruses because Adenovirus type 5 is a human adenovirus about such as vaccinia virus (Ridgeway, 1988; Baichwal and Sug- which a great deal of biochemical and genetic information is den, 1986; Coupar et al., 1988) adeno-associated virus (AAV) known, andit has historically been used for most construc- (Ridgeway, 1988; Baichwal and Sugden, 1986; Hermonat tions employing adenovirus as a vector. and Muzycska, 1984) and herpesviruses may be employed. The typical vector according to the present invention is 30 They offer several attractive features for various mammalian replication defective and will not have an adenovirus E1 cells (Friedmann, 1989; Ridgeway, 1988; Baichwal and Sug- region. Thus, it will be most convenient to introduce the den, 1986; Couparet al., 1988; Horwichetal., 1990). polynucleotide encoding the gene of interest at the position In order to effect expression of sense or antisense gene from which the El-coding sequences have been removed. However, the position of insertion of the construct within the constructs, the expression construct must be delivered into a adenovirus sequences is not critical to the invention. The cell. This delivery may be accomplishedin vitro, as in labo- polynucleotide encoding the gene of interest may also be ratory procedures for transformingcells lines, or in vivo or ex inserted in lieu of the deleted E3 region in E3 replacement vivo, as in the treatment of certain disease states. One mecha- vectors, as described by Karlssonet al. (1986), or in the E4 nism for delivery is via viral infection where the expression region where a helpercell line or helper virus complements 40 construct is encapsidated in an infectious viral particle. the E4 defect. Several non-viral methods for the transfer of expression Adenovirus vectors have been used in eukaryotic gene constructs into cultured mammalian cells also are contem- expression (Levrero et al., 1991; Gomez-Foix et al., 1992) plated by the present invention. These include calcium phos- and vaccine development (Grunhausand Horwitz, 1992; Gra- phate precipitation (Graham and Van Der Eb, 1973; Chen and ham and Prevec, 1991). Recently, animal studies suggested 45 Okayama, 1987; Rippeet al., 1990) DEAE-dextran (Gopal, that recombinant adenovirus could be used for gene therapy 1985), electroporation (Tur-Kaspaet al., 1986; Potteret al., (Stratford-Perricaudet and Perricaudet, 1991; Stratford-Per- 1984), direct microinjection (Harland and Weintraub, 1985), ricaudet, et al., 1990; Rich et al., 1993), Studies in adminis- DNA-loaded liposomes (Nicolau and Sene, 1982; Fraley et tering recombinant adenovirus to different tissues include al., 1979) and lipofectamine-DNA complexes,cell sonication trachea instillation (Rosenfeld et al., 1991; Rosenfeld et al., (Fechheimer et al., 1987), gene bombardment using high 1992), muscle injection (Ragotet al., 1993), peripheral intra- velocity microprojectiles (Yang et al., 1990), and receptor- venous injections (Herz and Gerard, 1993) and stereotactic mediated transfection (Wu and Wu, 1987; Wu and Wu,1988). inoculation into the brain (Le Gal La Salle et al., 1993). Someofthese techniques may be successfully adapted for in Retroviral vectors are also suitable for expressing inhibi- Vivo or ex Vivo use. tors of miR-15 family membersin cells. The retroviruses are 55 Once the expression construct has been delivered into the a group of single-stranded RNA viruses characterized by an cell the nucleic acid encoding the gene of interest may be ability to convert their RNA to double-stranded DNA in positioned and expressedatdifferent sites. In certain embodi- infected cells by a process of reverse-transcription (Coffin, ments, the nucleic acid encoding the gene may be stably 1990). The resulting DNAthenstably integrates into cellular integrated into the genomeofthe cell. This integration may be chromosomes as a provirus and directs synthesis of viral in the cognate location and orientation via homologous proteins. The integration results in the retention of the viral recombination (gene replacement)or it may be integrated in gene sequencesin the recipient cell and its descendants. The a random, non-specific location (gene augmentation). In yet retroviral genomecontainsthree genes, gag, pol, and env that further embodiments, the nucleic acid may be stably main- code tier capsid proteins, polymerase enzyme, and envelope tained in the cell as a separate, episomal segment of DNA. components, respectively. A sequence found upstream from Such nucleic acid segments or “episomes” encode sequences the gag gene contains a signal for packaging of the genome sufficient to permit maintenance andreplication independent into virions. Two long terminal repeat (LTR) sequences are of or in synchronization with the host cell cycle. How the US 9,078,919 B2 19 20 expression constructis delivered to a cell and where in the cell the selective uptake ofmacromolecules by receptor-mediated the nucleic acid remains is dependent on the type of expres- endocytosis in almostall eukaryotic cells. Because of thecell sion construct employed. type-specific distribution of various receptors, the delivery In another embodiment of the invention, the expression can be highly specific (Wu and Wu, 1993). construct may simply consist of naked recombinant DNA or Receptor-mediated gene targeting vehicles generally con- plasmids. Transfer of the construct may be performed by any sist of two components: a cell receptor-specific ligand and a of the methods mentioned above which physically or chemi- DNA-binding agent. Several ligands have been used for cally permeabilize the cell membrane. This is particularly receptor-mediated gene transfer. The most extensively char- applicable for transfer in vitro but it may be applied to in vivo acterized ligands are asialoorosomucoid (ASOR) (Wu and use as well. Dubensky et al. (1984) successfully injected Wu, 1987) and transferrin (Wagneret al., 1990). Recently, a polyomavirus DNAin the form ofcalcium phosphate precipi- synthetic neoglycoprotein, which recognizes the same recep- tates into liver and spleen of adult and newborn mice demon- tor as ASOR,has been used as a gene delivery vehicle (Ferkol strating active viral replication and acute infection. Ben- et al., 1993; Perales et al., 1994) and epidermal growth factor venisty and Neshif (1986) also demonstrated that direct (EGF)hasalso been used to deliver genes to squamous car- intraperitoneal injection of calctum phosphate-precipitated cinoma cells (Myers, EPO 0273085). plasmids results in expression of the transfected genes, it is In other embodiments, the delivery vehicle may comprise a envisioned that DNA encoding a polynucleotide of interest ligand and a liposome. For example, Nicolau et al. (1987) may also be transferred in a similar manner in vivo and employed lactosyl-ceramide, a galactose-terminal asialgan- express the gene product. glioside, incorporated into liposomes and observed an In still another embodimentof the invention for transfer- increase in the uptake of the insulin gene by hepatocytes. ring anaked DNA expression construct into cells may involve Thus, it is feasible that a nucleic acid encoding a particular particle bombardment. This method depends onthe ability to gene also maybespecifically delivered into a cell type (e.g. accelerate DNA-coated microprojectiles to a high velocity cardiac cell) by any numberof receptor-ligand systems with allowing them to pierce cell membranesandenter cells with- or without liposomes. For example, epidermal growth factor out killing them (Klein et al. 1987). Several devices for accel- (EGF) maybeusedas the receptor for mediated delivery of a erating small particles have been developed. One such device nucleic acid into cells that exhibit upregulation ofEGF recep- relies on a high voltage discharge to generate an electrical tor. Mannosecan be usedto target the mannose receptor on current, which in turn provides the motive force (Yanget al., liver cells. Also, antibodies to CD5 (CLL), CD22 (ym- 1990). The microprojectiles used have consisted of biologi- phoma), CD25 leukemia) and MAA (melanoma) can simi- cally inert substances such as tungsten or gold beads. 30 larly be used as targeting moieties. Selected organs includingthe liver, skin, and muscle tissue In particular example, the polynucleotide may be admin- a of rats and mice have been bombarded in vivo (Yanget al., istered in combination with a cationic lipid. Examples of 1990; Zelenin et al., 1991). This may require surgical expo- cationic lipids include, but are not limited to, lipofectin, sure of the tissueor cells, to eliminate any intervening tissue DOTMA, DOPE, and DOTAP. The publication of between the gun andthe target organ, i.e., ex vivo treatment. W00071096, whichis specifically incorporated by reference, Again, DNA encodinga particular polynucleotide of interest describes different formulations, such as a DOTAP:choles- maybedelivered via this method andstill be incorporated by terol or cholesterol derivative formulation that can effectively the present invention. be used for gene therapy. Other disclosures also discuss dif- In a further embodiment of the invention, the expression ferent lipid or liposomal formulations including nanopar- construct may be entrapped in a liposome. Liposomes are 40 ticles and methods of administration; these include, but are vesicular structures characterized by a phospholipid bilayer not limited to, U.S. Patent Publication 20030203865, membraneand an inner aqueous medium. Multilamellarlipo- 20020150626, 20030032615, and 20040048787, which are somes have multiple lipid layers separated by aqueous specifically incorporated by reference to the extent they dis- medium. They form spontaneously when phospholipids are close formulations and other related aspects ofadministration suspended in an excess of aqueous solution. The lipid com- 45 and delivery of nucleic acids. Methods used for forming ponents undergo self-rearrangement before the formation of particles are also disclosed in U.S. Pat. Nos. 5,844,107, 5,877, closed structures and entrap water and dissolved solutes 302, 6,008,336, 6,077,835, 5,972,901, 6,200,801, and 5,972, between the lipid bilayers (Ghosh and Bachhawat, 1991). 900, which are incorporated by reference for those aspects. Also contemplated are lipofectamine-DNA complexes. In certain embodiments, gene transfer may more easily be In certain embodimentsofthe invention,the liposome may 50 performed under ex vivo conditions. Ex vivo gene therapy be complexed with a hemagglutinating virus (HVJ). This has refers to the isolation of cells from an animal, the delivery of been shownto facilitate fusion with the cell membrane and a nucleic acid into the cells in vitro, and then the return of the promote cell entry of liposome-encapsulated DNA (Kaneda modified cells back into an animal. This may involve the et al., 1989). In other embodiments, the liposome may be surgical removalof tissue/organs from an animalorthe pri- complexed or employed in conjunction with nuclear non- mary culture of cells andtissues. histone chromosomalproteins (HMG-1) (Kato et al., 1991). The present invention also includes methods for scaveng- In yet further embodiments, the liposome may be complexed ing or clearing inhibitors ofmiR-15 family members follow- or employed in conjunction with both HVJ and HMG-1. In ing treatment. The method may comprise overexpressing that such expression constructs have been successfully hybridization sites for inhibitors of the miR-15 family mem- employed in transfer and expression of nucleic acid in vitro bers in cardiac tissue. In one embodiment, the method com- and in vivo, then they are applicable for the present invention. prises overexpression of hybridization sites for inhibitors of Where a bacterial promoter is employed in the DNA con- the miR-15 family members in cardiac muscle using a heart struct, it also will be desirable to include within the liposome muscle specific promoter (e.g. a-MHC). In another embodi- an appropriate bacterial polymerase. ment, the hybridization site may comprise a sequence of a Other expression constructs which can be employed to seed region from a miR-15 family member. In another deliver a nucleic acid encoding a particular geneinto cells are embodiment, the hybridization site may comprise the receptor-mediated delivery vehicles. These take advantage of sequence of SEQ ID NO: 18. In some embodiments, the US 9,078,919 B2 21 22 hybridization site may contain a sequencethat is complemen- however, where several days (2, 3, 4, 5, 6 or 7) to several tary to a sequence from the 3'UTRofone or moretargets of a weeks (1, 2, 3, 4, 5, 6, 7 or 8) lapse between the respective miR-15 family member, such as FGF2, TGFb-inducedfactor administrations. 2, BCL9I, BCL2L, CDC25A,cyclin E1, cyclin D1, or cyclin It also is conceivable that more than one administration of D2. either an inhibitor of a miR-15 family member, or the other In another embodimentofthe invention, an inhibitor ofone pharmaceutical agent will be desired. In this regard, various or more miR-15 family members is administered to the sub- combinations may be employed. By way of illustration, ject in combination with other therapeutic modalities. Cur- wherethe inhibitor ofamiR-15 family memberis “A” and the rent medical managementof cardiac hypertrophy in theset- other pharmaceutical agent is “B,” the following permuta- ting of a cardiovascular disorder includes the use of at least tions based on 3 and4 total administrations are exemplary: twotypes of drugs: inhibitors ofthe -angiotensin system A/B/A B/A/B B/B/A A/A/B B/A/A A/B/B B/B/B/A B/B/ and B- blocking agents (Bristow, 1999). Therapeu- A/B tic agents to treat pathologic hypertrophy in the setting of A/A/B/B A/B/A/B A/B/B/A B/B/A/A B/A/B/A B/A/A/B heart failure include angiotensin IT converting enzyme (ACE) B/B/B/A inhibitors and B- blocking agents (Eich- A/A/A/B B/A/A/A A/B/A/A A/A/B/A A/B/B/B B/A/B/B horn and Bristow, 1996). Other pharmaceutical agents that B/B/A/B have been disclosed for treatment of cardiac hypertrophy Other combinationsare likewise contemplated. include angiotensin II receptor antagonists (U.S. Pat. No. Treatment regimens would vary depending on the clinical 5,604,251) and neuropeptide Y antagonists (WO 98/33791). situation. However, long-term maintenance would appearto Non-pharmacological treatment is primarily used as an 20 be appropriate in most circumstances. It also may be desirable adjunct to pharmacological treatment. One means of non- to treat hypertrophy with inhibitors of miR-15 family mem- pharmacological treatment involves reducing the sodium in bers intermittently, such as within a brief window during the diet. In addition, non-pharmacological treatment also disease progression. entails the elimination of certain precipitating drugs, includ- Pharmacological therapeutic agents and methods of ing negative inotropic agents (e.g., certain calcium channel 25 administration, dosages, etc., are well knowntothoseof skill blockers and antiarrhythmic drugs like disopyramide), car- in the art (see for example, the “Physicians Desk Reference”, diotoxins (e.g., amphetamines), and plasma volume expand- Kiaassen’s “The Pharmacological Basis of Therapeutics”, ers (e.g., nonsteroidal anti-inflammatory agents and gluco- “Remington’s Pharmaceutical Sciences”, and “The Merck corticoids). Index, Eleventh Edition”, incorporated herein by reference in Thus,in additionto the therapies described above, one may 30 relevant parts), and may be combined with the invention in also provide to the subject more “standard” pharmaceutical tight of the disclosures herein. Somevariation in dosage will cardiac therapies with the inhibitor of one or more miR-15 necessarily occur depending on the condition of the subject family members. Examples of other therapies include, with- being treated. The person responsible for administration will, out limitation, so-called “beta blockers,” anti-hypertensives, in any event, determine the appropriate dose for the individual cardiotonics, anti-thrombotics, vasodilators, hormone 35 subject, and such individual determinations are within the antagonists, iontropes, diuretics, endothelin receptor antago- skill of those of ordinary skill in the art. nists, calcium channel blockers, phosphodiesterase inhibi- Non-limiting examples of a pharmacological therapeutic tors, ACE inhibitors, angiotensin type 2 antagonists and agent that may be used in the present invention include an cytokine blockers/inhibitors, and FIDAC inhibitors. The antihyperlipoproteinemic agent, an antiarteriosclerotic agent, combination therapy also may involve inhibiting the expres- 40 an /fibrinolytic agent, a blood coagulant, an sion or activity of additional miRNAsinvolved in cardiac antiarrhythmic agent, an antihypertensive agent, a vasopres- remodeling such as miR-499, miR-208, miR-208b and miR- sor, a treatment agent for congestive heart failure,an antiangi- 21. Combination therapy may also include overexpression of nal agent, an antibacterial agent or a combination thereof. particular microRNAs, such as miR-29. In addition, it should be noted that any ofthe following may Combinations maybe achieved by contacting cardiac cells 45 be used to develop newsets of cardiac therapy target genes as with a single composition or pharmacological formulation B-blockers were used in the present examples (see below). that includes an inhibitor of one or more miR-15 family White it is expected that many of these genes may overlap, membersanda standard pharmaceutical agent, or by contact- new genetargets likely can be developed. ing the cell with two distinct compositions or formulations,at In certain embodiments, administration of an agent that the same time, wherein one composition includes the inhibi- lowers the concentration of one of more blood lipids and/or tor of a miR-15 family memberand the other includes the lipoproteins, knownherein as an “antihyperlipoproteinernic,” standard pharmaceutical agent. Alternatively, the therapy may be combined with a cardiovascular therapy according to using an inhibitor ofa miR-15 family member mayprecede or the present invention, particularly in treatmentof atlierselero- follow administration of the other agent(s) by intervals rang- sis and thickenings or blockages ofvascular tissues. In certain ing from minutes to weeks. In embodiments where the stan- embodiments, an antihyperlipoproteinemic agent may com- dard pharmaceutical agent and the inhibitor of a miR-15 prise an aryloxyalkanoic/fibric acid derivative, a resin/bite family memberare applied separately to the cell, one would acid sequesterant, a HMG CoA reductase inhibitor, a nico- generally ensure that a significant period of time did not tinic acid derivative, a thyroid hormoneor thyroid hormone expire between the time of each delivery, such that the phar- analog, a miscellaneous agent or a combination thereof maceutical agent and inhibitor of a miR-15 family member examples of aryloxyalkanoic/fibric acid derivatives include would still be able to exert an advantageously combined beclobrate, enzafibrate, binifibrate, ciprofibrate, clinofibrate, effect on the cell. In such instances, it is contemplated that one clofibrate (atromide-S), clofibric acid, etofibrate, fenofibrate, would typically contact the cell with both modalities within gemfibrozil (lobid), nicofibrate, pirifibrate, ronifibrate, sim- about 12-24 hours of each other and, morepreferably, within fibrate and theofibrate. Non-limiting examplesof resins/bile about 6-12 hours ofeach other, with a delay time ofonly about acid sequesterants include cholestyramine (cholybar, ques- 12 hours being mostpreferred. In somesituations, it may be tran), colestipol (colestid) and polidexide. Non-limiting desirable to extend the timeperiodfor treatmentsignificantly, examples of HMG CoAreductase inhibitors include lovasta- US 9,078,919 B2 23 24 tin (mevacor), pravastatin (pravochol) or simvastatin (zocor). examples of Class IC antiarrhythmic agents include encam- Non-limiting examples of nicotinic acid derivatives include ide (enkaid) and flecamide (tambocor). nicotinate, acepimox, niceritrol, nicoclonate, nicomol and Non-limiting examples ofa beta Mocker, otherwise known oxiniacic acid. Non-limiting examples of thyroid hormones as a B-adrenergic blocker, a B- or a Class and analogs thereof include etoroxate, thyropropic acid and II antiarrhythmic agent, include (sectral), alpre- thyroxine. nolol, , , , , , Non-limiting examples of miscellaneous antihyperlipo- , , , bucurnolol, , proteinemics include acifran, azacosterol, benfluorex, f-ben- hufuralol, , , hydrochloride, zalbutyramide, camitine, chondroitin sulfate, clomestrone, , , , , , ceta- detaxtran, dextran sulfate sodium, 5,8,11,14,17-eicosapen- 10 molol, , dilevalol, , esmolol (brevibloc), taenoic acid, eritadenine, furazabol, megiutol, melinamide, , , , , , mytatrienediol, ornithine, y-oryzanol, pantethine, pentaeryth- , , , , , ritol tetraacetate, a-phenylbutyramide, pirozadil, probucol , , , , , prone- (oreico), B-sitosterol, sultosilic acid- salt, tiad- thalol, propanolol (inderal), (betapace), , enol, triparanol and xenbucin. A non-limiting example of an 15 tatinolot, , , and xibinolol. In cer- antiarteriosclerotic includes pyridinol carbamate. tain embodiments, the comprises an aryloxypro- In certain embodiments, administration of an agent that panolamine derivative. Non-limiting examples of arylox- aids in the removal or prevention of blood clots may be ypropanolamine derivatives include acebutolol, , combined with administration of a modulator, particularly in arotinolol, atenolol, betaxolol, bevantolol, bisoprolol, bopin- treatment of athersclerosis and vasculature (e.g., arterial) 20 dolol, bunitrolol, butofliolol, carazolol, carteolol, carveditol, blockages. Non-limiting examples of antithrombotic and/or cetaniolol, epanolol, indenolol, mepindolol, metipranolol, fibrinolytic agents include , metoprolol, moprolol, nadolol, nipradilol, oxprenolol, penb- antagonists, antiplatelet agents, thrombolytic agents, throm- utolol, pindolol, propanolol, , tertatolol, timolol and bolytic agent antagonists or combinationsthereof. In certain toliprolol. embodiments, antithrombotic agents that can be administered 25 Non-limiting examples of an agent that prolong repolar- orally, such as, for example, and wafarin (coumadin), ization, also known as a Class II antiarrhythmic agent, are preferred. include amiodarone (cordarone) and sotalol (betapace). Non-limiting examples of anticoagulants include aceno- Non-limiting examples of a calcium channel blocker, oth- coumarol, , , bromindione, , erwise known as a Class IV antiarrythmic agent, include an cournetarol, cyclocumarol, dextran sulfate sodium, dicuma- 30 arylalkylamine(e.g., bepridile, diltiazem, fendiline, preny- rol, , , ethylidene dicou- lamine, terodiline, verapamil), a dihydropyridine derivative marol, fiuindione, , , lyapolate sodium, (felodipine, isradipine, nicardipine, nifedipine, nimodipine, oxazidione, pentosan polysulfate, , phenproc- nisoldipine, nitrendipine) a piperazinde derivative (e.g., cin- ournon, phosvitin, , tioclomarol and . narizine, flunarizine, lidoflazine) or a micellaneous calcium Non-limiting examples of antiplatelet agents include aspi- 35 channel blocker such as bencyciane, etafenone, magnesium, rin, a dextran, (persantin), heparin, sulfinpyra- mibefradil or perhexyline. In certain embodiments a calcium none(anturane) andticlopidine (fiend). channel blocker comprises a long-acting dihydropyridine Non-limiting examples ofthrombolytic agents includetis- (nifedipine-type) calcium antagonist. sue plaminogen activator (activase), plasmin, pro-, Non-limiting examples of miscellaneous antiarrhythmic urokinase (abbokinase) streptase), anistre- 40 agents include adenosine (adenocard), digoxin (lanoxin), plase/APSAC (eminase). acecamide, ajmaline, amoproxan,aprindine, tosy- In certain embodiments wherein a patient is suffering from late, bunaftine, butobendine, capobenic acid, cifenline, dis- a hemorrhageoran increased likelihood ofhemorrhaging, an opyranide, hydroquinidine, indecainide, ipatropium bro- agent that may enhance blood coagulation may be used. Non- mide, lidocaine, lorajmine, lorcainide, meobentine, limiting examples of blood coagulation promoting agents 45 moricizine, pirmenol, prajmaline, , include thrombolytic agent antagonists and anticoagulant polygalacturonate, quinidinesulfite and viquidil. antagonists. Non-limiting examples of anticoagulant antago- Non-limiting examples of antihypertensive agents include nists include protamine and vitamine K1. , alpha/beta blockers, alpha blockers, anti-an- Non-limiting examples of thrombolytic agent antagonists giotensin II agents, beta blockers, calctum channel blockers, include amiocaproic acid (amicar) and tranexamic acid (am- 50 vasodilators and miscellaneous antihypertensives. stat). Non-limiting examples of include Non-limiting examples of an , also known as anagrelide, , cilstazol, daltroban, , an a-adrenergic blocker or an a-adrenergic antagonist, enoxaparin, fraxiparine, , lanioparan, ozagrel, include amosulalol, arotinolol, , , picotamide, plafibride, tedelparin, andtriflusal. mesylates, , , labetalol, nicer- Non-limiting examples of antiarrhythmic agents include 55 goline, , ,, and yohim- Class I antiarrhythmic agents (sodium channel blockers), bine. In certain embodiments, an alpha blocker may comprise Class II antiarrhythmic agents (beta-adrenergic blockers), a quinazoline derivative. Non-limiting examples of quinazo- Class III antiarrhythmic agents (repolarization prolonging line derivatives include , , doxazosin,pra- drugs), Class IV antiarrhythmic agents (calcium channel zosin, terazosin and trimazosin. In certain embodiments, an blockers) and miscellaneous antiarrhythmic agents. 60 antihypertensive agent is both an alpha and beta adrenergic Non-limiting examples of sodium channel blockers antagonist. Non-limiting examples of an alpha/beta blocker include Class IA, Class IB and Class IC antiarrhythmic comprise labetalol (normodyne, trandate). agents. Non-limiting examples of Class IA antiarrhythmic Non-limiting examples of anti-angiotensin I] agents agents include disppyramide (norpace), procainamide (pron- include angiotensin converting enzymeinhibitors and angio- estyl) and quinidine (quinidex). Non-limiting examples of tensin II receptor antagonists, Non-limiting examples of Class IB antiarrhythmic agents include lidocaine (xylocaine), angiotensin converting enzyme inhibitors (ACE inhibitors) tocamide (tonocard) and mexiletine (mexitil). Non-limiting include , (vasotec), , , US 9,078,919 B2 25 26 , , lisinopril, moveltopril, perin- zothiadiazine derivatives include althizide, bendroflumethi- dopril, and . Non limiting examples of an azide, benzthiazide, benzylhydrochlorothiazide, buthiazide, angiotensin II receptor blocker, also knownas an angiotensin chlorothiazide, chlorthalidone, cyclopenthiazide, cyclothiaz- II , anANG receptor blocker or anANG-II ide, diazoxide, epithiazide, ethiazide, fenquizone, hydrochlo- type-1 receptor blocker (ARBS), include angiocandesartan, rothizide, hydroflumethizide, methyclothiazide, meticrane, , , and . metolazone, paraflutizide, polythizide, tetrachlormethiazide Non-limiting examples of a sympatholytic include a cen- and trichlormethiazide. Non-limiting examples of N-car- trally acting sympatholytic or a peripherially acting sym- boxyalkyl(peptide/lactam) derivatives include alacepril, cap- patholytic. Non-limiting examples of a centrally acting sym- topril, cilazapril, delapril, enalapril, enalaprilat, fosinopril, patholytic, also known as an central nervous system (CNS) 10 lisinopril, moveltipril, , quinapril and ramipril. sympatholytic, include (catapres), Non-limiting examples of dihydropyridine derivatives (wytensin) (tenex) and (aldomet). include amlodipine, felodipine, isradipine, nicardipine, nife- Non-limiting examples of a peripherally acting sym- dipine, nilvadipine, nisoldipine and nitrendipine. Non-limit- patholytic include a ganglion blocking agent, an adrenergic ing examples of derivatives include , neuron blocking agent, a B-adrenergic blocking agent or a 15 debrisoquin, guanabenz, guanacline, , guanazod- alpha] -adrenergic blocking agent. Non-limiting examples of ine, , guanfacine, guanochlor, and a ganglion blocking agent include (inversive) . Non-limiting examples of hydrazines/phthala- and trimethaphan (arfonad). Non-limiting examples of an zines include budralazine, cadralazine, dihydralazine, adrenergic neuron blocking agent include guanethidine(is- endralazine, hydracarbazine, hydralazine, pheniprazine, melin) and (serpasil). Non-limiting examples of a 20 pildralazine and todralazine. Non-limiting examples of imi- B-adrenergic blocker include acenitolol (sectral), atenolol dazole derivatives include clonidine, , phentola- (tenormin), betaxolol (kerlone), carteolol (cartrol), labetalol mine, and . Non-limiting examples of (normodyne,trandate), metoprolol (lopressor), nadanol(cor- quanternary ammonium compoundsinclude azamethonium gard), penbutolol (levatol), pindolol (visken), bromide, chloride, , penta- (inderal) and timolol (blocadren). Non-limiting examples of 25 cynium bis(methylsulfate), pentamethonium bromide, pento- alpha] -adrenergic blocker include prazosin (minipress), dox- linium tartrate, phenactropinium chloride and trimethidinium azocin (cardura) and terazosin (hytrin). methosulfate. Non-limiting examples ofreserpine derivatives In certain embodiments a cardiovasculator therapeutic include , , , reserpine agent may comprise a vasodilator(e.g., a cerebral vasodilator, and . Non-limiting examples of sulfonamide a coronary vasodilator or a peripheral vasodilator). In certain 30 derivatives include ambuside, clopamide, furosemide, inda- preferred embodiments, a vasodilator comprises a coronary pamide, quinethazone, tripamide and xipamide. vasodilator. Non-limiting examples of a coronary vasodilator Vasopressors generally are used to increase blood pressure include amotriphene, bendazol, benfurodil hemisuccinate, during shock, which may occur during a surgical procedure. benziodarone, chloracizine, chromonar, clobenfurol, cloni- Non-limiting examples of a vasopressor, also known as an trate, dilazep, dipyridamole, droprenilamine, efloxate, eryth- 35 antihypotensive, include amezinium methyl] sulfate, angio- rityl tetranitrane, etafenone, fendiline, floredil, ganglefene, tensin amide, ditnetofrine, , etifelmin, etilefrin, herestrol bis(B-diethylaminoethy! ether), hexobendine,itra- gepefrine, , , , min tosylate, khellin, mannitol hexanitrane, medibazine, pholedrine and . nicorglycerin, pentaerythritol tetranitrate, pentrinitrol, per- Non-limiting examples of agents for the treatment of con- hexyline, pirnefyl line, , tricromyl, trimetazidine, 40 gestive heart failure include anti-angiotensin II agents, after- troInitrate phosphate and visnadine. load-preload reduction treatment, diuretics and inotropic In certain embodiments, a vasodilator may comprise a agents. chronic therapy vasodilator or a hypertensive emergency In certain embodiments, an animal patient that can not vasodilator. Non-limiting examples of a chronic therapy tolerate an angiotensin antagonist may be treated with a com- vasodilator include hydralazine (apresoline) and minoxidil 45 bination therapy. Such therapy may combine administration (oniten). Non-limiting examples of a hypertensive emer- of hydralazine (apresoline) and isosorbide dinitrate (isordil, gency vasodilator include nitroprusside (nipride), diazoxide sorbitrate). (hyperstat IV), hydralazine (apresoline), minoxidil (loniten) Non-limiting examples of a diuretic include a or and verapamil. benzothiadiazine derivative (e.g., althiazide, bendroftumet- Non-limiting examples of miscellaneous antihyperten- 50 hazide, benzthiazide, benzylhydrochlorothiazide, buthiazide, sives include ajmaline, y-amindbutyric acid, bufeniode, cicle- chlorothiazide, chlorothiazide, chlorthalidone, cyclopenthi- tainine, ciclosidomine, a tannate, , azide, epithiazide, ethiazide, ethiazide, fenquizone, hydro- flosequinan, , mebutamate, mecamylamine, meth- chlorothiazide, hydroflumethiazide, methyclothiazide, meti- yldopa, methyl 4-pyridyl ketone thiosemicarbazone, crane, metolazone, paraflutizide, polythizide, muzolimine, , , pinacidil, , pri- 55 tetrachlorornethiazide, trichlormethiazide), an organomercu- maperone, a protoveratrine, raubasine, rescimetol, ril- rial (e.g., chlormerodrin, meralluride, mercamphamide, mer- menidene, , sodium nitrorusside, ticrynafen, tri- captomerin sodium, mercumallylic acid, mercumatilin methaphan camsylate, tyrosinase and . dodium, mercurouschloride, mersaly]), a pteridine(e.g., fur- Incertain embodiments, an antihypertensive may comprise terene, triamterene), purines (e.g., acefylline, 7-morpholi- an arylethanolamine derivative, a benzothiadiazine deriva- 60 nomethyltheophylline, protheobromine, theobromine), ste- tive, a N-carboxyatkyl(peptide/lactam) derivative, a dihydro- roids including aldosterone antagonists (e.g., canrenone, pyridine derivative, a guanidine derivative, a hydrazines/ph- oleandrin, spironolactone), a sulfonamide derivative (e.g., thalazine, an imidazole derivative, a quanternary ammonium acetazolamide, ambuside, azosemide, bumetanide, butazola- compound,a reserpine derivative or a suflonamidederivative. mide, chloraminophenamide, clofenamide, clorexolone, Non-limiting examples of arylethanolamine derivatives 65 diphenylmethane-4,4'-disulfonamide, disulfamide, ethox- include amosulalol, , dilevalol, labetalol, proneth- zolamide, furosemide, indapamide, mefruside, methazola- alol, sotalol and sulfinalol. Non-limiting examples of ben- mide, piretanide, quinethazone, torasemide, tripamide, xipa- US 9,078,919 B2 27 28 mide), a uracil (e.g., aminometradine, amisometradine), a performing surgery on an organism, providing a cardiovas- potassium sparing antagonist(e.g., amiloride, triamterene) or cular mechanical prostheses, angioplasty, coronary artery a miscellaneous diuretic such as aminozine, arbutin, chlora- reperfusion, catheter ablation, providing an implantable car- zanil, ethacrynic acid, etozolin, hydracarbazine, isosorbide, dioverter defibrillator to the subject, mechanical circulatory mannitol, tnetochalcone, muzolimine, perhexyline, ticrnafen support or a combination thereof. Non-limiting examples ofa and urea. mechanical circulatory support that may be used in the Non-limiting examples of a positive inotropic agent, also present invention comprise an intra-aortic balloon counter- known as a cardiotonic, include acefylline, an acetyldigi- pulsation, left ventricular assist device or combination toxin, 2-amino-4-picoline, amrinone, benfurodil hemisucci- thereof. nate, bucladesine, cerberosine, camphotamide, convali- 10 In another embodiment, the present invention provides a atoxin, cymarin, , deslanoside, digitalin, methodoftreating or preventing a musculoskeletal disorder digitalis, digitoxin, digoxin, , dopamine, dopex- in a subject in need thereof comprising (a) identifying a amine, enoximone, erythrophleine, fenalcomine, gitalin, subject havingorat risk ofa musculoskeletal disorder; and (b) gitoxin, glycocyamine, heptaminol, hydrastinine, ibopatnine, increasing the expression and/oractivity ofone or more miR- a lanatoside, metamivam, milrinone, nerifolin, oleandrin, 15 15 family members in skeletal muscle cells of said subject. ouabain, , , proscillaridine, resibufoge- The disorder may be selected from the group consisting of nin, scillaren, scillarenin, strphanthin, sulmazole, theobro- muscular dystrophy, disuse atrophy, muscle wasting in mine and xarnoterol. response to anti-gravity and denervation. Increasing the In particular embodiments, an intropic agent is a cardiac expression and/or activity may comprise administering said glycoside, a beta- or a phosphodiesterase 20 one or more miR-15 family membersto said subject, option- inhibitor. Non-limiting examples of a cardiac glycoside ally comprised within a lipid vehicle, or may comprise includes digoxin (lanoxin) and digitoxin (crystodigin). Non- administering an expression vector that expresses said one or limiting examples ofa B-adrenergic agonistinclude albuterol, more miR-15 family members in said subject. The expression , , , , clorprenaline, vector may be viral expression vector, such as an adenoviral a denopamine,dioxethedrine, dobutamine (dobutrex), dopam- 25 expression vector, or a non-viral expression vector, such as ine (intropin), , , , ethylnore- one comprised within a lipid vehicle. The method mayfurther pinephrine, , , , ibopamine, comprise a non-miR-15 family membertherapy (i.e. another isoetharine, isoproterenol, , metaproterenol, meth- microRNAor other appropriate therapy). oxyphenamine, oxyfedrine, , , protoky- The present invention also encompasses a pharmaceutical lol, , , , soterenol, , tre- 30 composition comprising an inhibitor of one or more miR-15 toquinol, and . Non-limiting examples family members (e.g. miR-195, miR-497, miR-424, miR- of a phosphodiesterase inhibitor include amrinone(inocor). 15a, miR-15b, miR-16-1, and miR-16-2). The pharmaceuti- agents may comprise organonitrates, calcium cal composition may comprise any inhibitor of a miR-15 channel blockers, beta blockers and combinations thereof. family memberas described herein, such as an antagomir, an Non-limiting examples of organonitrates, also known as 35 antisense oligonucleotide, an inhibitory RNA molecule, anda nitrovasodilators, include nitroglycerin (nitro-bid,nitrostat), nucleic acid comprising one or more miR-15 bindingsites. isosorbide dinitrate (isordil, sorbitrate) and amy]nitrate (as- Whereclinical applications are contemplated, pharmaceuti- pirol, vaporole). cal compositions will be prepared in a form appropriate for Endothelin (ET) is a 21-aminoacid peptide that has potent the intended application. Generally, this will entail preparing physiologic and pathophysiologic effects that appear to be 40 compositions that are essentially free of pyrogens, as well as involved in the developmentofheart failure. The effects ofET other impurities that could be harmful to humansor animals. are mediated through interaction with two classes of cell Colloidal dispersion systems, such as macromolecule surface receptors. The type A receptor (ET-A)is associated complexes, nanocapsules, microspheres, beads, and lipid with and cell growth while the type B recep- based systems including oil-in-water emulsions, micelles, tor (ET-B) is associated with endothelial-cell mediated 45 mixed micelles, and liposomes, may be used as delivery vasodilation and with the release of other neurohormones, vehicles for the oligonucleotide inhibitors of microRNA such as aldosterone. Pharmacologic agents that can inhibit function or constructs expressing inhibitory nucleotides. either the production of ET orits ability to stimulate relevant Commercially available fat emulsions that are suitable for cells are known in the art. Inhibiting the production of ET delivering the nucleic acids ofthe inventionto tissues, such as involvesthe use ofagents that block an enzyme termed endot- 50 cardiac muscle tissue, include Intralipid®, Liposyn®, Lipo- helin-converting enzymethat is involved in the processing of syn® II, Liposyn® III, Nutrilipid, and other similar lipid the active peptide from its precursor. Inhibiting the ability of emulsions. A preferred colloidal system for use as a delivery ETto stimulate cells involves the use of agents that block the vehicle in vivo is a liposome(i.e., an artificial membrane interaction ofET withits receptors. Non-limiting examples of vesicle). The preparation and use of such systems is well endothelin receptor antagonists (ERA) include , 55 knownin the art. Exemplary formulations are also disclosed Enrasentan, , Darusentan, , Atrasen- in USS. Pat. No. 5,981,505; U.S. Pat. No. 6,217,900; U.S. Pat. tan, Avosentan, Clazosentan, Edonentan, sitaxsentan, TBC No. 6,383,512; U.S. Pat. No. 5,783,565; U.S. Pat. No. 7,202, 3711, BQ 123, and BQ 788. 227; U.S. Pat. No. 6,379,965; U.S. Pat. No. 6,127,170; U.S. In certain embodiments, the secondary therapeutic agent Pat. No. 5,837,533; U.S. Pat. No. 6,747,014; and WO03/ may comprise a surgery of some type, which includes, for 60 093449, which are herein incorporated by reference in their example, preventative, diagnostic or staging, curative and entireties. palliative surgery. Surgery, and in particular a curative sur- One will generally desire to employ appropriate salts and gery, may be used in conjunction with other therapies, such as buffers to render delivery vehicles stable and allow for uptake the present invention and one or more other agents. by target cells. Buffers also will be employed when recom- Such surgical therapeutic agents for vascular and cardio- 65 binant cells are introduced into a patient. Aqueous composi- vascular diseases and disorders are well known to those of tions ofthe present invention comprise an effective amount of skill in the art, and may comprise, but are not limited to, the delivery vehicle or cells, dissolved or dispersed in a phar- US 9,078,919 B2 29 30 maceutically acceptable carrier or aqueous medium. The action of microorganisms can be brought about by various phrases “pharmaceutically acceptable” or “pharmacologi- antibacterial and antifungal agents, for example, parabens, cally acceptable”refer to molecular entities and compositions chlorobutanol, phenol, sorbic acid, thimerosal, and the like. that do not produceadverse, allergic, or other untoward reac- In manycases,it will be preferable to include isotonic agents, tions when administered to an animal or a human. As used for example, sugars or sodium chloride. Prolonged absorp- herein, “pharmaceutically acceptable carrier” includes sol- tion of the injectable compositions can be brought about by vents, buffers, solutions, dispersion media, coatings, antibac- the use in the compositions ofagents delaying absorption, for terial and antifungal agents, isotonic and absorption delaying example, aluminum monostearate and gelatin. agents andthe like acceptable for use in formulating pharma- Sterile injectable solutions maybe prepared by incorporat- ceuticals, such as pharmaceuticals suitable for administration ing the active compounds in an appropriate amount into a to humans. The use of such media and agents for pharmaceu- solvent along with any other ingredients (for example as tically active substances is well known in the art. Except enumerated above) as desired, followedbyfiltered steriliza- insofar as any conventional media or agent is incompatible tion. Generally, dispersions are prepared by incorporating the with the active ingredients of the present invention,its use in various sterilized active ingredients into a sterile vehicle therapeutic compositions is contemplated. Supplementary which contains the basic dispersion medium andthe desired active ingredients also can be incorporated into the composi- other ingredients, e.g., as enumerated above. In the case of tions, provided they do not inactivate the vectors or cells of sterile powders for the preparation of sterile injectable solu- the compositions. tions, the preferred methods of preparation include vacuum- The active compositions of the present invention may drying and freeze-drying techniques which yield a powder of include classic pharmaceutical preparations. Administration 20 the active ingredient(s) plus any additional desired ingredient ofthese compositions accordingto the present invention may from a previously sterile-filtered solution thereof. be via any common route so long as the target tissue is The compositions of the present invention generally may available via that route. This includes oral, nasal, or buccal. be formulated in a neutral or salt form. Pharmaceutically- Alternatively, administration may be by intradermal, subcu- acceptable salts include, for example, acid addition salts taneous, intramuscular, intraperitoneal or intravenousinjec- (formed with the free amino groups ofthe protein) derived tion, or by direct injection into cardiac tissue. Pharmaceutical from inorganic acids (e.g., hydrochloric or phosphoric acids, compositions comprising miRNA inhibitors or expression or from organic acids (e.g., acetic, oxalic, tartaric, mandelic, constructs encoding inhibitory polynucleotides may also be and thelike. Salts formed with the free carboxyl groups ofthe administered by catheter systemsor systemsthat isolate coro- protein can also be derived from inorganic bases (e.g., nary circulation for delivering therapeutic agentsto the heart. 30 sodium, potassium, ammonium,calcium, or ferric hydrox- Various catheter systems for delivering therapeutic agents to ides) or from organic bases(e.g., isopropylamine, trimethy- the heart and coronary vasculature are known in the art. Some lamine,histidine, procaine andthe like. non-limiting examples of catheter-based delivery methods or Upon formulation, solutionsare preferably administered in coronary isolation methods suitable for use in the present a manner compatible with the dosage formulation and in such invention are disclosed in U.S. Pat. No. 6,416,510; U.S. Pat. amountas is therapeutically effective. The formulations may No. 6,716,196; U.S. Pat. No. 6,953,466, WO 2005/082440, easily be administered in a variety of dosage forms such as WO 2006/089340, U.S. Patent Publication No. 2007/ injectable solutions, drug release capsules and the like. For 0203445, U.S. Patent Publication No. 2006/0148742, and parenteral administration in an aqueous solution, for U.S. Patent Publication No. 2007/0060907, which are all example, the solution generally is suitably buffered and the herein incorporated by reference in their entireties. Such 40 liquid diluentfirst rendered isotonic for example with suffi- compositions would normally be administered as pharmaceu- cient saline or glucose. Such aqueoussolutions may be used, tically acceptable compositions, as described supra. for example, for intravenous, intramuscular, subcutaneous The active compounds mayalso be administered parenter- and intraperitoneal administration. Preferably, sterile aque- ally or intraperitoneally. By way ofillustration, solutions of ous media are employedas is knownto those of skill in theart, the active compounds as free base or pharmacologically 45 particularly in light of the present disclosure. By way of acceptable salts can be prepared in water suitably mixed with illustration, a single dose may be dissolved in 1 ml of isotonic a surfactant, such as hydroxypropylcellulose. Dispersions NaC]solution and either added to 1000 ml ofhypodermocly- can also be preparedin glycerol, liquid polyethylene glycols, sis fluid or injected at the proposedsite of infusion, (see for and mixtures thereofandin oils. Under ordinary conditions of example, “Remington’s Pharmaceutical Sciences” 15th Edi- storage and use, these preparations generally contain a pre- 50 tion, pages 1035-1038 and 1570-1580). Some variation in servative to prevent the growth of microorganisms. dosage will necessarily occur depending on the condition of The pharmaceutical forms suitable for injectable use or the subject being treated. The person responsible for admin- catheter delivery include, for example, sterile aqueous solu- istration will, in any event, determine the appropriate dosefir tionsor dispersions andsterile powders for the extemporane- the individual subject. Moreover, for human administration, ous preparation ofsterile injectable solutions or dispersions. preparations should meetsterility, pyrogenicity, general Generally, these preparationsare sterile and fluid to the extent safety and purity standards as required by FDA Office of that easy injectability exists. Preparations should be stable Biologics standards. under the conditions of manufacture and storage and should Any of the compositions described herein may be com- be preserved against the contaminating action ofmicroorgan- prised in a kit. In a non-limiting example, an inhibitor of one isms, such as bacteria and fungi. Appropriate solvents or or more miR-15 family members, such as an antagomur, is dispersion media may contain, for example, water, ethanol, included in a kit. The kit may contain two or more, three or polyol (for example, glycerol, propylene glycol, and liquid more, four or more, five or more, or six inhibitors for each polyethylene glycol, and the like), suitable mixtures thereof miR-15 family member. By way of example, the kit may and vegetable oils. The properfluidity can be maintained, for contain an inhibitor of miR-195 andan inhibitor ofmiR-15a. example, by the use of a coating, such as lecithin, by the All possible combinations of inhibitors for miR-15 family maintenance of the required particle size in the case of dis- members are contemplated by the invention. The kit may persion and by the use of surfactants. The prevention of the further include water and/or buffers to stabilize the inhibitory US 9,078,919 B2 31 32 polynucleotides. The kit may also include one or more trans- memberin the presence and absence of the candidate com- fection reagent(s) to facilitate delivery of the miRNA inhibi- pound. For example, a method generally comprises: tors to cells. (a) providing a candidate compound; The components of the kits may be packaged either in (b) admixing the candidate compound with a miR-15 fam- aqueous media or in lyophilized form. The container means ily member; of the kits will generally include at least one vial, test tube, (c) measuring activity of the miR-15 family member; and flask, bottle, syringe or other container means, into which a (d) comparingthe activity in step (c) with the activity in the component maybeplaced, and preferably, suitably aliquoted. absenceof the candidate compound, Wherethere is more than one componentin the kit (labeling wherein a difference between the measuredactivities indi- reagent and label may be packagedtogether), the kit also will 10 cates that the candidate compound is a modulator of a generally contain a second, third or other additional container miR-1.5 family member. into which the additional components may be separately Assays also may be conductedin isolated cells, organs, or in placed. However, various combinations of components may living organisms. be comprisedin a vial. The kits of the present invention also Assessing the activity or expression of a miR-15 family 15 will typically include a means for containing the nucleic member may comprise assessing the expression level of the acids, and any other reagent containers in close confinement miR-15 family member. Thosein theart will be familiar with for commercial sale. Such containers may include injection or a variety of methods for assessing RNA expression levels blow-moldedplastic containers into which the desired vials including, for example, northern blotting or RT-PCR.Assess- are retained. 20 ing the activity or expression of the miR-15 family member Whenthe componentsofthe kit are provided in one and/or may comprise assessing the activity of the miR-15 family moreliquid solutions, the liquid solution is an aqueous solu- member. In some embodiments, assessing the activity of the tion, with a sterile aqueous solution being particularly pre- ma-15 family member comprises assessing expression or ferred. activity of a gene regulated by the miR-15 family member. However, the components of the kit may be provided as 25 Genes regulated by miR-15 family members include, for dried powder(s). When reagents and/or componentsare pro- example, FGF2, TGFb-induced factor 2, BCL9I, BCL2L, vided as a dry powder, the powder can be reconstituted by the CDC25A,cyclin E1, cyclin D1, and cyclin D2. Thosein the addition ofa suitable solvent. It is envisioned that the solvent art will be familiar with a variety ofmethodsfor assessing the mayalso be provided in another container means. activity or expression of genes regulated by miR-15 family The container means will generally include at least one 30 members. Such methodsinclude, for example, northern blot- vial, test tube, flask, bottle, syringe and/or other container ting, RT-PCR, ELISA, or western blotting. means, into which the nucleic acid formulations are placed, It will, of course, be understood that all the screening preferably, suitably allocated. The kits may also comprise a methods of the present invention are useful in themselves second container meansfor containing a sterile, pharmaceu- notwithstanding the fact that effective candidates may not be tically acceptable buffer and/or other diluent. 35 Such kits may also include components that preserve or found. The invention provides methodsfor screening for such maintain the miRNA inhibitors or that protect against their candidates, not solely methodsoffinding them. degradation. Such components may be RNAse-freeor protect As used herein the term “candidate substance”refers to any against RNAses. Such kits generally will comprise, in suit- molecule that may potentially modulate the function ofmiR- able means,distinct containers for each individual reagent or 40 15 family members. Onewill typically acquire, from various solution. commercial sources, molecular libraries that are believed to A kit will also include instructions for employing the kit meetthe basic criteria for useful drugs in an effort to “brute components as well the use of any other reagent not included force” the identification of useful compounds. Screening of in the kit. Instructions may include variations that can be such libraries, including combinatorially-generated libraries implemented. A kit may also include utensils or devices for 45 (e.g., antagomir libraries), is a rapid and efficient way to administering the miRNA inhibitor by various administration screen large numberofrelated (and unrelated) compoundsfor routes, such as parenteral or catheter administration. activity. Combinatorial approaches also lend themselves to Tt is contemplated that such reagents are embodiments of rapid evolution of potential drugs by the creation of second, kits ofthe invention. Such kits, however, are not limited to the third, and fourth generation compounds modeled on active, particular items identified above and may include any reagent 50 but otherwise undesirable compounds. Non-limiting used for the manipulation or characterization of miRNA. examples of candidate compounds that may be screened The present invention further comprises methodsfor iden- according to the methods of the present invention are pro- tifying modulators of miR-15 family members. Identified teins, peptides, polypeptides, polynucleotides, oligonucle- inhibitors of the function of one or more miR-15 family otides or small molecules, Modulators of miR-15 family membersare useful in the prevention or treatmentor reversal 55 members may also be agonistsor inhibitors ofupstream regu- of cardiac hypertrophy or heart failure. Modulators (e.g. lators of any one of the miR-15 family members. inhibitors) of miR-15 family members may be included in A quick, inexpensive and easy assay to run is an in vitro pharmaceutical compositions tier the treatment of cardiac assay. Such assays generally use isolated molecules, can be disorders according to the methodsofthe present invention. run quickly and in large numbers, thereby increasing the These assays may comprise random screening of large 60 amountof information obtainable in a short period of time k libraries of candidate compounds; alternatively, the assays variety ofvessels may be usedto run the assays, including test may be used to focus on particular classes of compounds tubes, plates, dishes and other surfaces such as dipsticks or selected with an eye towards structural attributes that are beads. believed to make them more likely to inhibit the expression A technique for high throughput screening of compounds and/or function of miR-15 family members. 65 is described in WO 84/03564, whichis herein incorporated by To identify a modulator of a miR-15 family member, one reference in its entirety. Large numbers of small antagomir generally will determine the function of a miR-15 family compounds may be synthesized on a solid substrate, such as US 9,078,919 B2 33 34 plastic pins or some other surface. Such molecules can be prising contacting the cell with a modulator of a miR-15 rapidly screening for their ability to hybridize to miR-15 family member. In one embodiment, the expression ofFGF2, family members. TGFb-induced factor 2, BCL9I, BCL2L, CDC25A,cyclin The present invention also contemplates the screening of E1, cyclin D1, or cyclin D2 is decreased in the cell following compounds for their ability to modulate expression and/or administration of a miR-15 family agonist. In another function of one or more miR-15 family membersin cells. embodiment, the expression of FGF2, TGFb-induced factor Various cell lines, including those derived from skeletal 2, BCL9I, BCL2L, CDC25A,cyclin E1, cyclin D1, or cyclin muscle cells, can be utilized for such screening assays, D2 is increased in the cell following administration of a including cells specifically engineered for this purpose. Pri- miR-15 family inhibitor. mary cardiac cells also may be used, as can the H9C2cellline. An agonist of a miR-15 family member maybea poly- In vivo assays involve the use of various animal models of nucleotide comprising a mature sequenceor a star sequence heart disease, including transgenic animals, that have been from a miR-15 family member. In one embodiment,the poly- engineeredto have specific defects, or carry markers that can nucleotide comprises the sequence ofSEQ ID NO: 7, SEQ ID be used to measure the ability of a candidate compound to NO: 8, SEQ ID NO: 9, SEQ ID NO: 10, SEQ ID NO: 11, SEQ reach and affect different cells within the organism. Due to ID NO:12, SEQ ID NO:13, SEQID NO: 14, SEQID NO:15, their size, ease of handling, and information on their physi- SEQ ID NO:16, SEQ ID NO: 17, SEQ ID NO:20, or SEQ ID ology and genetic make-up, mice are a preferred embodi- NO: 21. In another embodiment, the agonist of a miR-15 ment, especially for transgenics. However, other animals are family member may be a polynucleotide comprising thepri- suitable as well, includingrats, rabbits, hamsters, guinea pigs, miRNAor pre-miRNA sequence for a miR-15 family mem- gerbils, woodchucks, cats, dogs, sheep, goats, pigs, cows, ber(e.g. pre-miR-195, pre-miR-497, pre-miR-424, pre-miR- horses and monkeys (including chimps, gibbons and 15a, pre-miR-15b, pre-miR-16-1, or pre-miR-16-2). For baboons). Assays for inhibitors may be conducted using an example, in one embodiment, the agonist may be a polynucle- animal model derived from any of these species. otide comprising a sequence of SEQ ID NO:1, SEQ ID NO: Treatmentofanimals with test compoundswill involve the 2, SEQID NO: 3, SEQ ID NO: 4, SEQ ID NO: 5, SEQ ID NO: administration of the compound, in an appropriate form, to 25 6, or SEQ ID NO:19. The polynucleotide comprising the a the animal. Administration will be by any route that could be mature sequence of miR-15 family member may be single utilized for clinical purposes. Determining the effectiveness stranded or double stranded. The polynucleotides may con- of a compound in vivo may involve a variety of different tain one or more chemical modifications, such as locked criteria, including but notlimited to alteration ofhypertrophic nucleic acids, peptide nucleic acids, sugar modifications, signaling pathways and physical symptoms of hypertrophy. such as 2'-O-alkyl (e.g. 2'-O-methyl, 2'-O-methoxyethyl), Also, measuring toxicity and dose response can be performed 2'-fluoro, and 4' thio modifications, and backbone modifica- in animals in a more meaningful fashion than in in vitro or in tions, such as one or more phosphorothioate, morpholino, or cyto assays. phosphonocarboxylate linkages. In one embodiment, the In one embodiment, the present invention provides a polynucleotide comprising a miR-15 family member method ofregulating cardiac cell survival comprising admin- 35 sequence is conjugated to cholesterol. In another embodi- istering to cardiac cells a modulator of one or more miR-15 ment, the agonist ofamiR-15 family membermay be an agent family members. In another embodiment, the modulator is an distinct from the miR-15 family memberthat acts to increase, agonist ofthe expressionor activity ofa miR-15 family mem- supplement, or replace the function of the miR-15 family ber. In another embodiment, cardiaccell survival is decreased member. In another embodiment, the miR-15 family agonist following administration of an agonist of a miR-15 family 40 may be expressed in vivo from a vector. member. In another embodiment, the modulator of a miR-15 In one embodiment, the present invention provides a family memberis an inhibitor of the expression or activity of method for treating pathologic cardiac hypertrophy, heart a miR-15 family member.In still another embodiment, car- failure, or myocardial infarction in a subject in need thereof diac cell survival is increased following administration of an comprising: identifying a subject having cardiac hypertrophy, inhibitor of a miR-15 family member. 45 heart failure, or myocardial infarction; and administering an In a further embodiment, the present invention provides a inhibitor of one or more miR-15 family membersto the sub- method of regulating apoptosis of cardiac cells comprising ject. In certain embodiments of the invention the miR-15 administering to cardiac cells a modulator of one or more family inhibitor may be identified by a method comprising: miR-15 family members. In another embodiment, the modu- (a) contacting a cell with a candidate compound;(b) assessing lator is an agonist of the expression or activity of a miR-15 50 activity or expression of a miR-15 family member; and (c) family member. In another embodiment, apoptosis of cardiac comparing the activity or expression in step (b) with the cells is increased following administration of an agonist of a activity or expression in the absence of the candidate com- miR-15 family member. In another embodiment, the modu- pound, wherein a reduction in the activity or expression ofthe lator ofamiR-15 family memberis an inhibitor ofthe expres- miR-15 family memberin the cell contacted with the candi- sion or activity of a miR-15 family member.In still another 55 date compound comparedto the activity or expression in the embodiment, apoptosis of cardiac cells is decreased follow- cell in the absence of the candidate compound indicates that ing administration of an inhibitor of a miR-15 family mem- the candidate compoundis an inhibitor of the miR-15 family ber. In some embodiments, the expression of FGF2, TGFb- member. induced factor 2, BCL9I, BCL2L, CDC25A, cyclin E1, A particular embodimentofthe present invention provides cyclin D1, or cyclin D2 is increased ina cell by contacting the transgenic animals that lack one or both functionalalleles of cell with a miR-15 family inhibitor. In other embodiments, a miR-15 family member. Also, transgenic animals that expression ofFGF2, TGFb-inducedfactor 2, BCL9I, BCL2L, express miR-15 family members under the control of an CDC25A,cyclin E1, cyclin D1, or cyclin D2 is decreased in inducible, tissue selective or a constitutive promoter, recom- a cell by contacting the cell with a miR-15 family agonist. binant cell lines derived from such animals, and transgenic Thus, the present invention includes a method ofregulating embryos may be useful in determining the exactrote that the expression ofFGF2, TGFb-inducedfactor 2, BCL9I, BCL2L, miR-15 family memberplays the development and differen- CDC25A,cyclin El, cyclin D1, or cyclin D2 in a cell com- tiation of cardiomyocytes and in the development of patho- US 9,078,919 B2 35 36 logic cardiac hypertrophy and heart failure. Furthermore, embryotransfer, the recipient females are anesthetized with these transgenic animals may provide an insight into heart an intraperitoneal injection of 0.015 ml of 2.5% avertin per development. The use of constitutively expressed miR-15 gram of body weight. The oviducts are exposed bya single family members provides a model for over or unregulated midline dorsal incision. An incision is then made through the expression. Also, transgenic animals that are “knocked out” body wall directly over the oviduct. The ovarian bursa is then for one or more miR-15 family members, in one or both torn with watchmakers forceps. Embryosto be transferred are alleles, are contemplated. placed in DPBS (Dulbecco’s phosphate buffered saline) and In a general embodiment, a transgenic animal is produced in the tip of a transfer pipet (about 10 to 12 embryos). The by the integration of a given transgene into the genome in a pipet tip is inserted into the infundibulum and the embryos mannerthat permits the expression of the transgene. Methods transferred. After the transfer, the incision is closed by two for producing transgenic animals are generally described by sutures. Wagner and Hoppe (U.S. Pat. No. 4,873,191; incorporated As used herein, the term “heart failure” is broadly used to herein by reference), and Brinsteret al. (1985; incorporated mean any condition that reduces the ability of the heart to herein by reference). pumpblood. Asa result, congestion and edema develop in the Typically, a gene flanked by genomic sequencesis trans- tissues. Most frequently, heart failure is caused by decreased ferred by microinjection into a fertilized egg. The microin- contractility ofthe myocardium,resulting from reduced coro- jected eggs are implantedinto a host female, and the progeny nary blood flow; however, many other factors may result in are screened for the expression of the transgene. Transgenic heart failure, including damage to the heart valves, vitamin animals may be produced from the fertilized eggs from a deficiency, and primary cardiac muscle disease. Though the number of animals including, but not limited to reptiles, 20 precise physiological mechanisms of heart failure are not amphibians, birds, mammals, andfish. entirely understood, heart failure is generally believed to DNA clones for microinjection can be prepared by any involve disorders in several cardiac autonomic properties, meansknowninthe art. For example, DNA clones for micro- including sympathetic, parasympathetic, and baroreceptor injection can be cleaved with enzymesappropriate for remov- responses. The phrase “manifestations of heart failure” is ing the bacterial plasmid sequences, and the DNA fragments used broadly to encompassall ofthe sequelae associated with electrophoresed on 1% agarose gels in TBE buffer, using heart failure, such as shortness of breath, pitting edema, an standard techniques. The DNA bandsare visualized by stain- enlarged tender liver, engorged neck veins, pulmonary rales ing with ethidium bromide, and the band containing the and the like including laboratory findings associated with expression sequences is excised. The excised band is then heart failure. placed in dialysis bags containing 0.3 M sodium acetate, pH 30 The term “treatment” or grammatical equivalents encom- 7.0. DNA is electroeluted into the dialysis bags, extracted passes the improvementand/or reversal of the symptoms of with a 1:1 phenol:chloroform solution and precipitated by heart failure (i.e., the ability of the heart to pump blood). two volumesof ethanol. The DNAis redissolved in 1 ml of “Improvementin the physiologic function” of the heart may low salt buffer (0.2 M NaCl, 20 mM Tris, pH 7.4, and 1 mM be assessed using any of the measurements described herein EDTA)andpurified on an Elutip-D™column. The column is (e.g., measurementof ejectionfraction, fractional shortening, first primed with 3 ml of high salt buffer (1 M NaCl, 20 mM left ventricular internal dimension, heart rate, etc.), as well as Tris, pH 7.4, and 1 mM EDTA) followed by washing with 5 any effect upon the animal’s survival. In use of animal mod- mlof low salt buffer. The DNA solutions are passed through els, the response of treated transgenic animals and untreated the column three times to bind DNAto the column matrix. transgenic animals is compared using any of the assays After one wash with 3 mloflow salt buffer, the DNA is eluted 40 described herein (in addition, treated and untreated non- with 0.4 ml high salt buffer and precipitated by two volumes transgenic animals may be included as controls). A com- of ethanol. DNA concentrations are measured by absorption pound which causes an improvement in any parameter asso- at 260 nm in a UV spectrophotometer. For microinjection, ciated with heart failure used in the screening methodsof the DNA concentrations are adjusted to 3 ug/ml in 5 mM Tris, pH instant invention may thereby be identified as a therapeutic 7.4 and 0.1 mM EDTA. Other methods for purification of 45 compound. DNA for microinjection are described in Palmiter et al. The term “dilated cardiomyopathy”refers to a type ofheart (1982); and in Sambrooket al. (2001). failure characterized by the presence of a symmetrically In an exemplary microinjection procedure, female mice six dilated left ventricle with poor systolic contractile function weeksofage are inducedto superovulate with a 5 IU injection and, in addition, frequently involves the right ventricle. (0.1 cc, ip) of pregnant mare serum gonadotropin (PMSG; The term “compound”refers to any chemical entity, phar- Sigma) followed 48 hourslater by a 5 IU injection (0.1 cc, ip) maceutical, drug, and the like that can be used to treat or ofhuman chorionic gonadotropin (hCG; Sigma). Females are prevent a disease, illness, sickness, or disorder of bodily placed with males immediately after hCG injection. Twenty- function. Compounds comprise both known and potential one hours after hCG injection, the mated females are sacri- therapeutic compounds. A compound can be determinedto be ficed by C02 asphyxiation or cervical dislocation and 55 therapeutic by screening using the screening methods of the embryos are recovered from excised oviducts and placed in present invention. A “known therapeutic compound”refersto Dulbecco’s phosphate buffered saline with 0.5% bovine a therapeutic compoundthat has been shown(e.g., through serum albumin (BSA; Sigma). Surrounding cumuluscells are animal trials or prior experience with administration to removed with hyaluronidase (1 mg/ml). Pronuclear embryos humans)to be effective in such treatment. In other words, a are then washed and placed in Earle’s balancedsalt solution known therapeutic compoundis not limited to a compound containing 0.5% BSA (EBSS) in a 37.5° C. incubator with a efficacious in the treatmentofheart failure. humidified atmosphere at 5% CO,, 95% air until the time of As used herein, the term “cardiac hypertrophy”refers to injection. Embryos can be implanted at the two-cell stage. the process in which adult cardiac myocytes respondto stress Randomly cycling adult female miceare paired with vasec- through hypertrophic growth. Such growth is characterized tomized males. C57BL/6 or Swiss mice or other comparable by cell size increases without cell division, assembling of strains can be used for this purpose. Recipient females are additional sarcomeres within the cell to maximize force gen- mated at the same time as donor females. At the time of eration, and an activation of a fetal cardiac gene program. US 9,078,919 B2 37 38 Cardiac hypertrophyis often associated with increased risk of Throughout this application, the term “about” is used to morbidity and mortality, and thus studies aimed at under- indicate that a value includes the standard deviation of error standing the molecular mechanismsof cardiac hypertrophy for the device or method being employed to determine the could have a significant impact on humanhealth. value. As used herein, the term “modulate”refers to a change or The use of the term “or” in the claims is used to mean an alteration in a biological activity. Modulation may be an “and/or” unless explicitly indicated to refer to alternatives increase or a decrease in protein activity, a change in kinase only or the alternatives are mutually exclusive, although the activity, a change in binding characteristics, or any other disclosure supportsa definition that refers to only alternatives changein the biological, functional, or immunological prop- and “and/or,” erties associated with the activity of a protein or other struc- 10 Asused in this specification and claim(s), the words “com- ture of interest. The term “modulator” refers to any molecule prising” (and any form ofcomprising, suchas “comprise” and or compound which is capable of changing oraltering bio- “comprises”), “having” (and any form of having, such as logical activity as described above. “have” and “has”’), “including” (and any form of including, The term “B-adrenergic receptor antagonist” refers to a such as “includes” and “include”) or “containing” (and any 15 chemical compound or entity that is capable of blocking, form of containing, such as “contains” and “contain’’) are either partially or completely, the beta (B) type of adrenore- inclusive or open-ended and do not exclude additional, unre- ceptors (1.e., receptors of the adrenergic system that respond cited elements or methodsteps. to catecholamines, especially norepinephrine). Some (-adr- The following examples are included to further illustrate energic receptor antagonists exhibit a degree of specificity for 20 various aspects of the invention. It should be appreciated by one receptor subtype (generally B,); such antagonists are those of skill in the art that the techniques disclosed in the termed “B,-specific adrenergic receptor antagonists” and examples which follow represent techniques and/or compo- “B.,-specific adrenergic receptor antagonists.” The term sitions discovered by the inventors to function well in the B-adrenergic receptor antagonist” refers to chemical com- practice of the invention, and thus can be considered to con- pounds that are selective and non-selective antagonists. 25 stitute preferred modes for its practice. However, those of Examples of f-adrenergic receptor antagonists include, but skill in the art should, in light of the present disclosure, are not limited to, acebutolol, atenolol, butoxamine,carteolol, appreciate that many changes can be made in the specific esmolol, labetolol, metoprolol, nadolol, penbutolol, pro- embodiments which are disclosed andstill obtain a like or panolol, and timolol. The use of derivatives of known (-adr- similar result without departing from the spirit and scope of energic receptor antagonists is encompassed by the methods 30 the invention. of the present invention. Indeed any compound, which func- EXAMPLES tionally behaves as a B-adrenergic receptor antagonist is encompassed by the methods of the present invention. Example 1 The terms “angiotensin-converting enzyme inhibitor” or 35 “ACEinhibitor”refer to a chemical compoundorentity that Regulation of Cardiac Hypertrophy and Heart is capable of inhibiting, either partially or completely, the Failure by Stress-Responsive miRNAs enzyme involved in the conversion of the relatively inactive angiotensin I to the active angiotensinI] in the rennin-angio- In light of their involvement in modulating cellular pheno- tensin system. In addition, the ACE inhibitors concomitantly 40 types, the inventors have hypothesized that miRNAs may inhibit the degradation of bradykinin, which likely signifi- play a role in regulating the response of the heart to cardiac cantly enhances the antihypertensive effect ofthe ACE inhibi- stress, which is knownto result in transcriptional and trans- tors. Examples ofACE inhibitors include, but are not limited lational changes in gene expression. To investigate the poten- to , captopril, enalapril, fosinopril, quiapril and tial involvement of miRNAs in cardiac hypertrophy, a side- ramipril. The use of derivatives of known ACEinhibitors is 45 by-side miRNA microarray analysis was performed in two encompassed by the methodsofthe present invention. Indeed established mouse models of cardiac hypertrophy, using a any compound,whichfunctionally behaves as anACEinhibi- microarray that represented 186 different miRNAs. Mice that tor, is encompassed by the methodsofthe present invention. were subjected to thoracic aortic banding (TAB), which As used herein, the term “genotypes”refers to the actual induces hypertrophy by increasedafterload on the heart (Hill genetic make-up of an organism, while “phenotype”refers to 50 et al., 2000), were compared to sham operated animals. In a physical traits displayed by an individual. In addition, the second model, transgenic mice expressing activated cal- “phenotype” is the result of selective expression of the cineurin (CnA) in the heart, which results in a severe, well- genome(1.e., it is an expression of the cell history and its characterized form of hypertrophy (Molkentin et al., 1998), response to the extracellular environment). Indeed, the were compared to wild-type littermates (FIG. 1A). RNA iso- human genomecontains an estimated 30,000-35,000 genes. 55 lated from hearts ofmice subjected to TAB showedincreased In each cell type, only a small (1.e., 10-15%)fraction ofthese expression of 27 miRNAs compared to sham-operated con- genes are expressed. trols, and CnA. Tg mice showed increased expression of 33 The use of the word “a”or “an” when used in conjunction miRNAs compared with non-transgenic littermate controls, with the term “comprising” in the claims and/or the specifi- of which 21 were up-regulated in both models. Similarly, cation may mean “one,” but it is also consistent with the 60 TAB and CnA-induced hypertrophy were accompanied by meaning of “one or more,” “at least one,” and “one or more reduced expression of 15 and 14 miRNAs,respectively, of than one.” which 7 miRNAs were down-regulated in common (FIG. It is contemplated that any embodiment discussed herein 1B). Northern analysis of expression of these miRNAs and can be implemented with respect to any method or composi- previous microarray analyses (Baradet al., 2004; Sempere et tion of the invention, and vice versa. Furthermore, composi- 65 al., 2004; Shingaraet al., 2005; Babaket al., 2004; Liu etal., tions andkits ofthe invention can be used to achieve methods 2004) indicate that they are expressed in a wide range of of the invention. tissues. US 9,078,919 B2 39 40 Based ontheir relative expression levels, conservation of the miR-195 Tg animals is specifically due to functional human, rat and mouse sequences, and levels of expression effects ofthis miRNA rather than a general non-specific effect during hypertrophy, the inventors focused on 11 up- and 5 resulting from miRNA over-expression. These results indi- down-regulated miRNAs. Northern blot analysis ofRNA iso- cate that increased expression of miR-195 induces hyper- lated from cardiac tissue from WT and CnA Tg animals trophic signaling leading to cardiac failure. Since miR-195 confirmed an increased expression ofmiR-21, miR-23, miR- belongs to a small family of related miRNAs, the miR-15 24, miR-125b, miR-195, miR-199a, and miR-214, and family, other family membersare also likely to participate in cardiac disease. decreased expression of miR-29c, miR-93, miR-150 and The ability of miR-195 to promote cardiac growth con- miR-181b (FIG. 1C). Collectively, these data indicate that trasts with that of miR-1, a muscle-specific miRNA that distinct miRNAsare regulated during cardiac hypertrophy, 10 inhibits cardiac growth by suppressing the expression of the suggesting the possibility that they might function as modu- bHLHprotein Hand2 (Zhaoet al., 2005). miR-1 is highly lators of this process. expressed in the adult heart, but miR-195 is apparently Ventricular hypertrophy develops in response to numerous capable of over-riding the inhibitory influence of miR-I on forms of cardiac stress and often leads to heart failure in cardiac growth. humans (Arad et al., 2002). Northern blot analysis of the 15 It is especially interesting that overexpression in cardi- hypertrophy-regulated miRNAsin idiopathic end-stagefail- omyocytes of the miRNAsthat were down-regulated during ing human hearts showed increased expression of miR-24, hypertrophy caused an apparent reduction in cell size, an miR-125b, miR-195, miR-199a and miR-214, while the effect opposite than that evoked by the upregulated miRNAs. expression for miR-23 appeared to be variable within the Oneinterpretation of these results is that these miRNAsnor- non-failing and failing groups (FIG. 2). No changein expres- 20 mally function to suppress growth and are therefore down- sion of miR-21, miR-27, miR-29c, miR-93, miR-150 and regulated to enhance hypertrophy. miR-181b was found (data not shown). Thus, the altered miRNA-195 belongs to a small family of microRNAs,the pattern ofmiRNAexpression in the failing human heart over- miR-15 family, which contains miR-195, miR-16-1, miR- lapped with that of the hypertrophic mouseheart, suggesting 15a, miR-15b, miR-16-2, miR-424, and miR-497. Four ofthe miR-15 family members are expressed as three clustered that these miRNAsrepresent a molecular signature ofadverse 25 cardiac remodeling. transcripts (FIG. 6A). Using a variety of bioinformatics approaches, potential mRNAtargets for raiR-195 were iden- Example 2 tified. Several ofthe identified target mRNAs encodeproteins involved in cell proliferation, survival and anti-apoptosis (FIG. 6B). Oneof the predicted targets for the miR-15 family Cardiac Over-Expression of miR-195 is Sufficient to 30 is FGF2, which has been shown to promote cardiac repair Drive Cardiac Hypertrophy (FIG.7). All members of the miR-15 family are up-regulated in failing human hearts, indicating that this family ofmicroR- MiR-24, miR-195 and miR-214 were overexpressed spe- NAsplays a key role in pathological cardiac remodeling cifically in the heart under the control of the a-myosin heavy (FIG.8). Since all the miR-15 family members share a con- chain (MHC)promoter, F1 offspring could not be obtained 35 served seed sequence, all miR-15 family members could be for miR-24, suggesting that cardiac over-expression of this inhibited simultaneously by targeting the seed sequence. One miRNA causes embryoniclethality. Since all offspring of the such approach entails overexpression of a nucleic acid con- miR-195 transgenic (Tg) line 3 died in the first two weeks taining multiple binding sites, which comprise a sequence after birth due to heart failure (FIG. 3), the Tg line 1 for complementary to the seed sequence. The nucleic acid would miR-195, which was viable, was used for further studies. 40 “scavenge” or “sponge” all separate members because of Northern blot analysis showed miR-195 to be expressed at their overlap in seed region and consequently overlap in target levels ~25-fold above normal in Tg line 1 (FIG. 3). Over- sequence (FIG. 8B) (Ebert et al., 2007). expression of miR-195 initially induced cardiac growth with All publications, patents, and patent applications discussed disorganization of cardiomyocytes, which progressed to a and cited herein are incorporated herein by reference in their dilated phenotype by 6 weeks of age. Although there were 45 entireties. All ofthe compositions and methods disclosed and some fibrotic lesions, the dramatic increase in size of indi- claimed herein can be made and executed without undue vidual myocytes in miR-195 Tg mice comparedto wild-type experimentation in light of the present disclosure. While the (WT) was morestriking (FIG.3). compositions and methods of this invention have been Echocardiography on 6 week-old animals showed that described in termsofpreferred embodiments,it will be appar- miR-195 Tg mice displayed thinning of the left ventricular 50 ent to those of skill in the art that variations may be applied to walls (AWs and PWs),an increase in left ventricular diameter the compositions and methods, and in the steps or in the (LVIDd and LVIDs) anda deterioration in cardiac function, as sequence of steps of the methods described herein without indicated by decreased fractional shortening (FIG. 4A). Heart departing from the concept, spirit and scope of the invention. weightto body weight ratios were also dramatically increased Morespecifically, it will be apparentthat certain agents which in miR-195 Tg animals compared to WTlittermates, indicat- 55 are both chemically and physiologically related may be sub- ing that overexpression of miR-195 was sufficient to stimu- stituted for the agents described herein while the same or late cardiac growth (FIG. 4B). Real-time PCR analysis on similar results would be achieved.All such similar substitutes cardiac tissue from miR-195 Tg animals comparedto their and modifications apparent to those skilled in the art are WTlittermates, revealed dramatic up-regulation ofthe hyper- deemed to be within the spirit, scope and concept of the trophic markersatrial natriuretic factor (ANF), b-type natri- 60 invention as defined by the appended claims. uretic protein (BNP) and B-myosin heavy chain (BMHC) in response to cardiac over-expression of miR-195 (FIG. 4C). X. REFERENCES In contrast to the dramatic effects of overexpression of miR-195 on cardiac structure, function, and gene expression, The following references, to the extent that they provide cardiac over-expression of miR-214 at levels comparable to 65 exemplary procedural or other details supplementary to those those of miR-195 had no phenotypic effect (FIGS. 4C and 5, set forth herein, are specifically incorporated herein by refer- and data not shown). Thus, the cardiac remodeling induced in ence. US 9,078,919 B2 41 42 . Pat. No. 4,873,191 Culotta and Hamer, Mol. Cell. Biol., 9:1376, 1989. . Pat. No. 5,604,251 Dandoloet al., J Virology, 47:55-64, 1983. . Pat. No. 5,844,107 DeVilliers et al., Nature, 312(5991):242-246, 1984. . Pat. No. 5,877,302 Deschampsetal., Science, 230:1174-1177, 1985. . Pat. No. 5,972,900 Dubensky et al., Proc. Natl. Acad. Sci. USA, 81:7529-7533, . Pat. No. 5,972,901 1984. . Pat. No. 6,008,336 Durandet al., Ann. Med., 27:311-317, 1995. . Pat. No. 6,077,835 Ebert et al., Nat. Methods, 4:721-726, 2007. . Pat. No. 6,200,801 Edbrookeet al., Mol. Cell. Biol., 9:1908, 1989. . Publn. 20020150626 10 Edgerton and Roy, J. Appl. Physiol., 89:1224-1231, 2000. . Publn, 20030032615 Edlundet al., Science, 230:912-916, 1985. . Publn. 20030203865 Eichhorn and Bristow, Circulation, 94:2285-2296, 1996. USS. Publ. 20040048787 EPO 0273085 Abraham etal., Mol. Med., 8:750-760, 2002. Fechheimer, et al., Proc Natl. Acad. Sci. USA, 84:8463-8467, Ambros, Cell, 113(6):673-676, 2003. 15 1987. Angel et al., Cell, 49:729, 1987b. Feng and Holland, Nature, 334:6178, 1988. Angel et al., Mol. Cell. Biol., 7:2256, 1987a. Ferkolet al., FASEB J., 7:1081-1091, 1993. Aradet al., Hum. Mol. genet., 11:2499-2506, 2002. Firak and Subramanian, Mol. Cell. Biol., 6:3667, 1986. Atchison and Perry, Ce//, 46:253, 1986. Fitts et al. J. Appl. Physiol., 89:823-839, 2000. Atchison and Perry, Ce//, 48:121, 1987. 20 Foecking and Hofstetter. Gene, 45(1):101-105, 1986. Babaket al., RNA 10:1813-1819, 2004. Fraley et al., Proc. Natl. Acad. Sci. USA, 76:3348-3352, 1979. Balchwal and Sugden, In: Gene Transfer, Kucherlapati (Ed.), Franzet al., Cardioscience, 5(4):235-43, 1994. Plenum Press, NY, 117-148, 1986. Friedmanet al., Genes Devel., 3:1314, 1989. Baldwin and Haddad, J. Appl. Physiol., 90:345-357, 2001. Fujita et al., Ced/, 49:357, 1987. Banerji et al., Ce//, 27(2 Pt 1):299-308, 1981. 25 Ghosh and Bachhawat,In: Liver Diseases, Targeted Diagno- Banerji et al., Ce//, 33(3):729-740, 1983. sis and Therapy Using Specific Receptors andLigands, Wu Barad et al., Genome Res. 14:2486-2494, 1997. et al. (Eds.), Marcel Dekker, NY, 87-104, 1991, Barneset al., J. Biol. Chem., 272(17):11510-11.1517, 1997. Ghosh-Choudhury et al., EMBO J, 6:1733-1739, 1987. Benvenisty and Neshif, Proc. Natl. Acad. Sci. USA, 83(24): Gilles et al., Cell, 33:717, 1983. 9551-9555, 1986. 30 Gloss et al., EMBO J,, 6:3735, 1987. Berkhoutet al., Ce//, 59:273-282, 1989. Godboutet al., Mol. Cell. Biol., 8:1169, 1988. Bhaysar et Genomics, 35(1):11-23, 1996. Gomez-Foix et al., J. Biol. Chem., 267:25129-25134, 1992, Blanar et al., EMBO J, 8:1139, 1989. Goodbourn and Maniatis, Proc. Natl. Acad. Sci. USA, Bodine and Ley, EMBO J., 6:2997, 1987. 85:1447, 1988. Boshart et al., Cell, 41:521, 1985. 35 Goodbournet al., Cell, 45:601, 1986, Bosze et al., EMBO J, 5(7):1615-1623, 1986. Gopal, Mol. Cell Biol., 5:1188-1190, 1985. Braddocket al., Ce//, 58:269, 1989. Gopal-Srivastavaet al., J. Mol. Cell. Biol. 15(12):7081-7090, Brenneckeet al., Ce//, 113:25-36, 2003. 1995, Brinster et al., Proc. Natl. Acad. Sci. USA, 82(13):4438-4442, Graham and Prevec, In: Methods in Molecular Biology: Gene 1985. 40 Transfer and Expression Protocol, Murray (Ed.), Humana Bristow, Cardiology, 92:3-6, 1999. Press, Clifton, N.J., 7:109-128, 1991, Bulla and Siddiqui, J. Virol., 62:1437, 1986. Graham and Van Der Eb, Virology, 52:456-467, 1973. Calin et al., Proc. Natl. Acd. Sci. USA, 99:15524-15529, Graham etal., J. Gen. Virl., 36(1):59-74, 1977. 2002. Greeneet al., Immunology Today, 10:272, 1989 Campbell and Villarreal, Mo/. Cell. Biol., 8:1993, 1988. 45 Grishoket al., Cell, 106:23-34, 2001. Campere and Tilghman, Genes and Dev., 3:537, 1989. Grossched] and Baltimore, Ce//, 41:885, 1985. Campoet al., Nature, 303:77, 1983. Grunhaus and Horwitz, Seminar in Virology, 3:237-252, Carrington et al. Science, 301(5630:336-338, 2003. 1992. Celander and Haseitine, J. Virology, 61:269, 1987. Harland and Weintraub, J. Cell Biol., 101(3):1094-1099, Celanderet al., J. Virology, 62:1314, 1988. 50 1985. Chandleret al., Cell, 33:489, 1983. Haslinger and Karin, Proc. Natl. Acad. Sci. USA, 82:8572, Chang and Karin, Nature, 410(6824):37-40, 2001. 1985. Changet al., Biochim. Biophys. Acta, 1092(4153-160, 1991. Hauber and Cullen, J. Virology, 62:673, 1988. Changet al., Mol. Cell. Biol., 9:2153, 1989. Henet al., Nature, 321:249, 1986, Changet al., Nature, 430(7001):785-789, 2004. 55 Henselet al., Lymphokine Res., 8:347, 1989. Chatterjee et al., Proc. Natl. Acad. Sci. USA, 86:9114, 1989. Herinonat and Muzycska, Proc. Natl. Acad, Sci. USA, Chen and Okayama, Mol. Cell. Biol., 7(8):2745-2752, 1987. 81:6466-6470, 1984. Chenet al., Mol. Cell Endocrinol., 162:45-55, 2000. Herr and Clarke, Ce//, 45:461, 1986. Chenetal., Science, 303(5654):83-86, 2004. Hersdorffer et al., DNA Cell Biol., 9:713-723, 1990. Choiet al., Cell, 53:519, 1988. 60 Herz and Gerard, Proc. Natl. Acad. Sci. USA, 90:2812-2816, Coffin, In: Virology, Fields et al. (Eds.), Raven Press, NY, 1993. 1437-1500, 1990. Hill et al., Circulation, 101:2863-2869, 2000. Cohenet al., J. Cell. Physiol., 5:75, 1987, Hirochikaet al., J. Virol., 61:2599, 11987. Costa et al., Mol. Cell. Biol., 8:81, 1988. Hirschet al., Mol. Cell. Biol., 10:1959, 1990. Couchet al., Am. Rev. Resp. Dis., 88:394-403, 1963. 65 Holbrooket al., Virology, 157:211, 1987, Couparet al., Gene, 68:1-10, 1988. Horlick and Benfield, Mol. Cell. Biol., 9:2396, 1989. Cripe et al., EMBO J., 6:3745, 1987. Horwichet al. J. Virol., 64:642-650, 1990. US 9,078,919 B2 43 44 Huanget al., Ce//, 27:245, 1981. Lusky et al., Mo/. Cell. Biol., 3:1108, 1983, Hug et al., Mol., Cell. Biol., 8:3065, 1988. Macejak and Sarnow, Nature, 353:90-94, 1991, Hutvagneret al., PLoS Biol., 2(4):E98, 2004, Majors and Varmus, Proc. Natl. Acad. Sci. USA, 80:5865, Hwanget al., Mol. Cell. Biol., 10:585, 1990. 1983. Imagawaet al., Ce//, 51:251, 1987. Mann etal., Ce//, 33:153-159, 1983, Imbra and Karin, Nature, 323:555, 1986. Mansenet al., Mol. Endocrinol., 15:2106-2114, 2001. Imleret al., Mol. Cell. Biol., 7:2558, 1987. Markowitz et al., J. Virol., 62:1120-1124, 1988, Imperiale and Nevins, Mol. Cell. Biol., 4:875, 1984. McNeall et al., Gene, 76:81, 1989. Ito and Roeder, Trends Endocrinol. Metab., 12:127-134, Meister and Tusehl, Nature, 431:343-9, 2004, 2001. 10 Miksiceket al., Cell, 46:203, 1986. Jakobovits et al., Mol. Cell. Biol., 8:2555, 1988. Molkentin et al., Ce// 93:215-228, 1998. Jameel and Siddiqui, Mo/. Cell. Biol., 6:710, 1986. Mordacq and Linzer, Genes and Dev., 3:760, 1989. Jayneset al., Mol. Cell. Biol., 8:62, 1988. Moreauetal., Nucl. Acids Res., 9:6047, 1981. Johnsonet al., Mol. Cell. Biol., 9:3393, 1989. Morkin, Microsc. Res. Tech., 50:522-531, 2000. Jones and Shenk, Ce//, 13:181-188, 1978. 15 Mossetal., Biol. Chem., 271(49):31688-31694, 19%. Kadesch and Berg, Mol. Cell. Biol., 6:2593, 1986. Muesinget al., Ce//, 48:691, 1987. Kanedaet al., Science, 243:375-378, 1989. Nayaet al., J Biol Chem, 275(7):4545-4548, 2000. Karin et al., Mol. Cell. Biol., 7:606, 1987. Nget al., Nuc. Acids Res., 17:601, 1989, Karlsson et al., EMBO J., 5:2377-2385, 1986. Nicolas and Rubinstein, In: Vectors. A survey of molecular Katinkaet al., Ce//, 20:393, 1980. 20 cloning vectors and their uses, Rodriguez and Denhardt Katinkaet al., Nature, 290:720, 1981. (Eds), Stoneham: Butterworth 494-513, 1988. Kato et al, J. Biol. Chem., 266:3361-3364, 1991. Nicolau and Sene, Biochim. Biophys. Acta, 721:185-190, Kawamotoet al., Mol. Cell. Biol., 8:267, 1988. 1982. Kelly et al., . Cell Biol., 129(2):383-396, 1995. Nicolau et al., Methods Enzymol., 149:157-176, 1987. Kiledjian et al., Mol. Cell. Biol., 8:145, 1988. 25 Ojamaaet al., Endocrinology, 141:2139-2144, 2000. Kimuraet al., Dev. Growth Differ. 39(3):257-265, 1997. Ondeket al., EMBO J, 6:1017, 1987, Kinugawaet al., Circ. Res., 89:591-598, 2001. Ornitz et al., Mol. Cell. Biol., 7:3466, 1987. Kinugawaet al., J. Clin. Endocrinol. Metab., 86:5089-5090, Palmiteret al., Nature, 300:611, 1982. 2001. Pantos et al., Horm. Metab. Res., 38:308-313, 2006. Kiriazis and Kranias, Annu. Rev. Physiol., 62:321-351, 2000, 30 Park et al., Mol. Cell., 19:643-653, 2005. Kiamutet al., Mol. Cell. Biol., 10:193, 1990. Paskindet al., Virology, 67:242-248, 1975. Klein et al., Nature, 327:70-73, 1987. Pasquinelli and Ruvkun, Ann. Rev. Cell Dev. Biol., 18:495- Kochet al., Mol. Cell. Biol., 9:303, 1989. 513, 2002, Kreket al., Nat. Genet., 37:495-500, 2005. Pavri et al., Mol. Cell., 18:83-96, 2005. Krenz and Robbins, J. Am. Coll. Cardiol. 44:2390-2397, 35 PCT Appin. WO 0071096 2004. PCT Appin. WO 84/03564 Kriegler and Botchan, In: Eukcuvotic Viral Vectors, Gluzman PCT Appln. WO 98/33791 (Ed.), Cold Spring Harbor: Cold Spring Harbor Labora- Pechet al., Mol. Cell. Biol., 9:396, 1989, tory, NY, 1982. Pelletier and Sonenberg, Nature, 334(6180):320-325, 1988. Kriegler et al., Ce//, 38:483, 1984. 40 Perales et al., Proc. Natl. Acad. Sci, USA, 91:4086-4090, Kriegler et al., Ce//, 53:45, 1988. 1994. Kriegler and Botchan, Mol. Cell. Biol., 3:325, 1983a. Perez-Stable and Constantini, Mo/. Cell. Biol., 10:1116, Kriegler et al., In: Gene Expression, Alan Liss (Ed.), Hamer 11990. and Rosenberg, New York, 198b. Physicians Desk Reference Kriitzfeldt et al., Nature, 438:685-689, 2005. 45 Picard and Schaffner, Nature, 307:83, 1984. Kuhl et al., Ce//, 50:1057, 1987. Pinkert et al., Genes and Dev., 1:268, 1987. Kunz et al., Nucl. Acids Res., 17:1121, 1989. Ponta et al., Proc. Natl. Acad. Sci. USA, 82:1020, 1985. Lagos-Quintanaet al., Science, 294(5543):853-858, 2001. Porton et al., Mol. Cell. Biol., 10:1076, 1990. LaPointeet al., Hypertension 273 Pt 2):715-22, 1996. Potter et al., Proc. Natl. Acad. Sci. USA, 81:7161-7165, 1984. LaPointeet al., J. Biol, Chem., 263(19):9075-8, 1988. 50 Queen and Baltimore, Ce//, 35:741, 1983. Larsenet al., Proc Natl. Acad. Sci. USA., 83:8283, 1986. Quinn et al., Mol. Cell. Biol., 9:4713, 1989, Laspiaet al., Cel/, 59:283, 1989. Racheret al., Biotechnology Techniques, 9:169-174, 1995. Latimeret al., Mol. Cell. Biol., 10:760, 1990. Ragotet al., Nature, 361:647-650, 1993. Lauet al., Science, 294(5543):858-862, 2001. Redondoet al., Science, 247:1225, 1990, Le Gal La Salle et al., Science, 259:988-990, 1993. Reisman and Rotter, Mol. Cell. Biol., 9:3571, 1989. Lee and Ambros, Science, 294(5543):862-864, 2001. Remington’s Pharmaceutical Sciences, 15” ed., pages 1035- Lee et al., Nature, 294:228, 1981. 1038 and 1570-1580, Mack Publishing Company, Easton, Lee et al., Nucleic Acids Res., 12:4191-206, 1984. Pa., 1980. Leunget al., Proc. Natl. Acad. Sci. USA, 48:18125-18130, Reran, Radiother. Oncol., 19:197-218, 1990. 2006. 60 ResendezJr. et al., Mol. Cell. Biol., 8:4579, 1988. Levinsonet al., Nature, 295:79, 1982. Rich et al., Hum. Gene Ther. 4:461-476, 1993. Levrero et al., Gene, 101:195-202, 1991. Ridgeway, In: Vectors: A Survey ofMolecular Cloning Vec- Lin et al., Mol. Cell. Biol., 10:850, 1990. tors and Their Uses, Rodriguez et al. (Eds.), Stoneham: Liu et al., Proc Natl Acad Sci USA 101:9740-9744, 2004. Butterworth, 467-492, 1988. Luria et al., EMBO J, 6:3307, 1987. 65 Ripe et al., Mol. Cell. Biol., 9:2224, 1989. Lusky and Botchan, Proc. Natl. Acad. Set. USA, 83:3609, Rippe, et al., Mol. Cell. Biol., 10:689-695, 1990. 1986. Riffling et al., Nuc. Acids Res., 17:1619, 1989. US 9,078,919 B2 45 46 Rosenet al., Ce//, 41:813, 1988. Thiesenet al., J. Virology, 62:614, 1988. Rosenfeldet al., Science, 252:431-434, 1991. Top et al., J. Infect. Dis., 124:155-160, 1971. Rosenfeld, et al., Ce//, 68:143-155, 1992. Treisman, Cell, 46(4):567-174, 1986 Rouxet al., Proc. Natl. Acad. Sci. USA, 86:9079-9083, 1989. Troneheet al., Mol. Biol. Med., 7:173, 1990. Sakai et al., Genes and Dev., 2:1144, 1988. Troncheet al., Mol. Cell. Biol., 9(11):4759-4766, 1989. Sambrook and Russell, Molecular Cloning: A Laboratory Trudel and Constantini, Genes and Dev., 6:954, 1987. Manual 3’ Ed., Cold Spring Harbor Laboratory Press, Tsika et al., Am. J. Physiol. Cell Physiol., 283:C1761-C1775, 2001. 2002. Satake et al., J. Virology, 62:970, 1988. Tur-Kaspaet al., Mol. Cell Biol., 6:716-718, 1986. Schaffneret al., 4 Mol. Biol., 201:81, 1988. 10 Tvndell et al., Nuc. Acids. Res., 9:62.31, 1981. Schuyler and Yarbrough, Basic Res. Cardiol., 85:481-494, Vadaszovaet al., Physiol. Res. 53(1):S57-61, 2004. 1990. van Rooij et al., Proc. Natl. Acad. Sci. USA, 103(48):18255- Searle et al., Mol. Cell. Biol., 5:1480, 1985. 18260, 2006. Sempereet al., GenomeBiol 5:R13, 2004. Vannice and Levinson, J. Virology, 62:1305, 1988. Sharp and Marciniak, Ce//, 59:229, 1989. 15 Varmuset al., Ce//, 25:23-36, 1981. Shaul and Ben-Levy, EMBO J., 6:1913, 1987. Vasseuret al. Proc Natl. Acad. Sci. USA, 77:1068, 1980. Sherman etal., Mol. Cell. Biol., 9:50, 1989. Wagneret al., Proc. Natl. Acad. Set. USA 87(9):3410-3414, Shingaraet al., RNA 11:1461-1470, 2005. 1990. Sleigh and Lockett, J EMBO, 4:3831, 1985. Wang, and Calame, Ce//, 47:241, 1986. Spalholz et al., Ce//, 42:183, 1985. 20 Weberet al., Cell, 36:983, 1984. Spandau and Lee, J. Virology, 62:427, 1988. Weiet al., J. Endocrinol. Invest., 28:8-11, 2005. Spandidos and Wilkie, EMBO J., 2:1193, 1983. Weinbergeret al. Mol. Cell. Biol., 8:988, 1984. Stephens and Hentschel, Biochem. J, 248:1, 1987. Winoto and Baltimore, Cel/, 59:649, 1989. Stratford-Perricaudet and Perricaudet, In: Human Gene Wonget al., Gene, 10:87-94, 1980. Transfer, Eds, Cohen-Haguenauer and Boiron, John 25 Wuand Wu, Adv. Drug Delivery Rev., 12:159-167, 1993. Libbey Eurotext, France, 51-61, 1991. Wuand Wu,Biochemistry, 27: 887-892, 1988. Stratford-Perricaudet et al., Hum. Gene. Ther, 1:241-256, Wu and Wu, / Biol. Chem., 262:4429-4432, 1987. 1990. Xu et al., Curr, Biol., 13:790-795, 2003. Stuart et al., Nature, 317:828, 1985. Yamauchi-Takihara, et. al., Proc. Nall. Acad. Sci. USA, Sullivan and Peterlin, Mol. Cell. Biol., 7:3315, 1987. 30 86(10):3504-3508, 1989. Swartzendruber and Lehman, / Cell. Physiology, 85:179, Yang and Russell, Proc. Natl. Acad. Sci. USA, 87:4144-4148, 1975. 1990. Takebe et al., Mol. Cell. Biol., 8:466, 1988. Yao and Eghbali, Circ. Res. 71:831-839, 1992. Tavernieret al., Nature, 301:634, 1983. Younget al., In: Handbook ofApplied Therapeutics, 7.1-7.12 Taylor and Kingston, Mol. Cell. Biol., 10:165, 1990a. 35 and 9.1-9.10, 1989. Taylor and Kingston, Mol. Cell. Biol., 10:176, 1990b. Yutzey et al., Mol. Cell. Biol., 9:1397, 1989. Taylor et al., J. Biol. Chem., 264:15160, 1989. Zelnin et al., FEBS Lett., 287(1-2):118-120, 1991. Temin, In: Gene Transfer, Kucherlapati (Ed.), NY, Plenum Zenget al., Cancer Res., 62(13):3630-3635, 2002. Press, 149-188, 1986. Zhaoet al., Nature, 436:214-220, 2005. The Merck Index, Eleventh Edition Ziober and Kramer, J. Bio. Chem., 271(37):22915-22, 1996.

SEQUENCE LISTING

<160> NUMBER OF SEQ ID NOS: 21

<210> SEQ ID NO 1 <211> LENGTH: 87 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 1

agcuucccug gcucuagcag cacagaaaua uuggcacagg gaagegaguc ugecaauauu 60

ggcugugeug cuccaggcag gguggug 87

<210> SEQ ID NO 2 <211> LENGTH: 112 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 2

ccaceceggu ccugcucceg ceccagcage acacuguggu uuguacggca cuguggecac 60

guccaaacca cacuguggug uuagagegag ggugggggag geaccgecga gg 112

<210> SEQ ID NO 3 US 9,078,919 B2 47 48 -continued

<211> LENGTH: 89 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 3 gucageagug ccuuagcage acguaaauau uggcguuaag auucuaaaau uaucuccagu 60 auuaacugug cugcugaagu aagguugac 89

<210> SEQ ID NO 4 <211> LENGTH: 81 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 4 guuccacucu agcagcacgu aaauauugge guagugaaau auauauuaaa caccaauauu 60 acugugcuge uuuaguguga ¢c 81

<210> SEQ ID NO 5 <211> LENGTH: 83 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 5 ccuuggagua aaguagcage acauaauggu uuguggauuu ugaaaaggug caggcecauau 60 ugugcugecu caaaaauaca agg 83

<210> SEQ ID NO 6 <211> LENGTH: 98 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 6 uugaggcecuu aaaguacugu agcagcacau caugguuuac augcuacagu caagaugcega 60 aucauuauuu gcugcucuag aaauuuaagg aaauucau 98

<210> SEQ ID NO 7 <211> LENGTH: 21 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 7 uageagcaca gaaauauugg c 21

<210> SEQ ID NO 8 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 8 ccaauauugg cugugcugcu cc 22

<210> SEQ ID NO 9 <211> LENGTH: 21 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 9 cagcagcaca cugugguuug u 21

<210> SEQ ID NO 10 <211> LENGTH: 22 US 9,078,919 B2 49 50 -continued

<212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 10 caaaccacac ugugguguua ga 22

<210> SEQ ID NO 11 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 11 uageagcacg uaaauauugg cg 22

<210> SEQ ID NO 12 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 12 ccaguauuaa cugugcugcu ga 22

<210> SEQ ID NO 13 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 13 ccaauauuac ugugcugeuu ua 22

<210> SEQ ID NO 14 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 14 uageagcaca uaaugguuug ug 22

<210> SEQ ID NO 15 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 15 caggccauau ugugcugecu ca 22

<210> SEQ ID NO 16 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 16 uageagcaca ucaugguuua ca 22

<210> SEQ ID NO 17 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 17

egaaucauua uuugcugcuc ua 22

<210> SEQ ID NO 18 US 9,078,919 B2 51 52 -continued

<211> LENGTH: 8 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 18 agcagcac

<210> SEQ ID NO 19 <211> LENGTH: 98 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 19 cgaggggaua cagcagcaau ucauguuuug aaguguucua aaugguucaa aacgugagge 60 gceugcuauac ccccucgugg ggaagguaga aggugggg 98

<210> SEQ ID NO 20 <211> LENGTH: 22 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 20 cagcagcaau ucauguuuug aa 22

<210> SEQ ID NO 21 <211> LENGTH: 21 <212> TYPE: RNA <213> ORGANISM: Homo sapiens

<400> SEQUENCE: 21 caaaacguga ggcgcugcua u 21

Whatis claimed is: 11. The method of claim 9, wherein the backbone modifi- 1. A methodof treating pathologic cardiac hypertrophy, cation is a phosphorothioate linkage. heart failure, or myocardial infarction in a subject in need 12. The method of claim 8, wherein said chemical modifi- thereofcomprising administering to said subject an antisense cation is a locked nucleicacid. oligonucleotide comprising a sequence that is at least par- 13. The method of claim 1, wherein said improved symp- tially complementary to a miR-15b sequence, wherein one or toms include increased exercise capacity, increased cardiac more symptomsofpathologic cardiac hypertrophy, heart fail- ejection volume, decreasedleft ventricular end diastolic pres- ure, or myocardial infarction is improved in the subject fol- sure, decreased pulmonary capillary wedge pressure, lowing administration of the antisense oligonucleotide. increased cardiac output, increased cardiac index, lowered 2. The method of claim 1, wherein the antisense oligo- 45 pulmonary artery pressures, decreased left ventricular end nucleotide comprises a sequence that is at least partially systolic and diastolic dimensions, decreased left and right complementary to the sequence of SEQ ID NO:16. ventricular wall stress, decreased wall tension, and reduction 3. The method of claim 2, wherein the antisense oligo- nucleotide comprises a sequencethat is fully complementary in infarct size. 14. The method of claim 1, wherein the antisense oligo- to the sequence of SEQ ID NO: 16. 50 nucleotide is administered by parenteral, oral, subcutaneous, 4. The method of claim 1, wherein the antisense oligo- intravenous, intramuscular, intradermal, intraperitoneal, nucleotide comprises a sequence that is at least partially transdermal, sustained release, controlled release, delayed complementary to the sequence of SEQ ID NO:6. 5. The method of claim 2, wherein the antisense oligo- release, suppository, catheter, or sublingual administration or direct injection into cardiac tissue. nucleotide comprises a sequencethat is fully complementary 55 to the sequence of SEQ ID NO:6. 15. The method of claim 1, further comprising administer- ing to the subject a second therapy. 6. The method of claim 1, wherein said antisense oligo- 16. The method ofclaim 15, wherein said second therapyis nucleotide is about 19 to about 25 nucleotides in length. 7. The method of claim 1, wherein said antisense oligo- selected from the group consisting of a beta blocker, an iono- trope, a diuretic, ACE-I, All antagonist, BNP, a Ca**-blocker, nucleotide is about 15 nucleotides in length. 60 8. The method of claim 1, wherein the antisense oligo- an endothelin receptor antagonist, and an HDAC inhibitor. 17. The method of claim 1, wherein the antisense oligo- nucleotide has at least one chemical modification. 9. The method of claim 8, wherein said chemical modifi- nucleotide is expressed from a vector under the control of a cation is a sugar modification or a backbone modification. promoter. 18. The method of claim 17, wherein the promoter is a 10. The method ofclaim 9, wherein the sugar modification 65 is a 2'-O-alkyl, 2'-O-methyl, 2'-O-methoxyethyl, 2'-fluoro, or cardiac-specific promoter. 4'thio modification. *****