CA2761248A1 - Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family - Google Patents

Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family Download PDF

Info

Publication number
CA2761248A1
CA2761248A1 CA2761248A CA2761248A CA2761248A1 CA 2761248 A1 CA2761248 A1 CA 2761248A1 CA 2761248 A CA2761248 A CA 2761248A CA 2761248 A CA2761248 A CA 2761248A CA 2761248 A1 CA2761248 A1 CA 2761248A1
Authority
CA
Canada
Prior art keywords
oligonucleotide
nucleic acid
expression
polynucleotide
antisense
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
CA2761248A
Other languages
French (fr)
Other versions
CA2761248C (en
Inventor
Joseph Collard
Olga Khorkova Sherman
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Curna Inc
Original Assignee
Opko Curna LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Opko Curna LLC filed Critical Opko Curna LLC
Priority to CA3185821A priority Critical patent/CA3185821A1/en
Publication of CA2761248A1 publication Critical patent/CA2761248A1/en
Application granted granted Critical
Publication of CA2761248C publication Critical patent/CA2761248C/en
Active legal-status Critical Current
Anticipated expiration legal-status Critical

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • A61K31/713Double-stranded nucleic acids or oligonucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • A61K31/7088Compounds having three or more nucleosides or nucleotides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P21/00Drugs for disorders of the muscular or neuromuscular system
    • A61P21/04Drugs for disorders of the muscular or neuromuscular system for myasthenia gravis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/28Drugs for disorders of the nervous system for treating neurodegenerative disorders of the central nervous system, e.g. nootropic agents, cognition enhancers, drugs for treating Alzheimer's disease or other forms of dementia
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/02Ophthalmic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P27/00Drugs for disorders of the senses
    • A61P27/16Otologicals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/04Inotropic agents, i.e. stimulants of cardiac contraction; Drugs for heart failure
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P9/00Drugs for disorders of the cardiovascular system
    • A61P9/10Drugs for disorders of the cardiovascular system for treating ischaemic or atherosclerotic diseases, e.g. antianginal drugs, coronary vasodilators, drugs for myocardial infarction, retinopathy, cerebrovascula insufficiency, renal arteriosclerosis
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N15/00Mutation or genetic engineering; DNA or RNA concerning genetic engineering, vectors, e.g. plasmids, or their isolation, preparation or purification; Use of hosts therefor
    • C12N15/09Recombinant DNA-technology
    • C12N15/11DNA or RNA fragments; Modified forms thereof; Non-coding nucleic acids having a biological activity
    • C12N15/113Non-coding nucleic acids modulating the expression of genes, e.g. antisense oligonucleotides; Antisense DNA or RNA; Triplex- forming oligonucleotides; Catalytic nucleic acids, e.g. ribozymes; Nucleic acids used in co-suppression or gene silencing
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • CCHEMISTRY; METALLURGY
    • C12BIOCHEMISTRY; BEER; SPIRITS; WINE; VINEGAR; MICROBIOLOGY; ENZYMOLOGY; MUTATION OR GENETIC ENGINEERING
    • C12NMICROORGANISMS OR ENZYMES; COMPOSITIONS THEREOF; PROPAGATING, PRESERVING, OR MAINTAINING MICROORGANISMS; MUTATION OR GENETIC ENGINEERING; CULTURE MEDIA
    • C12N2310/00Structure or type of the nucleic acid
    • C12N2310/10Type of nucleic acid
    • C12N2310/11Antisense
    • C12N2310/113Antisense targeting other non-coding nucleic acids, e.g. antagomirs
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y10TECHNICAL SUBJECTS COVERED BY FORMER USPC
    • Y10TTECHNICAL SUBJECTS COVERED BY FORMER US CLASSIFICATION
    • Y10T436/00Chemistry: analytical and immunological testing
    • Y10T436/14Heterocyclic carbon compound [i.e., O, S, N, Se, Te, as only ring hetero atom]
    • Y10T436/142222Hetero-O [e.g., ascorbic acid, etc.]
    • Y10T436/143333Saccharide [e.g., DNA, etc.]

Abstract

The present invention relates to antisense oligonucleotides that modulate the expression of and/or function of Dystrophin family, in particular, by targeting natural antisense polynucleotides of Dystrophin family. The invention also relates to the identification of these antisense oligonucleotides and their use in treating diseases and disorders associated with the expression of DMD family.

Description

Ti'RE.NTME`'T OF Ã1 YST.ROP.H.I.\ FAII1L ` RELATED DISEASES BY INHIBITION OF
NATURAL
ANTISENSE TRANSCRIPT TO QUIT) FAMILY
FIELD OF THE INVENTION
[0001) The present application claims the priority of U.S. provisional patent application No. 61/176,5,94 filed 8.
2009 and U.S. provisional patent application No. 61/317,350 lied. M arch 25, 2010 both of which are incorporated herein by reference in its entirety.
[0002] Embodiments of the invention comprise oligonucl:e3tides modulating expression and/or function of i3M1J
t :mild, and associated molecules.
BACKGROUND
[0003] DNA-RNA and RNA RNA hybridization are important to many aspects of nucleic acid function including DNA replication, transcription, and, translation. Hybridization is also central to a variety of technologies that either-detect a particular nucleic acid or alter its expression. Anti:sense nucleotides, for example, disrupt gene expression by hybridizing to target RNA, thereby interfering with RNA splicing, transcription, translation, and replication.. Antiseaase.
DNA has the added feature that DNA-R.,,\-.'A hybrids serve as a substrate for digestion by ribonuelease I-1., an activity that is present in most cell types. Arttisense molecules can be delivered into cells., as is the case for oligodeoxyntucleotides (ODNs), or they can be cxpressed from endogenous genes as RNA molecules. The FDA, recently approved an antisense drug, \`1TRAV EN.E'r',' (for treatment of cytome alovinis reti:nitis), reflecting that a aartisense has therapeutic unlit .
SUMMARY
[00041 this Summary is provided to present a scum ary of the invention to briefly indicate the nature arad substance of the 1-Mention. It is submitted with the understanding, that it will not be used to interpret or limit the scope or meaning of the claims.
[0005] In one emboditaaent, the invention prop ides methods for inhibiting the action of a natural antiisense transcript by using antisense oligonucleotid.e(s) targeted to any region of the natural int.
sense transcript resulting ill up-r'egulaÃioll of the corresponding sense gxene. It is also Contemplated herein that inhibition of the natural .anÃise nse transcript can be achieved. by siRNA, ribozymes and small molecules. which are considered to be within the scope of the present im carrion.
[0006] One embodiment provides a method of modul atin4g function atrd/or expression of an f3.MD taramily laolyarrreleotide in patient cells or tissues in vi-o or in vitro comprising contacting ? said. cells or tissues with an aanti.sense oligonuc cotide 5 to 30 nucleotides in length wherein said of gonui leotide has at least 5U?' sequence identity to a reverse complement of a polyraucleotide comprising 5 to 30 consecutive nucleotides within .nucleotides I to 378 of SEQ ID NO-3, 1 to 294 of SEQ ID NO, 4. 1 to 686 of SEQ ID NO,, 5, 1 to 4811 of SEQ ID NO: 6 and 1 to 501 of SEQ
I

I) \() 7 (1' figure 3) thereby modulating function aaad..or expression of the I)M.D fa m.ilk poi yn.ucleorl c in patent cells or tissues in vivo of in vitty.
[0007] In another preferred embodiment, an oli<gonucleot de targets a natural antisense sequence of UNID faarnily I?oly<nucleotides, for example, racleotides set forth in SEQ I) NO: to 7 and any varri.artts. alleles, ltorrtolo?s rnutants, derivatives fragments and co pleme nary sequences thereto. Examples of antiscnse oligonueleotides are set fol-th as S Q ID NOS: 8 to 22 (Figure 4 and 5).
[0008] Another embodiment provides a method of modulating function and/or expression of an DM) f amil .
polynuckotidc in patient cell- or tissues in vivo or in. vitro? comprising contacting said cells or tissues with an aratisetrse oligorucleotide S to 30 nucleotides in lerau h Wherein said olinonucleotide has at least 50% sequence identity to a reverse complement of the an antiscuse of the DMD fanidy polynucieotide;
thereby modulating function and/or expression of the DMD family polynrucleotide in Patient cells or tissues in Vivo or in -vitro.
[0009] Another embodiment provides a method of modulating function and/or expression cif an DMD family poly<nuclcotidc in patient cells or tissues in vivo or in vitro comprising contacting said cells or tissues with an. aartise rse.
?ligc?i?ucleoude 5 to 30 nucleotides in length wherein said of gonucleotide has at least 50% sequence identity to an antisetase oligonucleotide to xi. DMD family antisense pol na.aekt de, thereby modulating function and or express on of the DMD f oily poly, nucleotide in patient cells or tissues in v vo or itr vitro.
[0010] In a preferred embodiment, a composition comprises one or more autisense oli4gor?ucleotides which bind to sense and,or antisense DN-f family polynucleotides.
[0011] In another preferred embodiment, the oligonucleotides con-aprise one or more modified or substituted nucleotides [0012] In anotlterpref reed enth<?tiinaent, tl?t taliE onricleotides coaril7rise t a?e c?r nat?re modif ed hot?rls., [0013] In ,yet another eirrhodin?ea?t, the modified nucleotides comprise modified bases comprising phosphorot ioate, rarethy`lltl?crsl?hi?nxrte, peptide nucleic acids. 2'-O-methyl, Iluoro or carbon, methylene or other locked nucleic acid (LNA) molecules. Preferably, the modified nucleotides ate locked nucleic acid molecules, Including u.-LLN'A.
[0014] In another preferred ernlx?diment, the of gonueleotides are athniniistered to a patient subcutaneously, inttramuscularly>, intravenously or in. traaper itoneally.
[0015] In another preferred errtlaodimerit, the oli orr:aacleotides are administered in a pharmaceutical composition. A
treatment regimen comprises administering the anti-sense compounds at. least once to patient however, this treatment can be modified to include multiple doses over a period. of time. The treatment can be combined, with one or more other (-Ypcs of therapies.
[00 16] in another preferred embodi.a?gent, the oligonuclcotides are encapsulated in a liposome or attached to a carrier molecule tc.g. cholesterol l":'T' pci~t del,.
[001.7] Other aspects are described infix.

BRIEF DESCRIPTION OF THE DRAWfNGS
[0018] Figure 1:
Figure IA is a graph of real tine 1'(:'1:_ rcsults showing the t;_sld change standard deviation i DMD firmly mRN:
after treatment of 5 1 S 12 cells with phosphoroth oate oiigonucleotides :introduced using 1.ipofi ct< mine 2000, as compared to control. Real time PC'R a esul.ts show that the levels of DMD f mil mRNA in 51 SA.2 cells are significantly increased 48 h after treatment with two of the siR As designed to DMD family antisense B G108074.
Bars denoted as CUR-0636 to CUR-0654, correspond to samples treated with SEQ
ID NOS: 8 to 1 7 respectively.
l figure 113 is a grapl of teal time PCR results showing the fold change +
standard deviation. in IYNIt) fthni.ty to R- A
after treatment of 518A2 cells with phoslihc a tlaioatc. csli4?onaaeleotid.es introduced using Lipofectamine 2000, cis compared to control. Treatment with siRN`As to other antiserase molecules. BF$
3856.1, BF768753 and BF95064 3, did not elevate DMD family mRNA 1cyels, Bars denoted as CUR-0638, CUR-064$, CUR-0W4,-,6 and CUR-065' correspond to samples treated with SEQ ID NOS:: , 14, 13 and 16 respectively.
Figure IC is a -graph of real time TPC.R results shot ii:ng the fold change +
standard deviation in URN mRNA after treatment of MCF 7 cells with phosphorothioate oligonuelcotides introduced using Lipofe ctanxine -2000, as compared to control. Bars denoted as. CUR-1443 to CUR-1 7 correspond to samples treated with SEQ ID NOS: 18 to ' respectia~ ely.
[0019] Figure 2 shows ST:Q fD NO: 1: 1-lon:ac sapiens Dystrophin fiam ly, transcript variant Dp -27an, mR`tiA (INCBI Accession No.:
NM 004CK)6 }
2 SEQ ID NO: 2: Homo sapiens Utrophin (UTRN), a a:mRNA. (, CBI Accession No.:
NM 007124) [0020] Figure 3 shows SEQ ID NO: 3: Natural D VMD fan-illy antis nse sequence (BF838561) SEQ ID NO: 4: Natural DMD Ihmily antisense sequence (BO20l 074) SEQ ID NO: 5: Natural DMD lamfly antisense sequence (.13F950$43 ) SEQ ID NO: 6: Natural DM1) fbm lye afnisense sequence (B 768753 ) SEQ ID NO: 7-. Natural I TRN antisense sequence (..ENST0 0000431.309)..
[0021] Figure 4 shows DMD antisense oligonucleotides, SEQ ID NOs: 8 to 17, `a`
indicates RNA, [0022] Figure 5 shows the URN antisense oligo:nucteotides, SEQ ID NOs: 1.8 to 22- * indicates phosphotla:ioate bond, [0023] Figure 6 shows the D1t1:D sense oligonuckotides, SEQ ID Os: 23 to 321 The sense oligotmcleot.ides SEC) ID
NO: 23 to 32 are the reverse complements of the antiscnse oligonueleotides SEQ
ID NO: 8 to 17 reslpect >ely. 'r' indicates RNA.
DETAILED DESCRIPTION

[00214] Several aspects of the invention are describe l below with reference to exarripte applications for Illustration, It should be understood that ,:au e_rous s_pecitac dclails, relationships, and methods are set furth to provide a full understanding of the invention. one having ordinary skill in the relevant art, howy"cver:-, will read! recognize that the ir: ==entiori can be practiced without one or more of the specific details or with other methods. The present invention, is not l irarited. by the orderinL, of acts or events. as some acts niay occur in different orders andltrr concurrently with. other acts or events. 1 urthea.naore. not all iltusuaatecl acts or events are required to irralale#racnt a naetl e?clcalos in acco rdance with the present invention.
[0025] All genes gene names. and gene products disclosed herein are intended to correspond to homologs fron) any species for which the Compositions and methods disclosed herein are applicable. 'T'hus, the teimis Include, but are r1ot limited to Herres and ene products fioaxr humans and mice. It is understood that when a gene or gene product from a particular species is disclose :i, this disclosure is intended to be exemplary only, and is not to be interpreted as a limitation unless the context in which it appears clearly indicates. Thus, for example, for the genes disclosed herein, which in some embodiments relate to mars malian nucleic acid and amino acid sequences are intended to encompass homologous and/(?r ortholol ous genes and getic products from other animals including, but not limited to other nr anmials, fish. amphibians, reptiles, and birds. In preferred embodiments, the genes or nucleic acid sequences are human..

[0026] The terminolony used herein is for the purpose of describing particular embodiments only and is not intended to be limiting, of the invention. As used herein, the singular forms a". "aan"
and " t(re" a art; intended to include the plural forms as well, unless the context clearly indicates otherwise. Furthermore, to the. extent that the terms "including".
includes", "havi:ng", "has", "with", or variants thereof are used in either the detailed description and/or the claims, such ,_ .
terms are #.arte;n e to be inclusive in a niaaltrer similar to à ac term ,it~.r3ap3r'rstrr~,."
' [0027] The term "about" or "appruxiaaraately" means within an acceptable error range for the particular value as determined by one of orditraa % skill in the art, which will depend n part on how the value is measured or determined, 2S i.e., the .limitations of the measurement system. For example, "alxaut" can mean within 1. or more than 1. standard deviation, per the practice in the art.. Al.termatively, "about" can, mean a rani x. of tip to 20%, preferably up to .It %, more preferably up to 5%, and more preferably still up to 1% of a given value.
Alternatively. particularly with respect to biological systems or processes, the team Can mean -within an order of magnitude, preferably within 5-fend, and. more preferably within 2-fold, of a vala.e. Where. particular values are described in the application and claims, starless otherwise stated the term "about" meaning within an acceptable error range for the particular ti <~lue shoulel he assn med.
[0028] As used heroin, the terns niRINN-A" means the presently krio yu mRNA
tr:aanscript(s) of a targeted gene. and any firrtlier transcripts which may be elucidate 1.
[0029] By "aantisense olisaonuc eotides" or "antisensc compound" is :meant: an RNA or DNA molecule that binds to another RNA or DNA (taarnet RNA, DNA), For example, if it is an RNA
oligonucleotidc it binds to another R" A targct by means of RNA.-RNA inteaactions and alter. the activity of the target RNA (1 guchi cacti'., (U991) Ann. Ii . . Bt(xhem.
60, 631-652). An antisense obigon.uelcotide can upregtdate or dotiwnregulaite expression and/or function Ofa paarliculaar po yaauclcotfde. The definition is meant to include ally foreign RNA or DNA
molecule which is asefii f =on1 a therapeutic, diagnostic, or other vieiq)oint. Such molecules include, fbr example, aritiscnse RNA or DNA. molecules, interfere..nce RNA (RNA]), micro RNA, decoy RNA mole ;ales, siRl A, enzymatic RNA, therapeutic editing, RNA and agonist and antagonist RNA, aautisense oligomeric compounds, anÃiseanse o11gottucleoÃides. external guide sequence (EGS) oligonucleotides, alternate splicers, primers, Probes, and other oligomeric compounds that hybridize to at least a portion of the tar et nucleic acid. As such, these compounds may be introduced in the form of single-stranded, double-stranded, partially sinWe-stranded, or circular olio meric compounds.
[00301 In the context of this invention, the term "oligonaacieotide" refers to in ohgomcr or poly-mer of ribonucleic acid (R_N.A) or deoxyribonucleic acid (DNA) or mi etics thereof. The. term "oligoraucleoti.de", also includes linear or circular olifgomers of natural and/or modified monomers or linkages, includinLg deoxyribonucleosides, ribonucleosides, substituted and alplhaa-araonwric [tams thereof; peptide: nucleic acids (P:NA), locked nucleic acids ff.NA}, phosphorotlaioate, methylphosphoonate, and the like. Olrgon:ucleotides are capable of specifically binding to a target pea ymuclcotide by way of a regular pattern of raacaaaomer-to-raxonc rnrr in:teractioaas, such as W aÃYson-C:ricl Ã_ype of ba e laaairiaa Ho stem or reverse I-lo6gstce:n à lac s of base: paairiraa, or the like.
[0031 ] The olig nuclcoÃidc. may be "changer c", that is, composed of different regions. In the context of this in cntiora "chimeric" compounds are ol'igonucleotides, which contain two or more chemical regions, for example. DNA
re6on(s), .RNA region(s), PNA region(s) etc, Each chemical region is made up of at least one monomer unit, i.e., a nucleotide in the case of all oligormelcotidcs compound. These oligorn;aeleotides typically comprise at least one region wherein the oligonucleotide is modified in order to exhibit one or more desired properties. The desired properties of the oligonueleotid.e include, but are not limited, fbr exaumaple, to increased resistance to nuclease degradation, increased cellular uptake, and ,'or increased binding affinity for the target nucleic acid. Different regions of the oligonucleotide rnaati therefore have different properties. The chimeric oligonucleotides of the present invention can be f6ti-ned as mixed structures of two or more olit onucleotides, modified oligonucleotides, oligonucleosides and/or ol..igontacleotide analogs as described above.
[0032] The oligonucleotide can be composed of regions that can be linked in "register", that is, when the monomers are linked conscc,aa ely, as in .n 1tive. DNA, or linked via spacers. The spacers are intended to constitute a covalent "bridge" between the regions and have in preferred cases a length not exceeding about: 100 carbon. atoms. The spacers may carry different fiaractitaraalitier, for example, lraavirm positive or rrcgat:ive charge, carry special nucleic acid binding S

Properties tfratercalators, ;oc?t,e binder. to: ins, fiuorophorc etc.); being fipophil:ic, inda:tci:ng special sccondarr=
stnictures like, for example, alanirae containing peptides that induce alpha-helices.
[0031] As used hcreif "D 1.1) family". "Dystrophi:n farnil\v" and "dystr phin-related protein family : "d)strophi:n gene farilily" are inclusive of all fanail.y inenabers, mutants, alleles, .fagments, spxecies7 coding and noncoding sequences, sense and andserase polynucl :.otide sÃxanc1s, etc.
[00341 As t_#s;.d herein, the words Dystrophin, DMD, [3MD, (MD3B, L)\S142, DXS164-. D\S2O6, D` 5230.
13XS239, DXS268, DXS269, DXS2 and DXS272 are used Interchangeably in the present application, [0035] As used herein., the words T troph.in, UTRN, DMDL, DRP, DRP 1, Dystrop1i n-related proÃei a 1, FL 1236 8, are used intereiaangeabl in the present application, [0036] As used herein, the. words "d:ystrophin related protein 2", "dy,strophin-related protein 2" and. DRP2 are cased interchangeably in the present application.
[0037) As used herein, the words Dystrobrevin o-,d ~strohrevi.n, f"-dystrohrcvi:n, T3`TNA. and D1 13 are used interchangeably in the present application.
[0038] As used herein, the ter-nn "olipor ueleoti e specific for" or "oligontrcleotide which targets" refers to an ol.igonatcleotide having a sequence (1) capable of R main , a stable complex with a portion of the targeted genre, or (ii) capable of forming a stable duplex with a portion of a naRNA transcript of the targeted gene. Stability of the complexes and duplexes can he determined by theoretical Calculations and1or in vitro assays. Exemplary assays for determining stability of hybridization complexes and duplexes are described in the E
xar:rkples below.
[0039] As used herein, the term ""target nucleic. acid"' encompasses DNS., RNA. comprising preen NA and. in-RNA) transcribed from such DNA, and also eDNA derived from such RNA, coding, noncoding sequences, sense. or aratisense po ynucleotides. The specific hybridization of can oigomeri.c compound with its target nucleic acid interferes z ith the normal function of the nucleic acid. This modulation of function. of a target nucleic acid by commix>.urads, which specifically hybridize to it, is generally" referred to as "ant.isense ". The functions of DNS. to be interfered include, for example, replication and tratxsG:.l ti:'}fr. 1'l e tianctio:ns cif RNA to be interfered, include all vital functions such as, for example, translocation of the RNA to the site of protein translation, translation of protein from t: he RNA, splicing of the RNA to yield one or more raaRN.A species, and catalytic activity which may. be engaged in or facilit'ated by the RNA.
The overall effect of such int<.rfrrcnce with target nucleic acid function is modulation of the expression of an encoded product or oligonucleotides.
[0040] R.NA interference "RNAI" iis mediated by double stranded RNA (dsR.NA) molecules that have sequence-specific homology to their "target" nucleic acid sequences ({'aplen, N.: 1., et a[ (290l) .1'tm.>c_NVw/..<lc ml, Scf. I.fSA.
98:742-9747x. In certain embodiments of the present invention, the mediators are 5-25 nucleotide "small interfering"
RNA duplexes (siRNlAs). The siRNA.s are derived from the processing of dsRNA
by an Rl ase enzyme. known as Dicer (Bernstein, E., of al. (20Ã11) .Ncrtare 4093(i3-366}_ siRNA duplex products are recruited into ;a rn.u ti-protein siRN.A complex ternned RISC (RNA Taaduced Silencing Coanplex). Without wishing, to be bound by any particular theory, a RISC is then believed to be :aided to a target nucleic acid (suitably rRNA , where à e siRNA duplex interacts in a. sequence-specifflc way to nlgediaÃe cleavage in a catalytic fashion (Bernstein, E., cat a/. (2001) / iiure 409:363-366, Boudaa, A. et al. (2001) B//of. 11:170176-1784). Small interfering RNAs that can be used in accordance with the present invention can be synthesized and used according to procedures that are well known. in the art and that will be fan:ailiatr to the ordinarily skilled artisan. SrnAall r.nterl%ring RNAs for use in the methods of the present invention suitably comprise between about I to about 50 nucleotides (nt). In examples of lion 1111111 1"m embodiments, siR.NAs can comprise about 5 to about 40 tit, about 5 to about 30 nt, aboaat 10 to about 30 nt. about 15 to about 25 n t, or about 20-25 nucleeotides.
[0041] Selection of appropriate olilonucieotides is facilitated b us as c::ornputcr ptogruns that auÃonxat.ie all' align nucleic acid sequences and indicate. regions of identity or homology. Such rogranis are. used to compare nucle:.icc acid sequences obtained, for example, by searching databases such as GearBank or by sequencing PCR products.
Comparison of nucleic acid sequences from a ravage of species allows the selection of nucleic acid sequences that display an appropriate degree of identity between species. In the case of ,õe.:nes that have not beat sequenced Southern blots are perfixrmed to allow a detenniatation of the clegr : of identity between genes in. tar fet species and other species.
By perfibbr'nting Southern blots at varying degrees of stringency , as is well. known in the art, it is possible to obtain all approximate measure of identity. These procedures allow the selection of oli caiaaaelccttidcs that exhibit a high degree of complemerttarity to target r ucleic acid sequences in a stibiect to be controlled and a lower degree of conaplen-lentality to corresponding nucleic acid sequences in other species. One skilled in the art will realize that there is considerable latitude in selecting appropriate regions of genes for rase iax the present inventic n, [0042] By "enzymatic -RIBA" is meant an RN.A.:molecule with enzymatic activity (Cech, (I 98) J. Am c:on. Med.
f.~: c}c'. 260, 3030-3035). Enzymatic nucleic acids (ribozy mes) act by first binding to a target RNA. Such binding occurs through the target binding portion of an ertzyrnatic nucleic acid which is held in close pro,aimity to an enzymatic portion of the molecule that acts to cleave the target RNA. Thus, the er-tzyxnatic nucleic acid first recognizes and then binds a target RNA through base pairing, and once bound to the correct site, acts enzyrnta:ticallly to cut the target RNA-[00431 By "decoy RNA" is meant an RNA molecule that mimics the natural.
binding domain for a ligaaand. The decoy RNA therefore competes with natural binding target for the binding of a specific li and. For example, it has been shown that over-expression of HIV tratra-acÃi anion response (DU Z) RNA can act as a "decoy" and efficiently binds HJV tat protein, thereby preventing it from binding to TAR sequences encoded in the HIV RNA (Sullenger ct al.
(1990) Cell, 63, 6Ã11- 608). This is meant to be a specific emmiple. Those in the an will recognize that Ãh is is but one example, and other embodiments can be readily generated using techniques generally known in the art.
[0044] As used herein, the terxrx "rtaorrcraxrcrs" typically indicates monomers linked by phosphodiester bonds or analogs thereof to form oligonuc eotides ranging, in size 'h-ont a few monomeric units, e.., fa ml about 3-4, to about several hundreds of monomeric awaits, Analogs of phosphodicster linkages include:
phosphorothioate,. phiosphoroditl i:oate, methyiphosphornates, phosphoroselenoaate, phosphora.naidate,, and the like, as more fulls dehinbed below, I0045] The term. "nucleotide" covers nataarai y occurring nucicotidcs as well as nonnaturally occurring nucleotides. It should be clear to the person skilled in the art that various nucleotides wh:icla previously h avv e been considered "non-naturally occurring have subsequently been ffsund h nature. Thus, "nucleotides" includes not c raly< the knot n purina and pyrimidinc heterocycles-containing molecules, but also hcterocyclic analogues and tautoalacrs thereof: Illtrstaative examples of other types of nateleotides are mole _ules containing adenine, guanine, thvnnine, cytosine, aaaaa.eil, puriare, \anthi.ne, dianai_atopurine, l;-ono- N6-niethyladenine, 7-deaza_.xanth.inc, 'L
an ti uanme, ."44 ,N4- .thanocvtosiaa. N6,N6-ethano-2,6- diaaninopaaa'ine, 5-aaiethyicy"'tosine, IC _ .Cb)-alkyd lcytosianc, 5411wroliraaeil.. 5 bromou:racil, pseudoisocytosine, 2-iaydrtaxs- _z3astia l_ -trig c~lc~p tidaaa, isc ytosine..
isoguania, inosirae and the. ""nao.n-uatu ally occurring" nucleotide described ia-i Benner et as/.. U.S. Pat No. 5,432,272.
The term "nucleotide" is intended to cover every .rand all of these examples as well as analogues and. tautonaers thereof. Especially interesting nucleotides are those containing adenine, <guanine., th:y male, cytosine" and uraci.l, XvIlic:h are considered as the naturally occurring: ntieleoti:des in relation to therapeutic and diagnostic application in humans. Nucleotides include the natural 2 -deoxy and T-hydroxyl sugars, e,-,, as described in Kornberg and Baker, DNA Replication.
2nd Ed. (Freeman. San Francisco, 1992.) as well as their analo<gs.
[0046] "Analogs" in reference to a:aa .eieodde a includes synthetic nucleotides having modified base moieties and/or modified sugar moieties (see e.g., described generally by Sc heit, Nueleotide Analogs, John Wiley, New York, 1980;
Freier & Altmann, ( 1997) Vac/ Acid Ras., 25(22), 4429- 4443, 'rouhaa , ,J , (2001) N' ,iure Biotechnology 19:17-18-, Manoharan M,. (1999) .1 rc c arc ra.~ et Biaphtsica 4ctcr 1489:117-139;
F'reier S. M., (1997) n /er<. lc cf Resc ~r :fir, 25:442.9-4443, Uhlman, E., C2000) D rig . i seaver,t. d c'velo 3rr?t /rt. 3:
20>-2l a, Herdewin P., (2000) Aratiseosc c Vac/pie ,4cia;/ T)ror T ei' , 10:29 7-;10): 2"-0, 3'-Clinked J3.2.0J
l3icyrcloaarti inonucieosicles (sec c,rs. N,K t hristicn, ecn_ a:,i al, (199) ,1, Am. Chc'ia7 Soc , 12.0: 5458-5463, Prakash 1'1', Bhat .B.
(2007) (:urr 1'q) ed Chem, 7(7):64.1-9; Cho 1-=.,1, et at ('009) Annual Review at Aualviieal C hcmnhvi'v, 2, 211-264).
Such aalogs include synthetic nucleotides designeal to enhance binding pa-opcrtics, c. g., duplex or triplex stability:, specificitsy, or the like..
[00471 As used herein, "hy>bridizaation" means the pairing of substantially complementary strands of olio one ric ccanapoaands. One mechanism of pairing, involves hydrogen bay:ndi:n , which may be Watson-Crick, Ho gsteen or reversed i-1oOgstccn hydrogen bonding, between complementary nucleoside or nucleotide bases (nuclcotaeles) of the strands of oli ;omeric compounds, For example, adenine. and tlaymine. are complementary nucleotides which pair through the foram anon csfhydrogen bonds. Hybridization can occur under v ary-ing circumstances.
[00481 An antisense compound is "specifically hybridizable" when binding: of the compound to the target nucleic acid interferes .vjth the normal function of the target nucleic acid to cause a modulation of function and/or activity, and à here is a sufficient degree of complements city to avoid non-specific binding of the antiscrase compound to non-target na:aclei:c.

acid sequences under condition in which specific. binding is desired, i.e., under physiological conditions in the case of in vivo assays or therapeutic treatment, and under conditions in wt >hich assays are performed in the case of in vitro assays.
1 004 As used herein, the phrase "stringent h bridization conditions" or "strr:ttgettt conditions" refers to conditions tinder which a compound of the invention will hybridize to its target sequence. but to a ntinintal number of other sequences, Stringent conditions are sequence-dependent and will be different in different: circumstances and in the Context of this invention, stringent conditions" under which oligontcric co iipom ds hybridize to a target sequence are determined by the nature and composition of the oligcrnerie. compounds and the assays in which they are being investigated , In general, stringent hybridization conditions comprise 1,mv co:ttcentrations (<0 15M) of saalts with inorganic cation such as Na: or K:,--v- (i,c;., low ionic strength), teii paerature higher than 2O"C7 . _ 25' C:, below that. Tnx of the oligomeric conpound:target sequence complex, and the presence of denaturants such as formairiide, di:utethylforraramide., dimethvi sulfoxide, or the detergent sodium dodccyi sulfate. (SDS). For example, the hyrbridixation rate decreases .1.1% fo:r each foranamide. An example of a high stringency hybridization condition is 0. IX sodit.tata chloride-sodium citrate buffer # SSQ/0.1`} t= (w/v) SDS at 60 C. for 30 minutes.
[0050] "Complementary," as used herein, refers to the capacity for pn cise.
pairing betueet. two nucleotides on one or two oligonterie straands. For example, if a nucleobase at a cert.' in position of Ili antisense compound is capable of hydrogen bonding, with a nucleobase at a certain position of a target nucleic acid, said target nucleic acid being a DNA, RNA, or oligonucleotide molecule, then the position of hydrogen bonding between the oligot:aucleoti:d- and the target nucleic acid is considered to be a. complementary position, The oligome ric compound and the further _DNA, RNA. or oligonucl.eotide molecule are complementary to each other when a sufficient atirlaber of complementary positions in each molecule are. occupied by nucleotides which can hydrogen bond. with each other. Thus, "specifically hybrrdixab.le"
and teomplcmertÃar\=" are terms which are used to indicate as sutfi.cierat degree of precise paining or co-tuplen-telltarity over as sufficient number of nucleotides such that stable and specific binding occurs between the oligomeric compound and a fargct nucleic acid.
[0051] It is understood in the art that the sequence of an oli., omeric compound need not be 100% conmpleanentan, to that of its target nucleic acid to be specifically hybridizable. MMloreoowve r, an oliegonucleoÃide may hybridize over one or more segments such that intervening or adjacent segments are not involved in the hybridization event (e.g., :t loop structure, mismatch or hairpin structure-). The ohromeric compounds of the present invention comprise at least about 7t %. or at least about 75%. or at least about 80x'',, or at least about 85%, or at least about 90%, or at least about 95%, or at least about sequence complc:.mentarity to a target region. within the target nucleic acid sequence to which they are targeted. For example, an m o s e n s e compound in which 1 S of 20 nucleotides of the aantiseat.se compound are complementary to a target region., and wvo ld therefore specifically hybridize, would represent 90 percent complementarity. In this example, the rema:iuin noucomplementary nucleotides may be clustered or interspersed with complementary, rtrclcotidcs acid :need not be contiguous to each other or to complkiner t ary nuci:-otid. s. As such. an aarttisa nse compound tvhich is 18 nucleotides in length having 4 (four) noncomplementtuy nucleotides which are flanked by two regions of complete con plementarity yids the tatgct nucleic acid would have 77,8'?=i> overall cotrplerne ntarit with the target nucleic acid and vvould thus fall within the scope of the present i rvention. Percent com_plenrent:arity of an antisense compound with a region of a target nucleic acid can he detenttined routinely using BLAST programs (basic local alignment search tools) and PowerBLA.ST programs linoovn in the art (Aitsohall cat arl., 099Q) J Mral. Biol.< 215,403-410, Zhang and Madden, (1997) ljetxc)trae I cis,, 7, 649-656), Percent horar.olog , sequence identity or contplenteratarity. can be determined by, for example, the Gap prograrnr (Wisconsin Sequernce. Analysis Package, Version 8 for Unix, Genetics Computer Grotap, University Research Park., Madison. Wi.s. ), using default settings. Which uses the aal"Yon thm of Smith and \ `ate.rm an l: dv. Appt.
rtlcrth.. (1981) 2, 482- 89), 100521 As used herein, the term "Thermal Melting Point {`lar,)" refers to the temperature, under defined ionic strength, pK and nucleic acid concentration, at which 5(f'~'i, of the ohgoruclcotidcs complementary- to the target sequence hybridize to th taaget sequence at equilibrium. i`ypical.ly, stringent conditions will be those in which the salt concentration is at least about 0.01 to 1.0 M Na ion concentration or other salts) at pH 7.0 to 8.3 and the temperature is at least about YPC thr short oligona.rcleotides (e.g., 10 to 50 nucleotide), S
ringe:nt conditions may also be achieved with the addition of destabilizing agents such as forartartaide..
[0053) As used herein, "modulation" means either an increase (stimulation) or a decrease (inhibition) in the expression of a gene.
[0054] The ten-ii "variant" when used. in the context of as polynatclc:.otide seclitence, may encompass a polynucleotide sequence related to a wild type gene. 'This definition may also include, for example, "allelic," "splice," "species," or polymorphic" variants. A splice variant may .have significant identity to a reference r are?l.ecule, but will generally have a Yr-eater or lesser number of poly,rauc.leotides due to alternate splicing of eons during mRNA processing, The corresponding polypeptide irtaay possess additional ltanetionaal domains or anti absence of domains. Species variants are poly-nucleotide sequences that vary= from one species to another, Of particular utility in the invention are variants of wild type gene products. Variants may result from art least one mutation in the nucleic acid sequence and may result ill altered rare s or in poly rcpticles whose structure or function may or may not be altered. Any given nawral or recombinant gene may have nonce, one, or malty allelic forms. Common .mutational changes that give rise tea variants are en enaally! ascribed to natural deletions, additions, or substitutions of nucleotides. Each of these types of changes nmaÃ'occur alone., or ill combination with the others, one or more times in a given sequence.
[00551 The resulting poly-lx:ptides genc:mlly'uvill have significant atnuno acid ideiatity= relative to each other. A, polymorphic variant is a variation in the polynucleotide s quence of a paarticul<ar gene betwe~a n individuals of a given species. Polymorphic variants also may encompass "single nucleotide polymorphisaaas (SNPs, or single base mutations in AN hiCh the polynaaeleotide sequence varies by one base. The presence of SNPs may be indicativo of, for example, a certain. population with a propensity for a disease state, that is susceptibility versus resistance.
[0056] fact ivative poly nucleotides include nucleic acids sul-ajected to chemical modification, for example:., repiacemwn:t of hydro cn by an alktl aryl, or amino group. I erisati} cc e.., dcri "att e t?liÃot3taclcotidcs, may Comprise 1101)-naturally-occurriiaT.g portions, such as altered sugar moieties or inter-sugar lirakag~,cs. Exemplary among these are phosphorothioate and other Sulfur containing species which are known in the art, D ea i ative nucleic acids may also Contain labels, including radioraueleotides, enzvia es, fluorescent agents, the miltaminescent agents, chroaanaogenic agents, substrates, cofactors, inhibitors, niatwetie particles-, and the like.
[0057] A "derivative" polypeptide or peptide is one that is modified, for example, by 1glycosylatiori, pegylation, phosphorylatioia, sul:l .tion, reduction alkylat:iorn, acylatiori, cbetnical couplin;õ, or mild for -.txtl.:in treatnicnt, A derivative may also be modified to contain a detectable label, either directly or indirectly, including, but not limited to, a radioisotope, fluorescent, and enzymx label.
[0058] As used herein, the term "aniaaaal` or "Patient" is meant to include, for example, h:u 3aans, sheep, elks, deer, mule deer, minks, aa:maamamals, mioinkeys, horses cattle, pats, goats, dogs, cats, rats, mice, birds, chicken, reptiles, fish, insects and arachnids.
[0059] ".Niaaaa.aa ial" covers warm blooded mammals that are typically tinder medical care e..4~., humans and domesticated animals). Examples include feline, caanine, equine, bovine, and human, as well as just human.
1.0060.1 "Treating" or "treatment" covers the treatment of a disease-state in a maammal, and includes: (a) preventing the disease-state from occurring in as mananaal, in particular, When such mammal is predisposed to the disease-state but has not yet been diagnosed as halving it, (h) inhibiting the. disease-state, e.g., arresting it development; and or (C) relieving the disease-state, e .g., causing regression of the disease state until a desired endpoint is reached. Treating also includes the amelioration of a symptom of a disease. (e.f. lessen the pain or d:isc:.onitort), nhercin such :artieliora don inay or ma-,,,, not be directly aflec:tiaag the disease. (e,g. cause, trxnstrtissionn, expression, etc,).
[0061] As used. herein, "cancer" refers to all types of cancer or neoplasm or malignant tumors found In marnnili.s_ including, but not limited to: leukemias, lymphomas, melanomas, carcinomas and s:arcomaa. Thw cancer manifests itself as a "tumor" or tissue comprising malignant cells of the cancer.
Examples of tumors include sarcomas and carcinomas such as, brat: not limited to: fibrosarcoa:amaa, myxosareoma, lipos.arcoma, chondrosarcoma, ostconcnic sarcotamaa,, chordoma. a anwosatconaaaa cndothclosarco:ma, 1y mphitaa`g osaare.oraaa, 1y ml~h tng ocaat otliclir~sa:reoaaaa, sy"novionaa, a aaesothelioma, EwinW s tumor, leioniyosarcoraaa, rhabdonmyrosarcoma, coon careuaoma., pancreatic cancer, breast cancer, ovarian cancer, prostate cancer, squ amou,s cell carcinoma, basal cell carcinoma, a adenocaarcinonlaa, sweat gland carcinoma., sebaccous gland ca:rc.inon:ma., papillary carcinoma, papillary adenocarcinom as, c v,,t idenxocaarc.ia:noomn::aa, a:nedullary carcinoma, bronchotwenic carcialorama, anal cell carcinoma., he.patoma, bile duct caarcinoaira, chorioearcinoma, seaaaiaaoaaaa, criabi oral cyarcaaaotaaa il:taas' tumor, cervical cancer, testicular tumor. lung carcinoma small cell lung carcinoma, bladder carcinoma, epitlreliaal carcinon-aa, glionaa, aastroevlonaa, mcdulfobiastonaa4 cran:iopharya:a4giornaa, ependytnoma, pinealorna, henaarigio lasÃoma, acoustic :neuron;:a, oligodeadroglioma, meningio.ma, melanoma, neuroblastanaa, and rctinoblatstona. Additional cancers which can be treated by the disclosed composition according to the inkention include but not tbnited to, for exaampie, Hodgkin's Disease, Non-Hodgkins Lymphoma, multiple mvelomaa, neurohlastora breast cancer, ovarian cancer, lung cancer, rhahdomy' sarcoma, priin-m thromhocy to is.
primary macro lohuliner:nia.; simall-cell lung tumors, primary brain tun-.tors, stomach cancer, colon canter. mallyTant paricrea:tic insulationaaa, naaaliN, cant carcinoid, urinary bladder cancer, prean.atli xt.ant skin lesions, testicular cancer, lymphomas, thyroid. cancer, neuroblastoma-, esophageal cancer, genitourinary, tract caance.r, inalignant hypercalcenaiaa=
cervical cancer, endometriaal cancer, adrenal cortical cancer, and prostate cancer.
[0062] As used herein, a "amscle disease or disorder" includes but is not limited to n-auscular dystrophy (ID), a muscle-wasting disease, iatflatrtnriaÃory' n opa:thy or ran ositis (including, , for exaample dctniatomyositis, polyrny,t sins, inclusion. holy myotosis), n-ay=otubualar nay'op tlay, ncna.aihnc myopatlry. a dcsmin related racyopathy. \iarftaan myopaathy=, a mitoclaondrial myopathy etc.. As used .herein, muscular dystrophy refers to a group of genetic, hereditary muscle diseases that cause progressive muscle weakness and which may be characterized by progressive skeletal muscle weakness, defects in muscle proteins, and the death of muscle cells and tissue, Muscular dystrophy includes, but is not limited. to Duchenne (MID family). Recker (R'viD), Spinal Muscular Atrophy, Spinal bulbar muscular atrophy, dystrophiaaopathy, sartoglycaa_nopathy, limb girdle muscular dystrophy (LGMMD), congenital muscular dystrophy (CMD), faciosc rpuloh.umeral (FSHD), myotonic, oculophary ageal., distal, and Fmmaen - Dreiifuss.
[0063] "Ne.uroiocical disease or disorder" r fe_rs to any disease or disorder of the nervous system and., 'or visual system.
""Neurological disease or disorder" include disease or disorders that involve the central nervous system (brain, brainster and cerabel.lum), the peripheral nervous system (including; cranial ner' es), and the autonomic nervous s stern (parts of which are located in both. central and. peripheral rata Dias system). I xaaaal lc of neurologic <al disorders include but are not limited to, headache, stupor and coma, dementia, seizure, sleep disorders, trauma, infecti ins, neoplasms, neuroopÃhalniolcogy, movement disorders, dcmyclinaating diseases, spinal cord disorders, and disorders of peripheral. nerves, muscle and. neuromuscular, junctions. Addiction and mental illness, include, but are not limited to, bipolar disorder and schizophreniaa7 are also included in the defi.n.ition, of neurological disorder. The follow ing is a list of several neurological disorders', symptoms, signs and syndromes that can be treated using compositions and methods according to the present invention: acquired epileptiforni aphasia; acute disseminated enceplaalomyelitis;
adrentolcurkozlystrcpla:yy; age-related macular degeneration.; aageneesis of the corpus callosum; agnosia; Aicardi syndroa:ame;
Alexander disease; Alpers' disease; alternating hen--ii legia; Vascular dementia;; anayotrophic lateral sclerosis,, araerace alraly , Am geiman syndrome; angiomaatosis; anoxia; aphasia apraxia; aaaachnoid cysts;
arachnoiditis; Anroani-Chiar.i naaaifbrn anon; aateriovenous analformatiorr; Asperger syndrome; a ataxia telegiectasia; attention de-licit hyperactivity disorder; autism.; autonomic. d.ys inction; back pain; Batten disc:asses Beheet's disease, Bell's palsy; benign essential hlopharospaasnr benign fcal; anayotrophy; benign mu acarui al by>pertc tsion; Binswange ='s discase;
blepharospasm, Bloch Sulzberger syndrome; brachial ple^cus irljur ; !br:ai:n abscess; brtun .irlja.ary; brain tunxors (including glioblastort a multiforinc), spinal tuitror, Brown-Scqu lr{l sy'nndrome;
Canavan discasc; carpal Ãwiriel syndr'om e;
cau.salgia; central pain syndrenne; central on:t }e. i -ryei nolys s cephalic disorder; cerebral aneurysm; cerebral arteriosclerosis; cerebral atrophy, cerebral gigantism cc rebral palsy; Char-coÃ-Marie-Tooth d_ise ase: chem.otherapy-induced neuropathy and rreurop rth c pain; Chiani malfbrniation; chorea;
chronic nflamni aÃory de-mvelinitin<
polyncuropaathy; chronic pain; chronic regiottdl pain syndrome Coffin Lowry syndrome; eo iia, including persistent vegetative state; con(genital. facial dip eya.; cortreobasal. degeneration;
cranial artenitis; craniosy ostosis; Creutzeldt-Jal ob disease; cumulative trauma disorders; Cushing's syndronie; eytonicgalic inclusion body disease;
:o cytornegalovirus infection; dancing eyes-dancing feet syndrome;
.Dandy\\'alker syndronmre; Dawson disease; Dc %1oiaiet's stiYrrclrotrac De:jeriirc-Kla.rrxtkc palsy, dementia; d ;ririatonivosiÃis; diabetic ricur-opaÃ1ry: diffuse sclerosis-, d~;sautonoinix; d ss ;ralrlria; clysle ia; cly,toiaiiis; carly iiaftritile epileptic cttcc pli:alol atlay; r itlpty Sella saidioraae;
encephalitis; encephaloceles; cize~.lalaalc~tris*r nziraal arigio:matos s;
epilepsy; Er&s pzilsv; essential tremor; Fabry's disease; Faihfs syndrome: fainting; familial spastic paralysis; febrile seizures; Fisher syndrome, Friedreich`s ataxia, fronto-temporal dementia and other ` t iarcipratltir s", (i:auchers disease;
Ger Soria ml's syndrome; {giant cell arteritis; ;giant cell inclusion disease- globoid cell .leukodystaophy; Ciuillain-Barre syndrome; HY_ V-1-assoc..iatecd myeiopathy;
.Hallenorden-Spatz disease; head injury; headache; hentfa.cial spasm;
hereditary, spastic puaplcgia. her=edopathi.a atract c a lrolyrraetar=itill rmis; herpes zoster otieaas; herpes zoster;
.1Tirayattaa syndrome; .Hl assns:aced dementia atid ttcwopathy !also neurological n:iani.festations of AIDS); holoprosL.rrccpb a1 1-1tittingÃer r disease and other polyglutaf.raine repeat diseases; hydranencephaly; hydrocephalus;
hypercortisolrsm; hypoxia; irrtriataaae¾rircdiaÃed encephalor:nyelitis; inclusion body rrtyosrÃrs; incontinentia pignienti;
infantile phytaniic acid storage disease; infhnti.l refsr.arn disease; infantile spasms; inlani_matory< myopatho; intracranial cyst; inttacranial hyper=te tsion; Jotibert syndrome; Ke ams Sayre syndrome: Kennedy disease Kinsboume syndrome; Klippei Neil syndrome; Krahbe disease;
Kuugelberg-Welander disease lcuru; Lafora disease; Lambert-Eaton myasthenic syndrome; Landau-Me frier syndrome;
lateral medullar-y, (t `allenber4g) syndrome; leamiug disabilities; lxigh's disease, Lennox-Clustaut syridr=ome; Lesc.h-Ny<han syndrome; leukodysÃrophy; l.:ewy body dementia; .Lissencephaly; locked-in syndaoirase; Lou. Gehrig"s disease #i:c., motor neuron disease ctr tanayotrophic lateral sc.lerosin) funibar disc disease; Lyme di.seasa -ircaair logicÃal scclaaclte;
Machado-Joseph disease; ntacrcrieeph.aly; r:ttcgalcrrcepltarly; 1 1c11 crsscrra~l~rrsentlttil syndioattc, leriicaea disease;
meningitis; N'lenlres disease; mnevichromatic leukodystr-cphy; microcaphaly;
migraine; Miller Fisher syndrome; milli-strokes; mitochondrial myopathies; Mohiais syndrome, monomelic amyotrophy:
motor neuron disease; Moyanioya disease; mince?poly,~racchraridoses; milli-infarct dementia; maulÃif ?cal motor neur-opaÃlhay; multiple sclerosis and other clcrtaycl.irrtating disorders; a aulailrle. systeirt atrophy with Postural hypotension; P muscular dystrophy : rtas=asthenia gra is:
mvelinoclastic: diffuse sclerosi ; rtmyoclonic criceplraal.opathy of infants;
myoc.lorrus; myopathy. invotonia congenital;

taareolepsy; fCWOf:ibFOinwosis De1Uoleptic a ialigliart syndrome.
neurological, i- an.itestationc o' AIDS; neunol.ogi:cal sequelae oflupus; neuromyotonia; aaemonal ceroid ipof uscinpsis newonal migration disorders- alien-lann-.Pick disease;
O'Sullivan-McLeod syndrome; occipital nicrur4alg=ia: Occult spinal dysrap(iisrt sequence. Ohtalhaia syndrome oli`opooto>cerebellar atrophy; o soclo n is inyoclonus; optic neuritis;
orthostatic hypotension , overuse syndrome paresthesia, Nenrodctencxaative disease or disorder (Par'lcir: soax's disease, l htntinoton's disease, Alzheimer's diseaa e, atanyotro hic lateral sclerosis (ALS), den-tentia, multiple sclerosis and other diseases and disorders- associated. w ith aacuronaal cell death) paraunyototua coax<uca itaa1 p udn oplastic dist:aasts;
paroxysmal attacks, N wry Romberg s yndrotaxt.
11cl iiaeus-,Mcrzbachcr disease; periodic paralyses-, peripheral neur-op thy;
painful netrxopathy and ancuropa.thic pain;
Persistent o e<kctative state; Pervasive developmental disorders; photic sneeze reflex; phytranic acid storage disease;
:o Picks disease; pinched ate rze.. Pituitary tumors; polyymyositis.
poreneephaly; post-polio syndrome; posdierlaeÃic neuralgia Postinfectiotas eneephalomyclitis; postural hypotension; Praadcr-Willi syndrotne; primary lateral sclerosis-, ptioaa diseases; Progressive henaaithciaal atrophy; progressive antaltitocalletalcoeracephaalopaatlay; progressive sclerosing taoliodystrophy; progressive Suprantaelear palsy. pseudotaar:aaor cerebri; Rw saay-Htar2t sy,tidrome (types I and 11);
Rastnussen's ena cephalitis: reflex sympathetic dystrophy syndrome, Re.Csuan disease. repetitive motion disorders;
rc.laetttit e stress tlltatic s restless legs syndrome; reÃrc ~ it gas assctc iartt d myc.lop ltl' y; Rett syndrome, Reye's syndrome;
Saint Vitus dance; Sandhof disease., Sehilder's disease; schizencephaalyx;
septco-optic dysplaa.sia; shaken baby syndrottme;
shingles; Shy-l_?raager syra-adroma e; Sjogren's sy dro ne, sleet) apnea;
Soto'c sytatlrome. ,Paastit ity; spina hitidaa; spinal cord in jury: spiral cord armors: spinal aaaaasculaar )Brophy; Stiff-Pcrson syndrome; stn,) e. SÃarr.ge-\Vcber syndrome; subacute sclerosinfa pasnenecphallos; subcor-tic al artcriosdcrotic enc :.phaalopathy ;
Syrdeuhaam chorea.- syncope-ritz caaxay lira;
taardive dyskinesiaa- lay-Sachs disease; temporal a.rtcritis; tethered spinal cord syndrorm;; fhonisen disease; thoracic outlet syndrome; Tic Douloureux; '.Fodd's paralysis; Tourette sy adrome transient ischemic a a.ttaack; transmissibl spongi onn enceplxaalopauhaes; transverse myÃ;lms; iraum )tie. bruin ir:lltar y; tx lnor: Ãratetriinatl neuralgia; tropical Npastic pan, paresis: tuberous sclerosis; vascular- dementia (iamlti-infhrct dementia); vaasrtalitis including temporal arte.ritis; Von Hippel-Lindau disease; Waller:aberg's syndrome; U erduig-Hofnan disease; W
`cst syndrome; whiplash; Willi) mms syndrome; Wildon'S disease; and Zellw veeer syndrome. As user herein, a "rac:taac>a tasc:talaar' disease or disorder refers to anty disease ,adve\ely affecting both near Otis elements (brain, spi it cord, peripheral nen e) and muscle Ãsti iated. or smooth n tuscle}, including but not limited to involuntary movement disorders, dystoniaas, spinal cord imam or disease, multiple sclerosis, timvaasthcnia. gravis, Parkinsotn's disease, Ant otrophic Lateral Sclerosis (ALS), Hu ntington's disease, Lambert-".atom) Myraasthenic Syndrome (LES), Congenital Nlyaasthenie Syndromes (CMS), Chareot `Marie-Tooth Disease, (CMT'), Dc cnxie-Sottus Disease (1)5) Crcutzleldt-,lal ab disease, l ricereic:h's Ataxia., muscular dystrophy, spatstai ity from cerebral palsy and stroke.
[0064] A cardiovascular` disease or disorder includes those disorders that can either cause. ischemia or are caused by reperfusion of the heart. Examples include, but are not limited to, atherosclerosis, coronary artery disease, ' ranulonxatouc :nir=ocarditis, chronic myoeard tis (non grin 1oi atous ), primary 11~ acrtr phic cuidiflnl o nihy, peripheral artery disease WAD),, Stroke agrina pectoris, nnyo rdial infarction, cardiovascular tissue damage ca .used by cardiac arrest, cardiovascular tissue damage caused by cardiac bypass, cardio enc shock, and related conditions that would be known by those of ordinary skill in the art or which involve dys(hnetion of or tissue damage to the heart or vascular .cre, especially, but not ihnited. to, tissue damage related to Di\ID
family activation. CVS diseases include, but are next baited to, atherosclerosis. Yr~anulon:ra:tous r:uv>ocarditis, err cx<rrtliaB i:n rc Lion; myocardial fibrosis secondary to valvular heart disease, myocardial fibrosis Without itrlaarction, primary hypertrophic cardionayopathy, and chronic myocarditis (ncra-~?rarrrtilon atotrs).
[0065] As used herein. "cardionlyopathy`, refers to any disease or dysfunction of the myocardium (heart muscle) in.
1Ã0 which the heart is abnon-nally etilarged, thickened and/or stifie.ned, As a result, the heart muscle's ability, to pump blood is usually weakened. The disease or disorder can be, for example, inflanunatoty=, mctabolic, toxic, infiltratiVc, libroplastic, hematological, genctlc, or unknown in origin, Such cardiomyopathics may result from a lack of oxygen.
Other diseases include those that result from myocardial. injury which involves damage to the muscle or the myocardium in the wall of the heart as a result of disease or trauma.
Myocardial injury can be attributed to many brings such as, but not limited to, cardionryopathy, myocardial infarction. or congenital heart disease, Specific cardiac disorders to he treated also include con(gestive. heart failure:,, ventricular Or atrial sepal defect, congenital heart detect or ventricular aneurysm. The cardiac disorder may be pediatric in origin- Car-dion.yopathy includes but is not limited to, cardiomyopathy> ('dilated, hyper trophie ; restrictive, arrhythmogenic and unclassified cardiomyopaÃhy ), sporadic dilated cardiomyopatlry, X-linked Dilated Cardiomyopathy (XLD('), acute and chronic heart: failure,,. right heart failuue, left heart failure, biveratticular heart failure:, congenital heart defects. mitral valve stencsis, nutral valve insufficiency, aortic valve stenosis, aortic valve insufficiency, tricuspidal v alv-e. stenosis, tricuspidal valve insufclency, pulnaonal valve ste:uosis, pulmonal valve insufficiency, combined valve defects, .my ocardins, acute nlyocarditis, chronic myocarditis, viral myocaarditis, diastolic heart failure. systolic heart failure, diabetic heart failure and acccumulaation. diseases.
/ of tttr.,leofide and O1gonucleoride (' inposttlons and Malec ales [0006] i uge!s: in one embodiment, the taarcgets comprise nucleic acid sequences of Dystrophin family, including without limitation sense and;/or antisense noncorling and,or- coding sequences associated with DrvlD family.
[00/i7] Dy'sttophin has been known since 198 ;', (Hoffman eat al. (1987), (c li. 51:509-517) to be the protein that: is deficient itr Duchennc: muscular dystrophy (DMD). This is an elongated prvrtein present at the cytoplasmic surface of the vertebrate muscle cell membrane (Hof aran et al: (1987), (7e/l. 51:509-517). 'hree other dy strophin-relaatcd proteins, i.e. D.RPr (dystroplrin-rc.larted protein Type 1, or utiophin), D P
{clystropirin-i lafiteri protein Type 2), and dystrohrt ins, have also been identified as products of difkrcr t greases (ri'aTi el sal, (1998) Ihim 11 i (.enf_=.t. ': ? l ?8 ? }, Dystrophiras and utrophin have been detected in muscle of other mart malian species (Pons et at. (1994) Cir cul ation.
90:369-374 Wane cat cal. 41.998) Fatah.Ifo!' Genet. 7::581-588, Rafael cat a!.
(2000) Man .,146/ 9:1357-1367) and also in other tissues, such as the electric orl rua, that are derived from skeletal r:nuscle (Chang et al. (1 989) ,I Biol Chem.
264:2()$31-2(834), and in ner\ es (Rivicr cat cr/. (1999a) Histoch :rn;,1.
31:425.432 , ftivicr- cat at (19990d f fiasc 1, Res Cell. Ion/. 20:305-314) of : m.ar m.or=arta , i)ystrcsphin in taaanrtn al.
skeletal a1Itrsele natmacts with an associated protein complex (DA1'C) to to m a link between the cy toskeleton and the extracellul ar matrix (1bratghimov-Beskvnaya et al.
(1992) .\'ature.:355.t 96-702), This complex consists, of Ãxva d ystro lsc tta:; and f-j (f)tt=s or a', (19 5).I-1>w (:'! n<.
270:2596-25959), sarcoglycms l:Ir, and e) that are con plexe;d with :
arcospan. (Nigro Ot al. (1996 1:/err :A)! l net. S11791186; C'roslaie et al.
1997)1 B u! Ch(-,mi. 2723M2 1-31224; McNally c=t at (1998) F lS L,=u. 42-1-27-2), and three sy-ntrophins fil-, 112-) in muscle tissues (AIm et at (1996) J
Biol C iesn .2.71:27,224--7,30). .Howe -er, new isofornas of syntrophrras (''1- and 12-) have also been reported and were found expressed as lain-specific protein (Plluso et al. (2000) 1 Bial (: h(m. 275:15851-15860), Deficiency or variations in s me associated proteins gene r ate a.
diflierent muscle pathology, but the pat-lo :nesis of all of these related muscular dystrophics is still unclear, Ili is may be due to the heterogeneity of the data recorded, e LF., due to the existence of four- dystrophin-related proteins that all share homology with dystrophin's cyste.ine-rich and C-terminal domains and also because of the muscle type analyzed.
[0008] Dystrophin is a rod-shaped cytoplasraaie protein, and a vital part of a protein complex that connect the.
cytoskeleton of a muscle fiber to the surrounding extracellulaar tatatrix through the cell membrane. This complex is variously known as the costamere or the dystrophin- associated protein complex. 1%,111y muscle proteins, such as a-dy #rohra;rin syraacailir , syraenrin ara;c371ycan t strca lyc n, and 5arca~spart, cokxcai ac ~.udth dystrophin at The costamere.
[0069] Its deficiency is one of the root causes of muscular dystrophy. `y or-tmal tissue contains small amounts of dy,strophi:n (about 0,002% of total muscle protein), but its absence loads to both DMD family and fibrosis, a condition of muscle harde.:ninvg. A different mutation of the same gene causes defective dye strophin. leading to Becker's muscular dystrophy (BMD). Thus, it would be of great therapeutic value to t odulat, the expression andior function of dysÃrophin in cells, tissues or- organs ofpatients in need of such treatment.
[0070] 1_ ystrophin fanaily, the largest known human gene, encodes a protein called dystrophin. There are many different versions of dystrophin, some of which are specific to certain cell types. DyesÃrophin. is located chiefly in muscles used for movement (skeletal muscles) and the muscles of the heart (cardiac muscles). Small amounts of the protein are present in nerve cells in the brain.
In skeletal and cardiac muscles, dystrophin is part of a protein complex that strengthens muscle fibers and protects them from. injury as muscles contract and relax, The dystrophin complex acts as an anchor, connecting each a(l muscle cell's structural framework (cyrtoskelcton) with the lattice of proteins and other molecules outside the cell. The dystrophin complex may also playa. role in cell signaling by interacting with proteins that send and receive chen-rical, s gnals.

[0072] Little is known aborat the function of dystro hin in nerve cells and xw.ithout wishing to be bound by theory, dystrophin could be important for the normal str-a. Ctur-c aand function of synapses, [00731 1?uclhc ne an . Becker muscular dystrophy are caused, by mt:tt ations in the [)':L[) fan ily gene.
( Muscular dystrophy (INID) refers to a group of genetic disc de:as 3.1aos. Ma Pr symptom is muscle wasting.
There are two major forms of .D,, differing in severity and awe of onset. In Drrchern.re muscular dystrophy, syrtxpton.ts are noticeable in early childhood and quickly become debilitating. Becker muscular dystrophy. on the other hand, is of later onset and less severe. Both forms of MD ar : caused by mutations in the dystrrophin gene, a large (?.6 4b) gene comprised of 97 exons. The dystrophin protein pays an important structatral role as part of a large complex in muscle fiber membranes. When dystrophin is missing or non-f rnc-tionai, the entire complex is compromised. leading to lty degeneration of "ar:artyc le. tissue. 1rerr the ability to regenerate the muscle is exhausted, muscle t astiny occurs.
[007.5] l l.utations in the D1. ID fitmily gene also cause a fonaa of heart disease called X-linked dilated candioniyopathy.
I'lais condition enlarges and weakens the cardiac muscle, preventing it from pumping blood efficiently. Although dilated cardiomyopathy is a sign of Duchenne and Becker muscular- dystrophy, the isolated X-linked forin of this heat condition is not associated with weakness and 1. asting of skeletal muscles.
[00761 Utrophin is a 395 kDa Protein encoded by multiexonic 1 Mb U1'RN gene located on chromosome 6g24 (Pearce, ef ca/ (1993) M1111 Miri t 3< rrr. 2: 1765 1.7721). 'rhe structure of the gene bears large similarities to that of th .
dye#rc phiaa 4gerzc. In cor:rtrast to d\ trc phirr, t1le utrophin gene has a long 5' untnanshted re4g on, split oA Ã:r= two e eons, and it is preceeded by an l:l`i" FHisland.. In mouse, the gene maps to chromosome ill. 11tr-ophin is distributed throughout tine sareolemr-naa in fetal and regeneraÃing muscle, but is dozen-regulated in normal adult muscle and is restricted to the myotendinous and rnrti'uMrrrr.ascularsjunctions (Blake et al,, 1996). In the dystrophin deficient mclx mouse, uatrolihirl levels in muscle remain elevated. soon. after birth compared with normal mice., once the r:t#rophin levels have decreased to tile adult levels (about I week after birth) the first signs of muscle fibre necrosis are detected. However, there is evidence to suggest that in the small calibre: muscles, continual increased levels of utrophin can interact with the DGC complex (or an antigenically related complex) at the sarcolenanaa thus preventing loss of the complex with the result that these muscles appear norrr:tal. There is also a substantial body of evidence demonstrating that utrophin is capable of localising to the sarcoleairna in nonnal n:tuscle. During fetal muscle development (here is increased t.atrophin. expression, localised to the sarcolemnra: up until 18 weeks in the human and 20 days gestation in the mouse, After this time the utropbin sarcolenrr-nal staining steadily decreases to the significantly lower adult levels shortly before birth where utrophin is localised almost exclusively to the N MJ. The decrease in utrophin expression coincides with increased expression of dy strophin. See reviews Ubraghimov Beskrovnay r, et al. (1992) Nature? 355, 696 702, Blal e, et al. (191)4) Trends in Cell iliol, 4: 19 23, Tinsley , et al. (1993) CunrrC.r sin (Genet Del . 3-4,M%), [0077] DR-P2 is predicted to resernble certain short C-tern-ainal isofomis of dystrophin and dystrophit r iaated protein .1 (DRP1 or utroplniI ), DRP2 is a relatively saa-ta.ll protein, encoded in man by a 45 kb ~,genc ioe dizcd to X q22. It is expressed principally in the brain a3ad spinal cord, and is sinnilar in overall trtaCtaare to the t)pl 16 dysstiophin isof>rm.
[0078] l ystrobrer in, a member of the dystrophin family of proteins, was originally :identified from the i i ?e:ki :ell /i:'rnic'o electric organ as an 87-kDa phosphoprote.in associated with the c.vtt~plasnlic face of the postsynaptic membrane (\ 'aagtacr K R. el cr/, (! 993) :' evtvrt.. 10:511 522; OUT C. e-1 of. (1989) J t . 11 BaoL 109:17153-176,), It lt4 has been postulaatcd that the 87-kDaa protein plays a role In synapse formation or stability bectarase; it copurifl s with acetylclaoiirac. receptors from the e1ecta'ic organ taacaaah:raaacs. In anasaanranalian skeletal aanuscle, dysrrophin is found. in association with several mte,,., al and peripheral membrane proteins, forming a complex known as the dysti-ophin glycoprotein complex (UG(:) (pn. asti J A f aL (1991 ) C C//. 66A 121 .i irf I lbt .ghinio\ -Beskroz. tint :aa C), . ad. (1992) Nature (London). Yoshida M, el a/. (I990) .1-Biocherrr { l f> t x 10$:748 752).
[0079] In preferred embodiments, aaanti.sense oligonuaelc.otides are used to prevent or treat diseases or disorders associated with .1 :'~l.i :faatatal :membe s. Exemplary Dystrophin hmi.ly mediated diseases and, disorders which can be, treated with cell/tassaaes r generated from stem cells obtained using the antisense compounds con:aprise: a muscle disease or disorder (e.g Muscular dystrophy including i aaehenncs muscular dystrophy Becke-'s muscular- dystrophy, Spinal bulbar muscular atrophy, dy, trophiaaopathy, Satcoglycaanopathy, limb girdle nanscula:r dystrophy, congenital muscular dystrophy, congenital nnyolnathy, distal 111) pop h S\ mptotn>atic form of .t3 aa. e ttlsaa dystrophy ofDucbenne and Becker in :fe.male carriers n-tyotonic syndrome etc.; a a -auscle-wasting disease), a neurological disease or disorder (including a neuromuscular disease or disorder a fig., dysto~ ki, n-tvocionus-dystonia syndrome, etc) a disease or disorder associated with altered level of dystrophin or dystrophin I)APC-complex. Left ventricular noncoinpaction, cancer, a cardiovascular disease or disorder., eardiomyopathti (e.g.,. spx)radie dilated cardior yopathy, X-1inl c d Dilated Caardionayopathy (XLDC) etc.), atherosclerosis a cytoslaeletal disovkr, congenital stationary :night blindness and loss of he<arinr.
[0080_] In a preferred embodiment, the oligonucicotides are Specific for ltolyaaaacleotides of )~+4.i_ tantt.l~ which includes, sa:ithout limi ration noncoding regions. The DM1 f<anatly targets c.otnp}use'sariants of DNlf) fttnnil ; nna:ttaants of D: SID fiaoil >, in.Iudinu SNPs nonwwding sc:gttcncts of DM:t ttanily- :alts cs, fragn-tents and the like. PtcfLrably the <alig onucle tide is an aatrtlsense RNA. e aolecttlc .
[0081 ] In accordance with embodiments of the i:n vention= the taar4get.
nucleic. acid molecule is not limited to MID
ftunily polvnucleotides alone but extends to any of the isofornms, receptors, ho:molo;g-&, non-coding regions and the like of DM1 family.
[0082] In another preferred embodiment, in oligonucleotide targets a natural antisense sequence (natural aantisense to the coding and non-coding regions) of DIv1D f roily tra cts, including, without liimitaation,, vaariaants, alleles, hornologgs=

mutants, derivatives, fragnacnts and complen-tcntar sequences thereto.
Preferably the oligonucleotide :is an arrtisensc RNA or DNA t oiecule.
[0083] In another preferred enilsodiment, the oiigoineric compounds of the present. invention also include variants in which a different base is present at one or more of the nuclc otide position in the, compotr:trd. I or example, if the first tuicleotide is an adenine, variants may be produced xhich cc,.tmtait . t y:ty.iidi:tne. -uanosine., cytidinee or other naiwral or unnatural nucleotides at this position. This ma ,y be done at any of the positions of the auntisense cori pound.. These compounds are their tested using the methods described herein to determine their ability to irntliibit expression of a target nucleic acid, [(3084] In some ernbodimen s, homoloty, sequence identity or con:tple;n:tentarity, bet teen the antisensc compound and target is from about 50% to about 60'X',. In some e l hodime.nt r, homology, sequence identity or coniplernentat ity, is fi'orn about fiatt>. to about In some Intl cxlinrt:rrts, hor:nology, sequence identity or cor rplerrrentarity, is from about 70`>ir to about 80%. In some e trbodiments, homology, sequence identity or ccr rl lenre.nÃt~ tity, is from about 80% to about 90%. In some embodiments, homology., sequence identi.t., or compiement<
t-ity, is about 90%, about 9?`%% , about 941,'Zol, about 95%''a, about 96%42, about 9 7'1~%i%, about 98'X%, about 99%
fd or about .l00 %%.
[0085] An antisense compound is specifically hybridizable when binding of the compound to the target nucleic acid interferes with the normal function of the target nucleic acid to cause a loss of activity, and there is a sufficient degree of cornplementarity to avoid non-specific binding of the anti.sense compound to non-tar( et nucleic acid s quences under conditions in which specific binding is desired.. Such conditions include, i.e., physiological conditions in the case of in vivo assays or therapeutic treatncrtt, and conditions in which assays are performed in the case of in vitro assays.
[00861 An antise rse compound, whether DNA, R.\ A, ch_ià reric, substituted etc, is specific-ally hybridizable v Shen binding of the coa -tpou nd. to the target .DNA or RN A molecule interferes with. the nomial function of the target DNA. or RNA to cause a loss ofuÃility and there is a sufficient degree of complementarily to avoid non-specific binding of the antisense compound to non-target sequences under conditions in vg lrielr specific binding is desired, i.e., under-physiological conditions in the catse of in vivo assays or thentpct:ttic.
treat tment, and to the case of in vitro assay,s.; tinder conditions in which the assays. are perfoorined.
[0087] In another preferred etr:mbodiment, targeting of OM:l family including without limitation, an. tisense sequences which arc identified and expanded, using for example, PCR, hybridization etc., one or r core of the sequences set forth as SE ID NO: 3 to 7, and t ho like, modulate the expression or function of MID
family. In one emboditrment.
expression or function is up-regulated as compared to a control. In another preferred embodiment, expression or f inction is down-reg.tlatted. as compared. to a control.
[0088] In another preferred e xrbodinient, oligonucleotides comprise nucleic acid sequences set firth as SEQ ID }NOS:
8 to 22 including antisense sequences which are identified and expanded, using for example,, I'CR, by=bridization. ete.
These. oli ot3crc t: t tles can comprise one or more modified nucleotides, shorter or longer fragments, modified bonds and the like. Examples of modified bonds or itnernucleotide linkages comprise phosphorothioate, phosphorodithioate or the like. In another preferred embodiment, the nucleotides, comprise a phosphorus detivaÃi\e. The phosphorus derivative (or tnedfified phosphate group) which may be attached to the sugar or sugar analog moiety in the triodif_ cd.
oligonucleotides of the pres:rit invention mm-tay he :t nlonophostphate, diphosphaÃe, trtt5hosphate, alkyiphosplhate, S alkanephosphaÃ.e, pllc st lrtatotllitrate and the like. The preparat:ion of the above-noted phosphate analogs, an l their incorporation into nucleotides, modified nucleotides and olig onucleotides, per se, is also known and need not be described here.
[0089] The specificity, and sensitivity of antiserlse is also harnessed by those of skill in the art for therapeutic Uses.
:Antisense otigonucleotides have been. employed as therapeutic moieties in the treatment of disease states in. animals and matt, Antisense oligonuclootides have been. safely and effectively adruinistered to humans and numerous clinical trials are presently underway. It is thus established that oligonucleotides can be, useful therapeutic modalities that can be configured to be usefal in, treatrr-lent regimes for treatment of cells, tissues and wiimals, especially hhuulans, [0090] In embodiments of the present invention oligor-rmeric antisserrse coml ounds, particularly oligonucleotides, bind to target nucleic acid molecules and modulate the expression anci:'or function of molecules encoded by a target 1, 1 e.
The functions o:Ã DNA to be interfered coin prise, Ras example, replication and transcription. The functions of RNA to be interfered comprise all vital functions such as, for C"xample, tr-anslocation of the RNA to the site cif protein translation, tnuislation of protein from the RNA, splicing of the RNA to yield one or more inR.NA species, and catalytic actin its' which may be engaged in or facilitated by the RNA, The functions ma be upnregulate or inhibited depending on the functions desired, [0091] "I'he antisÃ:nse compounds, include, antisense oligorner-ic compounds, antiser se oligonucleotides, external guide sequence (EGS) caligcartlrcleotides, alternate splicers, primers, probes, and other ol:igomeric. compounds that hybridise to at least a portion of the target nucleic acid. As such, these compounds may be introduced in the form of single-stranded, dcauble-stranded, partially single-stranded, or circular oligonieric compounds.
[0092] Targeting an antisense compound to a particular nucleic acid molecule, in the context of this invention, can be a multistep process. The process usually begins with the identification of a Ã
art et nucleic acid who e function is to be modulated, This target nucleic acid may be, .feat example, a cellular gene (or r:n RN:A transcribed from the gene) whose expression is associated with a particular disorder or disease state, or a nucleic acid molecule from an infectious agent.
In the present invenÃion, the target nucleic acid encodes Dystrophin fancily, [0093) The targeting process usually also includes dete.miination of at least one target region, segment, or site within thee, target nucleic acid for the arrtisense interaction to occur such that the desired effect, c.gY., .n-rodulation of expression.
will result.. \ ithil the context of the present itwention, the term "region"
I
is de-fried as a portion. of the target nucleic acid having at least one identifiable structure, function, or characteristic.
Within red=ions of target nucleic acids are sei ialeiats. "Segments" are detaaed as smaller or sub-portions of regions withi:aa a target nucleic acid. "Sites," as used in the present invention, are defined as positions within a target nucleic acid.
[0094] In a preferred embodiment, the a:ntiseu.se oiigonticleoticles bind to the natural antisense sequences of Dystrophin family and modulate the expression andlor function ofDystrophin family (SEQ ID NO: I. and'D. Examples of aantisense sequences include SEQ ID NOS.- 3 to 21 [00951 In atiothei preferred eulbcxliment, the aà tiseuse ohgoi uclcoudes bid to one or more segnaeiaÃs of IDsÃrophin family polynticie-ifides and modulate the expression andlor function (if tistrophiÃi f inaily. The segments comprise at least five coasec:utive nucleotides of the l)ystrophin fatlaily sense, or ailtise":nse polynucleotides.
[0096] In another preferred embodiment, the antisense oiig=oÃaa.icleoticies are specific for natural antisense sequences of Dystrophin f:a:Ãaaily wherein binding of the oligonucleotides to the natural autisense sequences of DystrophiÃZ family modulate expression and/or function of Dystrophin fiarnily.
[0097] In another preferred embodiment, ol:igonucleotide compounds comprise sequences set forth as SEQ ID NOS: 8 to 22, antisen se sequences which are identified and expanded, using for example, P(_ R, hybridization etc These oligonucleotides can comprise one or more modified nucleotides, shorter or longer I,:aigments, modified bonds and the like. Examples of modified bonds or iiatemucleot:ide linkages comprise phosphoroÃ.hioatc, phosphorodith.ioaate or tine like. In another preferred embodiment, the nucleotides comprise a phosphorus derivative, The phosphorus derivative for modified phosphate group) t hich may be attached to the sugar or sugar analog .ialoict in the modified ohaonucleotidcs of the present: invention rimy lx a monophosphaÃ:e:, diphosphazÃ:e, trip'hosphatc.: alloy, lphospharc, allaanelahosphate, phosphorothioate and the like. The preparation of the above-noted phosphate analogs, and their incorporation into n.cicleotides, modified nuclef tides and oligonucleotides, per se, is also known and need not be described here.
[0098] Since, as is known in the art, the translation initiation colon is typically ?'-AUG On transcribed rri-RNA
aaaole:uies; 5'r' TG in the corresponding DNA Ãnole ule), the translation initiation colon is also referred to as the "AUG codon," the "start codoon" or the. "AUG start codo:n". A minority- of genes has a translation initiation codon haying the RNA. sequence 5'-GU G, 5' I I_ G or 5'-CUG; and 5'-AUA, 5r_ACG and 5'-C1JG have been shown to function in vivo. Thus, the t .mis "tanslation iÃaitiaation co don" and "start codon" can encompass i- aany codon sequences; even though the initiator amino acid in each instance is typically methionine (.in eukaryotes} or fornaylÃnethionine (in prokaryotes). Eukaryotic and prokaryotic genes may have two or more alternative start codons, any one of which may be prefl rentiaali.y utilized for translation initiation in a particular cell type or tissue, or under a particular set of conditions. I.n the context of the invention, "start cocoa"
and "translation initiation codon" refer to the codon or codons that are used in vivo to initiate translation of at). mR; A
transcribed from a genie encoding Dystrophin f ani ly, regardless of the sequence(s) of such colons. A translation termination codon (or "stop codon") of a gene may have one of three scque.:nces, i.e, 5 -l__ .AA. 5'-UAG and 5-[ GA (the.
corresponding DNA s ences are 5'-T AA 5'_ TAG and 5'-TGA, respectively). [0099] The terms "start codon region" and "translation initiation codon region" refer to a poi`tion of such an. RNA or erne That encompasses from about 25 to about 50 coati ?arotr_s nucleotides in either direction (i.c., 5' or 3') from a translation itaitiation. Codon, Similarly, the terms "stop code n reg-jon" and `"translation termination codon .region" refer to a portion of such an nR 1 A or gene that encompasses from about 15 to about 50 Corctiatio us nucleotides in either direction (i e., 5' or 3') from a translation term] nation coon.
C.onsequctiflyI the "s(art codon region" (or "translation initiation cod-on rewon") and the "stop codon region" or " translation wt-111111ation codon region") are all regions that inay be tar-geted effectively i "ith the antisense. compounds of the present invention.
[00001 The open reading frame (()RF) or "coding, region", which is known in the art to refer to the re ;ion between the translation initiation codon and the translation termination codon, is also a region which may be targeted effectively, . Within the context of the present invention, a targeted region is the intragenic region encompassing the translation initiation or teonination. codon of the open reading :fraric (ORF) of a gene.

[00011 Another target region includes the 5' untransl rted region known in the art to refer to the portion of an niRNS'A in the 5' direction Bona the translation initiation codon, and thus including, nucleotides between the 5' cap site and the translation initiation codon of an mR:NA (or corresponding nucleotides on the gene). Still another target regioll includes the :3" tints to laced region (3T TR), known in the art to refer to the portion of an mRN A in the 3' direction from the translation termination codon, and thits including nucleotides bec%veen the translation termination codon and 3' end of an mRNA (or con esfaondir g nucleotides on the 'gene:). 'f he 5' cap site of an mRNIA comprises an N ? iriethy laÃi:d guanosine residue Joined to the 5`-most residue of the aiRNA via a. 5'-S' triphosflhate linkage. The 5' cap region of an n-iR_N.A is considered to include the 5' cap structure itself as well, as the first 5() nucleotide ac iacent to the ca site.
Another target re{gioii for this invention is the 5' cap region.
[00102] Although some mtkaryotic mRN A transcripts arc directly translated, many contain one or more regions, known as "introits," which are excised from a transcript before. it is translated. The remaining (and therefore. ttntasla.tr:.el.;
regions are known as "exons" and are spliced. tot=ether to form, a Continuous rraRN:A sequence. In one embodiment, targeting splice sites, i.e.., mu on-exon ,junctions or exon-intron iiinctions, is particularly useful in situations wbere tibLnant splicing is implicated in disease:, or v here an over ).roduction ofrt p;.trticr,tl;.ir splice product is implicated in disease. An aberrant fission junction (file to rearrangement or deletion is another embodiment of a target site, n1RN,+
transcripts produced via the process of splicing of two ((Yr more) mRNAs from different gene sources are known as "fusion taanscrilats". ltxtaons can he effecÃxvcly< targeted. using antisense compounds targeted to, for example, DNA or pre-rnRNA.
[001031 In. another pre:.rrred embodiment, the antisense "11i;~anticlcr tides bind to coding and/or non-codrng regions of a target polynucl.eotidc and modulate the e pression and/or function of the target molecule.

[00104] hr another preferred t a bodia:nt nÃ, the antisc.nse oligonracieoticles bind to .n ataaral. azntiscrisc polya:aucieoticlcs and modulate the expression and/or function of the target nmolCcuie.
[00105] In another preferred embodiment-, the antigen-se ol.igonucleotide bind to sense polynuclleotides and. anoclulate the expression and/or function of the target n-tolecule.
[001061 Alternative RNA transcripts can, be produced fro.na the same geuornic region of D"` A. These alternative transcripts are generally 1\11(nkrt as "<<at-i .tits", N1c)re specs ic4lk "pre-rnRNA vari uts" are Ãraà scripts produced iirorn the same genainic DNA. that di .iLr f o:nr other transcripts produced from the samegenof is DNA in either their start or stop position and contain both intron.ic and. exot fc sequence.
[001Ã 7] Upon excision of one or more exon or introfr regions, or portions thereof d nringg Splicing, pre=nrR,NA e;,n i.an.ts produce smaller "rnRNA variants". Consequently, n-iR A variants are processed pare~rriR variants and each unique pre-armRNA variant must always produce a unique aaiR'v A variant as a result of splicing. These m RNA variants a are also known as "ailternativ=e splice Variants". If no splicing of tile pre-mRNA
variant occurs then the pre-mRNA variant is identical to the nRN:A. variant.
[00108] \'aarfants can be pr duced through the use of alternative signals to start or stop transcription. PremRNAs and.
rnRN ,; can possess more than one start colon or stop codon. Variants that originate from a pre-nmR_NA or rrmRNA that use alternative start codons are know as as ""alternative stark Marianas'" of that p e-nmR.NA or tR\A. Those transeri:pts that use an alternative stop codon. are .known as "after-native stop variants" of that pre mRNA or niR.\A.. One specific type of alternative ;stop variant is the "polyA variant" in which the multiple traanscripÃ:s produced result from the alternative selection of one of the "poly A stop signals" by the transcription machinery, thereby- producing transcripts that terminate at unique po.lyA sites, Within the context of the invention, the types of variants described herein arc. also embodiments of target nucleic acids.
[001Ã 9] The locations on the target nucleic acid to which the antisense compounds hybridize are defined as at least a S-nucleotide Jong portion of a tar get regFion to which an active antisense compound is targeted.
110] While the specific sequences of certain t erraI}tar target segments are set forth. herein, one of skill in the art.
will recognize that these serve to illustrate and describe particular embodiments within the scope of the present invention. Additional target segments are readily identifiable by one having, ordinary' skill in the art in view of this disclosure.

n c ( 5) consecuti e nucleotides [00111 1 -u g?et segments '+-.100 nucleotides in. length comYrprasing~ a Stretch at toast selected f =om within the illustrative pr e erred target segments are considered to he suitable .for targc:tianag as well.
3 tltl l l ]Target segnrerats can include DNA or RNA segt ences that comprise at least the 5 consecutive nucleotides fora the 4`-terraainus of one of the illustrative preferred target segments (tlae remaining nucleotides being, as consecut ve stretch of the saint L or .RNA beginning immediate y..pstreaara of the 5'-terrninaas of the target segment and.
contin ing until the DNA or RNA contains a about 5 to about lt)0 nuc.leotides). Simil<arly paeferred target segrrrents are:

represented by DNA or RNA sequences that comprise at least the 5 consecutive nucleotides from. the 3 temi: nt s of one of the illustrative preferred target segments (the rema.iaxirntg .thud eotides being a consecutive stretch of the same DNA or RNA hegiiin ., itzn t med.iatc_iv do nstt ar n of the 3'-tert minus of the target segment and continuirtg until the, DNA or RNA contains about 5 to about 100 nucleotides). One have skill in the art armed with the target segments illustrated here:irn % ill. be able. without undue expe .irnnentation, to identif further preferred t as g.et segments.
[001 13] Once one or more target regions, segments or sites have been identified, aarntisense compounds arc: chosen which are sufficiently complementary to the target, i.c., hybridize sufficierntl~ well carrel with sufficient: specificity, to gin e the desired of cct..
[0()114] In embodiments of the invention the ohgonucleotides bind to an.
atntisense strand of a particular mrget, Tile cnligornucleotides are at least 5 nucleotides in length and can l?t s nthesi ed so each oI:igonucleotide targets oaverlappin seequences Such that oligonuclentides are synthesized to cover the entire length of the target polyriuclcrotide. The targets also include coding as well as nc-arn coding regions.
[00115] In one emboditne:tnt, it is preferred to tyarjet sl ecif:ic nucleic acids by auntiscrnse oligonauci: aides. Targeting an antisense compound to a particular nucleic acid, is a .m ualtistep process.
The process usually begins with the identification of a nucleic acid sequence whose fwictiorn is to be modulated.
This may be, for example, a cellular gene (or ranR A transcribed. frown the genc) whose expr.:' 10ra is associated with a particular disorder or disease state, or a non coding polynucleotide such as for exa;:_mple. non coding RNA (nncRNA).
[0()116n] RNA can be classified into 0) messenger RNAs (mRNAS), which are translated into proteins, arid (2) non-pmtei n-coding RNAs (ncRNAs). ncRN As comprise micron lAs> antiscnse transcripts and other Transcriptional Units ('l U) containing a high density of stop codons and lacking aanv extensive "Open Reading Frame". Many nc.RNAs appear to start trearu initiation sites in 3' aaantraraslatesal regions (i'UTRs) of protein-coding loci.. ncRNAs are often rare and at least half of the ncRNAs that have been sequenced by the FANTOM
consortium seem not to be lx)lvadenylated.
Most researchers have fcnr obvious reasons focused on polvadenylated mRNAs that are processed and exported to the cytoplastnn. Recently. it was shown that the set of nc,rn-lnenl ad.en l atecl nuclear RN As may be very large, and that many such. transcripts arise from so-called i:tntergenic re ions (Chong, J. et a/.
(2005) c./`ewce 308 (5725), H49-1154'.
kapa anon, P..1 al. (20)5). tic:=fnwne.Rc>.s 15 (7), 987-997). The mechanism by which ncRNAs raaaay= rcgu.late gene expression is by base pairing sNrith target transcripts. The. RNA s that function b base pairing can be s rcnupa d .unto (1) cis encoded RNAs that are encoded at the same genetic location, but on the opposite strand to the RN As they act upon and therefore display perfect complennneritaarity to their target, and (2) trans-encoded R\As that are encoded at a ehroannosoann nl locatà cnn distinct from the RNAs they act upon, and generally do not exhibit perfect base-pairing potential.
with their targets.
[001171 Without ~~islnirng to be bound by theory., perturbation of an antisctnse lxnlynucleotide by the antrse;nse oli.tgonucieotides described herein can alter the expression of the corresponding sense messenger R.N ats, .However, this regulation can either be discordant (antisenso knockdowi results in messenger RNA cle-vadon.) or concordant Ontisense knockdown results in conconxitsant messenger RNA reduction). In these cases, aantisense oligonucleotides can be targeted to o erlapping or non-overlapping parts of the antisciase transcript resultin ; in its knockdown or sequestration. Codarig as well. as icon-coding antisense can be targeted in an identical manner and that either category is capable: of rC Uh1tili the corresponding sense transcripts - either in, a concordant or disconcordant nianner. The str, ategies that are employ ed in ickntifidntx nets= oliasonuc.leatides for use against a target i inn be based on the knockdown ofantiseirse l NA transcripts by antisensc oligonttcleotides or atny other means of mo-adulating the desia :d target [00116 ; ticaiec! /: 1.11 the case of discordant regulation. knocking down the antisense transcript elevates the expression of the conventional (sense) gene. Should that latter gene encode fora known or putative drug target, then.
knockdomvn of its antisense counterpart could conceivably mimic the action of a receptor agorust or an enzyme stimulant.
[001191 Stiuc< fi't' 2: In the case of concordant regulation, one could concomitantly knock down both aantisense a and.
sense to ranscripts and thereby achieve synergistic reduction of the conventional (sense) gene expression. If, for example an aantisense oligonucleotide is used to achieve knockdown, then this strategy can be used to apply one antisense ol.i:;=oniacleotide targeted to the sense transcript and another antisense oligonucleotide to the corresponding antisense transcript, r a single ea crg-cticaillR swilin-aetric antisense oltg_ontacleotide that si:aaaailtaaa et*usÃy targets o ed appins,) sense and antisense ta:ansi ripts.
[00120] According to the present invention, antisense compounds include anti-sense ol_itgonuclcotides. ribozyrr s, extenial guide sequence (EGS) oligonucleutides, siRNA compounds, single- or double-stranded RNA interfimnce (RN Ai) compounds such as siRNA compounds, and other oolipo eric. compounds which hybridize to at least a portion of the target nucleic acid and modulate its function. As such, they may be DNA, R .A. DNA-like, RNA-like, or mixtures thereof., or may be naiinetics of one or more of these. These compounds may be single-stranded, doublestranded, circular or hairpin oliggonaeric compounds and may contain structural elernents such as internal or terminal bulges, mismatches or loops. Antisense compounds are routinely prepared linearly but can Lie joined. or otherwise prepared to be circular and./or branched. Antisense compounds can include constructs such as. tsar example, two strands hybridized to form a wholly or partially double-stranded compound or a single strand with sufficient self-t<rnapic nxe aititaa ity` to allow for hybridization and formation of a fully or partially double-stranded compound. The two strands can be linked. internally leaving, f=ree 3' or 5` termini or can be linked to form a continuous hairpin structure or loop. The hairpin structure IIULi - contain aaaa ovetang on either the 5 or .' teruaiaius producing an extension of single stranded character. The double stranded compounds optionally can include o'v'erhangs on the ends. Further modifications can include conjugate groups attached to one of the tera3tini, selected nucleotide positions, sutg it positions or to one of the internucleoside linkages. Al.ternativc ly, the two strands can be linked via a non-nucleic acid moots or linker ;coup. When formed from only one strand, dSR'NA can take the form of a.
silt-coiaipleaaaentaa hairpin-i' e molecule that doubles back on itself to foma a duplex. `fhtas, the dsRNAs can be fiall:y or part1a11y double stranded.
Specificmodulation of gene expression can be achieved by stible expression of dsRNA hairpins in tr4aaasgenic cell lines, hovvev er, in sor-ne embodiments, the gene expression or function is tip regulated. \Vheaa formed from two stands, or a single strand that takes the firrrrr of a self-complernentar3. hairpin-type molecrrle doubled back on itself to form a dnplex, the two strands (or dnplex-forrrring regions of a single strand) are complementary RNA. str rnds that base pair in \VVatson-trick fashionn.
[00 1211 Once introduced. to a system, the compounds of the im-ct Ãort may elicit the action of one or more enzymes or structural proteins to of ect cleava4gc or other modification of tine target nucleic acid or may work via occu piney-based rnechani ms. In eneral., nucleic acids (ine1u1clrnt oligonucieotides) may be described as "DNA-likc " (r e.., generally"
lp having one or more 2`-deoxy sugars mid, generally, T. gather- than U bases) or "RNA-like" (Le, genenally having one or more ?'- hydroxyl or 2'-modified sugars and, generally U rather than `1"
bases). Nucleic acid helices can adept more than one type of structture, most commonly the A- and B-forms, it is believed that, in general, oligonucle.otide.s sv hich have B-foma-like structure are "DNA-like" and those which have A-fbnnlike structure are "R; A Like." In some (chimeric) embodiments: an antisense compound mnay contain both A- and B-forma regions.
[00122] In a another pre:Eerred embodiaa ent; the desired oligonucleotides or antisense compounds, eoraa.lprisc at least one of antisense RNA, antisen e DNA, chimeric anti.sense oligontic.leotides, antisense oltgonncl:eotides eomprisi:rag modified linkages, Interference RNA (RNAi), short interfering RNA (siRNA); a micro, interfering RNA (rniR.NA.); a small, temporal RNA (stRNA)-, or a short, hairpin RNA (shRNA); small RNA-induced gene activation. i RNAa ; small activating RNAs (saaRNAs), or combinations thereof [001.2 3] dsRNA can also activate gene expression, a mechanism that has been termed "small RNA-induced gene activation" or RNAa. dsR'N:A.s targeting gene promoters induce potent transcriptional activation of associated genes.
1Z :tat was demonstrated in human cells, using synthetic dsRNAs; termed "small activating RNAs" It is currently not known whether RNAa is conserved in other orgzuusms.
[00124] Small double-stranded RNA tdsRNA), such as small interfering RNA
(siRN:A) and microRNA (naiRNA)_ have been found to be the trig c.r of an e v calrati.canary< conserved mechanism. known as RNA interference (RN AO. RN.Ai invariably leads to gene silencing via remodeling chromatin to thereby suppress transcription, degrading complementary mRN,t, or blocking protein translation. How-ever, in instances described in detail in the examples section v Which follows, oligonuclcotides are shown to increase the expression andior- function of the Dystrophin family polynucleotides and encoded. products thereof dsRNAs may also act as small activating RNAs (saR:NA). Without wishing to be bound by theory, by targeting sequences in gene pronaoter:s, saRNAs would induce target gene expression in a phenomenon reef ,rred to as dsRNA--induced traaraseriptiona.l activation (RN.Aa ).
[001251 In a further embodiment, the. "preferred target set=meats" identified herein may be employed in a screen for additional compounds that modulate the expression of .t ystr~rlrlaira family laolyaatacle.otidc.s. "Modulators" ire those compounds that decrease or increase the expression of a :nucleic acid molecule encoding 13ystroph.in family and which comprise at least a 5 nucleotide portion that is Complementary to a preferred target seg=i3-.ent. The screening method comprises the steps of contacting a preferred target seõ-ment of a nucleic acid molecule encoding sense or natural antisense pohmucleotidcs of Dystrophira thiTaity with one or more candidate modulators, and selecting for one or more candidate modulators m.-hich decrease or increase the expression ofa nuclc i0.
acid molecule encoding Dystrophin fanily polyaaucleotides, e g SEQ ID NOS: 8 to 22, Once it is shown that the candidate modulator or modu'ators are capable of moduhttiang (e.g, either decreasing or incre.asi g) the oxpr ssion of a nucleic acid molecule encoding D=strophin family poh auel.eotides, the modulator may then be employed in further- investigative studies of the unction of 1.)ystrophin faanily polynucleotides, or for use as a research, diagnostic, or therapeutic agent in accordance with the present 19 invention.
[00126] Targeting the natural antisense sequence preferably modulates the function of the target gene. For example, the, I) 1:D fata-tily' gene (e.g. accession nuntbet NM WA006 Lind NM 007124, .Fig. 2), In a prefe -ed embodinment, the target is all antiseaase polynucleotide of the D MD f :n:ailr gene. Ina prefer-red emb. diment, an antisense oligona.ccleotide.-targets sense and/or n aural autisense sequences of Dystrophin family polynueleocides (e.g. accession number NMI 004006 arid N- 007124, Fig. 22), vasia nts, alleles, isoforms, homologs, mut{:ants, dem.-atives, fragments and complementary sequences thereto. .Preferably the oligonucleotide is an antisense molecule and the targets Include coding and oncodin , regions of antisense and/or sense DM1) fiamily pt)ly.,iiLuc.lceoatides_ [00127] The preferred target segments of the present. invention may be also be combined with their respective complementary antisa nse compounds of the present in ention to form stabilized double-strandi d (dttplexed) oligonuclcotides.
[00128] Such double stranded oligonueleotide moieties have been shown in the art to modulate target expression and regulate translation as well as RNA processing via an aratisc:nse :mesh anisraa. Moreover,, the double-strande l moieties m 4y, be subject to chemical modifications (Fire et t/.. (199$) i.atnre. 391.
806-811; Timmons and Fire, (1998) X-'ware, 395, 554; Timmons et al., (2001) Gene, 263, 103-112; Tabara ei all. (1998) c ierag e , 282, 430-4,31-, Montgomery- et al., (1998) Pr=coc. ' a i..hc ,d Si USA 95. 15502-15507; Tause. al cat al, (1999) Genes Dci;., 13, 3191--3197 Eflmshir el or/., (2001) Al ature, 411. 494-498; Elbashia- c'i cd_ (2001:) Genes.D ee. 15. 188-200). For example, si.tch double-stranded moieties have been shown to inhibit the target by the classical hybridization of antisense strand of the duplex to the target, thereby triggering enzymatic degrada.tiont of the target t. T
i@sterman ei all, (2002) It--~nee, 295, 694-697).
[00129] 111 a preferred embodiment, -all. antisense o.ligoaneleonde targets .F)ystrophin family polymiclcotides (Cg.
accession number NM 0 006 and Wt 071.24). variants. alleles, isof nns, homtlogs, mutaa-n_s, derivatives, fragments and complementary sequences thereto. Preferably the oligonuclecy i.de is an antiseiise rnol.ecule.

[00130] In accordance with embodiments of the in ve:ntion, the target nucleic acid rnolecule is not limited to Dystrophin family alone but extends to any of the isofornis, receptors, honaologs avid the U x of D strophiri f roily molecule.
[00131 ] In another preferred emhodiraierit, an oligoriucleotide targets a niaitur-al atitisense sequence of DMD family polyntrcleotrdcs, for example:, polvoucleotides set forth as S<. Q ID NO: 3 to 7. and ara \ atriarrts, alleles, .homology s, tnutmits, derivatives, fiagor:ienÃs and cor plementary sequences thereto.
E.xaniples of aaaatisense oli~t?nuefeoti lcs are set firth as SEQ ID NOS 8 to 22.
[00132] In one ernbodir:nent, the ohgonue.Ieotides are complementary, to or, bind to nucleic acid sequences of 3ystr'ophin .family aritiserasc, incltreli.ri without fi rotation noncodi:ng sense and./or atitisense sequences associated with D\sttoph.in faraiily potymiclcotidcs and modulate expression znrd."'o function of DYstrophill family molecules, [00 133] In another preferred enrlbsod:iment, the ohgonuclecot:ides are complementary- to or bind to nucleic acid sequences of DM1) family natural atatisense, set forth as SEQ ID NO: 3 to 7 and modulate expression and/or function of DMD :f mily molecules.
[00134] Iii a preferred embodiment, oligonuclcotidc.s comprise sequences of at least 5 consecutive:. nucleotides of SEQ
ID NOS 8 to '22 and modulate expression and or- function of Dystropliin family molecules.
[00135] The pol arucieotide targets comprise DMD family, including family members thereof variants of DMD
family-, mum ms of DM13 family:, including SNPs noncodmg sequences of DN,.I1 i:tnril y; alleles of DMD t,aniil' species variants, fragments and the like. Preferably the ohgonucleotids: is an antisense molecule.
[00136] In another preferred eriibodiment, the of gon.rrclcotide targeting ystr'ophin family poly~nr. cleotides. comprise, antisense RNA, interference RNA (RN-Ai), short mterf nng RNA (SIR A), micro interfering RNA (n nRNA); a small, temporal RNA stRNA); or a liort, hairpin RNA (shRN.A) small RNA-induced genre activation (RNA ,, or, small activating RNA (saaRNA).
[00137] In another lan fcra-ed emboditr ent, targeting of Uystroph.in family polvnucleoudes, e.g. SEQ 11) NO: 3 to 7, modulates the expression or function of those targets. In one embodiment, expression or fu ncti:on is up-regulated as compared to a control. In another preferred en bodiment, expression or function is clown-regrulaated as compared to a control.
[00138] In another preferred embodiment, antrser:rse compounds comprise sequences set forth as SEQ ID NOS: 8 to 22. "Rem oligonucleotides can comprise one or more modified nucleotides, shorter or longer fragments-1 modified bonds and the like, [00139] In another prefen-ed en:rbc iment. SEQ ID NOS: 8 to 22 comprise one or more L.NA uucÃeotides.
[00140] The modulation of a desired target nucleic acid can be carried out in several ways known in the art, or example, antisensc oligonucfcotides, siRNA. etc. Enzymatic nucleic acid molecules ribozynaes) are nucleic acid molecules capable of catalyrz:ing one, or more of a Variety of reactions, including the ability to repeatedly cleave other se13arate lucleie acid molecules in a nuclei tide base sequence-specific anaaaner. Such enzymatic nucleic acid. molecules can be used, for example, to target vinually any RNA. transcript (Zauõ rya a:
a 24. ! a? re 429 1986, Cechh, 260,J/1,1-L-1 3(i)3O I9SS; and. .fferics et al., 17 ;'tr ale ' Adds R s=eam-h 1371, 1989).
(011141) Because of their sequence-specificity, trans .leaving enzymatic aaa:ac eic acid molecules show promise as therapeutic agents for human disease (l_isanaan MISS igt en, t l 995) Ann.
Rep. ti(c'ci. Chem, 30, 285-994;
Christoffersen and :'vIaar, (1995) J .,:fed- Chem. 38, 2(x233-21.37).
Enzymatic satin ;ic acid naolecules can be desip, ed to cleave specific RNA targets within the baackgrowad of cellular RNA. Such a cleavage event renders the niRNA non functional and abrogates Protein expression from that RNA. In this manner, synthesis of a protein associated with a disease state can be scicctivel:y inhibited.
[031421 In general, enzymatic nucleic acids with RNA cleaving activity act by.
first binding to a target RNA. Such binding occurs through the target binding portion of a. en ymaatic nucleic acid which is held in close proximity to an enzymatic portion of the molecule that acts to cleave the target RNA. Thus, the enzyaaratic nucleic acid first recognizes and then binds a target RNA. through tom l.caaacartyar base l airn atatci aice houaaad, to the correct site, acts enzw aticaliy to cut the target RNA. Strategic cleavage of such a target RNA
will destroy its ability to direct synthesis of an encoded protein. After ata. enzymatic nucleic acid has bond and cleaved its RNA target, it is released from that RNA to search for another target and can repeatedly Intl and cleave new targets.
[00143] Several approaches such as in vitro selection (evolution) strategies (Orgel, (1979) Pm c. s.. R. &x.London, B
205, 435) have been used to evolve new nucleic acid catalysts capable of carÃ:alyr in a variety of reactions, such as cleavage and ligation of phos hodiester linkages and amide; linkages, (Joyce., (1989) Gene, 82, 83-87; Bcaudry et cal:, (1992) Science 25 7,. 635-641; Joyce, (1992) Sciettlc Amt.,,r :an 267, 90-97;
Breaker eat a!õ (1994) ÃIl~'l1 t f1 12, 268:
Bartel e< at, (1993) sit: i rrc e 261:14.11._ 1418; Szost ak. (19913) II S
17,,',9-93; Kumar el al (1995) E ASE R 1, 9. 1.183;
Wreaker, (1996) Cart r (o1 ? Biotech., 7, 44-21), [00144] The development of ribozyrnes that are optimal for catalytic activity would contribute significantly to any strategy that employs .RNA-cleavin4g rib zy mes for the purpose of regulating gene expression. The hammerhead rihozyme, for example, :functions With. a catalytic rate (kcat) of about I
rain-1 in the presence of saturating (10 anM) concentrations of Mg2+ cofactor. An artificial. "RSA !.lease" ribozyme has been shown to catalyze the corresponding seifntodification reaction with a ate of about 100 vain-1. In addition,, it is known that certain modified hammerhead ribozymes that have substrate binding arms made of DNA catalyze RNA cleavage with multiple turn}-over rates that approach 1 130 min-1. Finally, replacement of a specific residue within the catalytic core of the hammerhead with certain nucleotide analogues gives modified rit ozy mes that show as much as a 10-fold improvement in c tt .aly>tic rate. These findings den-aonstr ate that ribozynacs can promote chemical.
tr<ansforrraaation -k it:h catalytic rates that are significantly 1 4m, greater than those displayed in Vitro by most natural Self-cleaving ribozymes. It is then possible that the structures of certain selfelcavingg; ribo tees may be optimized to give maximal catalytic activity, or that entirely new RNA motifs can be made that display significantly faster rates for RNA phosphodiester cleavage.
[001 45] Intermolecular cleavage of an RNA substrate by an RNA. catalyst that fits the "hanm ierhead" model first shot%a'a in 1987 (Uh.lenbeck.., 0. C.. (1987) A'z_ihi e, :a 8: 596 6t3fl).
'The R catalyst M aas recover l and .reacted N ith multiple RN molecules, demonstrating that it was tr .sly ealal tic.
[001461 Catalytic R .As designed based on the ' h taaaraaealaeail" motif have been used to cleave specific tar=get sequences by r A-iniw tppropriaate base changes in the catailytis RNA to maintain aaeccssary lase pairing with the target sequences ( aselof and Gerlach, Ã1988) :Sctlar,<, 334, L'albcrt acrd F
ratenira (l E)S }.; c itt,~t 334, 196, Uhfe:nbeck, o. C. (1987) A'atute, 32S: 5966()Ã; Koiztrnai, :M1., et a'. (1988) F B5 L(?tl., 228, 2228-230)- 1"h:1s has allow d use of the :o cataly=tic RNA to cleave specific target sequences and indicates that catalytic RNAs designed according to the "parr nerhead" model may possibly cleave specific substrate R"-As in vivo, (see Haseloff'and Gerlach, (1988):'Vcri re, 3341,585; t `alhot and. Bruening, (1 3 ;5) .'~c7la,, > 34, 196, Uhlenbeek, 0.
C. (l9 ) \at ns> 1218: 596-600).
[00147] RNA interference (RN Ai) has become a powerful tool for modulating gone expression in mammals and mammalian cells. This approach requires the deli ery of small interfering RNA
tsiRN.A.) either as RNA itself or as DNA, usi:n an expression plasnud or virus maid the coding sequence for small hair rin RNAs that are processed to siRNAs, This system enables efficient transport of the lire:-siRNAs to the cytoplasm where they are act -e and permit the u so of regulated and tissue specific promotes for -one expression.
[001481 In a preferred embodiment, an oligonucleotidc: or antisense compound comprises an oligomer or polymer of ribonucleic acid (RNA) and/or deoxyribonucleic acid (DNA`), or as mimetic, chimera, analog or homolog thereof. "t"his tern-a includes oligonu:cfeotid.es composed of naturally occurring nucleotides, sugars and covalent internuelcoside (backbone) linkages as well as oligcmucleotidcs having non-naturally occurring portions which function similarly. Such modified or substituted oligonucleotides are often desired over native fumes because of desirable properties such as, for example, enhanced cellular uptake, enhanced affinity for a target nucleic acid and increased stability in the presence of nucleases.
[00149] According to the present irnvention, the oligonucleotides or "antisense compounds" include a a anÃrsense oli4gonucleotides (e g, RI A, DNA, mimetic. chimera. analog or homolog thereof), r ibozymes, external guide- sequence (EGS) oligonucleotides, siRNA compounds, single- or doublc-stranded RNA
interference (`NY-vi; compounds such as siRNA compounds, saRNA, aRNA, aand. other oligomeric. compounds which hybridize to at least a portion of the target nucleic acid and modulate its function. As such, they may be DNA, RNA, DNA-like. RNA-like, or mixtures thereof. or may be mimetics of one or more of these. These compounds may be single-stranded, double-stranded, circular or hairpin olipomeric compounds and may contain structural Clements such as internal or terminal bulge-s,, mismatches or loops. Antisense compounds are routinely prepared linearly but can be join d or other-vise prepared to be circular and/or branched.Antiserase compo nd.s can include constructs such. a. as. for example, two strands hybridized to form a -,w--holly or l artially double-sta artded canapaaand or a single strand with sufficient sell-cornpler:aa.entaarity to. allow for hybridization and fbrrnation of a fully or partially double-stranded compound.
The two strands can be linked internally leaving free 3' or 5' tctrrairri or can be finked to fonn a. Continuous hairpin structure or loop. 'floe hairpin structtare mas, contaaira an overhang on either the 5` or 3 to =u:ainus producing an extension of single stranded character. 'The doable stranded compounds optionally can include overhangs on the ends. Further mod:iflcations can include conjugate groups attached to one of the terniini, selected nucleotide positions, sugar positions or to one of the intermicle aside finktges.
Alternativc-ly, the two strands can be linked via a non-nucleic acid moiety or linker group. Wheat formed from orals, one strand, dsRNA can take the fbrr-ri of a self-c~~araptetraeartaa hairpin-type mmmolecule that doubles hack on itse.lt to form a duplex. =Fhus, the dsRNAs can be half= or partially double stranded. Specific araodu.lation of gene expression can be achieved by stable expression of &RNA hairlpins in transtgenie cell lines (l=iaii uond et al- , { 1991) ,'4'srf. Rsrv. Genr=i.. 22, 110-119 Matzke et a/., (2001) "a.rrr. () trr. Genet. Dev., 11, 221-227; Sharp, (201) Genes Der, 15, 485-49Ã1). When formed from two strands, or a shoe le strand that takes the fi arum of a self-eomtrplementaa , hairpin-type molecule doubled back on itself to fbrna a duplex, the two strands (or duple f rming :ae4 ie3aas of a si:aagle strand) are complementary RNA.
strands that base pair in Watson-Crick fashion.
[00150] Once introduced to a system, the compounds cat the invention may elicit the action cttctaa car more enzymes or str actural proteia>s to cl :ct c.lea aae e or other modification of the target nucleic acid ormay work via occupancy-based mechanisms. In _encral, nucleic acids (including oligonuc:leoÃide.s) may he described as "MA-like" (i e_, generally having one or more 2'-deoxy sugars and, generally, T rather than U bases) or "RNA-like" (a e.., generally having one or more -hydroxyl or 2'-modified sugars and, generally U rather than I bases), Nucleic acid helices can adopt more than one type of" structure, most con mostly the A- and 134-brtras. It is believed that, in t eneraal, ohgonucleotides ",hich have -fan.-n-like structure are "DNA-like" and those Which have A-formlike structure are "RNA-like." In some. (chimeric embodiments, an antisense compound may contain both A- and fl-form regions.
[00 151] The aratisense compounds in a accordance w vittt this invention can comprise an ant sense portion from about 5 to about 80 nucleotides (i.e. from about 5 to about 80 linked nucleosides) in length. This refers to the len4gtth of the antisense strand or portion of the ant sense compound. In other words, a single-stranded .anÃisense compound of the invention comprises from 5 to about 80 nucleotides, and a double-str anded antiserase compound of the invention. (such as a dsRNA, for example) comprises a sense and an ,mtisense strand or portion of 5 to about 80 nucleotides in length.
One of ordinary skill in the art will appreciate that this comprehends antiscnse portions of 5, 6, 7,8, 9. 10. 11, 12, ]3, 14, l5; 16, h7 18, 19.20 21, 1-2,23,24, 25, 26, 27, 28, 20, 3Ã ., M, 32, 3-1, 34 >5. 30, 37, 38, 39, 4d3, 41, 42, 4-1,4"4, 451 46, 47.48, 49, 50,51 . 52, 5 54, 55, 56 57, 58, 59, 6Ã1 61.6:2 e63, 64, 65, Ã
6, 67, 68, 69.,70- 71, 72, 73,74-7r', '76, 7-7., 78, 79, or 80 nucleotides in length, or any range there vitlhin.
[00152] In one e.n bodil"nent, the. antisense compounds of the invention have antiseaase portions of 11) to 5(1 nucleotides in .length. One having ordinary skill in the,, art will appreciate that this embodies oli,gonucleotides having antiscans...

portions of .l()1 11 U. 13, 14, 116. 17, 18, 19.2()7 21,22. ? t ?4.25, 26, 27 'S, 29 30, 31. 324 33. 34, 3 5.,36, .37 38, 39, 40, 41, 421, 43, 44, 45, 46, 47, 48, 49. or 50 nucleotides in length, or any range therewithin. In some embodiments.
the oligoatucleotid!es are 15 nucleotides in length.
(001 5:3] In one emhodimmnt, the antisense or oligonareleotide compounds of the :invention have intisealsc portions of 1.2 or 13 to 330 ancleotide in length. One having ordinary skill in the art will appreciate that this embodies aantisense compounds having antisea se portions of 12, U. 14. 15, 16. 17, 18, 19, 2() 21.
222-3, 24, 25, 26, 27, 28. 29 or 3() nucleotides in length, or any range therewithin.
[00154] In another preferred embodiment, the oigomeric compounds of the present in\ ention also include vanaants in which a different base is l aesetat at one or more of the nucleotide 1 ositions in the compound. For example, if the first nucleotide is an. adenosine variants may be produced which contain thyzaaidine, guanosiane or cyt.idine at this position, This may be clone at any of the positions of the aantisense or dsR` -A
compounds, These compounds are then tested using, the methods described herein to determine their ability to inhibit expression of a target nucleic acid.
[0015,S] In some embodiratents, homology, sequence identity or complenlentarity, between the antisense compound and target is from abou 4() i) to about 60%. in some embodiralents, hon:aology, sequence identity or complervientarity, is f om about 60`to about. 70" % . Ili. some emboditraents, homology, sequence identity or complementarit}, is from about 70% to about 80%. In some eanbaodiments, homology, sequence identit or compleatlealtarity, is from about 8W';*' to aboaat 90%. In some c al ltr cfianc n.Ã:s, Itoaatcalogyõ sequence icfealti.Ã
or eoataplenlealÃ<aa its . is about ()" riq about 0 2`>.+lq about 94%, about 95' . about 990e%, about 97%. about 98 F , above 991:%, or about [00156] Iii another prefWred embodiment, the. antiscnse oligonucl :otides, such as for exauatple, nucleic acid molecules set forth in SEQ ID NOS: 3 to 22 comprise one or more substitutions or modificit oons. In one eÃn.bodinaent, the rtaac:leotidea arc substituted with locked nucleic acids {I. ` ;A}.
[00157] In another preferred embodiment, the oligonucleotides target: one or more regions of the nucleic acid molecules sense and/`or antisense of coding and: or non-codin sequences associated with T 1v'i.Ã fianlily and the sequences set forth as S.1 Q .113 NOS: 1, ., and 3 to 7. The olig-onucleotieles are also targeted to overlapping regions of Sp ID NOS -:.1.2 and :3 to 7.
[00158] Certain preferred oligonucleotides of this invention are chimeric oligonucleotides. " 'hi.aaaeric oligonucleotides" or. "chimeras," in the context of this invention, are oligonEacleotides which contain two or more cheat tally distinct redoes-,, each made tap of at least one nucleotide. These ohg,onuclcotides typically contain it least one region of modified alaacleotides that confers one or more beneficial properties (such is, for example, increased nuclease resistance, increased uptake into cells, increased binding affinity for the target) and a rÃ:. ion that is a substrate for envy<mes capable of cleaving RICA:.DNA or RICA::RNA hybrids. By way of example, RNase 14 is a cellular eaadoatuclease Which cleaves the RNA strand of an RNA:DNA duplex. Activation of RNasc I-1, there.fore, results in cleavage of the RNA target, thereby greatly enhancing the efficiency of zantisense modulation of gene expression.

Consequently, comparable results can often be obtained with shorter oligonucleotides w. hen chimeric oligomicleotides are used, compared to phosphorot ioaÃ:e dieoxyoligoanucleotdes hzybt=.idizing to the sane target region Cleavage cat the RNA ta:tgct can be routinely detected by gel clccttophoresis and. if necessary., associated nucleic acid hybridization teclantrques known it). the. an. In one preferred. enmbodinaent, a client. Wig otrsrclc.otide comprises at least one region) arc?tlti e to increase target binding affinity, and, usually, a region that acts as a substrate for RNAse U. Affinity of an (Aigo ucleotide for its tar=get (in this case, a nucleic acid encoding ra.s) is routinely determined by measuring the Tin. of all ~liy7c?rattc le title 'taZr et pair, which is the temperature at which the ciligonueleotidc and target dissociat:4; dissociation :is detected spe .trophoton ett eally. The higher the Tm, the g:rcater is the aftttattz of the oiigc nuclleotidc for the target.
w target, [00 59] Chimeric antisense cot?pounds of the invention may be forr:aaed as composite structures of (tvo or more of gsonucleotides modified oligonucleotides of gonucleos des andor oligonuclÃ
otides :n.m)-.tctics as described above.
Such, compounds have also been referred to in the art as hybrids or gaapinc;t . Rcpmsentativc United States patents that teach the preparation of such hybrid strueftires comprise, but arc not limited to, US patent nos. 5,013,830, 5,141)-,797; 5, 220.007; 2567175 366,878; 5,4113,71 1; 5,491.,133, 57565,35Q, 5,623,065; 5,652-355-1 6152,356; and 5.71 0922 each of which is herein incorix)rated by reference.
[00 16] In another preferred cmbod ment, the region of the oligotaucleotide which is modified comprises at least otic nucleotide modified- at the T position of the su4gaar, most p:rcfi.ral ly a 2' Oalkyi, 'T-0-alkyl-0-alkyl or 22'41uoro-modifiod nucleotide, in other preferred embodiments, RN.A modifications include 2`-f uoro, 'T-ainim and 2' 0-methyl modifications on the ribose of pyrimidines. .basic residues or an inverted base at the 3' end of the RNA. Such modifications are routinely incorporated into oligonucleotides and these obi gonuclcotides have been shown to have a higher 1 m (i.e., higher ta3rgcct binding if mit4') than; ?' cle~as c?lt s?aarÃc.lcotÃclcti agnu nst a g en target' Theef-Tectofsuch increased affinity is to 4greaatly enhance .RN'Ai oligonucleotide. Inhibition of gene expression..RNAse H. is a cellular endonuclease that cleaves the RNA strand of RNA:DN.A. duplexes; activation of this en`ne therefore results in cleavage of the RNA target, and thus can greatly enhance the efficiency of RNAi inhibition. Cleavage of the RN.A.
target can be routinely demonstrated by gel electrophoresis. In another preferred embodiment, the chimeric oligonuc.leotide: is also modified to enhance nuclease resistance. Cells contain a variety of exo- and endo-nucleases, which cm, degrade nucleic acids. A number of nucleotide and nucleoside modi.fi.cations have been shown to make the olio onucleotide into which they are incorporated more resistant to nuclease digestion than the native oligodeoxvnucleotide, Nuclease: resistance is routinely measured by incubating oligonucleotides with cellular extaa.cts or isolated nuclease solutions and measuring the extent of intact oligonucleotide remaining over time, usually by gel electrophoresis. O.ligonatcles?t:ides which have been modified to enhance tans: it nuclease resistance survive intact for a longer tiarae than unmodified oli4Yc?tartcleotides, A variet : of c?1igomicleotide modifications have been demonstrated to eaahmice or confer nuclease resistance. O1igonuclcotides which comain at least one phosi?horothioate modification arc presently more. preferred. In some cases, oli4go:t uc.leotide modifications which enhance target binding affinity are also, independentl , able to enhance nuclease resistance. Sonic desirable modifications can be found in. De :'vlesinaeker ; =t a+.
# I995) Acc Clae.,'rn. Res., 28366-374.
[00161] Specific: examples of some preferred ohgona.taclColl des, ennvisionecl for this invention include those Comprising modified backbones, for example, phosphorothioates, phosphotriestcrsõ methyl.
phosphouates, short chain alkyl or cycloalkyl intersugar linkages oa- short chain hcteroatomic or hcteriocyclic iritersugar link -ages. Most preferred are oligonucleotides with phosphorothioate backbones and those with beteroatoin bickbornes,, par-t-icularlyy C'.1712 .,. [-1..Yt),.Y
CH22. C'11.~ C llv>) O-CI12 known as a mctlawlerrc(rire.tlayhrariria,) or MMI
backbone. CHI --O--N" (CH3)--CI-12, CH" -.';.N' (CH3)-.N (CH.3)--CH'_) and 0.._N (CH3j--CH2 C'H2 backbones, wherein the native phosphs_diester backbone is represented as O--P__O- _CH,). 'T`he amide backbones disclosed. by 1)e iesmaeker e/ a/- (1995) Ace- C'/fem.
RRes228:36(-371 are also preferred. Also preferred are ohgonucleotides ha ving r' orpholino backbone s ruetures (Sumrrmertora and Weller, U.S. Pat, No. 5,034.,50 6), In other prefeir d einbodiments, such as the peptide nucleic acid (PNA} backbone, the phosphodiester backbone of the oligonucleotide is replaced with a polyanaidc backbone, the nucleotides being bound directly or indirectly to the air nitrogen hors of the polvari ide backbone (Nielsen cm a+.
(1991) Science 251, 1497). Oligonucleotides may also coniprise one or more substituted sugar moieties. Preferred oligonucleotides c:onaprise One of the follow Firm at the 2' position: 01-1, SH: SC'1-13. 1, C?C \. OC1-13 ()C'H3. t)C1-13 O(CH2 n CH'), O(C142)n N E2 or O(GH21)n C.H 3 where n is fi~-am I to about 10 0 to CI.t) lower alkyl., alk=.oxyalkoxy, substituted lower alkyl, akin d or ar all~o l: Cl Hr; CN CF 3 tit"p: ; O.._. S--. or 0--- S. or N-alkenyl:
SOC11,3: S02 0113; ONO: N'02-, N3; N142; heteroc cloalkyl-, heterocyclorlkary1 anitaoalkylam.ino-, poh rlk)"larr-ai.no tituted sil ): an RNA cleaving g oup- a reporter group; all inte:rcalator. a group liar improving the pharmacok.in tic sub,, properties of an oh;õortucleo ide, or ri group for irr pro =irag the pharrraacudynarr-ric properties of an oh mnuclcotide and other strbstituents having, similar properties.. preferred modification includes 2'-methoxyrethoxy 12'-0-CH2 CH2 O013, also knows as 2'-O-(2-methoxyethyl)] (martin el at, (095) lfelv. C him.
Acia, 78, 486). Other preferred modifications include 2'-methoxy (2'.O-õC1-1+), 2'- propoxy d(?'-C}C112 C112C"113) and 2'nfItatrro (2'-F). Similar modifications mays also be made at other positions on the oligonucleotide, particularly the 3' position of the sugar on the :3' terminal iitrele:otide grad the 5' positioo t of S' tcrmi:nap nuel,..otide. Olwnueleotides may also have sugar, r imeti:ca such as cyclobaattiyls in place of the peratofirrtnaosyl group.
[00162] Clli onucleotides may also include, additionally or alternatively, nucl eobase (ofien referred to in the art simply as "base") modifications or substitutions, As used herein, "unmod_ified" or "natural" nucleotides include adenine (A), guanine. (Ci), thymine ('1), cytosine. (C and. ur acil (U), Modified nucleotides include nucleotides found only infrequently or transiently in natural nucleic acids, e.g., hy posraaathine. -irietlry lacfearane, 5-.i~fe pyn'j.nidinc,;, particularly -meth lcy>tos:irae (also referred to as 5-methyl-2' deoxvcytosirte and often -referred to in the art as 5-Me-C), 5-hvdroxy:tameth -lcytosine (11MC), glycosyl H 11\11C and gerttobiosyl 111 C, as well as synthetic nucleotidas, e ,g,, 2-artiinoadenine. ?-(ni;.tlts lari7arc)raclertre, 2 or other heterosuhstittated ai.kvladeni:nes, 2 tiaioauracii. 2-thioti yn ine 5-bromouracil, 5-Ihydr-oxymmmethyiuuricil, -az an:i c, 7-dea-z aguanine. N( Ã6-atntinohexyl)adeni.tte antd 2,6-diantinnopurine.
(Kornberg,, :1., DNA Replication, W. If, Freeman Co., San Francisco, 1980, pp75-7 r , Csebeyei-au, .. (1987) et at. , rcl. A(.-Os Res. 15:4513), A " attuversal" base known in the. art, e.;7., inosine, ril ay be included. 5-'+r1e-C suhstitutions have hee i shown to increase nucleic ae.icl duplex stability by 0.6-122T. (Sangli i, Y. S., in Crooke, S. T and Lchku, B., eds., Antisense Research and ApplicaÃions, CRC Press, Boca Raton, 1993, pp. 2 76-278) and are prtse.ndy preferred base substitutions.
[00 1631 Another mtaodi:fication of the oligonuclcotides of the invention Involves chemically linking to the olicnonucleotide one or inure moieties or conjugates vvhic.h enhance the activity or cellular uptake of the oli{?onuclcotide. Such moieties include but are not limited to .lipid.
moieties such, as a el oleste.rc-il moiety. a choleste:ryi rnoicty (Letsinger et al., (19189" 11roc. A'atl, Acad. &.1. t. 5,1 86, 655-30, choke acid (il'latnoih&a n et cit. (1994) Bioor .
11ed. (7ierra. Lef. 4, 1053), a thioether, C hexy l-S-trit}, thiol Ã
viariolharan et a/. (1992) Ann.:N'T Acx:rd : Sc. i660, 306:.
'i noharan. et al. (1993) l toor'..3 rt. Chem. Let. 3, 2765), a thiocholesterol (Oberhausen tt ai., (19`.:12>) _Ack!
Rc, 20, 533), an aliphatic chain, e.g-., dodecandiol or undecyl residues (Scion--f3chmoat is et a[ EM13C) J. I 99 i , 10, 1. 11, KaIvmov> Est at t l 9YU)17s. ;y Len, 2 5) 327, S' . inavchuk t..t a (1993) Biochwic 5, 49), t phuspholipid, c,g,, di-laesaclecy`1-a~ac-glycerol or triethylaammoni:un-a 1,2-di-0-1-icxadecyl-rac-glycet=o- 3-11-phosphon ttc (Nelanobar to car at.
(1995) l tr ah dron T:e tt. 36, 3651, Shea cat at. (1990) ,NNitel. .4c tdv Res. 18, 37 +7a polyanaine or a polyethylene glycol chain (Manoh<aran et al. (1995) hicleoshles & Mueleofd e , 14, 969), or adaa antane acetic acid (i4lanoharan at at.
(1995) Tetrahedron Left. 36. 3651 ). Oligonttc.leotides comprising lipophilic moieties, and methods for preparing such oligonucie:otides are known in the art, for example, U.S. Pat. Nos. 5j 18,()45. 5,218,1Ã15 and 51.45 r,25?
[00 164] It is not necessary for all positions in a given oligonttcleotide to be uniformly modified, and in fact more than one of the aft-are:mentioned naod.ific<ations u nay be incorg)orated in a Single oligonaatcleotide or even at within a single nucleoside within an oligonucleotide. The present invention also includes oligonucleotides which are chimeric olitaonuclcotides as hereinbefore defined.
[00165] In another e.armbodinment, the nucleic acid molecule of the present inverntion is cocaiugated with another aamo ety including, but not lirnited to abasic nucleotides, polyether, poly<anaine, 1 aalyanudes, peptides, carbohydrates., lipid, or polyhydrocarhon compounds. Those skilled in the tint will recognize that these molecules can be linked to t ne or more of any nuckkotides comprising the nucleic acid molecule at sever al positions on the sugar, base or phosphate group, [00166] The oligonucleotides used in accordance with this invention may be coact niently and routinely made through the well-kraowta. technique of solid phase synthes s. Equipment for such synthesis is sold by several vendors Including Applied Biosystenms. .Any other means for such synthesis may also be employed;
the actual synthesis of the olioonttcleotides is well within the talents of one of ordinary skill in the art. It is also well. known to use similar techniques to prepare other oligonucleotides such as the phosphorothioates and alkylated derivatives. It is also well known to use similar techniques and eo:ntnmercially available modified arnidites and controlled-pore glass (CIPG) products such as biotin, fluorescein, acridine or psoraiet modifie d aaraidite s and!or CPG (available from Glen Research, Sterling VA) to synthesize f uorescently labeled., b.iotinylated or other modified oligonucleotides such as Cholesterol-modified oliporrara.le.oticles.
100167] In accordance with the invention use of :nmodificaations such as the use of LNA. monomers to enhance the potency, specificity and. duration of action and broaden the routes of administration of oligonuclleotides, comprised of current chemistries such as ?a'lf)E, ANA. F NA. PS etc (Uhlman, et as/.
(20011) Current (?puntions in l r :rg f)t:scm erg, (V
i)ev k?p!nc ra.r. Vol, 3 No 2}, This can be ~at.hieved by substittating sonic of the monomers in the current oligonucleotid es by .l NA monomers. The LNA modified oligonucleotide may have. . size sin filar to the parent compound or may be larger or preferably smaller. It is preferred that such LNA-modi.fired oli gonucleotides contain less than about 70%, more preferably less than about 6(Y' . most preferably less than about 5W'/($ 1. N
monomer-, and that their sizes are be w.s>een about 5 and 25 nucleotides, tyrore preferably between about 12 x id 211 nucleotides.
[001681 Preferred modified oligonuclcctide backbones comprise, but not limited to, phosphorothi=gates, chiral phosphoroth.ioates, phosphorodithioates, phosphotriesters araaiaaoaalk ip o l lr triesters, methyl and other al.lt =l phosphon aces comprising 3`alk rlene phosphonates and claim phosphonatcs, l~ha~sl ltnxatch phosphoratnidates cotrmapr-isMg 3'-aar wino phosphoraar.ai.d rte and amino+alkylphosphor amidates, th.ionophosphorataaidates, thionoaalk lphosphonates, thiornoalkylpliosphotriesters, and bcsranophosphates having nonnal Y-5' linkages, ''-5' finked analogs ofahese, and those having inverted polarity wherein the adjacent pairs of nucleoside units are linked 3`-5' to 5'-3' or ? - to 5'-2'. Various :salt;, mixed salts nand free acid forms are also included.
[00169] Representative United States patents that teach the preparation of the above phosphorus contairillig linkages comprise, but are not limited to, US patent nos. 3,697,808: 4,469,863; 4,476, 301, 5.023,243; 5, 177,196; 5,188,897:
677;
5,.264,42-3; 5,276,019- 5,278,302, 5,"M,717- 5,321,131- 5,399,676 5,405,939-, 5,453.496, 5,455, 233, 5,466,677-, 5.476 925: 5,519,126; 536,821; 5,541,306- 5,550j 11 5,563. 253, 571,799;
587,361. and 5,61-5,050., each of which is herein incorporated by refe -cr cc.
[00170] Preferred modified oligonuclcotide backbones that do not include a phosphorus atom therein have backbones that are. formed by short chain alkyl or cycloalk <l intemucleoside linkages, mixed hetero<atom and alkyl or cycloalkyl irrterratacleosidi linkages, or one or more short charm heterc?aatoraric or la teroa clic itatcrratFeleca cle linka s. These comprise those having moil hohno linkages (tsomi d an part .from the sugar portion of a nucleoside); siloxane backbones; sulfide, sulfoxide. and sulfuric backbones, fortnaacetyl and thiofo rnacet ,l backbones; methylene f6rmacety4 and thioformactittyl backbones; alkene containing backbones; salftmate backbones, methyleneiniino and.
3 r iethylcnehytdrazino backbones; sulfonate and. stil.fi?namide backbones;
amide backbones; and otheas having mixed N, 0, S and CH2 component parts.
1711 Representative United. States patents that teach the preparation of the above ohgontrcleosides comprise, but ire. not limited to, US patent nos. 5,03 4.506, 5,166,315; 5,185,444, 5,214,1 34; 5,216,141, 5,235,033, 5,2(A, 562 5, 26475(-.Y4, 5,405.938; 6..434,257; 5,466,677, 5,470,967; 5,489,677; 5,541 307, 5_561,225; 5,596, Ã 8 , 5,602,240-5,610,289-15,602,2240-, 5,608,0465,610,289- 5,618,70 5,623. 070, 5,663,312;
5,633,160- õ 5,677,437- and 5,677,439, each of which is herein incorporated by reference, [00 172) In other preferred oligonucleotide mirnetics, both the sugar and the.
internucleoside linka- ;e. i.e., the backbone, of the nucleotide units are replaced with novel groups. The lase units are maintained for hybridization with an appropriate nucleic acid target compound. One such olicFcan-tenc compound, an oli.gonr.rc.lcotide mi-met c that has been shown to have excellent hybridization properties, is referred to as a peptide nucleic acid (I NA). In PNA compounds, the sugar-backbone of an oligoonueleotide is replaced with. an amide containing backbone, in particular an.
an linoethylglycine backbone. T.he raueleobases are retained and are bound directly or indirectly to a-z<a. nitrogen atoms of the amide portion of the backbone. Representative United States patents that teach the preparation of P l.A compounds comprise, but are not limited to, US patent nos, 5,539 0S': 5,714,331-1 and 5, ' 19,26,2, each of which is her in incorporated t y refers race.. Further teaching of PNA compounds can be found in Nielsen, et al. (1991) c/e:=nce 254, 1497-1500.

[001731 In another preferred embodiment of the invention the oligorrucleotidc s with phosphorothioate backbones and olia rrr.tcleosides with heteroatom back-bones, and is particular- CH2-NI =() CH2-,-1;12-N (CH3)-O-01-12-known as a naethyl.ene (methylinmino) or M.-M-11 backb ne,- C'H2-O-N Ã 1133-t .`.l t?- -C`T 1 N C:f l?}-l~ t :k1;33 C'H'2-and-O. N(CH3)-C112 C:1-12- wherein the native phosphodiester backbone is represented as-O-P-O-C:.1-12- of the above referenced US
patent no. 5,489,677, and the amide backbones of the above referenced US
patent no. 5,602,240. Also preferred are oligonucleotid:es having morpholino backbone structures of the above referenced US patent no. 5,034,506, [00174] Modified oli ~rrgtrcleotides ma also contain one or more substituted sugar moieties. Preferred oligonueleotrd; s comprise one of the folloaving at the '2' position: O1-1 F 0-, S-. or : alk 1; 0-. S-, or \-alkenyl; 0-, S-or N-alkyrryl; or 0 alkyl-0-alkyl, wherein the alkyl, alkenyl and alkyrayl .tray be substituted or unsubstituted C to CO
alkyl or C2 to CO alkenyl and alkynyl. Particularly preferred are Cl (012)n OmCl-13, O(CH n,OC-'f-13.0(C l-l2)rnNH2, 0(C H2)nC'H.3, O(C H2)nO NH2, and. C)(C.H2nON(CH2)nCH3)2 where n and m can be from I to about 10. Other preferred oligonucleotides comprise one of the following at the. 2` position:
C to CO, (lower alkyl, substituted lower alkyl, aalkaryl, gar alkyl, -alka_r =l or 0-aralkyl. SH., SCH.3, OCl , Cl.
13r, CN, CF3, OCF3, SOCH>, SO2C.H3, 0N02, N02, N3; NH-2, heterocycloalkyl, heterocycloalkaryl. aminoalkylarnino, polyaalkylaamino, substituted silyl, ,in RNA
.
cleaving group, a reporter group, an inte:rcalator , a group for improving the phannacokinetic. properties of an oligonueleotide. or a group for improving the phzannacodynamic properties of an oligotaucleotide., and other substituents having similar properties. A preferred modification comprises 2 --methoxyethoxy (2 -0-0I20-12O01 , also known as n-iethoxyethyl) or 2'-MC)E (Martin et 0/., (1995) fHeiv. ( !hurt. Acto, 78, 486-5,W) 04) i.., m aalkoxvalkoxy group. A l trthc.r preferred modification comprises 2'-dimetlt `laatrrirtrrt?~~r tlat?x , i.e. a 0 C N?)2C}N(C.H3)2 (group, also known. as 2'-D;MAtlf, as described in examples herein belmv, and '2'-ditncthylaniinoetlaoxyetlaoxy (also known in the art as 2'-O-flit:Mme.Ãihy1arni:tnoetlhoxyetlhyl or T- .D.ik'L AE,OE), i.e., ?',.0_ CH2-O-Cl-l2-N CF122)2.
[00175] Other prefeaared modifications comprise. ?'-medioxy (Y-0 C H3), '2"-anunopropoxy' Ã2'-0 C.H.2CH2CH.2NH2) and 2 fhaoro (2'-p'). Similar modifications may also be made at other positions on the oiigonucieotide, particularly the Y position of the sugar on the :' Ãerrimial nucleotide or Hi 2'-5' linked ol:igonucleoticles and the 5' posiÃion of ?' t rt~..mial trUcle tide. Oii oantcleotides taaay :also have sugar n:ainaetics such as c clobaatyl moieties in place of the pentofttranosyl sugar. Reptescntattv.e United States patents that teach the preparation of such modified sugar structures comprise, but are not limited to, US patent nos. 4,9S1,957, 5,118,800, 5,319{)X(), 359,041 5,44-6,137, 5,466,7786-1 5,514, 7S5; S ,51 < .1 5,567,811, 5,57 81 61 ' : 5591-7-2211-, 57r'97,909-15,610,300-1 5k'277053; 5,6397873; s,646 267, 5,658, 3;
5,670,633; and 5,704),920, each of w.vhlc a is herein incorporated by reference.
[00176] OligonueleoÃidc.s may also comprise ntacleobase often ref rred to in the art simply as "base") modifications or substitutions, As used herein, " unmodiffled" or "natural" nuelcotides comprise the ptarin bases adenine (A) and.
guanine (G), and the pyrimidinne bases th:ymine (T'), cytosine (C.) and uracil (U).. Modified aa.tacleotides comprise other synthetic and natural nucleotides such as 5-niethylo, tttsine (5-me-C), 5-li droxymeth >l cytosine, sautlairae., hypoxatathine, 2- anainoadoniale, 0-u-tethyl and other alkyl derivatives of adenine and guaÃaine, 2-propyl and other alkyl . .
derivatives of adenine and . ,unite, :2-Ãh:aou:racil, 2-tl-atothyaaaane and '-ttt.aoey'tostrae, 5-halouractl. and ctrtosine, 5-propynyl uracil and cytosine, 6-am uracii cytosine and thynaine, 5-uracil (pseudo-uracil), 4-thiouracil, 8-halo, 8-amino, S-tlaiol, 8-thin alkyl., 8-hydroxyl and other $-subsi i itcd adenines and guata_ines, 5-halo particularly 5-bromo, ?-trifluoromethyl and other 5-substituted aatacils and cytosincs, 7-raaethylcliaanine and. 7-methylaadenitic, S-aazaguanine and S-azaadeni:ne, 7-deazaguanitic and 7-de az aadeni.nc and 3-deazaguanine and l deazaadeninc.
[00177] Further, nucleotides comprise those disclosed in United States Patent No. 3,687,808, those disclosed in 'rile Concise Encyclopedia of Polymer Science And Engineering', pages 85 SEA, K.roschwitz, J.L, ed, John Wiley & Sons, 1990, those disclosed by Etighisch et at, 'An xc\\-andie Chenare, International Edition.', 1991, 30, page- 613, and those disclosed by Saanghvi, KS., Chapter 15, Aantisertsc Research and Applications', pages 289-302, Crooke, S.T. and Leblen, B. ca., CRC Press, 1933. Certain of these:nuucleotides are particularly easeful for increasing the binding affinity of the oligomeric compounds of the. invention. These comprise 5-substituted pyrim.idines, 6- azapyrimi:dines and N-2, N-6 and tt-ti substituted purines, comprising 5- props nyku rul and 5-propsnslcytos.ine. 5_ raaethyleytositie substitutions have been shown to increase nucleic acid duplex stability by 0,6-1.2T (Sanghv Y .S., Crooke, S.T. and Lebbicu, B., eds. 'AAnÃisetise. Research and. Applications'.
CRC Press, Boca Rt-aton, 1993, pp. 276-278) and are presently preferred. base substitutions, even more particularly when .
combined a ith 2 -Omehoxy ethyl sugar modifications.
[00178] Representative United States patents that teach the preparation of the above noted modified nucleo ides as well as other modified nucleotides comprise, but are not limited to, US patent nos. 3,687 801, as well a 4,815,205;

5130,302- 5,134,066: 5,17' 271' 5, 367,066, 5,432,27211- 5,457,187: 5,459,255-5,484,908: 5,502,177, 5,525,711-5,552,540-, 5,587,469; 5,596 091: 5,614,617- 5,750,692, and 5,681,941, each of which is herein incorporated by ret.rennce.
[00179) Another modification of the oligonucleotides of the invention involves chemically linking to the oligonucleotide one or -sore moieties or conjugates, which enhance the activity, cellular ciittri.butioaa, or cellular tuptake of the oliseonucleoude.
[00180) Such moieties comprise but are not innited to, lipid moieties such as a cholesterol naoicty, (Le.tsinger et ca/ =
(1989) .1='roe" 4:ccad. S, i USA 7 86, 6553-6556), cholic acid (Mataohara:n cat e d,7 (1994) B/ootg. ..ful. 'hem..1 el., 4, 1.053-1060), a th:ioether, e.g.. hexyl-S-nitylthiol (M'Iano.hataan et cal., (11992.) Ann. A,", 5 A.c. ad. 4i/., 660, 306-309;
Manoharan et 41[, (1993:) 1 tour' . 4A% 6'itcrrr, Let:,, 3. 2765-2770), a thiocholesterol (Oherhausea tit a!,, (1992) A"ilce.
Acids Rev.. 20, 533-538), an aliphatic chain, c.g., doclccandiol or undecyl residues (K< banov car a!., (1990) .l E BS Lc n., 259, 327-330-, Sri inarclhulc of tr1., (1993) f iuc panic 75, 49-54), a phospholipid, e,g,, Ali-lae. atlee.} 1-ras - I~ eerol or trie tlay<laraara2oniaataa 1.2-di-t 3-he adec 1-rac s?:l cea -: -F 1 phc spho as to (Matnoharan et a !.. (.1995) Jth'cahedr on Left., 36, 3651 3654; Shea c.t al., (1990) `r d. Ac w& Ia rw., 18, 3777-3783), a poiyat`a tnc or a polyethylene glycol chain (Tvlanncharata ca al., (1995) Nurkosid;s & Nuc.leot:ides, 14, 969-973), or aclarnant<ane acetic acid (Manoharan ci a1,, (1995) fefrcrfr~ .lri kr Len., 36, 3651 3654), a palmityl tnc etv (Mishra et cut., (1995) .fiioc/i/rrr, B'/ophv.,. Actct, 1264, 229-237), or an oc;tadecylanmine or hexSItaaniaac~-canal orgy 1-t ox =cholesterol moiety ('rooke ct ctL, (1996).41'harmacal. I ;i7:r.
Pier.. 277, 923-93).
[00181 ] Representative United. States patents that teach he preparation of such oligonucleotides conk agates comprise, but are not limited to, US patent nos. 4,828,979; 4,948,882, 5,218,105.
5525,465; 5,541,313; 5,545,730 5,552,538-5 578,717, 580,731; 5.580,731 5,591.584; 5,109,124: 5,118,802 5.138,045:
5,414,077, 5,486, 603, 5,5 12,439:
5 578,718: 5,608,(46; 4,587,044; 4,605,735; 4,667,0225: 4,762, 779' 4,789,73-7-' 4,824,941; 4,835,263- 4,876,13-5, 4,904.582: 4,958,013: 5.082, 830; 5,112,963: 5,214,136; 5,082,830; 5,112,963;
5,214,136; 4 245.022' 5,254,469;
5,258,506: 5,262,536, 5,2 72,250, 5,292,871, 5,31%,098: 5, 3 ' 1,241, 5,391, 723 5,416,203, 5,451,463; 55104-75-1 5.512,667; 5,514,185; 5, 565,552; 5,567,810, 5,574,142-1 57585,481:
5,5877371.; 5,595, 26; 5,5977,696-1 5_59992'3'1 5,599, 928 and 5.688,941, each of which is herein incorporated by reference.
[00182] I)rv,.g cl/sir var : The compounds of the present invention can also be applied in the areas of drug dIsc0%`crV
and mrget validation. The pa sent invention comprehends the 1ase:. of the compounds and preferred target segments identified herein in drug discover) efforts to elucidate relationships that exist between Dystrophin fa.nail 3 poly>nucleotides and a disease state, phenotype, or condition These methods include detecting or modulating Dystrophin family polvanucleotieles comprising contacting a sample, tissue, cell, or or xan.ism. with the compounds of the present itnveation, measuring the nucleic acid or protein level of Dystrophin family polynucleotides and/or a related phenoty1ric or chemical endpoint at some time after ttreatmeant, and optionally comparing the measured value to a non-treated sample or sample treated With a further compound of the inventior).
These :methods can also be performed 11.1 parallel or in combination -,vith other experiments v) determine the function of unknown genes for the process of target validation or to determine the validity of a Particular geile pioduct as a target for treatment or pcc \ ention of a particular disease, condition, or phenotype.

Assessing, flu-rq,%&W(,m or .Inhibition o Gene I R 7+d;>f/on.
[00 1831 Transfer of an exogenous nucleic: acid into as host cell or or +Yaaalisrrl can be assessed by directly detecting the presence of the nucleic acid in the cell or orga ilism. Such detection can be achieved by several methods well known ill the art. For example, the presence of the exogenous nucleic acid can be detected by Southern blot or by a polymerise chain reaction (PCR) technique using printers that specifically amplify nucleotide se trencd s associated with the nucleic acid. Expression of the exogenous nucleic acids can also be measured using, conventional methods including gene expression analysis. For instance, rnRNA produced from an exogenous nucleic acid can be detected and quantif ed using a Northern blot and reverse transcription PCR (RT-l'CR), [0018 ] Expression of RN.A from the exogenous nucleic acid. can also be detected by measuring an enzymatic activity or a reporter protein actitiity. For example, antisense modulatory activity can be measured indirectly as a decrease or increase 'in target nucleic acid expression as an indication that the exogenous nucleic acid is producing the effector RNA. Based on sequence conservation. p- mcr's can be designed and used to at)lplify color g mgiIosis of the target ;er)es. IrlitiAally, the most highly expressed coding rep ion from each gene can be used to build a model control gene, although any coding or non coding region can be drsr di Each d oa)tr of gene is assembled by ~ inserting d aach coding region bet vkwwee.n a reporter coding region and its poly 3;i signal. These plasnaids would produce an mR NA with a reporter gene in the upstream portion of the gene and a potential RNAi target in the 3' non-coding region. The c fleeÃaveness of individual antisense. ol#c.~C nucl4 ot.Edes would be assayed by modulation of the reporter gene. .Reporter genes usefu in the mct:hods of the pr :sent invention include, ac.eÃoliydrox 'acid synrhase (AI-IAS), alkaline: phosphataasc (AP), beta ga.lactosidase (LacZ), beta glucoronidase (GUS), chloramp a nicol acetylta ansferase (CAT), green fluorescent protein (GH'), red fluorescent protein (UP}, yellow fluorescent protein (YFP), cyan fluorescent protein) (CH"), horseradish peroxidase (F.RP), Itaciferase (Luc.), nopafane synthase ('SOS), octopine synthasc (OCS), and derivatives thereof Multiple selectable markers are available that conifer resistance to ampicillin, bleoaaryc.in, eh.lorampheni:col, gentaaat)ycin, hygronlycin; leanarnyein, lincomvcin, methotrcxate:, phosphinothricin, pur`ou1 -cin, and tetracycline.. lvicthods to determine modulation of a reporter gene are well known in the art, and include, but are not limited to, fluororrietric methods (0~g~ fluorescence spedrtroscopy, Fluorescence Activated Cell Sorting (FA..`5), fluorescence microscLTiy), antibiotic resistance determination.
[00185] MID ID family protein and InRNA expression can be assayed using methods known to those of skill in. the art:
and described elsewhere herein, For example, irr~anrtaada issa s such as the E.LISA can be used to measure protein levvels.

I).NI.T) family= antibodies for .ELISAs are available commercially, e.g., from Abnova, (Walnut, CA), Abcam, Cambridge, MA.
[00186] In embodiments, MID fan.aily, expression (e.g., tnl .NA or protein) in a sample te..;., cells or tissues Ill Vi%vo or in vitro) treated using an antisense ohgonucleotide of the invention is evaluated by comparison with D M.I) fam:ily expression in ra control s.tnalple. For eaan ple, express ?n of the protein or aauelelc acid can be ionapau d using as a;thods k:nof;>n to those of skill in the art with that in a n:aock treated or untreated sample. Altmatively, co.a ptrisor e ith a sample treated W1ith a control ant.rsense oligonucleoticle (e.g., one laaving an altered or different scqucnce) can be made depending on the. information desired. In another embodiment, a deference in the expression of the t)N1t) far3aily protein or nucleic aced in a treated vs. an untreated sample can be compared e Vitli the difference in expression of a different nucleic acid (includin and standard deemed appropriate by the researcher, e.g., a housekeeping `ene) in a treated sample vs, an untreated sam le.
[00 1871 Observed differences can be expressed as desired, e.g., in the form of a ratio or fraction, for use in a comparison With control. in embodin-aents, the level of D%TD family na.RNA or protein, in a sample treated with all antisense olifonucleotide of the present Mention, is increased or decreased by about I.25-fold to about ..104old or more relative to an untreated sample or a sample -reated with a control nucleic acid. In embodiments, the level of DM:D
family mRNA or protein is increased or decreased by at least about 1 ?5-fi-ald, at least about 13-fold, at least about 1.4-fold, at least about 1.5-fold, at. least about 1.Ãi-fold, at least about I .7-fold, at least about 1.8-tbld, at least about 2-fold, at least about 2.5-fold, at least about 3-fold, at least about 3.5-fold, at least about 4-fold, at least about 4.5-fold, at least about 5-fold, at least about 5.5-fold, at least about 6-field, at least about 6.5-fold, at least about 7-fold, at least about 7,5-fold, at least about 8-fold, at least abut 8,5-:fold, at least about 9-fold, at least about 9.5-fold, or at least about 1Ã.I-fold or more.
Kits, Research 1 ergx ,=,,em , Diva tnos ties, and Plerapt wilc s [00188] The compounds of the present invention can be utilized for diagnostics, ther rpeutics, and prophylaxis, and as research reagents and components of kits. Furthermore- antisense oligonuclcotides, which are able to inhibit gene expression with exquisite specificity, are often used by those of ordinary, skill to elucidate the function of particular genes or to di tinguish between :functions of various members ofa biological, pathway.
[00189] For use in kits and diagnostics and in various biological systems, the cornpounds of the present insvention, either alone or in combination With other compounds or therapeutics, are useful as tools in differential and/or combinatorial analyses to elucidate expression patterns of a portion or the entire complement of genes expressed within.
cells and tissues.
[001901 As used herein the term "biological: System" or `system" is defined as any organis a1, cc-It., cell culture or tissue, that expresses, or is made competent to express Products of the Dvstrophiaa family genes. These include, but are not limited. to, humans, trarasgcnic animals, cells, cell cultures, tissues, xenografls, transplants and combinations thereof.

[0019 ] As one non limiting example, expression patterns ZviÃ.hi-n pelts or tissues treated with one or more mim,"etise eotaportndS are compared to control cells or tissues not treated with antisetrse compounds and the patterns produced are analyzed for diffea-ential levels of Beare expression as they pea-trin, for example., to disease association, signaling pathway, cellular localization, expression level., size, structure or function of the genes examined. These analy=ses can he performed on stimulated or uristamulated cells and in the presence or absence of other comlxmrid that affect expression patterns".
[00192) 'Examples of methods of gene expression analysis known in the an.
include DNA. arrays or microarrays (Braznaa and Vilo, (2000) Lett., 480, 17-24, Colas, et al., (2000) I i B:S
Len_ 48(1, 246), SAGE (serial analysis of ,gene e\prc lion) (Madden, > alr, (2000) Drug I_)iseoi 5, 415- 42-5), READS
(restriction enzyme alilplific"Ition :.o of du csted c.DN \.s) (Prash;.u and NN`eissman, f l.c 9S3) l-kIht)ils l:nzyr ful.. 3Ã3 , 258_'2), TOGA (tot i gene e: pre lion aaraal sis) (Sutc.litl' ., et cal., (2000) I'mc. N`mL A<.wi Si c. t..:S. 1.<
97, 1976-81), protein aara s and protcomics (Cells, et aL, (2000) I'BS'LeÃr., 480, 2-16; JunghluÃ, eà at, Electrophoresis, 1999, 20, 21{10-10), expressed sequence tag (p.ST) sequencing t{"clis et al., FEBS Lett-, 2000, 00, 2.1.6; l.:arssorr, et <a.l.y J. 13ioteelanol., '?ttfl, Ã). 143 ), saa_btractiv-e R:` A fingerprinting (Su1 i) 41 uchs, c>t c i. (?t tlt:t): tral. }3iocl~etxr.
286, J 1-~3 ,Larson, c t cxl , (200f11 d otrtcl!r;j` 41, w0'3 _ 208), subtr.a .tiuc clonmu, differential display. ( (1313) {;Jut .tic and Belmont, (2000) ('a're, O1?rr , ;llacrc? ?ac>i; 3, 316-21), comparative genoraric hybrid ration (Carulli, eat at,. (1.998),1. cell Biochem.:Su pL, 31 286-96 ), FISH (fluorescent in situ hyb idization) techniques (Going i nc1 (raster sir, (1999) liar .. Cancer 35, 1895-904. and mass spectromem methods (To, Comb Q N00) C/w n, High I hr ot. glrpt c Screen, 3 235-41.), [00193] The compounds of the invention are useful for research and diagnostics, because these compounds hybridize to nucleic acids encoding I)ystroph:n fytnnily. For example, oligonueleotdes that hybridize with such efficiency and under such conditions as disclosed. .herein as to be effective 13ystrophin family modulators are effective primers or probes under conditions favoring Yene amplification or detection, respectisely. These primers and probes are useful in methods requiring the specific detection of nucleic acid molecules encoding, Dystrophira firstly and in the amplification of said nucleic acid molecules fix detection or thr use in farther studies of Dystrophin family. Hybridization of the antisctre oli4?onttelecrtides, particularly the primers and pra_obes, of the invention with a nucleic acid encoding;
I3ystroph:in frnaily can be detected by means known in the art. Such means may include conjugation of an enzyme to the ohgonucleo ide, radiolaheling of the oligonucleotide, or any other suitable detection means. Kits us nsg such detection paeans for detecting the level of Dystrophin family in a sample may also be prepared.
[00194] The specificityy and sensitivity of aar.tisense are also harnessed by those of skill in the art for therapeutic uses.
Antisense compounds have been employed as therapeutic moieties in the treatment of disease states in animals, including humans. Antisense oliconuc;lec?title drugs have been safely and efk'ct?sely administered to humans and numerous Clinical trials are presently, underway. It is thus established that antasense compounds can be easeful therapeutic modalities that can be configured to be useful in treatment yet}-ianes for the treatment of cells. tissues and nrrn atl.s, especially Imman , [0019'] for therapeutics, an amnial, preferably, a human= suspected of having, a disease Or disorder~ which can be treated ley modulating the expression of Dystrophi:n family polynucicotides is tn-cated, by administe:ring arrtiser3.se:-compounds in accordance with this inventiora. For example. in one non-l:imitin errnbodirtnent, the methods cornprise the step of administering to the animal in need of treatment, a therapeutically of c.ti e amp-iount of Dystr coin. farnil modulator, The Dystrophin fiaanily modulator of the present invention effeectively, modulate the activ=ity of the i) stroph_in family, oor modulate the expression of the: E)ystrophin fanail:y protein, .h one embodiment, the activity or expression of Dystrophin family in arr. animal is inhibited by about 101;i, as compared to a control. Preferably, the activity or expression of Dystrophin family in an animal is inhibited by about ')W-;%, More pre.l-er.tbly, the activity or expression of Dystrophin family in an a animal is inhibited by 50% or more.
Thus, the oligome:.ric compounds n iodulaÃe expression of Dystrophin family mRN:A b .= at least 10%, by at least 50" 0, by at least 25%, by at least 30 ., by at least 40%, by at least 5Ã)t' , by at least 60 by at least 70%, by at least 75,1',07 by at least 80%, by at least by at least 90' . by at least 95'N',, by at least 981'.'f,, by at least 99%, or by 10t'' as compared to a control.
[00 196] In one crnbodimertt, the activity or- expression of Dystroplihi family and/or in an animal is increased by about 10% as compared to a control. Preferably, the activity, or expression of Dystrophin family in an animal is increased by about 3t i"'<,. More preferably, the activity or expression of'l yy>ytrc? lai r f tnaily in an :tariraz:tl is increased by of IIIom Thus, the oligomeric compounds modulate expression of T, ystrt3phira t ttx~i;ly n-fl ..\. by at least by at' least 50'!,k;, by at least 25'/n, E/i yt t3 E~: t~,: :t~, by at lesrst by at least 4t. "~by at least fit) : ~, by at cast blf: u, by at least by Mfr : ,:,, at least 75%, by at least 8l)'''i,, by at least 85%, by at least 9t its , by at least by at least 98%, by at least 99`%`o, or by 100% as compared to a control.
[00197] For exararple, the reduction of tile expression of Dystrophin .fancily may be measured in serum, blood, adipose tissue, liver or any other body fluid, tissue or organ of the animal, Preferably, the cells contained within said fluids, tissues or organs being analyzed contain a rrarcleie acid molecule encoding, 13y strophitr fa.mi,ly peptides and= or the Dystroph_in family protein itself:
[00198] The compounds of the invention can be utilized in phara iaceutical.
compositions by adding an effective amount of a compound to a suitable pharmaceutically acceptable diluent or carrier. Use of the c 3mpournds and methods of the invention rnay also be useful prophylactically.

[001991 Another modification of the oligonucleotides of the invention involves chem ically linking, to the oligonucleotide one or more moieties or conjugates that enhance the activityv, cellular distribution or cellular uptake of the oligonucleotide. These moieties or conjugates Can include conjugate groups eo. ales fly bound to Imictional, Lumps such as primary or secondary hydroxyl groups. Conjugate groups of the invention include. arteresa sators, reporter molecaales. po:lya miaaes, polvaaaaides, l olycthyleaae sahccok, polyethersõ
groups that enhance the phammeodynami:c properties of tali caaxacrs, and coups that enhance the lily irm ec k:iixetic properties of oll~womers. T picakon ut to dips include cholesterols, lipids, phospholipids, biotin, phenazine, Mate, phenatrthridine, aritlaraq inaone, acridine, fluorescei:ns, rhodatnines, coutaaaari:ns, and dyes. Groups that enhance the pharaaaacodynataaic properties, in the context of this ins e:ntion, include groups that improve uptake, enhance resistance to degradation, and/or stre:ngthe 3 secluence-specific. hybridization with the iarget nucleic acid. Groups that enhance the pharax-aacokinetic properties., in the context of this iaavention, iaacliacle groups that iaa.iprove upt aloe, distribbaution, metabolism or excretion of thc coanlpounds of the present invention. Representative coraj'agate groups are disclosed in la3tea aa:tiona.l Patent lapl etatie>n. No.
PC:'1l US92''09196 filed Oct. 23. 1992, and U.S. Pat. No. 6,287 86{3, \ Which are i:ncoiporated herein by reference.
Conjugate moieties include. but are not limited to, lipid moieties such as a cholesterol moietyt cholic acid, a lhioether, e.g., hcxyl=5e tritylthiol, a thioc;.holestcrol, all aliphatic chain, e.g..
dodecandiol or tandecyi residues, a phos holipid, e.g., cli lac~ac ecu1 rta<: ly'cercrl car triethylammon uata 1;? cll t l h<:~+itdccy l tae ly'cca` llphospl iactte:, a polvamme or a.
holy=ethy<lcne -glycol chain., or adamantane acetic acid, a pahni.M moicty, or an octadecyrlamine or hezylamiazo-c arhonyl-oxycholesterol iaaoiety. Ol gcniuc.leotides of the indention nary also be conÃuigated to active drug subst ices, for exataaple, aspirin, warf:aadn, plienyllautazonc, ilauprofcaa, suprokn, tenlaulfen, l e-toprofen, (S)-(4.)-pranoprofe:1), carprofen, dansylsarcosinc 2, 3,5-triio.dolaeaazoic acid, Ãlufenamic acid, folinic acid, a benzothiadiazade, chlorothiazidc, a diazcpine, indoaa:iethicin, a barbiturate, a cephalosporin, a sulfa drug, an antidiabetic, an antibactea al or an antibiotic.
[00200] Representative United States patents that teach the preparation of such ohgonueleoti:ales conjugates include, but are not limited to, U.S. Pat, Nos. 828,979; 4,948,882. 5,218,105, 5,525,465 5,541,313; 5,545,730 5,5521538:.
5,578,717, 5.580,731; 5,580,731. 5,591,584; 5,109,124; 5,11802- 5,138,045-5,414,03+7; 5486,603, 5,512,439;
5,578,718: 5,608,0346; 4,587,()44, 4,605,735, 4,667,0215- 4,7 62 _ 779, 4.789,737- 4,824,941, 4,835,263; 4,876,335-4,904,5822; 4,958,013; 5,082,83(), 5,112.9633; 5.214,136; 5,082,830; 5.11-2-963- 5,214,136; 5,245-022, 5,2'54,469;
5,258,506- 5262,536; 5,2 2.250: 5.29 .873; 53171,098, 5.371,241. 5,391-723, 5.416,2013 5,451,463; 5,510,475 5,512,667; 5514,785; 5,565,552. 5,567,810, 5.574,142; 5,585,481, 5,587,371 5 595,726; 5,597;696, 5,59 92?' 5,599.928 and 5,688,94.1.

F 7f'mula/ions [00201] The compounds of the invention may also be admixed, encapsulated, conjugated or otherwise associated with other i}:tolecules, molecule structures or mixtures of compounds, as fore aaaaple, liposomes, receptor-t4ai eted.
aaatile Talcs oral, rectal.. topical or oilier fix-n ulations, for assisting in uptake, distribution aadlor absorption.
Representative United States patents that teach the Preparation of such uptake, distribution and/or absorption-assisting 1o.nnulations, include, heat are not limited: to, U.S. P t.Nt?s. 5,108,021;
5,354,"-44; 5,416,0101; 5459,12. 5,521.291;
5.543,165; 5,547,932-1 5,583,020.; 5,591,721; 4,426,330; 4,5334.899;
5.013,556; 5,108.921 5,21-1,804-1 5 227,170;

5,264,221, 5,356,633; 5,39-5,619-7 5,416,016; 5.417,978; 5,4627854; 5.469,851;
5,512,295õ 5.5-1,528; 5,534,259;
5,543,1.52: -5,5561948- 5,580,575:. and 5,595.756, each of which is herein incorporated by reference, [00202] A.1tltoup-13, the antisense oli4gonucleotides do not :need to is administered in the context of a vector in order to modulate a target expression and/or :ftrnctiora, embodiments of the inveaa.titrra r~laates to expression vector constructs for the. expression of antisense oligornucleotides comprising prc .mote , hybrid pr mote,. gene sequences and possess a strong, cot stitut.ive , romot r- actie its`; or a promoter actin ity which can be induced in the desired case.

[00203] Ira an embodiment, invention practice in:volves administering at least one of he foregoing anrisense oliaonue.leotides with i suitable nucleic acid deliver-r, systsem. In one embodiment, that system includes a 310.11-tip iral vector operably linked to the polynucleotide. Examples of such :nonvira:l vectors include the oligonucleotide alone (e.g.
any one or more of SEQ ID NOS 8 to 22) or in combination with a suitable protein.. poly sacceliaride. or lipid tfb i-rrulation.
[00204] Additionally suitable nucleic acid delivery systems include viral vector, typically> sequence from at least one of an axdeno vi:rus aderao~i:rers assraciarted virus (AAV), helper-dependent adenovirus, retrovirus, or he:mavgglutir:ratin virus of Japan-liposome (1 VJ) complex. Preferably, the vital vector comprise s a strong eukarrvotic pro oter operably linked to the poly>rrucleotide e.g.. a eye tornegalu irus t { "'~~1V f promoter.
[00205] Additionally preferred vectors include viral. vector-s, fusion proteins and chemical eoniugates. Retrot:iral vectors include Moloney murine leukemia viruses and 1-11V-based viruses. One preferr-ed. 111 f-based viral vector comprises at least two vectors wherein the gag and po1 genes are fromn-.a an 1-V 1;e.norne and the cnv gene is from another virus. DN viral vectors are pretearccl, These vector include pox vectors such as orthopox or avrpox vectors, herpesvirus Vectors such as a herpes simplex 1 virus (HSV) vector lGcller, A.1. er' at, (1995).#::~e t.r~r.~s lrcrr 64: 487;
Lim, F. el at, in DNA (lon r~ :.l lArraanrtal.i zn Sy step s. D. Glover. Ed.
(t) .for d l rii~ _ Pre ss Oxford _Englaand) (1995).', Geller, Al. e at., (1993) Pr oc :' iaiL Acad. Sc).: 1:.5....90 7 603 Geller, A.l., et at.. ('1990) Pr m, . d e/. Acad. S ,i 1SA:
87:11401, Adenovirus Vectors (LeGal LaSalle et al.,. Science, 259:988 (1993);
Davidson, ci at.. (1993) ,fiat, Gcnei, 31-219; Yang, et al, { l }51.1: l irrrz. 69: 2004) and Adeno-associated Virus Vectors (Ka Litt, M,G., et at, ( 1994) ,Naat.
Ã<ic'neL 8:1.48).
[00206] The aantisense compounds of the invention encompass any pharmaceutically acceptable salts, esters, or salts of such esters, or any other compound which, upon administration to an animal, including a hunmaan, is capable of pro. idrngg (directly or indirectly) the biologically acti ve me ah;,rlite or residue thereof [00207] The terra "pharrraaaceuticzally acceptable salts" refers to physiologically and pharmaceutically acceptable salts of he compounds of the invention: i.e,, salts that retain the desired biological aacti,~ ity of the parent compound and do not impart undesired toxicological effects tftereto, For oligonuele:otides, preferred examples of pharmaceutically acceptable salts and their uses are further described. in U.S. Pat. No.
6,287,860, which is incorporated herein by reference.

[00208] The present invention also includes pharnaaceutieaal compositions and for-n: uiadons that include the arldsense con-pounds of the invention. The phaarniaaceu.ticaal compositions of the present invention matt be aditinistÃ.rcd in a number of ways depending upon whether local or systemic treatment is desired and 'upon the area to be treated.
Administration may be topical (including oplathaalmic and to inueous menibraties including vaginal and rectal delivery), pul-nxo:nary, e.&7 by inhaalaation or insuftlation of pow-clears or ae=rosols, including h\ ne.buli7er; i:nuatt;achcail, intumasal, epidermal and transde.rtnal), oral or parentetal. Pare.nteraal administration includes intravenous, .iiiÃriaalaÃc.riaal, subc lt4at eou , intiia eritolaeal or intranmscular injection or infusion; or intractanial, e,g., intratheeaal or Tatra-ventricular, adm inistt atiotu.
[()0209] For trea:tinaa tissues in the central nervous system, adimtii-nistration can he made by, e.g. injection or infusion :o into the cerebrospinal fluid, Mministration of antisense RNA into cerebrospinal fluid is described, U.S. Pat.
App, Pub. No. 2007/0117772, `Methods for slowing fbtamilial ALS disease progression," incorporated herein by, reference in its entirety, [00'210] When it is intended that the antisense oliaonucleotide of the present itav'eftion be administered, to cells in the.
central nervous system, administration can be with one or more agents capable of promoting penetration of the subject antisense oligonucleotide across the blood-brain harrier. f atjection can be madee. f., in the entorhinal cortex or hippoeaaaapus. Delivery of neurotrophie factors by administration of an adenovirus 'ector to Motor neurons i:n muscle tissue is described in, e.g.. U.S. Paat. No. 6,632,427 `Adenei iral-y cctor-nicdiate gene transfer into medullary motor neurons,' incorporated herein by reference. Delivery of vectors directly to the brain, e.g., the striatum, the Lhal amus, the hipposcanapus, or the substa;Lntia ntgr'a, is known in the art and. described, e.g., in U.S. Pat, No. 6,7%,523, "Aclenovirus vectors for the traanst r of foreign genes into cells of the central nervous system particularly in brain," incorporated herein by referznee Administration can be rapid as by injection or rriaade over a period of time as by slow infusion or administration of slow rele tse forriaulations.
[00211] The subject aritisense oligonucleotides can also he linked or conjugated with agents that provide desirable pharmaceutical or ph-Innacodynaa.naic properties. For example, the antisense oligonuclcotide can be coupled to any substance, known in the art to promote penetration or transport across the blood-brain Bari ier. such as an aantil,odv to the transferrin receptor, and administered by intravenous injection. The antisense compound can he linked with ,a viral vec.tor for example, that makes the antise ise compound axiore effective .and."or increases the transport of the axitisellw'' compound across the blood-brain barrier. Osmotic blood brain barrier disruption can also be accomplished by =
e.Yg.=
infusion of sugarss- including. but not limited to, n-teso et)thritol, xylitol, D(:) galactose, D(+) lactose, D(4- ) :.lose, du-lc:itol, myo.-inositol, L(-) fructose, f)(=-) n unni.tal, D(+) glucose, D(-) a:r tbinose, D(-) :uahinose, cellob:io c, D(+) maltose, D(-) ra:ftinosc, L(+) rhamnose:; D(-') Taelibiose, D(-) rabuse:, adonitol, D("-) aat ahi.Ã:ol. U-) tii ibitol, D(+) -lULvse, Lt- fireÃase, D(-) l^ \O L, L(1-) lyxose, arid L(-) l rose, or amino aacicls Including, but not thuited to-, glutasume, lysine..
atrtginine.,, aasparaa:ai_ne, aspartic acid. cysteine, glutaa:naffc acid, glyc.irie, histidine, leucine, niethioninc, phenylaa.laani:nee.

proline, serine, threonine, tyrosine, Valine, and taurine. Methods and :materials for enhancing, blood brain barrier penetration are described, e.g., in U. S. Patent No. 4,866 (.42, Method for the delivery of tgenetic material across, the blood brain barrier," 6,294,521, "Material for passage through the blood-brain barrier," < nd 6936589, "l'arenietal delivery systems.õ all incorporated herein by reference in their entirety.
[002121 The subject aatisense compounds i :tay be admixed, encapsulated, 4oujuLgated or otherwise associated with other molecules, anolecale structures or mixtures of compounds, for example, liposomes, re ceptor tar ~ a <tecl naa lccaalc s, oral, rectal, topical or other formulations, for assisting in uptake, distribution and/or absorption, For example, cationic lipids may be. included in the formulation to facrhtate oh onucleotide uptabe.
One such composition shown to facilitate uptake is UPOFEC IN (available from GIBCO-BRI., Bethesda, MD).
[00213] OligarivicleoÃid s with at least one 2 -0-riceÃhoxyethyl modification are believed to be fear Ãic-ularly useful for oral administration. Pharmaceutical cor:npositions and formulations for topical administration may include transden-nal patchesõ ointn}erns; lotions, creams, gels, drops, suppositories, sprays, liquids and pen-dcrs, Conventional pharmaceutical carriers., aqueous, powder or oily bases, thick ener:s and the like may be necessary or desirable. Coated condoms, -loves and the like may also be useful.
[00214] The, pharmaceutical formulations of the present invention. which n my conveniently be presented in unit dosage forma., may be prepared according to conventional techniques well known in the pharmaceutical Industry, Such t claraiilraes include the step of bringing p into association the active ingredients with the pharmaceutical. carrier(s) or excipient(s), In general, the formulations are prepared by uniformly and intimately bringing into association the active ingredients with liquid carriers or finely divided solid carriers or both, and then, if necessary, shaping the product.
[002 151 The compositions of the present invention may he formulated into any of many possible dosage forms such as, but not limited to, tablets, capsules., gU Capsules, liquid sy>rarps soft gels, ;upposi.tories, and menu . The compositions of the present invention may also be formulated as suspensions in aqueous, non aqueous or mixed media.
Aqueous suspensions may further contain substances that increase the viscosity of the suspension including, for example, sodium carboxyaaaethy. lcellulose, sorbitol and.'or dextrin, 'The suspension rzrayalso contain stabilizers.
[00216] Pharmaceutical compositions of the present invention include, but are not limited to, solutions, emulsions, foams and liposo ne-containing .formulations. The pharmaceutical compositions and formulations of the present invention may comprise one or more penetration enhancers, carriers, excipients or other active or inactive ingredients.
[00217] Emulsions are typically heterogeneous systems of one liquid dispersed in another in the form of droplets usually exceeding 0.1 lam in diameter. Emulsions may contain additional components in addition to the dispersed phases., and the actu e. ding that may be present as as solution in either the aqueous phase., oily phase or itself as a separate phase.:Microemulsions are included as an embodiment of the present invention. Emulsions and their arses are well known in the art and as further described in U.S. Pat. No, 6,287 860.

[00218] Formulations of the present inve-ution include 1ipo:aonaal formulations. As used its the present invention, the term 11posom " means a vesicle con posed of an phi'philic 11pids ar angeti in a spherical ilayir or bilayers. Liposomm]es are Lill. ila ilaellar or nlta.lt,larriellar vesicles vv~hach .hare a 1114"m 3braile form cd from a lipop itic mat .'E it and ~1 aqueous ulterior that contains the composition to be delivered. Cationic liposomes are positively charged. liposonaes that are believed to interact With. aievatively charted. D: A mo.lecules to forma stable complex.. Liposonaes that are pH-se_nsttiv e.
or negatively-charted are believed to entrap DNA rather than complex vv th i.Ã. Both. cationic. and noncatiollic liposo es have been used to deliver'DNA to cells.
[00219] t..iposaanaes also include "stcricaliy staabilized`t hloasonles, a to m-i which, as used herein, refer to liposomes comprising otne or more specialized lipids. When incorlxorated into liposonlec_ these sp iaize.d lipids result 111 tiposotraes v4=itl) e ilunLed circul ttiozn lifer mes relat-v-e to l po ouiesla o ng Fuch speÃ:.ialize.d lipids. : ample sol`
stetically stabilized liposo ales arc those i11 which part of the vesicle-forming lipid portion of the liposorrte comprises otac or rnott l.y%et}lipicls of is dcaivatizecl v3itla oral. of more 1lydrophilic polymers, such as a polyethylene glycol (PEG) moiety<. LiposoÃtlcs and their uses are further described in U.S, Pat. No 6,287,860.
[00'220] The phartuaeeutic. tl ft tvulations atnd compositions of the present invetntion mays also include surfacta<tnts. The use of surf. acta nts in drug products, flormula:tions artd i11 emulsions is well krao wn in the art. Surtzaetants and their uses are. further described in U S. Pat. No. 6,287,860, which is incorporated he.rei:tn by re.f rence.
[00221 ] hi one embodiment, the present invention employ: s various penetration e ahancc r a to effect the efficient delivery of nucleic acids, particularly ohgonucleotides. in addl.Ãi.on to aiding the di_tiusion of non-lipoph.ilic druts tcross cell membranes, penetration enhancers also e1:allance the perrmeability of lipophilic drugs. Penetration criftaricers rilay be classified as belonging to oone of five broad cate Tories, i.e., surfactaants, t :tty' acids, bile salts, chetatin g. agents,. and rion-chelating nonsurfactants, Penetration etahancers and. their uses are further described in U. S. Pat_ to 6,287 860, vvhich is incorporated herein by reference.
[00222] One of skill ir.t the art will recognize that fforaraulaations arc routinely designed according, to their intended use, i.e. route of administration.
[0022 3] Preferred formulations for topical administration include those in which the ol.igouucleotides of the _inventi.on aarc in a admixture s ith as tolvicaal delivery agent: such aia lipids, lipo;io acs, .fatty acids, f itrt amid esters, steroids, cheldtmng atge nts and surfactants. Prcferred lipids acid liposotaaes include neutral (e.g. dioleoyl~phosphatidyl DOP ethanolamine, diiaay istcy lphosphatid4l. eholine D-.M'Pt'r , distearo.lvphos_phat:idyl chohne) negative (e.g. ciiat3}a i\ru\ hallo lihaticty 1.
<glycerol D-IPG) and cationic e.s . ~liolet~ ttetrataletly tanlinnpr~~l 1 DOTAP aaild dial yl-phosphaatidy%l ethaanolata:iirte DOT IA).
[00224] For topical or other administration, oligormcleotides of the invention may be encapsulated within l posonies or may fbrarl complexes thereto, in particular to cationic lilxosoaae.s.
Alternatively, oliwonticleottdes may be conlplcxed.

to 11pids2 in particular to cationic lipids. Preferred fatty acids and esters, pharrraaceuticaily acceptable salts thereof, and their uses are fu flier described in U. S. Pat. No. 6,28 7,860.
[00225] Compositions and foruru.latiors for oral administration include powders or granules, rrricroparti:ertlates, naraopartierrlates, suspensions or solutions in water or non-aqueous rrrcdia, capsules, del capsules, sachets, tablets or minitablets. Thiclceoers, flavoring agents, diluents, enru sifters, dispersing aids or binders triav be desir=able, Preferred oral fora ulattions are those in which ol.igotiucleotides of the invention are administered in conjunctlorr with ore or more penCtratiorr enhancers surfactants and chelators. Preferred surfiatctants include Batty acids arid./err esters or salts thereof, bile acids and/or salts thereof. Preferred bile acids/salts and. Bitty acids and. their uses are further described in U.& Pat.
No. 6,257,860, which is incorporated herein by:reference. Also preferred are combinations of penetrat=ion enhancers, for exartrple, fatty acicls.Isahs in combination with bile aerds"salts. A
particularly preferred con-ibb ation is the sodium salt of lauric acid, capric acid and UDC A, Fur her perldratioir enhancers include pcit oxy ettlryiene-9-tarrrvI ether, polyox ethy lerr.e 2 -e.et I
etire.r. li rsuelt. rid .s of tyre in errÃion rrra be deli erect ors 11 , in xrarrtrla f rrrr inc ludin sprayed dried particles, or conrplexed to form micro or nanoparticlcs.
Oligoruclcotide co:mplexing agents and their uses are lumber described in U` S. Pat. No, 6,2K7,860, which is incorporated he=re=in by re.fereuce.
[00226] Compositions and formulations for l areuteral, intrathecal or intraventricular administration may include sterile aqueous solutions that iriay also contain. buffers, diluents and other suitable additives such as, but not limited to, Penetration enhancers., carrier compounds and other pharmaceutically acceptable carriers or x:cipierits, [00227] Certain embodiments of the invention provide pharmaceutical compositions containing one or more oligonaerie compounds and one or more other eherriotherapeutic agents that function by a nonyantiserrsc mechanism.
Examples of such chemotherapeutic agents include but are not limited to cancer-chemotherapeutic drugs such as daunoruhicin, datum:mycin, dactinomycin, doxoruhicin, epinibicin, idar .tbicin, esoruhicin, bleomyciri, mafosfamide, ifosfarnide, cytosine ar:rabinosidc, bischloroedivi nitrosurea, burulfarr, ruitomycin C. actirnrora ycin D mithr rrza cirr prednisone, hy>droxyprogesteronc, testosterone, tan-roxi.fen, daearbaxine, procarbazine:, hexcira ee thy lnre'l:traaine, per t~ttr:rc.tlryirtrclrinzirrc> it itoxzttrtrorre, tttrmsaaeriric, chlorairibucil> nre (ryle_yclcolrexvinitrostirea, n fro en xratrstards, melphal.an, c clophosphariude. 6-ramereaptopurine, 6-thicvt anirne, cytatabine, 5- azacytidine., hydnixyurca, deoxycofornayein, 4-hydroxyl?,eroxvcyclo-plaoslilroraraaide, 34luorouracil (5.Ft ), -trrcrrcrd rsy rricli:uc (5-F[JciR) methotrexate (I ITX), eolchicine, tatxol, vineristrne, virrbl rstirre, etoposide (VP-1 6), trimetrcxate, ir-inotecan, topotecan, e.rneital~ine, Ãeniposide, cisplatia and diethylstilbestrol (DES). When used with the compounds of the invention, such chemotherapeutic agents. may be used individually (e g,, 5 -FU and oliuonucleotide), sequentially 5-FU and oligonucleotide for a period of time fo.llo ved by -TT- and ol.igonticleotide), or in combination %ifh one or more other such chemotherapeutic agents (e,g 5-FU . , LMMTX and olio onucleotide, or 5411, radiotherapy and oligE>.rrr_iclcotide ..Anti inflammatory= drugs, including h=ut not limited to nonsteroidal a anti-inflzurirn.atory drugs and corticosteroids, and antiviral drugs, including but not liarit::.d to ribivi:rin.,., vidarabinc, acycl-wir and ganciclovir, may also be combined in Compositions of the invention. Combinations of aratiserase compounds and other s aratiscaase drugs are also within the scope of this invention. Two or more combine compounds may be used toggether or sequentially, t00228] In another related e bodiment, compositions of the invention may contain one cgs- more antisense compounds, particularly oligonucleotides, targeted to a first nucleic acid and. one or rraorc additional antisense compounds targeted to a second nucleic acid target. For example, the first target may be a particular autisense sequence of Dystrophin family, and the second target may he a region from another nucleotide sequence, Alternatively- , Compositions of the inv erntiorr may Contain two or more antisense compounds t4rargctc;d to di fco nt reg,ioiis of the same Dystrophin fin-lily nucleic acid taar'get. Iwlurarerous e anaples of antisense compounds are illtastrated herein and others maybe selected fi'oni among suitable coaaa aoartrds .know ra in the art. Two or more combined compounds rmay be used together or sequentiall.
[00229] The formulation of therapeutic compositions and their subsequent administration (dosing) is believed to be within the skill of those in the art. Dosing is dependent on severity and responsiveness of the disease state to be treated, with the course of treatraaent lastin from several days to sever-al montias, or until a cure is effected or a dira:ainution of the disease state is achieved. Optimal dosing schedules can be calculated from measurements of drug accumulation in the body of the patient. Persons of ordinary skill can e.asilydetermine opt:itaautaa dosages, . dosing methodologies and repetition rates. Optimum dosages may vary depending on the relative. potency of individual oligonueleoti:des, and Can generally be estimated based on EcStis found to he effective in in vitro and.
in vivo animal models. In general, dosage is from 0.01 lag to 100 g per kg of body weight, and may he given once ormore daily, d e kly, monthly or yearly, or even once every 2 to 2 ) years. Persons of ordinary skill in the art can easily estimate,, repetition rates for dosing based on measured residence times and concentrations of the drug in bodily fluids or tissues. Following successful treatment., it may be desirable to have the patient undergo maintenance therapy to prevent the recurrence of the disease state, wherein the oligonucleotide is administered in naairrtenanCe doses, ranging from 0.01 ttg to 100 1, per kg of body weight, once or more daily, to once every 20 years.
[00230] In embodiments, a patient is treated with a dosage of drug that is at least about .1, at least about 2. at least about 3., at least about at least about 5, at least about 6, at. least about 7, at least about , at lea. st ailx sat at least about 8. 9, 10, at least about 15, at least about 20, at least about 25, at least about )0, at least about 35, at least about 40, at least about 45, at least about 50, at least about 60, at least about 70, at least about 80, at least about 90, or at least about 100 nay/`kg body weight. Certain injected dosages of aaartasense oliggoraucleotides are, described, e.g., in U.S. Pat. No, 7,563,884, "Antisense modulation of I'' l'1 l e press on, ' incorporated.
herein by reference in its entirety.
[002311 While various embodiments of the present invention ha ve been described above.. it should be understood that they have been presented b.y way of example only, and not l.imitaation.
Numerous changes to the disclosed err lacaeliments can be made in accordance with the disclosure herein without departing from the spirit or scope of the irav ention. Thus, the breadth and scope of the present invention should not be limited by any of the above described canal odiments.

[00232] All documents mentioned herein are incorporated herein b reference.
All publications and patent documents cited in this application are incorporated by rck-rcnc-e for all purposes to the sane extent as if each individual publication or patent document were so individually denoted. By their citation of v::arious references in this document=
Applicants do not admit any particular ref-ere rice is "prior art" to their invention, Ernbodlmcjits of inventive compositions and methods are illustrated in the following e; anmples.
EXAMPLES
[002 33] The following uon-liataiting Examples serve to :i l:ustaate selected embodiments of the invention. It will be appreciated that variations in proportions and alternatives in elements of the components shoNvil will be apparent to those skilled in the art and are within the scope of embodiments of the present invention.

Example I: ] )sign of waivelue o//g 1tm it?F:1 1des w 'ecif1 ?r a n7. clei`c aacitdmolecule an//sense to a D strl.)phin t1 717 ;`
4711(116r (7 sense sfnmui [002'-;4] As indicated above the tern "oligonucleotide specific fog" or "oligonucleotide targets" refi.rs to an oligoaaucleotide having a sequence (i) capable of forming a stable complex with a portion of the targeted gene, or (ii) capable of forming aa. stable duplex with a portion of an niRNA transcript of the targeted gene.
[00235] Selection of appropriate oligonucleotides is facilitated by using computer programs that automatically align nucleic acid sequences and indicate regions of identity or homology. Such programs are used. to compare nucleic acid sequences obtained, for example, by searching databases. such, as GenBaank or by sequencing PCR. products.
Comparison of nucleic acid sequences from a range of species allows the -election of nucleic acid sequences that display an appropriate degree of identity bets een species, In the case of genes that have not been sequenced, Southern blots are, performed to allow a detemuination of the degree of identity' between genes in target species and other species.
By performing Southern blots at varying degrees of stringency as is well, known in the art, it is possible to obtain an approximate measure of identity. These procedures allow the selection of oligonucleotides that exhibit a bigh degree of et?raalalc:raacraÃaarit}` to target a :nucleic acid sequences in a subject to be controlled and a lower degree ofcomplcmentmitV
to corresponding nucleic acid sequences in other species. One skilled in the art. will realize that there Is considerable latitude in selecting appropriate regions of genes for use in the, present inv=ention.
[00236] An antiseaase compound is "spa,eci.ficaally hybridizable" iv-hen binding of the compound to the target nucleic aae.id intcafcars E itla tlae notaaaal fxaaactioaa cifÃlae target nucleic acid ÃÃi cause a antaeltal afion of ttani:tio-1 aaaad/oa <activits, aaacl there is a sufficient degree of conmplementaarity to avoid nonspecific binding of the antiseaasc compound to aaola-ta:rg :t nucleic acid sequences under conditions in which specific binding is desired, i.e., under physmaalogacaal conditions in the case of in vivo assays or therapeutic treatment, and under conditions in which assay,; arc performed in the case of in vitro assays [002-17) The hybridization properties of the oligonucl.eot:idly described herein cant be determined by one or Edmore in vitro ass." y-s as known in the a ii, For example, the properties of the oligonucleotides described herein call be obtained by determination of bindiaag strength between the target natural antis ise and a 1 tcntia.l drt.tg molecules using, melting curve assay.
I00238] 'l'he bi. dl >'~-' strength between the target :natural. autisense:
and a potential drug molecule ('Molecule) can be estimated usin any of the established methods of aneasurina the st}-enati of intermolecular Interactions. for example, a naeltim., curve assay.
[00'23 1 melting curve assay dcte:rnlines the temperature at which a rapid transition from cobble-stranded to single-stranded confirmation occurs for the natural a~ratisears, ylolecarlc complex.
This temperature is widely accepted as a reliable measure of the interaction strenr.th het teen the two molecules.
[00240] A meltinf curve assay c< .n he :=formed using a eDNA copy of the actual natural antisense RNA molecule or a synthetic DNA or RNA nucleotide corresponding to the binding site of the ~.lolecule. Multiple kits containing all necessary reagents to perform this assay are available (c.4g. Applied Biosystems Inc. Melt doctor kit). These kits include a suitable buffer solution containing one of the double strand DNA (dsDN A) binding dyes (such as A*BI HRM dyes.
SYBR Green. SYTO, etc.). The properties of the dsDNA dyes are such that they emit almost no fluorescence in free.
fi=rm: but are highly fluorescent when bound to dsDNA.
[00241] To perform the assay the eDNA or a corresponding ohigraanuclcoude are mi. ed with Molecule in.
concentrations defined. by the particular manufacturer's protocols. The mixture is heated to 95 'C to dissociate al pre-fonned dsDNA complexes, then slowly cooled to room temperature or other lower temperature defined by the kit manufacturer to allow" the DNA molecules to anneal. The newly formed complexes are then slowly heated to 95 `.C
wN- ith simultaneous continuous collection of data on the amount of fluorescence that is produced by he reaction. The fluorescence intensity is inversely proportional to the amounts of dsDNA
present in the reaction. The data can be collected using a real time PCR instrument compatible with the kit (e t;.:
lit's StepOne Plus Real Tinae PCR System or LisghtTy>per instrument, Roche Diagnostics, Lewes, UKK:).
[00242] Melting peaks are constructed by plotting the negative demat:ive of .fluorcscence. with respect to temperature (-d(Fi.uorescencejr `l') on the y-a-is) against temperature (x-axis) using appropriate software (forexan. plc LightTvper (Roche) or SDS Dissociation Carve, ABI). T.he data, is analyzed to identify the temperature of the rapid transition from.
dsDNA complex to single strand molecules, This temperature is called Tan and is directly proportional to the strength of interaction between the two r aolecules, Typically, Tm will exceed 40'C, P-awnp1e 2: 1-lo u ear=nrt c / I7 !~}rrr gift3 xai rtrrclzc>t i _-! f-czu1T?C'+1a' 0/ j 18,42 f eik '-vd `? anti{ nTte 01rg )rinucleotide [002431 51 lA2 cells obtained from Albert Einstein- iontefioae Cancer Center, Y were grown in growth media (MEMI`L-',BSS (Flyclorte cat #S1-130024, or Mediatech cat 4 MT-10-010-CV) 1+:113ts l'f3S Mediatech catty` MT35- 011-CV1 i enicillitr trcptsrtray eita (Nlediatech cat4 -.N4T3O-Ã 02-C1)) at 37C
and 5% C02. One day before the experiment the cells were replated at the density of L5 ` 105"..1111 into 6 well plates and incubated. at 37CC and 5 ,% C02 . On the day of the experiment the media in the 6 well plates was changed to fresh growth media. .. All antisense oligo micleotides were diluted to [tae concentration of 70 ttM. Two ld of this solution was incubated with 4(X) ItI of O1-1E'1 .medi a (Ciibco e P 319854)70) and 4 td of Lipofect,:at13it3e 20W (Iri itrogen cat,' 11668019) at rood; to 1apca azure for 20 mini a and applied to each w 'ell of the 6 well plates with 51$A2 cells, A Similar mixture inelaading2 pi of water instead of the otivonucleotide solution, was used. for the mock-transtecte . controls. After :;-18 h of incubation at 37'CC and 5'14 C02) 4.
the media was changed to fresh growth media. 48 h after addition of antis rtse ohgonucl codes the media was re.nio\ d and RNA was extracted fiom the cells using SV Total NA Isolation System from Proinega (cat 4, .3105) or RNeasy Total RICA Isolation kit fro in Qiaa4. en (cats 74181) folloi ing the ma31t factttre.rs, instructions, 600 nog of.RNA a~,as added to the reverse transcription reaction performed using Verso cl)NA kit from Thermo Scientific (catr{-AB 145313) or 1144b Capacity eDN 4 Reverse Transcription Kit (cat# 4368813 as described in the rnanufaeturer's protocol. The cDNA
'roan this revers transcription reaction was used to monitor gõ ene expression by real tim PC.R. using ,-BI Tasman Gene .Expression :Mix (eat#43fi9510) and primer /probes designed by% CBI
(Applied Biosystems Tafnnaan Gene Expression Assay: Hs001878Ã)5 n l bye Applied T3iosyste.nas inc., Foster C; it y CA). ".she .following PC: R cycle evas used:.
30 C for 2 miia, 95 C for 1 0 ialin, 40 cycles of (95 C:. filr 1 5 seconds, 60 C' .for I mi.n) using StepOne Plus Real Tinme PCR Machine (Applied T3iosy's(ea3as), Fold change in gene expressioon after tte.atnient with antisense.
oligonucleotides vas calculated based can the diference iln I8S-normalized dCt values between. treated and mock-transfec.ted samples.
[00244] Results: Reaal time PCR results show that the levels of DMO faanily aR` IIA in 518A2 cells are sign.ificantly increased 48 h afer treatment with two of the siRNAs designed to DMD fa ily aritisense 90208074. Treatmrlent with siRNAs to other aa3.tisense molecules, 9F838561, BF 768753 and BF9:50Ãi4>, did not elevate L MD family mRN A
levels (Fig I A and .B).
7r'e atrrrena o T C l cceiil.s twtth= r~irt ens ro/igii tau41e ofddc [00245] MCF-7 cells from ATCC (ca(# -1TB-22) were growtn tit growth media (M
!EBSS (Hycione cat =SH30024, or Mediatech cat # .MT-l0-010-CV) FT3S (Mediatech catii kfl'35- 0.1 t .CV) laenicilimist3etafonl}:cit7 (Med.iatech cat# 1T30-002-(=.1.)) at l7 C and w% COr. tine day, befog : the experiment the cells were replated. aat the density of 1.5 x 10'`!ml unto 6 well l fates and incubated at :37 C" and 5% CO-). On the days of the experiment the media in the 6 vell plates kzs changed to .fresh gro %Ih media. All aantisense oli onucleotides wve.re diluted to the concentration of 20 ltM. T"vo Ill of this solution was in cuba:ted with 400 tai ofOlptl TN4EN4 media (Gibco cat *'_ 19t45-070) and 4 Gal of Lipofecttamine 2000 (Inz i.trogen caatii 1166 019) at i~,aoai temperat,tre for 20 mini and applied to each well of the 6 well plates with MCF-7 cells. A Similar mixture including.2 tit of water instead.
of t e oligonueleotide olutio.n waas used. for the mock-ta ansfected conÃrols. After 3-18 h of incubation at 37 C` and 5' 'f C O t:l e median waas c hanf ed to rc h. rowth media, 48 ti ofÃer addition of antiscnse oli{gouucleotides the media was remo ed and RNA wa= extracted mean the cells using S\' Total RNA Iso.latioon System from Proniega (cat # .r' 105) or RNeasy Total RNA Isolaation kit from Qi.aagen (cat- 741.81) following the manufacturers' instructions. 600 ng of RN IA was added. to the reverse transcription reaction perfortied using Verso eDNA kit from Thermo Scientific (c at AB145 313) or High Capacity eDN'1 Reverse Transcription Kit (cat/ 4368813) as described in the manufacturer's protocol, The CDN A from this reverse transcription reaction was used to monitor gene expression by, real time PCR
using ABI "T'aclna.ata Gene Expression Mix (c:at#4-60:SlO) and primers,, probes designed by A_BI (Applied Biocssteans Itquian Gene Expression Assay, HsOl 126016 ml by Applied i3iosystems Inc_ .Foster City CA). The ffalloazing,PCR cycle was used; WC for 2 min, 95 C for 10 raaita> 40 cycles of (95"f.,: for 15 seconds, 60 C for 1 naira3 using lV x40(10 then mtaal cycler { tratta e,rac) or StepOrre Plus Real'Time.PC. f_;Maclaine (Applied Riosysteraa:s).
[00246] Fold change in gene expression after treatment with antiserase oligonucleotides was calculated based on the difference in 1$S-noramraali.zed dCt values between treated and naocle transfected samples.
[00247] Result.: Real time PCR results show that the levels of T.;TR-N in-RNA
in MC1~-7 cells are significantly increased. 48 h after treatment with siR.NAs designed to UTNN finally anttisense ENST0000t1431:319.
[00248] Although the invention has been illustrated and described with resp ct to one or more imple aaentations, equivalent alterations and a a:aodilications will occur to others skilled in the art upon the reading and understanding of this specification and the annexed drmvi.ngs. In addition, while a particular feature of the invention may have been disclosed with respect to only one of several iraaplenie:titations, such feature may be combined with one or more other features of the other implementations as may be desired and. advantageous for any given or particular application.
[00249] The Abstract of the disclosure will allow the reader to quickly ascertain the nature of the technical disclosure.
It is submitted with the understanding that it will not be used to interpret or limit tho scope or meaning of the following claims,

Claims (37)

What is claimed is:
1. A method of modulating a function of and/or the expression of a Dystrophin family polynucleotide in patient cells or tissues in vivo or in vitro comprising:
contacting said cells or tissues with at least one amiscnsc oligonucleotide 5 to 30 nucleotides in length wherein said at least one oligonucleotide has at least 50% sequence identity to a reverse complement of a polynucleotide comprising 5 to 30 consecutive nucleotides within nucleotides 1 to 378 of SEQ ID NO: 3, 1 to 294 of SEQ ID NO: 4, 1 to 686 of SEQ ID NO: 5, 1 to 480 of SEQ ID NO: 6 and 1 to 501 of SEQ IDNO: 7 (Figure 3); thereby modulating a function of and/or roe expression of the Dystrophin family polynucleotide in patient cells or tissues in vivo or in vitro.
2. A method of modulating a function of and/or the expression of a Dystrophin family polynucleotide in patient cells or tissues in vivo or in vitro comprising:
contacting said cells or tissues with at least one amiscnsc oligonucleotide 5 to 30 nucleotides in length wherein said at least one oligonucleotide has at least 50% sequence identity to a reverse complement of a natural antiscnse of a Dystrophin family polynucleotide; thereby modulating a function of and/or the expression of the Dystrophin family polynucleotide in patient ceils or tissues in vivo or in vitro.
3. A method of modulating a function of and/or the expression of a Dystrophin family polynucleotide in patient cells or tissues in vivo or in vitro comprising:
contacting said cells or tissues with at least one antisensc oligonucleotide 5 to 30 nucleotides in length wherein said oligonucleotide has at least 50% sequence identity to an antiscnse oligonucleotide to the Dystrophin family polynucleotide; thereby modulating a function of and/or the expression of the Dystrophin family polynucleotide in patient cells or tissues in vivo or in vitro.
4. A method of modulating a function of and/or the expression of a Dystrophin family polynucleotide in patient cells or tissues in vivo or in vitro comprising:
contacting said cells or tissues with at least one antisensc oligonucleotide that targets a region of a natural antiscnse oligonucleotide of the Dystrophin family polynucleotide; thereby modulating a function of and/or the expression of the Dystrophin family polynucleotide in patient cells or tissues in vivo or in vitro.
5. The method of claim 4, wherein a function of and/or the expression of the Dystrophin family is increased in vivo or in vitro with respect to a control
6. The method of claim 4, wherein the at least one antisensc oligonucleotide targets a natural antiscnse sequence of a Dystrophin family polynucleotide.
7. The method of claim 4, wherein the at least one antisensc oligonucleotide targets a nucleic acid sequence comprising coding and/or non-coding nucleic acid sequences of a Dystrophin family polynucleotide.
8.The method of claim 4, wherein the at least one antiscnse oligonucleotide targets overlapping and/or non-overlapping sequences of a Dystrophin family polynucleotide.
9. The method of claim 4, wherein the at least one antiscnse oligonucleotide comprises one or more modifications selected from: at least one modified sugar moiety, at least one modified imcmucicoside linkage, at least one modified nucleotide, and combinations thereof.
10. The method of claim 9, wherein the one or more modifications comprise at least one modified sugar moiety selected from: a 2'-O-mcthoxycthyi modified sugar moiety, a 2'-methoxy modified sugar moiety, a 2'-O-alkyl modified sugar moiety, a bicyclic sugar moiety, and combinations thereof.
11. The method of claim 9, wherein the one or more modifications comprise at least one modified intrnuclcoside linkage selected from: a phosphorothioate, 2- Omethoxyethyl (MOE), 2'-fluoro, alkylphosphonate, phosphorodithioate, alkylphosphonothioate, phosphoramidate, carbamate, carbonate, phosphate tricster, acetamidate, carboxymethyl ester, and combinations thereof .
12. The method of claim 9, wherein the one or more modifications comprise at least one modified nucleotide selected from: a peptide nucleic acid (PNA), a locked nucleic acid (LNA), an arabino-nucleic acid (PANA), an analogue, a derivative, and combinations thereof.
13. The method of claim 1, wherein the at least one oligonucleotide comprises at least one oligonucleotide sequences set forth as SEQ ID NOS: 8 to 22.
14. A method of modulating a function of and/or the expression of a Dystrophin family gene in mammalian cells or tissues in vivo or in vitro comprising:
contacting said cells or tissues with at least one short interfering RNA
(siRNA) oligonucleotide 5 to 30 nucleotides in length, said at least one siRNA oligonucleotide being specific for an antiscnse polynucleotide of a Dystrophin family polynucleotide, wherein said at least one siRNA
oligonucleotide has at least 50%
sequence identity to a complementary sequence of at least about five consecutive nucleic acids of the antisense and/or sense nucleic acid molecule of the Dystrophin family polynucleotide;
and, modulating a function of and/or the expression of Dystrophin family in mammalian cells or tissues in vivo or in vitro.
15. The method of claim 14, wherein said oligonucleotide has at least 80%
sequence identity to a sequence of at least about five consecutive nucleic acids that is complementary to the antisense and/or sense nucleic acid molecule of the Dystrophin family polynucleotide.
16. A method of modulating a function of and/or the expression of Dystrophin family in mammalian cells or tissues in vivo or in vitro comprising:
contacting said cells or tissues with at least one antisense oligonucleotide of about 5 to 30 nucleotides in length specific for noncoding and/or coding sequences of a sense and/or natural antisense strand of a Dystrophin family polynucleotide wherein said at least one antisense oligonucleotide has at least 50% sequence identity to at least one nucleic acid sequence set forth as SEQ ID NOS: 1 , 2 and 3 to 7;
and, modulating the function and/or expression of the Dystrophin family in mammalian cells or tissues in vivo or in vitro.
17.A synthetic, modified oligonucleotide comprising at least one modification wherein the at least one modification is selected from: at least one modified sugar moiety; at least one modified internuclcotide linkage; at least one modified nucleotide, and combinations thereof: wherein said oligonucleotide is an antisense compound which hybridizes to and modulates the function and/or expression of a Dystrophin family gene in vivo or in vitro as compared to a normal control.
18. The oligonucleotide of claim 17, wherein the at least one modification comprises an internucleotide linkage selected from the group consisting of: phosphorothioate, alkylphosphonate, phosphorodithioate, alkytphosphonothioate, phosphoramidate, carbamate, carbonate, phosphate triester, acetamidate, carboxymethyl ester, and combinations thereof.
19. The oligonucleotide of claim 17, wherein said oligonucleotide comprises at least one phosphorothioate internuclcotide linkage.
20. The oligonucleotide of claim 17, wherein said oligonucleotide comprises a backbone of phosphorothioate internucleotide linkages.
21. The oligonucleotide of claim 17, wherein the oligonucleotide comprises at least one modified nucleotide, said modified nucleotide selected from: a peptide nucleic acid, a locked nucleic acid (LNA), analogue, derivative, and a combination thereof.
22. The oligonucleotide of claim 17, wherein the oligonucleotide comprises a plurality of modifications, wherein said modifications comprise modified nucleotides selected from:
phosphorothioate ,alkylphospbonate, phosphorodithioate, alkylphosphonothioate, phosphoramidate, carbamate, carbonate, phosphate triester, acctamidate, carboxymethyl ester, and a combination thereof.
23. The oligonucleotide of claim 17, wherein the oligonucleotide comprises a plurality of modifications, wherein said modifications comprise modified nucleotides selected from: peptide nucleic acids,locked nucleic acids (LNA), analogues, derivatives, and a combination thereof .
24.The oligonucleotide of claim 17, wherein the oligonucleotide comprises at least one modified sugar moiety selected from: a 2'-O-methoxyethyl modified sugar moiety, a 2'-methoxy modified sugar moiety, a 2'-O-alkyl modified sugar moiety, a bicyclic sugar moiety, and a combination thereof.
25. The oligonucleotide of claim 17, wherein the oligonucleotide comprises a plurality of modifications, wherein said modifications comprise modified sugar moieties selected from: a 2'-O-methoxycthyl modified sugar moiety, a 2'-methoxy modified sugar moiety, a 2'-O-alkyl modified sugar moiety, a bicyclic sugar moiety, and a combination thereof.
26. The oligonucleotide of claim 17, wherein the oligonucleotide is of at least about 5 to 30 nucleotides in length and hybridizes to an antiscnsc and/or sense strand of a Dystrophin family polynucleotide wherein said oligonucleotide has at least about 20% sequence identity to a complementary sequence of at least about five consecutive nucleic acids of the antisense and/or sense coding and/or noncoding nucleic acid sequences of the Dystrophin family polynucleotide.
27. The oligonucleotide of claim 17, wherein the oligonucleotide has at least about 80% sequence identity to a complementary sequence of at least about five consecutive nucleic acids of the antisense and/or sense coding and/or noncoding nucleic acid sequence of the Dystrophin family polynucleotide.
28. The oligonucleotide of claim 17, wherein said oligonucleotide hybridizes to and modulates expression and/or function of at least one Dystrophin family polynucleotide in vivo or in vitro, as compared to a normal control.
29. The oligonucleotide of claim 17, wherein the oligonucleotide comprises the sequences set forth as SEQ ID
NOS: 8 to 22.
30. A composition comprising one or more oligonucleotides specific for one or more Dystrophin family polynucleotides, said polynucleotides comprising antisense sequences, complementary sequences, alleles, homologs, isoforms, variants, derivatives, mutants, fragments, or combinations thereof.
31. The composition of claim 30, wherein the oligonucleotides have at least about 40% sequence identity as compared to any one of the nucleotide sequences set forth as SEQ ID NOS: 8 to 22.
32. The composition of claim 30, wherein the oligonucleotides comprise nucleotide sequences set forth as SEQ ID
NOS: 8 to 22.
33. The composition of claim 32, wherein the oligonucleotides set forth as SEQ
ID NOS: 8 to 22 comprise one or more modifications or substitutions.
34. The composition of claim 33, wherein the one or more modifications are selected from: pbosphorothioate, methylphosphonate, peptide nucleic acid, locked nucleic acid (LNA) molecules, and combinations thereof.
35. A method of preventing or treating a disease associated with at least one Dystrophin family polynucleotide and/or at least one encoded product thereof, comprising:
administering to a patient a therapeutically effective dose of at least one antisense oligonucleotide that binds to a natural antisense sequence of said at least one Dystrophin family polynucleotide and modulates expression of said at least one Dystrophin family polynucleotide: thereby preventing or treating the disease associated with the at least one Dystrophin family polynucleotide and/or at least one encoded product thereof.
36. The method of claim 35, wherein a disease associated with the at least one Dystrophin family polynucleotide is selected from: a muscle disease or disorder (eg., Muscular dystrophy including Duchenne's muscular dystrophy, Becker's muscular dystrophy, Spinal bulbar muscular atrophy, dystrophinopathy, sarcoglycanopathy, limb girdle muscular dystrophy, congenital muscular dystrophy, congenital myopathy, distal myopathy, Symptomatic form of muscular dystrophy of Duchennc and Becker in female carriers myotonic syndrome etc.; a muscle-wasting disease), a neurological disease or disorder (including a neuromuscular disease or disorder e.g., dystonia, myoclonus-dystonia syndrome, etc.) a disease or disorder associated with altered level of dystrophin or dystrophin DAPC-complcx, Left ventricular noncompaction, cancer, a cardiovascular disease or disorder, cardiomyopathy (e.g., sporadic dilated cardiomyopathy, X-linked Dilated Cardiomyopathy (XLDC) etc.), atherosclerosis a cytoskektal disorder, congenital stationary night blindness and loss of hearing.
37. A method of identifying and selecting at least one oligonucleotide for in vivo administration comprising:
selecting a target polynucleotide associated with a disease state; identifying at least one oligonucleotide comprising at least five consecutive nucleotides which are complementary to the selected target polynucleotide or to a polynucleotide that is antisense to the selected target polynucleotide; measuring the thermal melting point of a hybrid of an antisense oligonucleotide and the target polynucleotide or the polynucleotide that is antisense to the selected target polynucleotide under stringent hybridization conditions; and selecting at least one oligonucleotide for in vivo administration based on the information obtained
CA2761248A 2009-05-08 2010-05-07 Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family Active CA2761248C (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
CA3185821A CA3185821A1 (en) 2009-05-08 2010-05-07 Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US17659409P 2009-05-08 2009-05-08
US61/176,594 2009-05-08
US31735010P 2010-03-25 2010-03-25
US61/317,350 2010-03-25
PCT/US2010/034026 WO2010129861A2 (en) 2009-05-08 2010-05-07 Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family

Related Child Applications (1)

Application Number Title Priority Date Filing Date
CA3185821A Division CA3185821A1 (en) 2009-05-08 2010-05-07 Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family

Publications (2)

Publication Number Publication Date
CA2761248A1 true CA2761248A1 (en) 2010-11-11
CA2761248C CA2761248C (en) 2023-03-14

Family

ID=43050898

Family Applications (2)

Application Number Title Priority Date Filing Date
CA2761248A Active CA2761248C (en) 2009-05-08 2010-05-07 Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family
CA3185821A Pending CA3185821A1 (en) 2009-05-08 2010-05-07 Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family

Family Applications After (1)

Application Number Title Priority Date Filing Date
CA3185821A Pending CA3185821A1 (en) 2009-05-08 2010-05-07 Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family

Country Status (8)

Country Link
US (2) US9012139B2 (en)
EP (1) EP2427554B1 (en)
JP (1) JP5931720B2 (en)
KR (1) KR101742334B1 (en)
CN (1) CN102459597B (en)
CA (2) CA2761248C (en)
ES (1) ES2618572T3 (en)
WO (1) WO2010129861A2 (en)

Families Citing this family (31)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2012065143A1 (en) 2010-11-12 2012-05-18 The General Hospital Corporation Polycomb-associated non-coding rnas
US9920317B2 (en) 2010-11-12 2018-03-20 The General Hospital Corporation Polycomb-associated non-coding RNAs
JP6188686B2 (en) * 2011-06-09 2017-08-30 カッパーアールエヌエー,インコーポレイテッド Treatment of FXN-related diseases by inhibition of natural antisense transcripts to frataxin (FXN)
BR112014028644A2 (en) 2012-05-16 2017-08-15 Rana Therapeutics Inc COMPOSITIONS AND METHODS FOR MODULATION OF ATP2A2 EXPRESSION
EA201492121A1 (en) 2012-05-16 2015-10-30 Рана Терапьютикс, Инк. COMPOSITIONS AND METHODS FOR MODULATING THE EXPRESSION OF THE FAMILY OF HEMOGLOBIN GENES
CN104540946A (en) 2012-05-16 2015-04-22 Rana医疗有限公司 Compositions and methods for modulating UTRN expression
US10837014B2 (en) 2012-05-16 2020-11-17 Translate Bio Ma, Inc. Compositions and methods for modulating SMN gene family expression
KR102028784B1 (en) 2012-05-16 2019-10-04 트랜슬레이트 바이오 인코포레이티드 Compositions and methods for modulating gene expression
US20150152410A1 (en) 2012-05-16 2015-06-04 Rana Therapeutics, Inc. Compositions and methods for modulating mecp2 expression
DK2850186T3 (en) 2012-05-16 2019-04-08 Translate Bio Ma Inc COMPOSITIONS AND PROCEDURES FOR MODULATING SMN GENFAMILY EXPRESSION
WO2014039189A1 (en) 2012-08-01 2014-03-13 Mcnally Elizabeth Mitigating tissue damage and fibrosis via latent transforming growth factor beta binding protein (ltbp4)
CN103405774A (en) * 2012-10-24 2013-11-27 尹海芳 Novel drug targeting delivery adjuvant
KR101485576B1 (en) * 2013-05-29 2015-01-21 고려대학교 산학협력단 A bacterium salmonella harboring siRNA against syntrophin beta and antitumoral composition thereof
KR101666618B1 (en) * 2013-08-16 2016-10-14 아주대학교산학협력단 Animal cell transfected with nucleotide which suppress expression of dystrophin polynucleotide
GB201410693D0 (en) 2014-06-16 2014-07-30 Univ Southampton Splicing modulation
KR102620328B1 (en) 2014-10-03 2024-01-02 콜드스프링하버러보러토리 Targeted augmentation of nuclear gene output
CA2966044A1 (en) 2014-10-30 2016-05-06 The General Hospital Corporation Methods for modulating atrx-dependent gene repression
WO2016149455A2 (en) 2015-03-17 2016-09-22 The General Hospital Corporation The rna interactome of polycomb repressive complex 1 (prc1)
TW201722439A (en) 2015-10-09 2017-07-01 波濤生命科學有限公司 Oligonucleotide compositions and methods thereof
SG11201802870RA (en) 2015-10-09 2018-05-30 Univ Southampton Modulation of gene expression and screening for deregulated protein expression
CN109312343B (en) 2015-12-14 2022-09-27 冷泉港实验室 Antisense oligomers for the treatment of autosomal dominant mental retardation type 5 and Dravet syndrome
CN106248953A (en) * 2016-07-14 2016-12-21 武汉璟泓万方堂医药科技股份有限公司 A kind of immune chromatography test card and its production and use
SG10202108375XA (en) 2017-08-25 2021-09-29 Stoke Therapeutics Inc Antisense oligomers for treatment of conditions and diseases
CA3108289A1 (en) * 2018-08-02 2020-02-06 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating facioscapulohumeral muscular dystrophy
JP2022514778A (en) 2018-12-21 2022-02-15 ノースウェスタン ユニバーシティ Use of annexin in the prevention and treatment of fascial damage
WO2020139977A1 (en) 2018-12-26 2020-07-02 Northwestern University Use of glucocorticoid steroids in preventing and treating conditions of muscle wasting, aging and metabolic disorder
US20220296631A1 (en) * 2019-05-06 2022-09-22 Antisense Therapeutics Ltd Methods for treating muscular dystrophy using inhibitory oligonucleotides to cd49d
IL298063A (en) 2020-05-11 2023-01-01 Stoke Therapeutics Inc Opa1 antisense oligomers for treatment of conditions and diseases
US11638761B2 (en) 2021-07-09 2023-05-02 Dyne Therapeutics, Inc. Muscle targeting complexes and uses thereof for treating Facioscapulohumeral muscular dystrophy
WO2023138502A1 (en) * 2022-01-20 2023-07-27 Ractigen Therapeutics Oligonucleotide modulators activating utrophin expression
CN115820642B (en) * 2022-11-11 2023-10-10 昆明理工大学 CRISPR-Cas9 system for treating Dunaliella muscular dystrophy

Family Cites Families (226)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
NZ207394A (en) 1983-03-08 1987-03-06 Commw Serum Lab Commission Detecting or determining sequence of amino acids
US4754065A (en) 1984-12-18 1988-06-28 Cetus Corporation Precursor to nucleic acid probe
US5034506A (en) 1985-03-15 1991-07-23 Anti-Gene Development Group Uncharged morpholino-based polymers having achiral intersubunit linkages
US5506337A (en) 1985-03-15 1996-04-09 Antivirals Inc. Morpholino-subunit combinatorial library and method
US4683202A (en) 1985-03-28 1987-07-28 Cetus Corporation Process for amplifying nucleic acid sequences
US4683195A (en) 1986-01-30 1987-07-28 Cetus Corporation Process for amplifying, detecting, and/or-cloning nucleic acid sequences
US4800159A (en) 1986-02-07 1989-01-24 Cetus Corporation Process for amplifying, detecting, and/or cloning nucleic acid sequences
US5288512A (en) 1987-12-15 1994-02-22 The Procter & Gamble Company Reduced calorie fats made from triglycerides containing medium and long chain fatty acids
NL8800756A (en) 1988-03-25 1989-10-16 Vereniging Voor Christelijk Wetenschappelijk Onderwijs GENETICALLY MANUFACTURED PLANT CELLS AND PLANTS AND USEABLE RECOMBINANT DNA.
US6203976B1 (en) 1989-07-18 2001-03-20 Osi Pharmaceuticals, Inc. Methods of preparing compositions comprising chemicals capable of transcriptional modulation
US5457189A (en) 1989-12-04 1995-10-10 Isis Pharmaceuticals Antisense oligonucleotide inhibition of papillomavirus
US5852188A (en) 1990-01-11 1998-12-22 Isis Pharmaceuticals, Inc. Oligonucleotides having chiral phosphorus linkages
US5459255A (en) 1990-01-11 1995-10-17 Isis Pharmaceuticals, Inc. N-2 substituted purines
US6034233A (en) 1990-05-04 2000-03-07 Isis Pharmaceuticals Inc. 2'-O-alkylated oligoribonucleotides and phosphorothioate analogs complementary to portions of the HIV genome
IE66205B1 (en) 1990-06-14 1995-12-13 Paul A Bartlett Polypeptide analogs
US5650489A (en) 1990-07-02 1997-07-22 The Arizona Board Of Regents Random bio-oligomer library, a method of synthesis thereof, and a method of use thereof
US5610289A (en) 1990-07-27 1997-03-11 Isis Pharmaceuticals, Inc. Backbone modified oligonucleotide analogues
US5218105A (en) 1990-07-27 1993-06-08 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5138045A (en) 1990-07-27 1992-08-11 Isis Pharmaceuticals Polyamine conjugated oligonucleotides
US5432272A (en) 1990-10-09 1995-07-11 Benner; Steven A. Method for incorporating into a DNA or RNA oligonucleotide using nucleotides bearing heterocyclic bases
CA2096222C (en) 1990-11-13 1998-12-29 Stephen D. Lupton Bifunctional selectable fusion genes
US6307040B1 (en) 1992-03-05 2001-10-23 Isis Pharmaceuticals, Inc. Sugar modified oligonucleotides that detect and modulate gene expression
US5474796A (en) 1991-09-04 1995-12-12 Protogene Laboratories, Inc. Method and apparatus for conducting an array of chemical reactions on a support surface
US5576302A (en) 1991-10-15 1996-11-19 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating hepatitis C virus having phosphorothioate linkages of high chiral purity
US5661134A (en) 1991-10-15 1997-08-26 Isis Pharmaceuticals, Inc. Oligonucleotides for modulating Ha-ras or Ki-ras having phosphorothioate linkages of high chiral purity
EP0538194B1 (en) 1991-10-17 1997-06-04 Novartis AG Bicyclic nucleosides, oligonucleotides, their method of preparation and intermediates therein
US6335434B1 (en) 1998-06-16 2002-01-01 Isis Pharmaceuticals, Inc., Nucleosidic and non-nucleosidic folate conjugates
US5605662A (en) 1993-11-01 1997-02-25 Nanogen, Inc. Active programmable electronic devices for molecular biological analysis and diagnostics
US5573905A (en) 1992-03-30 1996-11-12 The Scripps Research Institute Encoded combinatorial chemical libraries
IL101600A (en) 1992-04-15 2000-02-29 Yissum Res Dev Co Synthetic partially phosphorothioated antisense oligodeoxynucleotides and pharmaceutical compositions containing them
US5288514A (en) 1992-09-14 1994-02-22 The Regents Of The University Of California Solid phase and combinatorial synthesis of benzodiazepine compounds on a solid support
US6710174B2 (en) 2001-09-13 2004-03-23 Isis Pharmaceuticals, Inc. Antisense inhibition of vascular endothelial growth factor receptor-1 expression
ATE223485T1 (en) 1992-10-15 2002-09-15 Toray Industries METHOD FOR PRODUCING RECOMBINANT MHCII PROTEIN IN MICROORGANISMS
NZ266386A (en) 1993-05-11 1997-11-24 Univ North Carolina Use of antisense rna oligonucleotides in regulating gene expression
AU6953394A (en) 1993-05-21 1994-12-20 Targeted Genetics Corporation Bifunctional selectable fusion genes based on the cytosine deaminase (cd) gene
JP3484197B2 (en) 1993-09-03 2004-01-06 アイシス・ファーマシューティカルス・インコーポレーテッド Amine derivatized nucleosides and oligonucleosides
US5491084A (en) 1993-09-10 1996-02-13 The Trustees Of Columbia University In The City Of New York Uses of green-fluorescent protein
JPH09506253A (en) 1993-11-30 1997-06-24 マクギル・ユニヴァーシティ Inhibition of DNA methyltransferase
US5908779A (en) 1993-12-01 1999-06-01 University Of Connecticut Targeted RNA degradation using nuclear antisense RNA
US5519134A (en) 1994-01-11 1996-05-21 Isis Pharmaceuticals, Inc. Pyrrolidine-containing monomers and oligomers
US5593853A (en) 1994-02-09 1997-01-14 Martek Corporation Generation and screening of synthetic drug libraries
AU1925195A (en) 1994-02-22 1995-09-04 Dana-Farber Cancer Institute Nucleic acid delivery system, method of synthesis and uses thereof
US5539083A (en) 1994-02-23 1996-07-23 Isis Pharmaceuticals, Inc. Peptide nucleic acid combinatorial libraries and improved methods of synthesis
US5902880A (en) 1994-08-19 1999-05-11 Ribozyme Pharmaceuticals, Inc. RNA polymerase III-based expression of therapeutic RNAs
US6551618B2 (en) 1994-03-15 2003-04-22 University Of Birmingham Compositions and methods for delivery of agents for neuronal regeneration and survival
US6015880A (en) 1994-03-16 2000-01-18 California Institute Of Technology Method and substrate for performing multiple sequential reactions on a matrix
US5807522A (en) 1994-06-17 1998-09-15 The Board Of Trustees Of The Leland Stanford Junior University Methods for fabricating microarrays of biological samples
US5525735A (en) 1994-06-22 1996-06-11 Affymax Technologies Nv Methods for synthesizing diverse collections of pyrrolidine compounds
US5549974A (en) 1994-06-23 1996-08-27 Affymax Technologies Nv Methods for the solid phase synthesis of thiazolidinones, metathiazanones, and derivatives thereof
US6645943B1 (en) 1994-10-25 2003-11-11 Hybridon, Inc. Method of down-regulating gene expression
GB9501465D0 (en) 1995-01-25 1995-03-15 King S College London Nucleoside phosphorothioate derivatives,synthesis and use thereof
DE19502912A1 (en) 1995-01-31 1996-08-01 Hoechst Ag G-Cap Stabilized Oligonucleotides
IT1275862B1 (en) 1995-03-03 1997-10-24 Consiglio Nazionale Ricerche ANTI-SENSE TRANSCRIPT ASSOCIATED WITH SOME TYPES OF TUMOR CELLS AND SYNTHETIC OLIGODEOXYNUCLEOTIDES USEFUL IN DIAGNOSIS AND TREATMENT
IT1276642B1 (en) 1995-03-03 1997-11-03 Consiglio Nazionale Ricerche ANTI-SENSE TRANSCRIPT PRESENT IN B LYMPHOCYTES AND SYNTHETIC OLIGODEOXYNUCLEOTIDES USEFUL FOR INHIBIRING THEIR ACTION
US5739311A (en) 1995-06-07 1998-04-14 Gen-Probe Incorporated Enzymatic synthesis of phosphorothioate oligonucleotides using restriction endonucleases
US5569588A (en) 1995-08-09 1996-10-29 The Regents Of The University Of California Methods for drug screening
US20070173465A9 (en) * 1995-10-11 2007-07-26 Monahan Sean D Expression of zeta negative and zeta positive nucleic acids using a dystrophin gene
CZ243498A3 (en) 1996-02-14 1999-09-15 Isis Pharmaceuticals, Inc. Oligonucleotides with a gap and modified sugar
PT888385E (en) 1996-03-14 2004-01-30 Genentech Inc UTILIZATIONS OF GDNF AND GDNF RECEPTORS
AU2733697A (en) 1996-04-17 1997-11-07 Aronex Pharmaceuticals, Inc. Antisense inhibitors of vascular endothelial growth factor (vegf/vpf) expression
US5786213A (en) 1996-04-18 1998-07-28 Board Of Regents, The University Of Texas System Inhibition of endogenous gastrin expression for treatment of colorectal cancer
US5756710A (en) 1996-06-05 1998-05-26 The Trustees Of Columbia University In City Of New York Phosphorothioate oligonucleotides that bind to the V3-loop and uses thereof
US5898031A (en) 1996-06-06 1999-04-27 Isis Pharmaceuticals, Inc. Oligoribonucleotides for cleaving RNA
US5849902A (en) 1996-09-26 1998-12-15 Oligos Etc. Inc. Three component chimeric antisense oligonucleotides
US5739119A (en) 1996-11-15 1998-04-14 Galli; Rachel L. Antisense oligonucleotides specific for the muscarinic type 2 acetylcholine receptor MRNA
US7008776B1 (en) 1996-12-06 2006-03-07 Aventis Pharmaceuticals Inc. Compositions and methods for effecting the levels of high density lipoprotein (HDL) cholesterol and apolipoprotein AI very low density lipoprotein (VLDL) cholesterol and low density lipoprotein (LDL) cholesterol
US7235653B2 (en) 1996-12-31 2007-06-26 Isis Pharmaceuticals, Inc. Oligonucleotide compositions and methods for the modulation of the expression of B7 protein
JP3756313B2 (en) 1997-03-07 2006-03-15 武 今西 Novel bicyclonucleosides and oligonucleotide analogues
US6013786A (en) 1997-08-22 2000-01-11 Hybridon, Inc. MDM2-specific antisense oligonucleotides
DE69829760T3 (en) 1997-09-12 2016-04-14 Exiqon A/S BI- AND TRI-CYCLIC-NUCLEOSIDE, NUCLEOTIDE AND OLIGONUCLEOTIDE ANALOG
US7572582B2 (en) 1997-09-12 2009-08-11 Exiqon A/S Oligonucleotide analogues
US6794499B2 (en) 1997-09-12 2004-09-21 Exiqon A/S Oligonucleotide analogues
US7285288B1 (en) 1997-10-03 2007-10-23 Board Of Regents, The University Of Texas System Inhibition of Bcl-2 protein expression by liposomal antisense oligodeoxynucleotides
US6034883A (en) 1998-01-29 2000-03-07 Tinney; Charles E. Solid state director for beams
US6175409B1 (en) 1999-04-02 2001-01-16 Symyx Technologies, Inc. Flow-injection analysis and variable-flow light-scattering methods and apparatus for characterizing polymers
US7321828B2 (en) 1998-04-13 2008-01-22 Isis Pharmaceuticals, Inc. System of components for preparing oligonucleotides
US20040186071A1 (en) 1998-04-13 2004-09-23 Bennett C. Frank Antisense modulation of CD40 expression
US6221587B1 (en) 1998-05-12 2001-04-24 Isis Pharmceuticals, Inc. Identification of molecular interaction sites in RNA for novel drug discovery
US6833361B2 (en) 1998-05-26 2004-12-21 Ribapharm, Inc. Nucleosides having bicyclic sugar moiety
HUP0102152A3 (en) 1998-05-26 2002-04-29 Icn Pharmaceuticals Inc Costa Nucleosid and oligo nucleotid analogues having bicyclic sugar derivative
US20030139359A1 (en) 2001-12-04 2003-07-24 Isis Pharmaceuticals Inc. Antisense modulation of phospholipid scramblase 3 expression
US6100090A (en) 1999-06-25 2000-08-08 Isis Pharmaceuticals Inc. Antisense inhibition of PI3K p85 expression
US6867294B1 (en) 1998-07-14 2005-03-15 Isis Pharmaceuticals, Inc. Gapped oligomers having site specific chiral phosphorothioate internucleoside linkages
US6242589B1 (en) 1998-07-14 2001-06-05 Isis Pharmaceuticals, Inc. Phosphorothioate oligonucleotides having modified internucleoside linkages
US6214986B1 (en) 1998-10-07 2001-04-10 Isis Pharmaceuticals, Inc. Antisense modulation of bcl-x expression
US6376541B1 (en) 1998-11-06 2002-04-23 Alcon Manufacturing, Ltd. Upregulation of endogenous prostaglandins to lower intraocular pressure
US5985663A (en) 1998-11-25 1999-11-16 Isis Pharmaceuticals Inc. Antisense inhibition of interleukin-15 expression
DE60021700T2 (en) 1999-01-27 2006-06-01 Becker, David, Dr., Abbots Langley FORMULATIONS CONTAINING ANTISENSE NUCLEOTIDE SPECIFIC FOR CONNEXINE
CN1273478C (en) 1999-02-12 2006-09-06 三共株式会社 Novel nucleosides and oligonucleotide analogues
CA2850318A1 (en) 1999-02-26 2000-08-31 The University Of British Columbia Trpm-2 antisense therapy
US20040137423A1 (en) 1999-03-15 2004-07-15 Hayden Michael R. Compositions and methods for modulating HDL cholesterol and triglyceride levels
EP1100895B1 (en) 1999-03-15 2007-06-13 University of British Columbia Abc1 polypeptide and methods and reagents for modulating cholesterol levels
KR20020007331A (en) 1999-03-18 2002-01-26 추후제출 Detection of mutations in genes by specific LNA primers
US7084125B2 (en) 1999-03-18 2006-08-01 Exiqon A/S Xylo-LNA analogues
US6734291B2 (en) 1999-03-24 2004-05-11 Exiqon A/S Synthesis of [2.2.1]bicyclo nucleosides
US6639059B1 (en) 1999-03-24 2003-10-28 Exiqon A/S Synthesis of [2.2.1]bicyclo nucleosides
BR0009333A (en) 1999-03-26 2002-01-08 Aventis Pharm Prod Inc Composition to reduce lipg gene expulsion and the enzymatic activity of the lipg polypeptide, to inhibit the enzymatic activity of the lipg polypeptide, to increase the level and enzymatic activity of the lipg polypeptide in a patient and methods to increase the cholesterol level of high density lipoprotein and apolipoprotein ai, to reduce the level of very low density lipoprotein cholesterol and ldl cholesterol and vldl cholesterol in a patient, to diagnose a predisposition to low levels of hdl cholesterol and apolipoprotein ai, and to determine whether a test compound can inhibit the enzymatic reaction between lipg polypeptide and hdl cholesterol and apolipoprotein ai and can increase the enzymatic reaction between lipg polypeptide and vldl and vdl cholesterol
ES2300261T3 (en) 1999-04-08 2008-06-16 Novartis Vaccines And Diagnostics, Inc. POTENTIAL OF THE IMMUNE RESPONSE FOR APPLICATIONS OF VACCINES AND GENETIC THERAPY.
US6977295B2 (en) 1999-04-21 2005-12-20 Invitrogen Corporation Locked nucleic acid hybrids and methods of use
DK1178999T3 (en) 1999-05-04 2007-08-06 Santaris Pharma As L-ribo-LNA analogues
US20030233670A1 (en) 2001-12-04 2003-12-18 Edgerton Michael D. Gene sequences and uses thereof in plants
US6525191B1 (en) 1999-05-11 2003-02-25 Kanda S. Ramasamy Conformationally constrained L-nucleosides
DE19925073C2 (en) 1999-06-01 2001-07-19 Stefan Weiss Nucleic acid molecules with specific recognition of native PrP · S ·· c ·, production and use
US6656730B1 (en) 1999-06-15 2003-12-02 Isis Pharmaceuticals, Inc. Oligonucleotides conjugated to protein-binding drugs
EP1189940A2 (en) 1999-06-25 2002-03-27 Genset A bap28 gene and protein
US20040006031A1 (en) 2002-07-02 2004-01-08 Isis Pharmaceuticals Inc. Antisense modulation of HMG-CoA reductase expression
US6147200A (en) 1999-08-19 2000-11-14 Isis Pharmaceuticals, Inc. 2'-O-acetamido modified monomers and oligomers
EP1218411A4 (en) 1999-09-20 2004-09-01 Millennium Pharm Inc Secreted proteins and uses thereof
US6617442B1 (en) 1999-09-30 2003-09-09 Isis Pharmaceuticals, Inc. Human Rnase H1 and oligonucleotide compositions thereof
WO2001025488A2 (en) 1999-10-06 2001-04-12 Quark Biotech, Inc. Method for enrichment of natural antisense messenger rna
US6986988B2 (en) 1999-10-06 2006-01-17 Quark Biotech, Inc. Method for enrichment of natural antisense messenger RNA
AU2778801A (en) 2000-01-07 2001-07-24 Baylor University Antisense compositions and methods
DE60122285D1 (en) 2000-01-14 2006-09-28 Us Gov Health & Human Serv METHONOCARBACYCLOALKYLANALOGA OF NUCLEOSIDES
US6303374B1 (en) 2000-01-18 2001-10-16 Isis Pharmaceuticals Inc. Antisense modulation of caspase 3 expression
WO2005070136A2 (en) 2004-01-12 2005-08-04 The Trustees Of The University Of Pennsylvania Up-regulation of bone morphogenetic protein (bmp) gene expression in bone cells by electromagnetic signals
JP2001247459A (en) 2000-03-03 2001-09-11 Oakland Uniservices Ltd Combination therapy for cancer
US6936467B2 (en) 2000-03-27 2005-08-30 University Of Delaware Targeted chromosomal genomic alterations with modified single stranded oligonucleotides
AU4948801A (en) 2000-03-27 2001-10-08 Univ Delaware Targeted chromosomal genomic alterations with modified single stranded oligonucleotides
US7402434B2 (en) 2000-05-08 2008-07-22 Newman Stuart A Splice choice antagonists as therapeutic agents
WO2002062840A1 (en) 2000-06-29 2002-08-15 Pharma Pacific Pty. Ltd. INTERFERON-α INDUCED GENE
WO2002009700A1 (en) 2000-07-28 2002-02-07 Cancer Research Technology Limited Cancer treatment by combination therapy
EP1314734A1 (en) 2000-08-29 2003-05-28 Takeshi Imanishi Novel nucleoside analogs and oligonucleotide derivatives containing these analogs
JP2004507263A (en) 2000-09-02 2004-03-11 グリュネンタール・ゲゼルシャフト・ミト・ベシュレンクテル・ハフツング Antisense oligonucleotides to VR1
US6444464B1 (en) 2000-09-08 2002-09-03 Isis Pharmaceuticals, Inc. Antisense modulation of E2F transcription factor 2 expression
AU2001289085A1 (en) 2000-09-20 2002-04-02 Isis Pharmaceuticals, Inc. Antisense modulation of flip-c expression
EP1325121A2 (en) 2000-10-13 2003-07-09 Institut de Cardiologie de Montreal Antisense oligonucleotide directed toward mammalian vegf receptor genes and uses thereof
US20030228618A1 (en) 2000-11-24 2003-12-11 Erez Levanon Methods and systems for identifying naturally occurring antisense transcripts and methods, kits and arrays utilizing same
US20050222029A1 (en) 2001-01-04 2005-10-06 Myriad Genetics, Incorporated Compositions and methods for treating diseases
US7423142B2 (en) 2001-01-09 2008-09-09 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of anti-apoptotic genes
WO2002068470A2 (en) 2001-02-26 2002-09-06 Pharma Pacific Pty Ltd Interferon-alpha induced gene
US20020147165A1 (en) 2001-02-22 2002-10-10 Isis Pharmaceuticals, Inc. Antisense modulation of calreticulin expression
AUPR497101A0 (en) 2001-05-14 2001-06-07 Queensland University Of Technology Polynucleotides and polypeptides linked to cancer and/or tumorigenesi
IL143379A (en) 2001-05-24 2013-11-28 Yissum Res Dev Co Antisense oligonucleotide against the r isophorm of human ache and uses thereof
US7053195B1 (en) 2001-06-12 2006-05-30 Syngenta Participatious Ag Locked nucleic acid containing heteropolymers and related methods
US7153954B2 (en) 2001-07-12 2006-12-26 Santaris Pharma A/S Method for preparation of LNA phosphoramidites
CA2453183C (en) 2001-07-12 2016-05-10 University Of Massachusetts In vivo production of small interfering rnas that mediate gene silencing
US7425545B2 (en) 2001-07-25 2008-09-16 Isis Pharmaceuticals, Inc. Modulation of C-reactive protein expression
US20030096772A1 (en) 2001-07-30 2003-05-22 Crooke Rosanne M. Antisense modulation of acyl CoA cholesterol acyltransferase-2 expression
US7259150B2 (en) 2001-08-07 2007-08-21 Isis Pharmaceuticals, Inc. Modulation of apolipoprotein (a) expression
WO2003020739A2 (en) 2001-09-04 2003-03-13 Exiqon A/S Novel lna compositions and uses thereof
US20040214766A1 (en) 2001-10-01 2004-10-28 Kari Alitalo VEGF-C or VEGF-D materials and methods for treatment of neuropathologies
CN1635895A (en) 2001-10-10 2005-07-06 雀巢产品有限公司 Coffee plant with reduced alpha-D-galactosidase activity
US7125982B1 (en) 2001-12-05 2006-10-24 Frayne Consultants Microbial production of nuclease resistant DNA, RNA, and oligo mixtures
US6965025B2 (en) 2001-12-10 2005-11-15 Isis Pharmaceuticals, Inc. Antisense modulation of connective tissue growth factor expression
CA2365811A1 (en) 2001-12-21 2003-06-21 Institut De Cardiologie A new gene therapy using antisense strategy to estrogen receptors (er .alpha. and/or er .beta.) to optimize vascular healing and cardioprotection after vascular injury
KR20030056538A (en) 2001-12-28 2003-07-04 주식회사 웰진 EFFECTIVE INHIBITION OF TRANSFORMING GROWTH FACTOR-β1 BY A RIBBON-TYPE ANTISENSE OLIGONUCLEOTIDE
US20030191075A1 (en) 2002-02-22 2003-10-09 Cook Phillip Dan Method of using modified oligonucleotides for hepatic delivery
AU2003212729A1 (en) 2002-02-25 2003-09-09 Gunnar Norstedt Vitamin d upregulated protein 1 (vdup-1) methods and uses thereof
AU2003225701A1 (en) 2002-03-08 2003-09-22 Glen Research Corporation Fluorescent nitrogenous base and nucleosides incorporating same
GB2386836B (en) 2002-03-22 2006-07-26 Cancer Res Ventures Ltd Anti-cancer combinations
US7169916B2 (en) 2002-04-01 2007-01-30 Isis Pharmaceuticals, Inc. Chloral-free DCA in oligonucleotide synthesis
US20050215504A1 (en) 2002-04-02 2005-09-29 Bennett C F Antisense modulation of sterol regulatory element-binding protein-1 expression
JP4338527B2 (en) 2002-04-05 2009-10-07 サンタリス ファーマ アー/エス Oligomer compounds that regulate HIF-1α expression
US7569575B2 (en) 2002-05-08 2009-08-04 Santaris Pharma A/S Synthesis of locked nucleic acid derivatives
US6808906B2 (en) 2002-05-08 2004-10-26 Rigel Pharmaceuticals, Inc. Directionally cloned random cDNA expression vector libraries, compositions and methods of use
US7199107B2 (en) 2002-05-23 2007-04-03 Isis Pharmaceuticals, Inc. Antisense modulation of kinesin-like 1 expression
US7148342B2 (en) 2002-07-24 2006-12-12 The Trustees Of The University Of Pennyslvania Compositions and methods for sirna inhibition of angiogenesis
US20040033480A1 (en) 2002-08-15 2004-02-19 Wong Norman C.W. Use of resveratrol to regulate expression of apolipoprotein A1
EP1555870A4 (en) 2002-09-10 2006-11-02 Samuel Roberts Noble Found Inc Methods and compositions for production of flavonoid and isoflavonoid nutraceuticals
JP2006500070A (en) 2002-09-25 2006-01-05 ファルマシア・コーポレーション Antisense regulation of farnesoid X receptor expression
US7229976B2 (en) 2002-09-26 2007-06-12 Isis Pharmaceuticals, Inc. Modulation of forkhead box O1A expression
KR20120038546A (en) 2002-11-01 2012-04-23 더 트러스티스 오브 더 유니버시티 오브 펜실바니아 Compositions and methods for sirna inhibition of hif-1 alpha
GB2394658A (en) 2002-11-01 2004-05-05 Cancer Rec Tech Ltd Oral anti-cancer composition
US20040152651A1 (en) 2002-11-01 2004-08-05 Rana Tariq M. Regulation of transcription elongation factors
US7892793B2 (en) * 2002-11-04 2011-02-22 University Of Massachusetts Allele-specific RNA interference
EP1562971B1 (en) 2002-11-05 2014-02-12 Isis Pharmaceuticals, Inc. Polycyclic sugar surrogate-containing oligomeric compounds and compositions for use in gene modulation
WO2004044132A2 (en) 2002-11-05 2004-05-27 Isis Pharmaceuticals, Inc. Modified oligonucleotides for use in rna interference
US20060009410A1 (en) 2002-11-13 2006-01-12 Crooke Rosanne M Effects of apolipoprotein B inhibition on gene expression profiles in animals
CA2506576C (en) 2002-11-18 2018-03-06 Santaris Pharma A/S Antisense gapmer oligonucleotides
US7144999B2 (en) 2002-11-23 2006-12-05 Isis Pharmaceuticals, Inc. Modulation of hypoxia-inducible factor 1 alpha expression
US7713738B2 (en) 2003-02-10 2010-05-11 Enzon Pharmaceuticals, Inc. Oligomeric compounds for the modulation of survivin expression
WO2004083432A1 (en) * 2003-03-21 2004-09-30 Academisch Ziekenhuis Leiden Modulation of exon recognition in pre-mrna by interfering with the secondary rna structure
US7598227B2 (en) 2003-04-16 2009-10-06 Isis Pharmaceuticals Inc. Modulation of apolipoprotein C-III expression
US7339051B2 (en) 2003-04-28 2008-03-04 Isis Pharmaceuticals, Inc. Compositions and methods for the treatment of severe acute respiratory syndrome (SARS)
WO2004108081A2 (en) 2003-06-02 2004-12-16 Isis Pharmaceuticals, Inc. Oligonucleotide synthesis with alternative solvents
AU2004253455B2 (en) 2003-06-03 2011-03-03 Eli Lilly And Company Modulation of survivin expression
US7825235B2 (en) 2003-08-18 2010-11-02 Isis Pharmaceuticals, Inc. Modulation of diacylglycerol acyltransferase 2 expression
AR045937A1 (en) 2003-09-18 2005-11-16 Lilly Co Eli MODULATION OF THE EXPRESSION OF THE EIF4E EUCARY INITIATION FACTOR
US8258105B2 (en) 2003-10-07 2012-09-04 Isis Pharmaceuticals, Inc. Antisense oligonucleotides optimized for kidney targeting
WO2005045034A2 (en) 2003-10-23 2005-05-19 Sirna Therapeutics, Inc. RNA INTERFERENCE MEDIATED TREATMENT OF PARKINSON DISEASE USING SHORT INTERERING NUCLEIC ACID (siNA)
DK1706489T3 (en) 2003-12-23 2010-09-13 Santaris Pharma As Oligomeric Compounds for the Modulation of BCL-2
US7468431B2 (en) 2004-01-22 2008-12-23 Isis Pharmaceuticals, Inc. Modulation of eIF4E-BP2 expression
GB0403041D0 (en) 2004-02-11 2004-03-17 Milner Anne J Induction of apoptosis
EP1566202A1 (en) 2004-02-23 2005-08-24 Sahltech I Göteborg AB Use of resistin antagonists in the treatment of rheumatoid arthritis
US7402574B2 (en) 2004-03-12 2008-07-22 Avi Biopharma, Inc. Antisense composition and method for treating cancer
US20080199960A1 (en) * 2004-05-13 2008-08-21 Juliano Rudolph L Methods for the Delivery of Oligomeric Compounds
US8394947B2 (en) 2004-06-03 2013-03-12 Isis Pharmaceuticals, Inc. Positionally modified siRNA constructs
WO2006085987A2 (en) 2004-07-09 2006-08-17 University Of Iowa Research Foundation Rna interference in respiratory epitheial cells
US7427675B2 (en) 2004-08-23 2008-09-23 Isis Pharmaceuticals, Inc. Compounds and methods for the characterization of oligonucleotides
WO2006050734A2 (en) 2004-11-09 2006-05-18 Santaris Pharma A/S Potent lna oligonucleotides for the inhibition of hif-1a expression
US7220549B2 (en) 2004-12-30 2007-05-22 Helicos Biosciences Corporation Stabilizing a nucleic acid for nucleic acid sequencing
WO2007002718A2 (en) 2005-06-27 2007-01-04 Alnylam Pharmaceuticals, Inc. Rnai modulation of hif-1 and theraputic uses thereof
US20070213292A1 (en) 2005-08-10 2007-09-13 The Rockefeller University Chemically modified oligonucleotides for use in modulating micro RNA and uses thereof
EP1937312B1 (en) 2005-08-30 2016-06-29 Ionis Pharmaceuticals, Inc. Chimeric oligomeric compounds for modulation of splicing
EP2325315B1 (en) 2005-10-28 2014-05-07 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of huntingtin gene
CA2626584A1 (en) 2005-11-04 2007-05-18 Alnylam Pharmaceuticals, Inc. Compositions and methods for inhibiting expression of nav1.8 gene
AU2006336624B2 (en) 2005-11-17 2010-11-25 Board Of Regents, The University Of Texas System Modulation of gene expression by oligomers targeted to chromosomal DNA
CA2631713A1 (en) 2005-12-02 2007-09-13 Sirtris Pharmaceuticals, Inc. Modulators of cdc2-like kinases (clks) and methods of use thereof
CN101378764A (en) 2005-12-09 2009-03-04 贝勒研究院 Diagnosis, prognosis and monitoring of disease progression of systemic lupus erythematosus through blood leukocyte microarray analysis
WO2007071824A1 (en) 2005-12-20 2007-06-28 Oy Jurilab Ltd Novel genes and markers associated with high-density lipoprotein -cholesterol (hdl-c)
CN100356377C (en) 2005-12-20 2007-12-19 无锡永中科技有限公司 Document display method
EP1976567B1 (en) 2005-12-28 2020-05-13 The Scripps Research Institute Natural antisense and non-coding rna transcripts as drug targets
WO2007133812A2 (en) * 2005-12-30 2007-11-22 Philadelphia Health & Education Corporation, D/B/A Drexel University College Of Medicine Improved carriers for delivery of nucleic acid agents to cells and tissues
WO2007089607A2 (en) * 2006-01-26 2007-08-09 University Of Massachusetts Rna silencing agents for use in therapy and nanotransporters for efficient delivery of same
DK2314594T3 (en) 2006-01-27 2014-10-27 Isis Pharmaceuticals Inc 6-modified bicyclic nucleic acid analogues
US7569686B1 (en) 2006-01-27 2009-08-04 Isis Pharmaceuticals, Inc. Compounds and methods for synthesis of bicyclic nucleic acid analogs
ES2544861T3 (en) 2006-03-31 2015-09-04 Alnylam Pharmaceuticals Inc. Compositions and methods to inhibit the expression of the Eg5 gene
EP2023939B1 (en) 2006-05-05 2012-06-27 Isis Pharmaceuticals, Inc. Compounds and methods for modulating expression of pcsk9
NZ572666A (en) 2006-05-11 2010-11-26 Alnylam Pharmaceuticals Inc Compositions comprising double stranded rna and methods for inhibiting expression of the pcsk9 gene
WO2007134181A2 (en) 2006-05-11 2007-11-22 Isis Pharmaceuticals, Inc. 5'-modified bicyclic nucleic acid analogs
US7666854B2 (en) 2006-05-11 2010-02-23 Isis Pharmaceuticals, Inc. Bis-modified bicyclic nucleic acid analogs
EP1867338A1 (en) 2006-05-30 2007-12-19 Université Libre De Bruxelles Pharmaceutical composition comprising apolipoproteins for the treatment of human diseases
WO2008066672A2 (en) 2006-11-06 2008-06-05 Beth Israel Deaconess Medical Center Identification and use of small molecules to modulate transcription factor function and to treat transcription factor associated diseases
WO2008057556A2 (en) 2006-11-06 2008-05-15 Beth Israel Deaconess Medical Center Identification and use of small molecules to modulate ese-1 transcription factor function and to treat ese-1 transcription factor associated diseases
US8093222B2 (en) 2006-11-27 2012-01-10 Isis Pharmaceuticals, Inc. Methods for treating hypercholesterolemia
US20100169996A1 (en) 2007-01-19 2010-07-01 Lionel Navarro Methods and compositions for modulating the sirna and rna-directed-dna methylation pathways
WO2008124660A2 (en) 2007-04-06 2008-10-16 The Johns Hopkins University Methods and compositions for the treatment of cancer
US20080293142A1 (en) 2007-04-19 2008-11-27 The Board Of Regents For Oklahoma State University Multiple shRNA Expression Vectors and Methods of Construction
CA2688240A1 (en) * 2007-05-24 2008-12-04 The United States Government As Represented By The Department Of Veterans Affairs Intranuclear protein transduction through a nucleoside salvage pathway
WO2009044793A1 (en) * 2007-10-02 2009-04-09 Alphagen Co., Ltd. siRNA TARGETING ONCOGENE
EP2304030B1 (en) 2008-07-01 2015-11-25 Monsanto Technology LLC Recombinant dna constructs and methods for modulating expression of a target gene
US8153606B2 (en) 2008-10-03 2012-04-10 Opko Curna, Llc Treatment of apolipoprotein-A1 related diseases by inhibition of natural antisense transcript to apolipoprotein-A1
EP2177615A1 (en) 2008-10-10 2010-04-21 Fraunhofer-Gesellschaft zur Förderung der angewandten Forschung e.V. Method for a genome wide identification of expression regulatory sequences and use of genes and molecules derived thereof for the diagnosis and therapy of metabolic and/or tumorous diseases
US8606289B2 (en) 2008-11-10 2013-12-10 Qualcomm Incorporated Power headroom-sensitive scheduling
EP2358382A2 (en) 2008-11-22 2011-08-24 The University of Bristol NOVEL USES OF VEGFxxxB
ES2573981T3 (en) * 2009-04-10 2016-06-13 Association Institut De Myologie Tricycle-DNA antisense oligonucleotides, compositions, and methods for the treatment of diseases

Also Published As

Publication number Publication date
CN102459597B (en) 2020-06-30
EP2427554A4 (en) 2013-05-22
CN102459597A (en) 2012-05-16
US9533004B2 (en) 2017-01-03
CA3185821A1 (en) 2010-11-11
US20120046345A1 (en) 2012-02-23
ES2618572T3 (en) 2017-06-21
WO2010129861A3 (en) 2011-05-12
WO2010129861A2 (en) 2010-11-11
US20150094356A1 (en) 2015-04-02
KR101742334B1 (en) 2017-06-01
US9012139B2 (en) 2015-04-21
EP2427554B1 (en) 2016-11-16
KR20120013425A (en) 2012-02-14
CA2761248C (en) 2023-03-14
EP2427554A2 (en) 2012-03-14
JP5931720B2 (en) 2016-06-08
JP2012525852A (en) 2012-10-25

Similar Documents

Publication Publication Date Title
CA2761248A1 (en) Treatment of dystrophin family related diseases by inhibition of natural antisense transcript to dmd family
RU2611186C2 (en) TREATMENT OF TUMOR PROTEIN 63 (p63) RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO p63
JP6026482B2 (en) Treatment of tumor suppressor gene-related diseases by suppression of natural antisense transcripts against tumor suppressor genes
US10519448B2 (en) Treatment of brain derived neurotrophic factor (BDNF) related diseases by inhibition of natural antisense transcript to BDNF
JP6049623B2 (en) Treatment of IDUA-related diseases by inhibition of natural antisense transcripts to α-L-iduronidase (IDUA)
RU2639550C2 (en) Treatment of diseases connected with site-1 membrane-impacted peptidase of transcription factors (mbtps1), by inhibiting natural antisense transcript to mbtps1
US9834767B2 (en) Treatment of interferon regulatory factor 8 (IRF8) related diseases by inhibition of natural antisense transcript to IRF8
JP5934106B2 (en) Treatment of HGF-related diseases by inhibition of natural antisense transcripts against hepatocyte growth factor (HGF)
RU2608496C2 (en) Treatment of pyrroline-5-carboxylate reductase 1 (pycr1) related diseases by inhibition of natural antisense transcript to pycr1
RU2611192C2 (en) TREATMENT OF RNase H1 RELATED DISEASES BY INHIBITION OF NATURAL ANTISENSE TRANSCRIPT TO RNase H1
ES2629339T3 (en) Treatment of diseases related to paraoxonase 1 (pon1) by inhibition of natural antisense transcript to pon1
RU2620978C2 (en) Treatment of diseases associated with methionine sulfoxide reductase a (msra), by msra natural antisense transcript inhibition
US20170226516A1 (en) Treatment of glial cell derived neurotrophic factor (gdnf) related diseases by inhibition of natural antisense transcript to gdnf
RU2612884C2 (en) Treatment of diseases associated with colonystimulating factor 3 (csf3) by inhibition of natural antisense transcript to csf3
KR20120026622A (en) Treatment of collagen gene related diseases by inhibition of natural antisense transcript to a collagen gene

Legal Events

Date Code Title Description
EEER Examination request

Effective date: 20150421

EEER Examination request

Effective date: 20150421

EEER Examination request

Effective date: 20150421

EEER Examination request

Effective date: 20150421

EEER Examination request

Effective date: 20150421