Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
INHIBITORS OF POLYNUCLEOTIDE REPEAT-ASSOCIATED RNA FOCI AND USES THEREOF
Document Type and Number:
WIPO Patent Application WO/2015/042685
Kind Code:
A1
Abstract:
Compounds which inhibit the formation and/or accumulation of RNA foci, such as those due to polynucleotide repeats (e.g., trinucleotide repeats) are described herein. Also described herein are uses of such compounds, such as for the inhibition of the formation and accumulation such RNA foci, as well as for the treatment of polynucleotide repeat disorders (e.g., trinucleotide repeat disorders), such as myotonic dystrophy (e.g., DM1). Such compounds include compounds of formula 1, 1a, 1b, 2, 2a and (3) described herein.

Inventors:
CHARTRAND PASCAL (CA)
QUERIDO EMMANUELLE (CA)
Application Number:
PCT/CA2013/050730
Publication Date:
April 02, 2015
Filing Date:
September 25, 2013
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
VALORISATION RECH (CA)
International Classes:
C07D231/14; A61K31/18; A61K31/352; A61K31/36; A61K31/4155; A61K31/4406; A61K31/4453; A61K31/496; A61K31/5375; A61P21/00; C07C311/46; C07D213/82; C07D217/08; C07D265/30; C07D295/26; C07D311/84; C07D317/68; C07D409/12; G01N33/50
Foreign References:
CA2618370A12007-02-15
Other References:
JIANG, M ET AL., NEUROPROTECTIVE ROLE OF SIRTL IN MAMMALIAN MODELS OF HUNTINGTONS'S DISEASE THROUGH ACTIVATION OF MULTIPLE SIRTL TARGETS, vol. 18, 2012, pages 153 - 158, XP055291939
JEONG, H ET AL., SIRT1 MEDIATES NEUROPROTECTION FROM MUTANT HUNTIGNTON BY ACTIVATION OF THE TORC1 AND CREB TRANSCRIPTIONAL PATHWAY, vol. 18, 2012, pages 159 - 165, XP055291959
See also references of EP 3049393A4
Attorney, Agent or Firm:
GOUDREAU GAGE DUBUC (McGill College#220, Montréal Québec H3A 3H3, CA)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1. A method for treating a polynucleotide repeat disorder, said method comprising administering to a subject in need thereof an effective amount of a compound of formula (1 ), (2), or (3):

wherein: R1 is:

aryl, heteroaryl, arylmethyl, or heteroarylmethyl, said aryl, heteroaryl, arylmethyl, and heteroarylmethyl being optionally substituted on the aryl ring or on the methyl group by alkyl, alkenyl, or alkynyl, and said heteroaryl and heteroarylmethyl comprising only oxygen and/or nitrogen atoms as heteroatom(s), or

alkyl, alkenyl or alkynyl, said alkyl, alkenyl and alkynyl being substituted by cycloalkyl, cycloalkenyl or cycloalkynyl, R2 and R3 are independently:

a hydrogen atom,

alkyl, alkenyl, or alkynyl,

cycloalkyl, cycloalkenyl, cycloalkynyl,

an aryl free of heteroatom,

aralkyl, or

an aliphatic heterocycle,

with the proviso that at least one of R2 and R3 is not a hydrogen atom or a C-|-C2 alkyl, or R2 and R3, together with the nitrogen atom to which they are attached, form a 6- or more membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen atom,

R4 and R5 are independently:

a hydrogen atom,

alkyl, alkenyl, or alkynyl,

cycloalkyl, cycloalkenyl, cycloalkynyl,

an aryl free of heteroatom,

aralkyl, or

an aliphatic heterocycle,

with the proviso that at least one of R4 and R5 is not a hydrogen atom or a Ci-C2 alkyl, or R4 and R5, together with the nitrogen atom to which they are attached, form a 6 membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen or oxygen atom, R6 is H, halogen, or CrC8 alkyl optionally substituted with one or more halogen atoms,

R7 is Ci-C8 alkyl optionally substituted with one or more halogen atoms,

R8 and R9 are independently alkyl, alkenyl, or alkynyl, said alkyl, alkenyl, and alkynyl being substituted by optionally substituted aryl or optionally substituted thioaryl, or

R8 and R9, together with the nitrogen atom to which they are attached, form a 5- or more membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen atom, and substituted with optionally substituted aryl, with the proviso that when R7 is C-i alkyi, R8 and R9 are independently alkyi, alkenyl, or alkynyl, said alkyi, alkenyl, and alkynyl being substituted by aryl or thioaryl, or a pharmaceutically acceptable salt thereof.

The method of claim 1 , wherein R1 in formula (1 ) is a substituted aryl.

The method of claim 2, wherein R1 is

The method of claim 1 , wherein R1 in formula (1 ) is a substituted or unsubstituted arylmethyl.

The method of claim 4, wherein R1 is

The method of claim 4, wherein R

The method of claim 1 , wherein R1 in formula (1 ) is an unsubstituted or substituted heteroaryl.

16. The method of any one of claims 1 to 10, wherein R2 is H and R3 is a C3-C12 cycloalkyl.

17. The method of claim 16, wherein NR2R3 is: 18. The method of any one of claims 1 to 10, wherein R2 is H and R3 is an unsubstituted arylalkyl.

19. The method of claim 17, wherein NR2R3 is 20. The method of any one of claims 1 to 10, wherein R2 and R3 form a substituted or unsubstituted 6- to 12-membered aliphatic heterocycle.

21 . The method of claim 20, wherein R2 and R3 form a substituted or unsubstituted 6-membered aliphatic heterocycle.

22. The method of claim 21 , wherein NR2R3 is 23. The method of claim 1 , wherein R1 in formula (2) is a substituted aryl.

24. The method of claim 23, wherein R1 is 25. The method of any one of claims 1 , 23, and 24, wherein R4 and R5 form a 6-membered aliphatic heterocycle comprising one additional oxygen atom. The method of claim 25, wherein R4 and R5 form H3

The method of claim 1 , wherein R6 is H or a halogen. The method of claim 27, wherein R6 is CI.

The method of any one of claims 1 , 27 and 28, wherein R7 is a CrC8 alkyl optionally substituted with one or more halogen atoms.

The method of claim 29, wherein R7 is a C1-C3 alkyl optionally substituted with one or more halogen atoms.

The method of claim 30, wherein R7 is methyl, trifluoromethyl, or propyl.

The method of any one of claims 1 , and 27 to 31 , wherein R8 and R9 are independently alkyls substituted by aryl or thioaryl.

The method of claim 32, wherein said alkyls are C1-3 alkyls.

The method of claim 32 or 33, wherein said aryl is phenyl and said thioaryl is 2-thienyl.

or

The method of any one of claims 1 , and 27 to 31 , wherein R8 and R9, together with the nitrogen atom to which they are attached, join to form a substituted 5- or more membered aliphatic heterocycie.

The method of claim 36, wherein the heterocycie is substituted with an optionally substituted aryl.

38. The method of claim 37, wherein NR8R9

39. The method of claim 1 , wherein said compound of formula (1) is:

or a pharmaceutically acceptable salt thereof.

The method of claim 1 , wherein said compound of formula (2) is r a pharmaceutically acceptable salt thereof.

or a pharmaceutically acceptable salt thereof.

42. The method of any one of claims 1 to 41 , wherein said polynucleotide repeat disorder is myotonic dystrophy. 43. The method of claim 42, wherein said myotonic dystrophy is myotonic dystrophy type 1 (Steinert's disease).

44. Use of the compound defined in any one of claims 1 to 41 for treating a polynucleotide repeat disorder in a subject.

45. Use of the compound defined in any one of claims 1 to 41 for the manufacture of a medicament for treating a polynucleotide repeat disorder in a subject.

46. The use of claim 44 or 45, wherein said polynucleotide repeat disorder is myotonic dystrophy.

47. The use of claim 46, wherein said myotonic dystrophy is myotonic dystrophy type 1 (Steinert's disease). 48. The compound as defined in any one of claims 1 to 41 for treating a polynucleotide repeat disorder in a subject.

49. The compound as defined in any one of claims 1 to 41 for the manufacture of a medicament for treating a polynucleotide repeat disorder in a subject. The compound of claim 48 or 49, wherein said polynucleotide repeat disorder is myotonic dystrophy.

The compound of claim 50, wherein said myotonic dystrophy is myotonic dystrophy type 1 (Steinert's disease).

A compound, wherein said compound is the compound defined in any one of claims 1 to 41.

A composition comprising the compound of claim 52 or 53 and a pharmaceutically acceptable carrier.

A method for reduce the formation of RNA foci or aggregates in a cell, said method comprising contacting said cell with the compound defined in any one of claims 1 to 41 .

The method of claim 55, wherein said method is in vitro.

57. A method for determining whether a test compound may be useful for treating a polynucleotide repeat disorder, said method comprising (i) contacting said test compound with a cell expressing a reporter construct, said reporter construct comprising a first domain encoding a reporter transcript and a second domain comprising a plurality of polynucleotide repeats located downstream of said first domain; and (ii) determining the aggregation of said reporter transcript in said cell, wherein a decrease in the aggregation of said reporter transcript in said cell in the presence of said test compound, relative to the aggregation in the absence of said test compound, is indicative that said test compound may be useful for treating a polynucleotide repeat disorder. 58. The method of claim 57, wherein the decrease in the aggregation of said reporter transcript in said cell is determined by quantifying the number of reporter transcript foci in the nucleus of said cell.

59. The method of claim 57 or 58, wherein said reporter transcript is a luciferase or beta- galactosidase transcript. 60. The method of any one of claims 57 to 59, wherein said reporter construct is under inducible expression.

61 . The method of claim 60, wherein said reporter construct is operably linked to a tetracycline- responsive element (TRE).

62. The method of claim 62, wherein said cell further expresses the tetracycline-responsive transcriptional activator (tTA) from the strong immediate early promoter of cytomegalovirus

63. The method of claim 61 or 62, wherein said expression is induced by culturing said cell in the absence of tetracycline (Tc), or a derivative thereof.

64. The method of claim 63, wherein said tetracycline (Tc) derivative is doxycycline (Dox). 65. The method of any one of claims 61 to 64, wherein said method comprises:

(i) culturing said cell in the absence of tetracycline (Tc), or a derivative thereof to induce expression of the reporter transcript; and (ii) contacting said test compound with said cell in the presence of tetracycline (Tc), or a derivative thereof to inhibit expression of the reporter transcript.

Description:
INHIBITORS OF POLYNUCLEOTIDE REPEAT-ASSOCIATED RNA FOCI AND USES

THEREOF

CROSS REFERENCE TO RELATED APPLICATIONS

The present application claims the benefits of U.S. provisional application Serial No. 61/705,833, filed September 26, 2012, which is herein incorporated by reference in its entirety.

TECHNICAL FIELD

The present invention generally relates to polynucleotide repeat disorders such as trinucleotide repeat disorders, and more particularly to compounds and uses thereof such as for the treatment of diseases associated with the presence of polynucleotide repeats, such as myotonic dystrophy.

BACKGROUND ART

Polynucleotide repeat disorders are a set of genetic disorders caused by polynucleotide repeat expansions (typically trinucleotide repeat expansions). The expanded polynucleotide/trinucleotide repeats have been shown to cause the retention of transcripts in the nucleus, where it accumulates in numerous foci (RNA aggregates). More and more RNA aggregates or foci have been identified in different pathologies, for example Myotonic Dystrophy type 1 (DM1 ) (Davis et al., 1997) and type 2 (DM2) (Liquori et al., 2001 ), Fragile X-associated tremor/ataxia syndrome (FXTAS) (Tassone et al., 2004), Spinocerebellar ataxia type 8 (SCA8) (Daughters et al., 2009) and Huntington's disease-like 2 (HDL2) (Rudnicki et al., 2007). All these diseases are characterized by microsatellite expansions of CNG or CCTG repeats in specific genes, leading to the accumulation of their transcripts as nuclear RNA foci (Ranum and Cooper, 2006).

Myotonic dystrophy is a chronic, slowly progressing, highly variable, inherited multisystemic disease. Two types of myotonic dystrophy exist. Type 1 (DM1 ), also known as Steinert disease, has a severe congenital form and a milder childhood-onset form. Type 2 (DM2), also known as proximal myotonic myopathy (PROMM), is rarer and generally manifests with milder signs and symptoms than DM1. DM1 is the most common adult form of muscular dystrophy with a prevalence of up to 1 in 7,000 worldwide. DM1 is highly prevalent in the Saguenay region of Quebec where its carrier rate reaches 1 in 550, making DM1 a Canadian-specific disease. There is no current treatment for the progressive myopathy, which eventually kills the patients, highlighting the urgent medical need for therapeutics. It is a multisystemic disorder (FIG. 1 ), caused by an expansion of CUG trinucleotide repeats in the 3' untranslated region (UTR) of the protein kinase DMPK mRNA (Brook et al. 1992, Buxton et al. 1992, Fu et al. 1992, Mahadevan et al. 1992). The expanded CUG repeats have been shown to cause the retention of this transcript in the nucleus, where it accumulates in numerous foci (FIG. 2; Taneja et al. 1995, Davis et al. 1997). The current toxic RNA hypothesis posits that the retention of mutant DMPK (dystrophia myotonica protein kinase) mRNAs in the nucleus alters the function of RNA-binding proteins, such as the alternative splicing factors MBNL1 and CUGBP1. As a consequence, mRNA mis-splicing has been reported for several genes in DM1 (reviewed in Wheeler and Thornton 2007). One of the mechanisms proposed is that these nuclear RNA foci sequester essential proteins that normally interact with CUG nucleotides in mRNAs and interfere with their normal function in the cell. Disrupting these nuclear RNA foci and promoting the nuclear export of the CUG-rich transcripts should reduce the alteration of splicing factor function and prevent the development of symptoms in patients with DM1 (Wheeler 2008).

There is thus a need for the development of novel strategies to inhibit the aggregation of RNA with expanded tracts of triplet repeats, for the treatment of diseases associated with the presence of triplet repeats

The present description refers to a number of documents, the content of which is herein incorporated by reference in their entirety.

SUMMARY OF THE INVENTION

The present invention relates to compounds, as well as to uses thereof, such as to treat a polynucleotide repeat disorder (such as a trinucleotide repeat disorder, such as myotonic dystrophy).

In a first aspect, the present invention provides method for treating a polynucleotide repeat disorder, said method comprising administering to a subject in need thereof an effective amount of a compound of formula (1 ), (2), or (3):

(1 ), wherein: R 1 is:

aryl, heteroaryl, arylmethyl, or heteroarylmethyl, said aryl, heteroaryl, arylmethyl, and heteroarylmethyl being optionally substituted on the aryl ring or on the methyl group by alkyl, alkenyl, or alkynyl, and said heteroaryl and heteroarylmethyl comprising only oxygen and/or nitrogen atoms as heteroatom(s), or

alkyl, alkenyl or alkynyl, said alkyl, alkenyl and alkynyl being substituted by cycloalkyl, cycloalkenyl or cycloalkynyl,

R 2 and R 3 are independently:

a hydrogen atom,

alkyl, alkenyl, or alkynyl,

cycloalkyl, cycloalkenyl, cycloalkynyl,

an aryl free of heteroatom,

aralkyl, or

an aliphatic heterocycle,

with the proviso that at least one of R 2 and R 3 is not a hydrogen atom or a Ci-C 2 alkyl,

or R 2 and R 3 , together with the nitrogen atom to which they are attached, form a 6- or more membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen atom,

R 4 and R 5 are independently: a hydrogen atom,

alkyi, alkenyl, or alkynyl,

cycloalkyl, cycloalkenyl, cycloalkynyl,

an aryl free of heteroatom,

aralkyl, or

an aliphatic heterocycle,

with the proviso that at least one of R 4 and R 5 is not a hydrogen atom or a Ci-C 2 alkyi,

or R 4 and R 5 , together with the nitrogen atom to which they are attached, form a 6 membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen or oxygen atom.

R 6 is H, halogen, or CrC 8 alkyi optionally substituted with one or more halogen atoms,

R 7 is Ci-C 8 alkyi optionally substituted with one or more halogen atoms,

R 8 and R 9 are independently alkyi, alkenyl, or alkynyl, said alkyi, alkenyl, and alkynyl being substituted by optionally substituted aryl or optionally substituted thioaryl, or

R 8 and R 9 , together with the nitrogen atom to which they are attached, form a 5- or more membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen atom, and substituted with optionally substituted aryl,

with the proviso that when R 7 is C-i alkyi, R 8 and R 9 are independently alkyi, alkenyl, or alkynyl, said alkyi, alkenyl, and alkynyl being substituted by aryl or thioaryl,

or a pharmaceutically acceptable salt thereof.

In embodiments of Formula (1 ), R 1 is substituted aryl, such as aryl substituted with one to three alkyi groups, preferably methyl groups. In embodiments, the substituted aryl is phenyl substituted with two methyl groups, such as:

In embodiments of Formula (1 ), R 1 is an arylmethyl, preferably:

(benzyl). In embodiments, the arylmethyl, such as benzyl, is substituted by an alkyl group, preferably a C 1-3 alkyl group, such as ethyl. In embodiments, the alkyl group substitutes the methyl group of the arylmethyl. In embodiments, R 1 is:

In embodiments of Formula (1 ), R 1 is an unsubstituted or substituted heteroaryl. iment, the heteroaryl comprises one or two oxygen atoms such as: :

,3-Benzodioxole) or (xanthene).

In other embodiments, the heteroaryl comprises one or two nitrogen atoms, preferably one nitrogen atom, such as:

The heteroaryl may be substituted, for example with one to three alkyl groups, preferably methyl groups. In embodiments, the substituted heteroaryl is 1 -phenyl-1 H-pyrazole substituted with one methyl group, such as:

In embodiments of Formula (1 ), R 1 is an alkyl substituted cycloalkyl, preferably Ci -6 alkyl substituted with C 5 - 6 cycloalkyl. In a further embodiment R 1 is:

In embodiments, one of R 2 and R 3 is hydrogen or alkyl (preferably C1-C3 alkyl), and the other is alkyl comprising 3 or more carbon atoms, aralkyi (preferably phenyl-alkyl) or cycloalkyi (preferably cyclohexyl or adamantyl).

In an embodiment, R 2 and R 3 are C 3-6 alkyl, preferably propyl.

In an embodiment, R 2 is H and R 3 is a C 3 -C 6 alkyl, in a further embodiment, R 3 is a C 4 alkyl, such as n-butyl, ferf-butyl, or sec-butyl.

In an embodiment, R 2 is H and R 3 is a C3-C12 cycloalkyi. In a further embodiment, R 3 is adamantyl, such as adamant-1-yl:

In an embodiment, R 2 is H and R 3 is an arylalkyl, such as benzyl.

In an embodiment, R 2 is alkyl and R 3 is alkyl comprising more than 3 carbon atoms.

In another embodiment, R 2 and R 3 form a substituted or unsubstituted 6- to 12-membered aliphatic heterocycle, which optionally comprises one or more additional nitrogen atom. In a further embodiment, said heterocycle is 6-membered. In yet a further embodiment, said heterocycle is 6- membered heterocycloalkyl comprising 0 or 1 additional nitrogen atom. In embodiments, the substituents are one to three Ci -6 alkyl, preferably methyl. In embodiments, R 2 and R 3 form:

In embodiments of Formula (2), R 1 is substituted aryl, such as aryl substituted with one to three alkyl groups. In embodiment, the aryl is phenyl. In embodiments, the alkyl groups are methyl. In embodiments, there are two methyl groups. In embodiments, R 1 is: In embodiments, R 4 and R 5 , together with the nitrogen atom to which they are attached, form a 6-membered, optionally substituted, aliphatic heterocycle, optionally comprising one or more additional nitrogen or oxygen atom. In further embodiments, the heterocycle comprises one further oxygen atom. In embodiments, the heterocycle is substituted with one to three alkyl groups, preferably two methyl groups. In embodiments, R 4 and R 5 , together with the nitrogen atom to which they are attached, form:

In an embodiment, R 6 is H or a halogen, in a further embodiment the halogen is CI.

In an embodiment, R 7 is a C-|-C 3 alkyl optionally substituted with one or more halogen atoms, such as three fluoride atoms. In embodiments, R7 is methyl, ethyl, propyl, or -CF 3 , preferably methyl, propyl or -CF 3 , more preferably propyl.

In an embodiment, R 8 and R 9 are independently alkyls, such as Ci -3 alkyls, preferably methyl groups, substituted by aryl or thioaryl, preferably phenyl and/or 2-thienyl. In a further embodiment NR 8 R 9 is:

In another embodiment, when R 7 is not a C-i alkyl, R 8 and R 9 , together with the nitrogen atom to which they are attached, join to form a substituted 5- or more membered aliphatic heterocycle. In embodiments, the aliphatic heterocycle may be pyrrolidinyl or piperidinyl. In a further embodiment, the heterocycle is substituted with an optionally substituted aryl, preferably optionally substituted phenyl. Substituents for the aryl include C^e alkyl and alkyloxy. In yet further embodiment, NR 8 R 9 is:

In further embodiments, the compound is:

10

or a pharmaceutically acceptable salt thereof.

In another aspect, the present invention provides the compound defined above for treating a polynucleotide repeat disorder in a subject. In another aspect, the present invention provides the compound defined above for the manufacture of a medicament for treating a polynucleotide repeat disorder in a subject.

In another aspect, the present invention provides the use of the compound defined above for treating a polynucleotide repeat disorder in a subject. In another aspect, the present invention provides the use of the compound defined above for the manufacture of a medicament for treating a polynucleotide repeat disorder in a subject.

In an embodiment, the polynucleotide repeat disorder is myotonic dystrophy, in a further embodiment myotonic dystrophy type 1 (Steinert's disease). In another aspect, the present invention provides a compound, wherein said compound is the compound defined above. In another related aspect, the present invention provides a composition comprising the above- mentioned compounds and a pharmaceutically r embodiments of

In another aspect, the present invention provides a method for reducing the formation of RNA foci or aggregates in a cell, said method comprising contacting said cell with the compound defined above. In an embodiment, the method is in vitro. In another embodiment, the method is in vivo. In an embodiment, the RNA is a mutated mRNA comprising polynucleotide repeats, such as trinucleotide (or triplet) repeats.

In another aspect, the present invention provides a method for determining whether a test compound may be useful for treating a polynucleotide repeat disorder, said method comprising contacting said test compound with a cell expressing a reporter construct, said reporter construct comprising a first domain encoding a reporter transcript and a second domain comprising a plurality of polynucleotide repeats located downstream of said first domain, and determining the aggregation of said reporter transcript in said cell, wherein a decrease in the aggregation of said reporter transcript in said cell in the presence of said test compound, relative to the aggregation in the absence of said test compound, is indicative that said test compound may be useful for treating a polynucleotide repeat disorder.

In an embodiment, said second domain of said reporter construct comprises at least 100 polynucleotide repeats, in a further embodiment, at least 200 polynucleotide repeats, in a further embodiment, at least 300 polynucleotide repeats, in a further embodiment, 100-1500 polynucleotide repeats, in further embodiments, 100, 200, 300 or 1250 polynucleotide repeats.

In an embodiment, the polynucleotide repeat is a trinucleotide repeat.

In an embodiment, the decrease in the aggregation of said reporter transcript in said cell is determined by quantifying the number of reporter transcript foci in the nucleus of said cell.

In an embodiment, the above-mentioned reporter transcript is a luciferase or beta- galactosidase transcript.

In an embodiment, the above-mentioned reporter construct is under inducible expression. In an embodiment, the above-mentioned reporter construct is operably linked to a tetracycline-responsive element (TRE).

In an embodiment, the above-mentioned cell further expresses the tetracycline-responsive transcriptional activator (tTA) from the strong immediate early promoter of cytomegalovirus (PCMV)- In an embodiment, the above-mentioned expression is induced by culturing said cell in the absence of tetracycline (Tc), or a derivative thereof. In a further embodiment, the tetracycline (Tc) derivative is doxycycline (Dox).

In an embodiment, the above-mentioned method comprises:

culturing said cell in the absence of tetracycline (Tc), or a derivative thereof to induce expression of the reporter transcript; and

contacting said test compound with said cell in the presence of tetracycline (Tc), or a derivative thereof to inhibit expression of the reporter transcript.

Other objects, advantages and features of the present invention will become more apparent upon reading of the following non-restrictive description of specific embodiments thereof, given by way of example only with reference to the accompanying drawings. BRIEF DESCRIPTION OF DRAWINGS

In the appended drawings:

FIG. 1 shows the symptoms of myotonic dystrophy; FIG. 2 shows DM1 patient fibroblasts with a 2000 CUG-triplet repeat expansion in the 3'UTR of the DMPK mRNA. This mutant mRNA forms nuclear foci which can be detected by fluorescent in situ hybridisation (FISH) (arrows);

FIG. 3 shows the structure of two compounds (compounds #15 and 32) identified in the first screening experiments;

FIG. 4 shows that compounds #15 and 32 reduce the formation of mutant DMPK mRNA foci in DM1 patient myoblasts. Myoblast cells with 1250 CTG-repeats in the 3'UTR of the DMPK gene were treated with compounds 15, 32 or DMSO for 7 days. The formation of (CUG)n RNA foci was detected by FISH. The surface occupied by CUG-repeat RNA foci in the nucleus of the DM1 myoblasts was quantified with the Imaris™ program. Each data point represents one nucleus and the median value is shown on the graph. ***p < 0,001 ;

FIG. 5 shows a structure-activity relationship (SAR) analysis of analogues of compounds 15 and 32. A screen of 49 analogues of compounds 15 and 32 was performed in C2C12 myoblasts expressing a LacZ-145CUG reporter mRNA. Sixteen analogues increase the beta-galactosidase activity in the reporter cell line (table). EC 50 assays of the analogue compounds show that they increase the expression of the beta-galactosidase expressed from the LacZ-145CUG mRNA, but not from the LacZ-5CUG mRNA. The cells were treated with 0.2, 1 , 2, 5 and 25 μΜ concentrations of each compound for 24 hrs. The EC 50 and maximal induction of beta-galactosidase (Max) were measured for each compound; and

FIG. 6A and B shows the structure of analogues of compounds #15 (Fig. 6A) and 32 (Fig.

6B) identified in the SAR analysis described in FIG. 5, FIG. 6C shows further analogs (no. 102, 195, 196, 213, 214, 215, 216, 217, 218, 219, 220, 221 ) of these compounds;

FIG. 7 shows the effect of compounds #15 and 32 and representative analogues thereof on the levels of the CUGBP1 alternative splicing factor. (A) Western blot on the CUGBP1 protein in normal human myoblasts (WT) and DM1 patient myoblasts treated with compound 15 (and its analogues 8 and 12), or compound 32 (and its analogue 43) at a concentration of 20 μΜ for 7 days. (B) Quantification of the bands in panel (A) acquired with the ChemiDoc™ MP imaging system (Bio-Rad). GAPDH is used as a loading control;

FIG. 8 shows the effect of compound 15 on the alternative splicing of three transcripts mis- spliced in DM1 . Normal human myoblasts (WT) or myoblasts from a DM1 patient were treated with compound 15 at 20 μΜ or DMSO during 7 days. (A) RT-PCR on endogenous SERCA1 , TNNT2 and MBNL2 mRNA (hnRNPA2/B1 is used as a negative control). Quantification of the percentage of exon inclusion tested for SERCA1 (B), TNNT2 (C) and MBNL2 (D). *** P < 0,001. FIG. 9 shows the effect of compounds #15 and 32 and representative analogues thereof on the alternative splicing of the endogenous MBNL2 transcript. Normal human myoblasts (WT) or myoblasts from a DM1 patient, were treated with DMSO, compound 15 (or analogues 8 or 12), or compound 32 (or analogue 43) at 20 μΜ concentration during 7 days. Pentamidine treatment was done at a 50 μΜ concentration for 16 hours or at 75 μΜ for only 12 hours, due to the toxicity of this compound. (A) RT-PCR on the MBNL2 mRNA. (B) Quantification of the percentage of inclusion of exon 7 (band 7+ / band 7+ + band 7-)* 100;

FIG. 10 shows the effect of compounds #15 and 32 and representative analogues thereof on the alternative splicing of the endogenous MBNL2, MBNL1 and Sercal transcripts. Myoblasts from a DM1 patient were treated with DMSO, compound 15 (or analogs 8, 12, 19, 21 , 22, 23, 24, 27, 28, 29, 31 , 32n, 34), or compound 32 (or analogs 43, 46) at 20μΜ concentration during 7 days. Correction of mis-splicing is reported in %, where 0% means no correction, while 100% reflects complete splicing correction. Toxicity of compounds is indicated by the ratio of viable cell number (where 1 corresponds to no toxicity), and the percentage of dead cells (%). Toxicity was measured by treating the cells for 7 days with 20μΜ of compound or DMSO. All the values were obtained from the average of 3 independent experiments;

FIG. 11 shows the effect of analogs of compounds 15 and 32 on the alternative splicing of the endogenous MBNL2, MBNL1 , Sercal and TNNT2 transcripts. Myoblasts from a DM1 patient were treated with DMSO, analogues of compound 15 (8, 32n), or compound 32 (and analogues 102, 195, 196, 213, 214, 215, 216, 217, 218, 219, 220, 221 ) at 20μΜ concentration during 7 days. Correction of mis-splicing is reported in %, where 0% means no correction, while 100% reflects complete splicing correction. All the values were obtained from the average of 3 independent experiments;

FIGs. 12A and 12B shows a dose-response effect of compound 32 on the splicing of exon 7 of MBNL2 mRNA (FIG. 12A) and exon 7 of MBNL1 mRNA (FIG. 12B) in normal (M908) or DM1 myoblasts (DM1 ). UNT: untreated cells. All the values were obtained from the average of 3 independent experiments;

FIG. 13 shows the effect of various analogues of compounds 15 and 32 on the alternative splicing of the exon 2 of hnRNPA2/B1 mRNA, which is not mis-spliced in DM1 myoblasts. M908: normal myoblasts; DM1 : DM1 patient myoblasts. All the values were obtained from the average of 3 independent experiments;

FIGs. 14A-D shows a dose-response assay of compounds 32 (FIGs. 14A-B) and 196 (FIGs. 14C-D) on the ratio of viable cell number and the percentage (%) of cell death. Both normal (M908) and DM1 myoblasts (DM1 ) were treated for 7 days at concentrations varying from 5 to 60 μΜ.

DISCLOSURE OF INVENTION

In the studies described herein, the present inventors have demonstrated that compounds having the structure of formula (1 ) and (2) defined below reduce the formation of mutant DMPK mRNA foci in DM1 patient myoblasts, have the capacity to correct the stabilization of the CUGBP1 alternative splicing factor in DM1 myoblasts, and correct the mis-splicing of specific mRNAs in DM1 cells. These compounds may thus be useful for the treatment of diseases associated with the accumulation of RNA foci (RNA aggregates), such as expanded polynucleotide (e.g, trinucleotide) repeat disorders.

Accordingly, in a first aspect, the present invention provides method for treating a polynucleotide repeat disorder, said method comprising administering to a subject in need thereof an effective amount of a compound of formula (1 ), (2), or (3):

wherein: R 1 is:

aryl, heteroaryl, arylmethyl, or heteroarylmethyl, said aryl, heteroaryl, arylmethyl, and heteroarylmethyl being optionally substituted on the aryl ring or on the methyl group by alkyl, alkenyl, or alkynyl, and said heteroaryl and heteroarylmethyl comprising only oxygen and/or nitrogen atoms as heteroatom(s), or

alkyl, alkenyl or alkynyl, said alkyl, alkenyl and alkynyl being substituted by cycloalkyl, cycloalkenyl or cycloalkynyl,

R 2 and R 3 are:

a hydrogen atom,

alkyl, alkenyl, or alkynyl,

cycloalkyl, cycloalkenyl, cycloalkynyl,

an aryl free of heteroatom,

aralkyl, or

an aliphatic heterocycle,

with the proviso that at least one of R 2 and R 3 is not a hydrogen atom or a C-|-C 2 alkyl,

or R 2 and R 3 , together with the nitrogen atom to which they are attached, form a 6- or more membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen atom,

R 4 and R 5 are:

a hydrogen atom,

alkyl, alkenyl, or alkynyl,

cycloalkyl, cycloalkenyl, cycloalkynyl,

an aryl free of heteroatom,

aralkyl, or

an aliphatic heterocycle,

with the proviso that at least one of R 4 and R 5 is not a hydrogen atom or a Ci-C 2 alkyl,

or R 4 and R 5 , together with the nitrogen atom to which they are attached, form a 6 membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen or oxygen atom,

R 6 is H, halogen, or C-|-C 8 alkyl optionally substituted with one or more halogen atoms,

R 7 is C-|-C 8 alkyl optionally substituted with one or more halogen atoms, R 8 and R 9 are independently alkyl, alkenyl, or alkynyl, said alkyl, alkenyl, and alkynyl being substituted by optionally substituted aryl or optionally substituted thioaryl, or

R 8 and R 9 , together with the nitrogen atom to which they are attached, form a 5- or more membered, optionally substituted, aromatic or aliphatic heterocycle, optionally comprising one or more additional nitrogen atom, and substituted with optionally substituted aryl,

with the proviso that when R 7 is C-i alkyl, R 8 and R 9 are independently alkyl, alkenyl, or alkynyl, said alkyl, alkenyl, and alkynyl being substituted by aryl or thioaryl,

or a pharmaceutically acceptable salt thereof.

As used herein, "alkyl" or the prefix "alk" refers to an optionally substituted straight or branched chain saturated hydrocarbon group. Examples of straight or branched chain alkyl groups include, but are not limited to, methyl, trifluoromethyl, ethyl, 1 -propyl, 2-propyl, 1 -butyl, 2-butyl,

2- methyl-1 -propyl, 2-methyl-2-propyl, 1 -pentyl, 2-pentyl, 3-pentyl, 2-methyl-1 -butyl, 3-methyl-1 - butyl, 2-methyl-3-butyl, 2, 2-dimethyl-1 -propyl, 1 -hexyl, 2-hexyl, 3-hexyl, 2-methyl-1 -pentyl,

3- methyl-1 -pentyl, 4-methyl-1 -pentyl, 2-methyl-2-pentyl, 3-methyl-2-pentyl, 4-methyl-2-pentyl, 2,2-dimethyl-1 -butyl, 3, 3-dimethyl- 1 -butyl, 2-ethyl-1 -butyl, 1 -heptyl, and 1 -octyl. A substituted alkyl can be substituted with one or more (e.g., 2, 3, 4, 5, 6, or 7) substituent groups such as -halogen, -NH 2 , -NH(C Ci 2 alkyl), -N(C Ci 2 alkyl) 2 , -OH, -0-(C Ci 2 alkyl), or C 6 -Ci 0 aryl groups, such as phenyl or naphthyl groups, or any other substituent group described herein. In an embodiment, the alkyl group contains 1 -12 carbons, in further embodiments 1 -8, 1 -6 or 1 -3 carbons.

As used herein, "aryl" refers to an optionally substituted monocyclic or polycyclic structure wherein all rings are aromatic, either fused together (e.g. naphthalene) or linked together (e.g. biphenyl) and formed by carbon atoms. Exemplary aryl groups include phenyl, naphthyl, and biphenyl. Where an aryl group is substituted, substituents can include any substituent groups described herein. In an embodiment, the aryl comprises from 6 to 15 carbons (C 6 -Ci 5 aryl).

As used herein, "heteroaryl" or "heteroaromatic" refers to an aryl where one or more carbon atom has been replaced by an heteroatom, such as N, O, or S. In an embodiment, the heteroaryl is 5-6 membered.

As used herein, "arylalkyl" refers to an aryl group attached to the parent molecular group through an alkyl group. In an embodiment, the "arylalkyl" is a "C 7 -C 14 arylalkyl" having the formula -(C x -alkyl)-(C y -aryl) wherein (x+y) is an integer between 7 and 14 and x is at least 1 . Where an arylalkyl group is substituted, substituents can include any substituent group described herein.

As used herein, "arylalkenyl" and "arylalkynyl" refer to an aryl group attached to the parent molecular group through an alkenyl and alkynyl group, respectively. As used herein, "cycloalkyi" refers to an optionally substituted, aliphatic, saturated or unsaturated monocyclic or polycyclic (e.g., bicyclic or tricyclic) hydrocarbon ring system. Polycyclic cycloalkyls may be linear, fused, bridged, or spirocyclic. In an embodiment, the cycloalkyi contains 3-12 carbon atoms (C 3 -C 12 cycloalkyi).

As used herein, "cycloalkylalkyl" refers to a cycloalkyi group attached to the parent molecular group through an alkyl group. As used herein, "cycloalkylalkenyl" and "cycloalkylalkynyl" refer to a cycloalkyi group attached to the parent molecular group through an alkenyl and alkynyl group, respectively.

As used herein, "alkenyl" refers to an optionally substituted unsaturated, straight or branched chain hydrocarbon group containing at least one carbon-carbon double bond. In an embodiment, the alkenyl comprises from 2 to 8 carbon atoms "C 2 -C 8 alkenyl", in a further embodiment from 2 to 6 or 2 to 4 carbon atoms.

As used herein, "alkynyl" refers to an optionally substituted unsaturated, straight or branched chain hydrocarbon group containing at least one carbon-carbon triple bond. In an embodiment, the alkynyl comprises from 2 to 8 carbon atoms "C 2 -C 8 alkynyl", in a further embodiment from 2 to 6, or 2 to 4 carbon atoms.

As used herein, "halogen" refers to -F, -CI, -Br, or -I.

As used herein, a "heterocycle" or "heterocyclyl" is an optionally substituted aromatic or aliphatic monocyclic or bicyclic ring system that includes one or more carbon atoms and heteroatoms (e.g., 1 , 2, 3, or 4 heteroatoms), such as oxygen, nitrogen, and sulfur. Aliphatic heterocycles may have one or more double bonds. Examples of double bonds include carbon- carbon double bonds (C=C), carbon-nitrogen double bonds (C=N), and nitrogen-nitrogen double bonds (N=N). Examples of 3- to 9-membered heterocycles include, but are not limited to, aziridinyl, oxiranyl, thiiranyl, azirinyl, diaziridinyl, diazirinyl, oxaziridinyl, azetidinyl, azetidinonyl, oxetanyl, thietanyl, diazinanyl, piperidinyl, tetrahydropyridinyl, piperazinyl, morpholinyl, azepinyl or any partially or fully saturated derivatives thereof, diazepinyl or any partially or fully saturated derivatives thereof, pyrrolyl, oxazinyl, thiazinyl, diazinyl, triazinyl, tetrazinyl, imidazolyl, benzimidazolyl, tetrazolyl, indolyl, isoquinolinyl, quinolinyl, quinazolinyl, pyrrolidinyl, purinyl, isoxazolyl, benzisoxazolyl, furanyl, furazanyl, pyridinyl, oxazolyl, benzoxazolyl, thiazolyl, benzthiazolyl, thiophenyl, pyrazolyl, triazolyl, benzodiazolyl, benzotriazolyl, pyrimidinyl, isoindolyl, and indazolyl. Where an heterocycle is substituted, substituents can include any substituent group described herein. In an embodiment, the heterocycle is a "3- to 9-membered heterocycle".

As used herein, "heterocyclylalkyl" refers to a heterocyclic group attached to the parent molecular group through an optionally substituted alkyl group. As used herein, "aromatic" refers to a cyclic ring system having (4n +2) π electrons in conjugation, where n is 1 , 2, or 3.

Any group described herein may be substituted or unsubstituted. When substituted, they may be with any desired substituent or substituents that do not adversely affect the desired activity of the compound. Examples of preferred substituents are those found in the exemplary compounds and embodiments disclosed herein, as well as substituents such as: halogen (chloro, iodo, bromo, or fluoro); Ci -6 alkyl; C 2-6 alkenyl; C 2-6 alkynyl; hydroxyl; Ci -6 alkoxyl; amino (primary, secondary, or tertiary); nitro; thiol; thioether; imine; cyano; amido; carbamoyl; phosphonato; phosphine; a phosphorus (V) containing group; carboxyl; thiocarbonyl; sulfonyl; sulfonamide; ketone; aldehyde; ester; oxo; haloalkyl (e.g., trifluoromethyl); cycloalkyl, which may be monocyclic or fused or non- fused polycyclic (e.g., cyclopropyl, cyclobutyl, cyclopentyl, or cyclohexyl), or aliphatic heterocyclic, which may be monocyclic or fused or non-fused polycyclic (e.g., pyrrolidinyl, piperidinyl, piperazinyl, morpholinyl, or thiazinyl); and aromatic carbocyclic or heterocyclic, monocyclic or fused or non- fused polycyclic (e.g., phenyl, naphthyl, pyrrolyl, indolyl, furanyl, thiophenyl, imidazolyl, oxazolyl, isoxazolyl, thiazolyl, triazolyl, tetrazolyl, pyrazolyl, pyridinyl, quinolinyl, isoquinolinyl, acridinyl, pyrazinyl, pyridazinyl, pyrimidinyl, benzimidazolyl, benzothiophenyl, or benzofuranyl). Specific substituent groups includes benzyloxy; -N(CH 3 ) 2 ; O-alkyl (0-CH 3 ); O-aryl; aryl; aryl-lower alkyl; - C0 2 CH 3 ; -OCH 2 CH 3 ; methoxy; -CONH 2 ; -OCH 2 CONH 2 ; -S0 2 NH 2 ; -OCHF 2 ; -CF 3 ; and -OCF 3 . A substituted group may have 1 , 2, 3, 4, 5, 6, 7, or 8 substituent groups. These substituent groups may optionally be further substituted with a substituent listed herein. Substituents may also be optionally substituted by a fused-ring structure or bridge, for example -OCH 2 0-. In other embodiments, these substituents are not further substituted.

As used herein, "pharmaceutically acceptable salt(s)," includes but are not limited to salts of acidic or basic groups that may be present in compounds used in the present compositions. Exemplary pharmaceutically acceptable salts are described in Berge et al., J. Pharm. Sci. 1977; 66:1 -19 and in Pharmaceutical Salts: Properties, Selection, and Use, (Eds. P.H. Stahl and CG. Wermuth, Wiley-VCH, 2008). Compounds included in the present compositions that are basic in nature are capable of forming a wide variety of salts with various inorganic and organic acids. The acids that may be used to prepare pharmaceutically acceptable acid addition salts of such basic compounds are those that form non-toxic acid addition salts, i.e., salts containing pharmacologically acceptable anions, including, but not limited to, sulfuric, citric, maleic, acetic, oxalic, hydrochloride, hydrobromide, hydroiodide, nitrate, sulfate, bisulfate, phosphate, acid phosphate, isonicotinate, acetate, lactate, salicylate, citrate, acid citrate, tartrate, oleate, tannate, pantothenate, bitartrate, ascorbate, succinate, maleate, gentisinate, fumarate, gluconate, glucaronate, saccharate, formate, benzoate, glutamate, methanesulfonate, ethanesulfonate, benzenesulfonate, p-toluenesulfonate, mesylate, hydroxymethylsulfonate, hydroxyethyl sulfonate, and pamoate (i.e., 1 ,1 '-methylene-bis- (2-hydroxy-3-naphthoate)) salts. Similarly, compounds of the invention that include ionizable hydrogens can be combined with various inorganic and organic bases to form salts. Representative alkali or alkaline earth metal salts include sodium, lithium, potassium, calcium, magnesium and the like, as well as nontoxic ammonium, quaternary ammonium, and amine cations, including, but not limited to, ammonium, tetramethylammonium, tetraethylammonium, methylamine, dimethylamine, trimethylamine, triethylamine, ethylamine, and the like.

In embodiments of Formula (1 ), R 1 is substituted aryl, such as aryl substituted with one to three alkyl groups, preferably methyl groups. In embodiments, the substituted aryl is phenyl substituted with two methyl groups, such as:

In an embodiment, R 1 is an arylmethyl. In embodiments, R 1 is an unsubstituted arylmethyl. In embodiments, the aryl group in the arylmethyl is phenyl. In a further embodiment, R 1 is:

In other embodiments, R 1 is an arylmethyl substituted by an alkyl group, preferably a C 1-3 alkyl group, such as ethyl. In embodiments, the alkyl group substitutes the methyl group of the arylmethyl. In a further embodiment, R 1 is:

In another embodiment, R 1 is unsubstituted or substituted heteroaryl. . In an embodiment, the heteroaryl group is unsubstituted.

In embodiments, the heteroaryl has an aryl ring fused to a 5-membered ring comprising one or two heteroatoms, preferably two oxygen atoms. In a further embodiment, R 1 is:

In other embodiments, the heteroaryl has two aryl rings fused to a 6-membered ring comprising one or two heteroatoms, preferably two oxygen atoms.. In a further embodiment, R 1 is:

In another embodiment, R 1 is a heteroaryl comprising one or two heteroatoms. In an embodiment, the heteroaryl is unsubstituted. In an embodiment, the heteroaryl is 6-membered.. In an embodiment, the heteroaryl has one heteroatom. In a further embodiment, the heteroatom is a nitrogen atom. In a further embodiment, R 1 is:

In other embodiments, the heteroaryl is substituted, for example with one to three alkyl groups, preferably methyl groups. In an embodiment, the heteroaryl comprises a 6-membered ring linked to a 5-membered ring. In embodiments, the 5-membered ring comprises one or two heteroatoms, preferably two nitrogen atoms. In embodiments, the substituted heteroaryl is 1 -phenyl-1 H-pyrazole substituted with one methyl group, such as:

In another embodiment, R 1 is alkyl substituted with cycloalkyl. In a further embodiment R1 is otherwise unsubstituted alkyl substituted with unsubstituted cycloalkyl. In an embodiment, the alkyl group in the alkylcycloalkyl is a C 1-6 alkyl, such as a alkyl. In a further embodiment, the alkyl is an ethyl group. In an embodiment, the cycloalkyl is a 5-membered cycloalkyl. In a further embodiment, R 1 is:

In an embodiment, R 1 is not:

In embodiments, one of R 2 and R 3 is hydrogen or alkyl (preferably C C 3 alkyl), and the other is alkyl comprising 3 or more carbon atoms, aralkyi (preferably phenyl-alkyl) or cycloalkyl (preferably cyclohexyl or adamantyl).

In an embodiment, R 2 is H and R 3 is a C 3 -C 6 alkyl. In an embodiment, R 3 is linear. In another embodiment, R 3 is branched. In a further embodiment, R 3 is a C 4 alkyl. In a further embodiment, NR 2 R 3 is:

In an embodiment, NR R is:

In an embodiment, NR 2 R 3 is:

In an embodiment, NR 2 R 3 is:

In an embodiment, R 2 and R 3 are C 3-6 alkyi, preferably propyl.

In another embodiment, R 2 is H and R 3 is a C 3 -C 12 cycloalkyl, in a further embodiment a Cio cycloalkyl. In a further embodiment 3 is adamantly, and NR 2 R 3 is for example adamant-1 -yl:

In another embodiment, R 2 is H and R 3 is a C7-C14 arylalkyl, such as a C7-C1 0 arylalkyl, in a further embodiment an unsubstituted arylalkyl. In a further embodiment NR 2 R 3 is:

In another embodiment, R 2 and R 3 together with the nitrogen atom to which they are attached form an optionally substituted 6- to 9-membered non heterocycle. In an embodiment, the heterocycle comprises zero or one additional heteroatom, in a further embodiment the additional heteroatom is nitrogen. In an embodiment, the heterocycle is a 6-membered aliphatic heterocycle. In another embodiment, the heterocycle is substituted, in a further embodiment substituted with an alkyi, such as a C-|-C 6 or C1-C4 alkyi, preferably methyl. In a further embodiment, the substituent is methyl. In a further embodiment, 2 R 3 is:

In an embodiment, NR 2 R 3 is: In an embodiment, NR 2 R 3 is:

In an embodiment, NR 2 R 3 is not:

or NH-CO-CH 3 .

In an embodi (1 a):

( 1 a) wherein R 1 is as defined above, or a pharmaceutically acceptable salt thereof.

In another embodiment the compound is a compound of formula (1 b):

wherein R 2 and R 3 are as defined above, or a pharmaceutically acceptable salt thereof.

In a further embodiment, the compound is: 26

, or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

, or a pharmaceutically acceptable salt thereof. In an embodiment the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

υ or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

O cm or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment the compound is:

or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is: or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

a pharmaceutically acceptable salt thereof.

or a pharmaceutically acceptable salt thereof.

In embodiments of Formula (2), R 1 is a substituted or unsubstituted aryl, in a further embodiment a substituted aryl. In an embodiment, the aryl is substituted with an alkyl, for example one or three alkyl, in a further embodiment a Ci-C 6 or C1-C4 alkyl, such as a methyl. In embodiments, the substituted aryl is phenyl substituted with two methyl groups, such as:

In embodiments, R 4 and R 5 , together with the nitrogen atom to which they are attached, form a 6-membered, optionally substituted, aliphatic heterocycle, optionally comprising one or more additional nitrogen or oxygen atom. In further embodiments, the heterocycle comprises one further oxygen atom. In embodiments, the heterocycle is substituted with one to three alkyl groups, preferably two alkyl groups, preferably methyl groups. In embodiments, R 4 and R 5 , together with the nitrogen atom to which they are attached, form:

In an embodiment, the compound is:

or a pharmaceutically acceptable salt thereof. In an embodiment, R 6 is H or a halogen, in a further embodiment the halogen is CI.

In an embodiment, R 7 is a C 3 -C 8 alkyl, in a further embodiment a C 3 -C 6 alkyl. In embodiment, the alkyl is substituted with one or more halogen atoms, for examples one to three such atoms, for example three fluoride atoms.

In an embodiment, R 8 and R 9 are independently alkyls, such as Ci -3 alkyls, preferably methyl groups, substituted by optionally substituted aryl or optionally substituted thioaryl. In embodiment, substituted preferably monocyclic aryl or thioaryl, for example phenyl and/or 2-thienyl, which are preferably unsubstituted. In a further embodiment, NR 8 R 9 is:

In another embodiment, when R 7 is not a Ci alkyl, R 8 and R 9 , together with the nitrogen atom to which they are attached, join to form a 5- or more membered substituted aliphatic heterocycle. In embodiments, the aliphatic heterocycle may be substituted pyrrolidinyl or piperidinyl:

wherein Z 5 is a substituted or unsubstituted aryl. In a further embodiment, Z 5 is a substituted aryl, in a further embodiment the substituent is alkyloxy, such as a methoxy (-0-CH 3 ). In a further embodiment, NR 8 R 9 is:

In a further embodiment, the compound is: 36

or a pharmaceutically acceptable salt thereof.

In an embodiment, the compound is:

or a pharmaceutically acceptable salt thereof. is:

rmaceutically acceptable salt thereof.

In an embodiment, the compound is:

or a pharmaceutically acceptable salt thereof.

or a pharmaceutically acceptable salt thereof.

d is:

or a pharmaceutically acceptable salt thereof. In another aspect, the present invention provides a compound of formula 1 , 1 a, 1 b, 2, 2a or (3) as defined above. In particular embodiments, the compound is:

In an embodiment, the above-mentioned compound reduces the formation of mRNA foci or aggregates in a cell.

It will be appreciated that the above-defined compounds can contain one or more asymmetric carbon atoms which give rise to enantiomers. The compounds can be prepared as racemates or can be made from enantiomeric intermediates. Both racemates and enantiomers form part of the present invention.

The present invention also provides prodrugs of the compounds of the invention. Prodrugs include derivatives of the compounds of the invention that can hydrolyze, oxidize, or otherwise react under biological conditions (in vitro or in vivo) to provide an active compound of the invention. Examples of prodrugs include, but are not limited to, derivatives and metabolites of a compound of the invention that include biohydrolyzable moieties such as biohydrolyzable amides, biohydrolyzable esters, biohydrolyzable carbamates, biohydrolyzable carbonates, and biohydrolyzable phosphate analogues. In certain embodiments, prodrugs of the compounds of the invention with carboxyl functional groups are the lower alkyl esters of the carboxylic acid. The carboxylate esters are conveniently formed by esterifying any of the carboxylic acid moieties present on the molecule. Prodrugs can typically be prepared using well-known methods, such as those described by Burger's Medicinal Chemistry and Drug Discovery 6 th ed. (Donald J. Abraham ed., 2001 , Wiley) and Design and Application of Prodrugs (H. Bundgaard ed., 1985, Harwood Academic Publishers Gmfh). Biohydrolyzable moieties of a compounds of the invention 1 ) do not interfere with the biological activity of the compound but can confer upon that compound advantageous properties in vivo, such as uptake, duration of action, or onset of action; or 2) are biologically inactive but are converted in vivo to the biologically active compound. Examples of biohydrolyzable esters include, but are not limited to, lower alkyl esters, alkoxyacyloxy esters, alkyl acylamino alkyl esters, and choline esters. Examples of biohydrolyzable amides include, but are not limited to, lower alkyl amides, a-amino acid amides, alkoxyacyl amides, and alkylaminoalkylcarbonyl amides. Examples of biohydrolyzable carbamates include, but are not limited to, lower alkylamines, substituted ethylenediamines, aminoacids, hydroxyalkylamines, heterocyclic and heteroaromatic amines, and polyether amines.

The above-mentioned compounds can be obtained via standard, well-known synthetic methodology, see e.g. March, J. Advanced Organic Chemistry; Reactions Mechanisms, and Structure, 4 th ed., 1992. Illustrative methods are described below. Starting materials useful for preparing the compounds of the invention and their intermediates are also commercially available or can be prepared from commercially available materials using known synthetic methods and reagents.

The compounds of formula 1 , 1 a and 1 b may be synthesized, for example, based on the methods disclosed in PCT publication No. WO 99/36398.

Because of their activity, the above-defined compounds are advantageously useful in veterinary and human medicine. The compounds of formulas 1 , 1 a, 1 b, 2, and 3 described herein are useful for the treatment of polynucleotide repeat disorders. In an embodiment, a compound described herein is formulated in a pharmaceutical composition, in association with one or more carrier(s). As used herein, "carrier", "pharmaceutical carrier" or "pharmaceutically-acceptable carrier" refers to a diluent, adjuvant, excipient, or vehicle with which a compound of the invention is administered. Such pharmaceutical carriers can be liquids, such as water and oils, including those of petroleum, animal, vegetable or synthetic origin, such as peanut oil, soybean oil, mineral oil, sesame oil and the like. Saline solutions and aqueous dextrose and glycerol solutions can also be employed as liquid carriers, particularly for injectable solutions. The pharmaceutical carriers can be saline, gum acacia, gelatin, starch paste, talc, keratin, colloidal silica, urea, and the like. In addition, auxiliary, stabilizing, thickening, lubricating and coloring agents may be used. Suitable pharmaceutical carriers also include excipients such as starch, glucose, lactose, sucrose, gelatin, malt, rice, flour, chalk, silica gel, sodium stearate, glycerol monostearate, talc, sodium chloride, dried skim milk, glycerol, propylene, glycol, polyethylene glycol 300, water, ethanol, polysorbate 20, wetting or emulsifying agents, or pH buffering agents.

As used herein, and as well understood in the art, "treatment" is an approach for obtaining beneficial or desired therapeutic results, such as clinical results. Beneficial or desired results can include, but are not limited to, alleviation or amelioration of one or more symptoms or conditions (e.g., for myotonic dystrophy, one or more of the symptoms depicted in FIG. 1 , such as muscle weakness, muscle wasting); diminishment of extent of disease, disorder, or condition; stabilization (i.e., not worsening) of state of disease, disorder, or condition; prevention of spread of disease, disorder, or condition; delay or slowing the progress of the disease, disorder, or condition; amelioration or palliation of the disease, disorder, or condition; and remission (whether partial or total). "Treatment" can also mean prolonging survival as compared to expected survival if not receiving treatment.

The term "polynucleotide repeat disorders" (or "polynucleotide repeat expansion disorders") refers to disorders characterized based upon the presence of unstable and abnormal expansions of DNA-repeats, which in turn typically results in abnormal expression of one or more genes. The most common types of repeats are trinucleotide repeats (often CAG, but also CCG, CTG, CGG, GCC or GAA). Accumulation of RNA foci (or RNA aggregates) is a hallmark of polynucleotide repeat disorders. Examples of polynucleotide repeat disorders include Huntington's disease (HD), Huntington's disease-like 2 (HDL2), spinocerebellar ataxias (types 1-8, 12 and 17), fragile X syndrome, fragile X-E syndrome, fragile X-associated tremor/ataxia syndrome, myotonic dystrophy (type 1 or type 2), juvenile myoclonic epilepsy, Dentatorubral-pallidoluysian atrophy (DRPLA), Friedreich's ataxia (FRDA), spinal and bulbar muscular atrophy (SBMA, also known as Kennedy's disease), chromosome 9p21 amyotrophic lateral sclerosis-frontotemporal dementia (ALS-FTD, associated with an hexanucleotide (GGGGCC) repeat expansion). In an embodiment, the trinucleotide repeat disorder is myotonic dystrophy, in a further embodiment myotonic dystrophy type 1 (DM1 ) (Steinert's disease).

In another aspect, the present invention provides a method for inhibiting the accumulation of RNA foci (RNA aggregates) in a cell, said method comprising contacting said cell with one or more compounds of formula 1 , 1 a, 1 b, 2 and/or 3 defined above, or a composition defined above. In an embodiment, the method is an in vitro method. In another embodiment, the above-mentioned method is an in vivo method.

In another aspect, the present invention provides a method for treating a disease associated with accumulation of RNA foci (RNA aggregates), said method comprising administering to a subject in need thereof an effective amount of one or more compounds of formula 1 , 1 a, 1 b, 2 and/or 3 defined above, or a composition defined above. Diseases associated with accumulation of RNA foci (RNA aggregates) include, for example, the polynucleotide repeat disorders noted above.

The patient or subject is an animal, including, but not limited to, a human, mammal, e.g., cow, horse, sheep, pig, cat, dog, mouse, rat, rabbit, mouse or guinea pig, or other animal such as a chicken, turkey, or quail. In an embodiment, the patient or subject is a human.

The present compositions, which comprise an effective amount of one or more of the above-defined compounds, can be administered by any convenient route, for example by infusion or bolus injection, or by absorption through epithelial or mucocutaneous linings (e.g., oral mucosa, rectal, intestinal mucosa, etc.), and can be administered together with another biologically active agent. Administration can be systemic or local. Various delivery systems are known, e.g., encapsulation in liposomes, microparticles, microcapsules, capsules, etc., and can be used to administer a compound. In certain embodiments, more than one compound is administered to a patient. Methods of administration include but are not limited to intradermal, intramuscular, intraperitoneal, intravenous, subcutaneous, intranasal, epidural, oral, sublingual, intranasal, intracerebral, intravaginal, transdermal, rectally, by inhalation, or topically to the ears, nose, eyes, or skin. The preferred mode of administration is left to the discretion of the practitioner.

In specific embodiments, it may be desirable to administer one or more compounds locally to the area in need of treatment. This may be achieved, for example, and not by way of limitation, by local infusion during surgery, topical application, e.g., in conjunction with a wound dressing after surgery, by injection, by means of a catheter, by means of a suppository, or by means of an implant, said implant being of a porous, non-porous, or gelatinous material, including membranes, such as sialastic membranes, or fibers.

Pulmonary administration can also be employed, e.g., by use of an inhaler or nebulizer, formulating with an aerosolizing agent, or via perfusion in a fluorocarbon or synthetic pulmonary surfactant. In certain embodiments, the compounds can be formulated as a suppository, with traditional binders and carriers such as triglycerides. In another embodiment, the compounds can be delivered in a vesicle, in particular a liposome. In yet another embodiment, the compounds can be delivered in a controlled-release system. The present compositions may include an effective amount of one or more of the above-defined compounds and a pharmaceutically acceptable carrier.

The present compositions can take the form of solutions, suspensions, emulsion, tablets, pills, pellets, capsules, capsules containing liquids, powders, sustained-release formulations, suppositories, emulsions, aerosols, sprays, suspensions, or any other form suitable for use.

Compounds, included in the present compositions, which are acidic in nature are capable of forming base salts with various pharmacologically or cosmetically acceptable cations. Examples of such salts include alkali metal or alkaline earth metal salts and, particularly, calcium, magnesium, sodium, lithium, zinc, potassium, and iron salts. In another embodiment, the compounds of the invention are formulated in accordance with routine procedures as a pharmaceutical composition adapted for intravenous administration to human beings. Typically, compounds for intravenous administration are solutions in sterile isotonic aqueous buffer. Where necessary, the compositions may also include a solubilizing agent. Compositions for intravenous administration may optionally include a local anesthetic such as lignocaine to ease pain at the site of the injection. Generally, the ingredients are supplied either separately or mixed together in unit dosage form, for example, as a dry lyophilized powder or water free concentrate in a hermetically sealed container such as an ampoule or sachette indicating the quantity of active agent. Where the compound is to be administered by infusion, it can be dispensed, for example, with an infusion bottle containing sterile pharmaceutical grade water or saline. Where the compound is administered by injection, an ampoule of sterile water for injection or saline can be provided so that the ingredients may be mixed prior to administration.

Compositions for oral delivery may be in the form of tablets, lozenges, aqueous or oily suspensions, granules, powders, emulsions, capsules, syrups, or elixirs, for example. Orally administered compositions may contain one or more optional agents, for example, sweetening agents such as fructose, aspartame or saccharin; flavoring agents such as peppermint, oil of wintergreen, or cherry; coloring agents; and preserving agents, to provide a pharmaceutically palatable preparation. Moreover, where in tablet or pill form, the compositions may be coated to delay disintegration and absorption in the gastrointestinal tract thereby providing a sustained action over an extended period of time. Oral compositions can include standard carriers such as mannitol, lactose, starch, magnesium stearate, sodium saccharine, cellulose, or magnesium carbonate. Such carriers can be of pharmaceutical grade.

The amount of the compound that will be effective in the treatment of a particular disorder or condition will depend on the nature of the disorder or condition, and can be determined by standard clinical techniques. In addition, in vitro or in vivo assays may optionally be employed to help identify optimal dosage ranges. The precise dose to be employed in the compositions will also depend on the route of administration, and the seriousness of the disease or disorder, and may be determined for a given case. Suitable effective dosage ranges for intravenous administration are generally about 0.01 to about 5 g, preferably about 0.01 to about 1 g of the compound per kilogram body weight. In specific embodiments, the i.v. dose is about 0.005 to about 0.5 g/kg, about 0.01 to about 0.3 g/kg, about 0.025 to about 0.25 g/kg, about 0.04 to about 0.20 g/kg, or about 0.05 to about 0.20 g/kg (or the equivalent doses expressed per square meter of body surface area). Alternatively, a suitable dose range for i.v. administration may be obtained using doses of about 1 to about 2000 mg, without adjustment for a patient's body weight or body surface area. Suitable dosage ranges for intranasal administration are generally about 0.01 pg/kg body weight to 10 mg/kg body weight. Suppositories generally contain 0.5% to 20% by weight of one or more compounds of the invention alone or in combination with another therapeutic agent. Oral compositions can contain about 10% to about 95% by weight of one or more compounds of the invention alone or in combination with another therapeutic agent. In specific embodiments of the invention, suitable dose ranges for oral administration are generally about 0.1 to about 200 mg, preferably about 0.5 to about 100 mg, and more preferably about 1 to about 50 mg of a compound of the invention per kilogram body weight or their equivalent doses expressed per square meter of body surface area. In specific embodiments the oral dose is about 0.25 to about 75 mg/kg, about 1.0 to about 50 mg/kg, about 2.0 to about 25 mg/kg, about 2.5 to about 15 mg/kg, or about 5.0 to about 20 mg/kg (or the equivalent doses expressed per square meter of body surface area). In another embodiment, a suitable dose range for oral administration, from about 10 to about 4000 mg, without adjustment for a patient's body weight or body surface area. Other effective doses may be extrapolated from dose-response curves derived from in vitro or animal model test systems. Such animal models and systems are well known in the art.

The invention also provides pharmaceutical kits comprising one or more containers containing one or more of the above-defined compounds. Optionally associated with such container(s) can be a notice in the form prescribed by a governmental agency regulating the manufacture, use or sale of pharmaceuticals or biological products, which notice reflects approval by the agency of manufacture, use or sale for human administration. In certain embodiments, the kit may also contain one or more agents useful for treating the polynucleotide repeat disorder to be administered in combination with one or more of the above-defined compounds.

MODE(S) FOR CARRYING OUT THE INVENTION

The present invention is illustrated in further details by the following non-limiting examples. Example 1 : Materials and Methods

C2C12 myoblast reporter cell line assays.

C2C12 murine myoblast cells and Phoenix retrovirus packaging cells were cultivated in DMEM supplemented with 10% FBS. Phoenix cells were transfected with the desired retroviral vector by the Calcium-Phosphate method and, 48 hours after transfection, retrovirus-containing supernatant was supplemented with polybrene and deposited on cells to infect. C2C12 Tet-Off cell lines were generated by infecting cells with the retrovirus pTet-Off IN (Clontech) and selecting for Doxycycline (Dox)-regulated clones. Cell lines expressing the LacZ-5CUG, LacZ-145CUG, LUC- 5CUG or LUC-200CUG transcripts were generated by infecting C2C12 Tet-Off cells with the desired reporter construct in the pRevTRE plasmid (Clontech) and selecting for stable clones with inducible expression. The 3'UTR of human DMPK containing 5 CTG repeats or 200 CTG repeats was cloned downstream of the luciferase or beta-galactosidase open reading frames. The C2C12 cell line(s) are cultivated in media containing 1 ng/ml Dox to repress transcription of the reporter constructs.

For the beta-galactosidase assay with the LacZ-145CUG and LacZ-5CUG cell lines, Dox- containing media is removed and plates are washed twice with 1X PBS, and incubated for 4hrs in media without Dox. The cells are then trypsinised and 5,000 cells per well are plated in 100μΙ of media containing 0.1 ng/ml of Dox in black 96-well plates. The next day, media is replaced with fresh media containing the compounds at the desired concentration in this manner: the media is put in a screw-cap tube and heated at 37°C. The 20mM compound stock in 100% DMSO is pipetted directly into the media, Dox is also added to 0.1 ng/ml, and the tube is mixed well by inversion and gentle vortexing, and immediately overlayed on the cells. Pure DMSO is used for the control cells. The final concentration of DMSO is never higher than 0.5 %. Stocks of compound are prepared in 100% ultra-pure tissue culture tested DMSO (SIGMA D2650), aliquoted in single use volumes to avoid freezing-thawing, and kept at -20°C. Beta-galactosidase expression is determined with the Fluorescein di(Beta-D-galactopyranoside) FDG assay. The lysis buffer is prepared as follows (for 50 ml): 15μΙ Triton X-100, 5ml HEPES 100mM pH 7.8, and 400μΙ MgS0 4 8mM. The FDG substrate (Invitrogen F1 179) stock is prepared at 20mM in a solution of dH20/DMSO/EtOH 8:1 :1 . Cells are washed in 1X PBS, then lysed in 90μΙ of lysis buffer, and 10μΙ of 1 mM FDG (diluted in dH20) is added for a final concentration of 100μΜ. The plate is incubated at room temp for 2 hrs (protected from light) and read in fluorescence at 490nm/515nm.

Seven day treatment of DM1 patient myoblasts with compounds Human myoblasts were obtained from the Myosix institute associated with the Association Francaise contre les Myopathies (AFM). Five primary myoblast lines were obtained as controls, and one line from a DM1 patient with a 3.8 Kb CTG triplet amplification corresponding to approximately 1250 CTG repeats. The human myoblasts are cultivated in HAM-F10 media (SIGMA N6635) with 15% FBS, 1 % chick embryo extract, and 390 ng/ml of dexamethasone. One day before adding compounds, 2 X 10 5 cells were plated in 100mm plates (for CUGBP1 western blot or RT-PCR alternative splicing assay) or 2 X 10 4 cells/well are seeded in 12-well plates containing 12mm round glass coverslips (for FISH to detect RNA foci). One day after plating, media is replaced with fresh media containing the compounds at the desired concentration in this manner: the media is put in a screw-cap tube and heated at 37°C. The 20mM compound stock in 100% DMSO is pipetted directly into the media and the tube is mixed well by inversion and gentle vortexing, and immediately overlayed on the cells. Pure DMSO is used for the control cells. Final concentrations on myoblasts are never higher than 0.5 %. This procedure is repeated twice during the seven days of treatment, the media is thus replaced with fresh media containing compound every 2-3 days. Cells are collected or fixed for FISH after one week.

For Western blots, CUGBP1 was detected with the 3B1 -3D1 1 monoclonal antibody (mAb) (SIGMA C51 12) and GAPDH was detected with the 6C5 mAb (Millipore MAB374). The enhanced chemiluminescence signal (ECL) was acquired and quantified with the ChemiDoc™ MP imaging system (Bio-Rad). For alternative splicing assays, total RNA was collected with Trizol™ (Invitrogen), and 1 μg was used for reverse transcription with random primers with the RevertAid™ H Minus M-MuL RT (Fermentas) according to manufacturer's instructions. For MBNL2, PCR amplification was performed with primers 5'-ACAAGTGACAACACCGTAACCG-3' (SEQ ID NO:1 ) and 5'-TTTGGTAAAGGATGAAGAGCACC-3' (SEQ ID NO:2). SERCA1 was amplified with primers 5'-ATGATCTTCAAGCTCCGGGC-3' (SEQ ID NO:3) and 5'-CAGCTCTGCCTGAAGATGTG-3' (SEQ ID NO:4). For TNNT2 (cardiac Troponin T), PCR was carried out with primers 5'- AT AG AAG AG GTG GTG G AAG AGT AC-3' (SEQ ID NO:5) and 5'-

GTCTCAGCCTCTGCTTCAGCATCC-3' (SEQ ID NO:6). The alternative splicing of the hnRNPA2/B1 transcript was detected with the primers 5'-CTGAAGCGACTGAGTCCGCG-3' (SEQ ID NO:7) and 5'-ACAGTCTGTAAGCTTTCCCC-3' (SEQ ID NO:8). PCR was performed with Taq using standard conditions but the number of amplification cycles was optimized to be in the quantitative range for each gene. The products of PCR amplification were resolved on 3.5 % agarose gels containing Gelred™ (Biotium) and images were acquired and quantified with the ChemiDoc™ MP imaging system (Bio-Rad). Detection and Quantification of CUG-triplet repeat RNA foci

For fluorescent in situ hybridization (FISH), cells grown on glass coverslips were fixed in PBS containing 4% paraformaldehyde, dehydrated for 2 hours in 70% ethanol, and incubated overnight with 10 ng of probe in 2XSSC 40% formamide at 37 ° C. The next day, coverslips were washed twice in 2XSSC 40% formamide, treated with DAPI, and mounted in glycerol:PBS:PPD mounting media. To visualize CUG-triplet repeat mRNA foci, a single probe with the sequence (CAG)io was labelled with Cy3 and used for FISH. A control Cy3-labelled probe with the sequence (CTG)io gave no hybridization signal in DM1 patient myoblasts. Images for FISH were acquired on an epifluorescence light microscope with a 1.40 NA 60X oil objective and a CCD camera. For each field of cells, six image Z-stacks with 0.5 μιη steps were acquired with Cy3 and DAPI filter cubes. To quantify the CUG repeat RNA aggregates, the 3D images of the foci were first processed by maximum intensity projection with the MetaMorph™ software (Molecular Devices Inc.), resulting in a 2D image containing all the foci in each cell. A script was developed with MetaMorph™ to first outline the nuclei using the DAPI images, then threshold the CUG-repeat RNA foci in the nuclei to define them, and finally sum the surface area covered by the nuclear RNA foci in each nucleus. The threshold value for the CUGn RNA foci was determined by obtaining the average value for background nucleoplasm^ fluorescence when WT human myoblasts are hybridized with the (CAG)io-Cy3 probe (this background fluorescence signal is a combination of non-specific hybridization of the probe, autofluorescence of the cells and camera background). The MetaMorph™ program then calculates the surface area covered in each nucleus for all the foci in the Cy3 image that have a) pixels with a fluorescence value above the threshold, and b) a size larger than 3 pixels (to remove single pixel background). For each treatment condition, 60 to 80 cells were analyzed and all the data points were presented, with the median value indicated. Each experiment was repeated at least three times.

A high-throughput screen (HTS) of chemical compounds was performed in order to identify and validate drug candidates that relieve the nuclear retention of CUGn mRNAs and dislocate nuclear foci formation in a DM1 cell culture model. For that screen, a reporter mRNA containing a luciferase ORF with the DMPK 3' untranslated region containing 200 CUG repeats was used as a model system for DM1 . This allowed the screening of a comprehensive library of 1 10,000 small molecules (Maybridge™ Screening collection and Specs™ screening library) for effectors on CUGn mRNA nuclear retention and RNA foci formation. In a secondary screen, fluorescent in situ hybridization (FISH) was used to visualize the disruption of the nuclear CUG-rich RNA foci. From these screens, two chemical compounds which: 1 ) increase nuclear export and translation of reporter mRNAs containing expanded CUG triplet repeats, and 2) decrease the number and intensity of nuclear CUGn RNA foci, were identified.

Example 3: Characterization of the two "hit" compounds

The two compounds identified by the screening assay described above, which were named compound 15 and 32, have the structure depicted in FIG. 3. Compound 15 has a para- sulfonamide structure, and compound 32 is a benzyl methylthiophene amide. Both are small molecules which fit the Lipinski's "rule of five" for drug-like properties (C.A. Lipinski; F. Lombardo; B.W. Dominy and P.J. Feeney (2001 ). Adv Drug Del Rev 46: 3-26) and should thus demonstrate good ADME (absorption, distribution, metabolism and excretion) profiles. The dose-response effect of candidate drugs 15 and 32 on the nuclear export of luciferase-CUG200 mRNA was assessed in the C2C12 myoblast cell line. The amount of CUG repeat RNA that get exported and properly translated into luciferase in response to treatment with varying concentrations of target drugs was measured in order to determine the dose-response curves. The data show that, for compound 15, the plateau of maximal effect was reached at 15 μΜ (EC 50 = 6.8 μΜ), while a maximal effect peak was reached at 5 μΜ (EC 50 = 2.5 μΜ) for compound 32.

Example 4: Reduction of CUG-rich RNA foci formation in DM1 patient myoblasts

To confirm the activity of these compounds on an endogenous target, the ability of the two chemical compounds to disrupt RNA foci formation in myoblasts from a patient with DM1 was verified. The DM1 myoblasts with approximately 1250 CTG triplets in the DMPK gene were treated for 7 days with 15 μΜ of compound 15 or 5 μΜ of compound 32, compared to the control (DMSO only). Using DAPI staining and fluorescent in situ hybridization (FISH) with a probe against CUG- repeat RNA, the surface occupied by RNA foci in the nucleus was measured with the MetaMorph™ software, which provides a quantitative measurement of the amount of CUG-repeat mRNA aggregated and retained in a given nucleus. A detailed description of the FISH and foci quantification procedures is included in the section entitled "Detection and quantification of CUG- triplet repeat RNA foci" of Example 1 (Materials and methods). As shown in FIG. 4, a more than two-fold decrease in RNA foci formation was observed in DM1 myoblasts for both compounds. These experiments were repeated several times, with similar results. These results confirm the inhibitory effect of the hit compounds on the formation of CUG-rich RNA foci in myoblasts from DM1 patient, which express endogenous DMPK transcripts with a large expansion of CUG triplets.

Example 5: Structure-activity relationship (SAR) analysis of compounds 15 and 32 A screen of small-molecules structurally similar to compounds 15 and 32 isolated in our original high-throughput screen was performed. 49 compounds that are structural analogues of compound 15 or 32 were chosen. These are shown in the tables below:

These analogous compounds allowed testing the structure-activity relationship of compounds 15 and 32. From this screen, 15 new hit compounds that increased β-galactosidase activity nearly two-fold in C2C12 myoblasts expressing the LacZ-145CUG reporter mRNA, and had no effect on activity from the LacZ-5CUG mRNA control cell line, were identified (FIG. 5). Twelve of these new compounds are related to drug candidate 15, and two are similar to drug candidate 32 (chemical structures shown in FIG. 6A and B).

Example 6: Synthesis of further analogs

Other analogues as depicted in FIG. 6C, including a "chimera" of both compounds 15 and

32, were synthesized.

Synthetic methods for preparing these compounds are illustrated below. Starting materials are commercially available or may be prepared according to procedures known in the art or as illustrated herein.

All temperatures are in degrees Celsius. Mass spectra (MS) were measured by electrospray ion-mass spectroscopy (ESI) on an Agilent 6120 Quadrapole™ MS coupled to an Agilent 1 100™ series HPLC instrument. NMR spectra were recorded on a Varian Mercury spectrometer at 400 MHz for 1 H and 376 MHz for 19 F. The following abbreviations have the indicated meanings:

AcOH acetic acid

Alk alkyl

Ar aryl

atm = atmosphere

BINAP 2,2'-bis(diphenylphosphino)-1 ,1 '-binaphthalene

Boc tert-butoxycarbonyl

n-BuLi n-butyllithium

Cbz carboxybenzyl

CH2CI2 = dichloromethane

DBU 1 ,8-diazabicyclo[5.4.0]undec-7-ene

DEAD diethyl azodicarboxylate

DIPEA N,N-diisopropylethylamine

DMAP 4-(dimethylamino)pyridine

DMF N,N-dimethylformamide

DMSO dimethyl sulfoxide

ESI electrospray ionization

Et 3 N triethylamine

Et 2 0 diethylether

EtOAc or EA = ethyl acetate

EtOH ethyl alcohol

h hour(s)

H 2 hydrogen

HATU 0-(7-azabenzotriazol-1-yl)-N,N,N',N'-tetrameth} hexafluorophosphate

HCI hydrochloric acid

HPLC High Pressure Liquid Chromatography iPrOH 2-propanol

KF potassium fluoride

LC-MS Liquid Chromatography Mass Spectrometry

LiOH lithium hydroxide

MeCN acetonitrile

MeMgBr = methylmagnesium bromide

MeOH methyl alcohol MeTHF : 2-methyltetrahydrofuran

MgS0 4 = magnesium sulfate

min = = minute(s)

MS = mass spectroscopy

MTBE : methyl fert-butyl ether

N 2 = nitrogen

NaBH 4 = sodium borohydride

NaHCOa = sodium bicarbonate

NaOH = sodium hydroxide

Na 2 S0 4 = sodium sulfate

NH 3 = ammonia

NH 4 CI = ammonium chloride

NH 4 OH = ammonium hydroxide

NMP : /V-methyl 2-pyrrolidinone

NMR = nuclear magnetic resonance spectrosc

Moc : = methoxycarbonyl

P = pressure

Pd/C = palladium on charcoal

PG = protecting group

Ph = phenyl

Pyr = pyridine

rbf = round bottom flask

Rf = retention factor on silica gel rt = room temperature

TBDMS = tert-butyldimethylsilyl

Ts = toluene-4-sulfonyl

TFA = trifluoroacetic acid

TFAA = trifluoroacetic anhydride

TFA-NHS = trifluoroacetic /V-hydroxysuccinimide

THF = tetrahydrofuran

TLC = thin layer chromatography

TMEDA = Λ/,Λ/,Λ/',Λ/'-tetramethylethylenediamine

T3P = Propylphosphonic anhydride General Sequence 1 :

Step 1 : Thionyl chloride (0.90 ml_, 12 mmol, 5.0 eq.) was added dropwise to a stirred suspension of 5-methyl-1 -phenyl- //-/-pyrazole-4-carboxylic acid (0.5 g, 2.5 mmol; prepared according to Dycman et al. WO 2004/099156) in DCM (5.0 mL) at 0°C. The reaction was subsequently warmed to rt and stirred for an additional 6 hours before being concentrated to dryness under high vacuum. The residue was used directly in the next step.

Step 2: DIPEA (2.0 eq.) was added to a solution of the product from Step 1 , an appropriate secondary amine (1 .2 eq.) and DMAP (0.1 eq.) in DCM (0.2 M). The reaction was stirred at rt for 3 hours before being concentrated to dryness and the residue purified directly by flash chromatography on silica gel eluting with an increasing proportion of EtOAc in hexanes. yVjyV-Dibenzyl-S-methyl-l -phenyl-iH-pyrazole^-carboxamide (Compound 214)

Prepared as a colorless gum using dibenzylamine in General Sequence 1.

1 H NMR (CDCI 3 ) δ 7.58 (1 H), 7.52-7.21 (15H), 4.70 (4H), 2.50 (3H). MS ESI: 382.2 [M

H] + . lndolin-1 -yl(5-methyl-1 -phenyl-fH-pyrazol-4-yl)methanone (Compound 220)

Prepared as a colorless gum using 2, 3-dihydro- //-/-indole in General Sequence 1.

1 H NMR (CDCI 3 ) δ 7.83 (1 H), 7.54-7.49 (2H), 7.48-7.44 (3H), 7.25-7.18 (3H), 7.04 (1 H), 4.31 (2H), 3.19 (2H), 2.52 (3H). MS ESI: 304.2 [M + H] + .

^-(^(WjW-Dipropylsulfamoy phenylJ-S-methyl-l -phenyl-iH-pyrazole^-carboxamide

(Compound 196)

Reaction Sequence:

Step 1 : Dipropylamine (1.9 ml_, 14 mmol, 3.0 eq.) was added dropwise to a suspension of 4-nitrobenzenesulfonyl chloride (1 .0 g, 4.5 mmol) in DCM (15 mL) at 0°C. After 10 minutes at 0°C, the reaction was stirred at rt for 2 hours before being partitioned between water and DCM. The layers were separated and the aqueous layer was extracted with DCM (2X). The combined organic layers were dried (MgS0 4 ), filtered and concentrated under reduced pressure and the residue was purified by flash chromatography on silica gel eluting with an increasing proportion of EtOAc in hexanes.

Step 2: A saturated solution of NH 4 CI (0.5 mL) and iron powder (0.59 g, 10 mmol, 10 eq.) were added to a solution of the product from Step 1 (0.30 g, 1 .0 mmol) in EtOH (4.0 mL). The mixture was heated at 65°C for 18 hours before being cooled to rt and partitioned between EtOAc and water. The layers were separated and the aqueous layer extracted with EtOAc (3X). The combined organics layers were dried (MgS0 4 ), filtered, concentrated and dried under suction and high vacuum to afford 4-amino-/V,/V-dipropylbenzenesulfonamide which was used directly without further purification.

Step 3: The product from Step 2 (42 mg, 0.16 mmol, 1 .2 eq.) was added to a solution of 5- methyl-1 -phenyl- //-/-pyrazole-4-carbonyl chloride (30 mg, 0.14 mmol) and DIPEA (48 μΙ_, 0.27 mmol, 2.0 eq.) in DCM (0.68 mL). The reaction was stirred at rt for 18 hours before being concentrated to dryness. The residue was purified by flash chromatography on silica gel eluting with an increasing proportion of EtOAc in hexanes to afford the title compound as a yellow solid.

1 H NMR (DMSO-d6): δ 10.19 (1 H), 8.35 (1 H), 7.96 (2H), 7.76 (2H), 7.61-7.49 (5H), 3.03- 2.99 (4H), 2.56 (3H), 1 .48-1 .40 (4H), 0.85-0.77 (6H). MS ESI: 463.2 [M + Na] + .

W-Benzyl-1 ^henyl-W-(thiophen-2-ylmethyl)-5-(trifluoromethyl)-iH^yrazol e-4-carboxamide (Compound 102)

Reaction Sequence:

Step 1 : Benzylamine (3.8 mL, 35 mmol, 1.05 eq.) and acetic acid (9.6 mL, 170 mmol, 5 eq.) were added to a solution of 2-thiophenecarboxaldehyde (3.1 mL, 34 mmol) in DCE (1 10 mL) with stirring at rt for 18 hours. Sodium triacetoxyborohydride (8.95 g, 42.3 mmol, 1.2 eq.) was then added with continued stirring at rt for an additional hour. The reaction vessel contents were then cooled to 0°C and a saturated solution of NaHC0 3 was added slowly to pH 8. Following cessation of the evolution of hydrogen gas, the layers were separated and the aqueous layer was extracted with DCM (3X). The combined organic layers were dried (MgS0 4 ), filtered and concentrated under reduced pressure. The residue was purified by flash chromatography of the residue on silica gel eluting with an increasing proportion of EtOAc in hexanes. Step 2: DIPEA (96 μΙ_, 0.55 mmol, 3.0 eq.) was added to a suspension of 1 -phenyl-5- (trifluoromethyl)-i/-/-pyrazole-4-carboxylic acid (50 mg, 0.20 mmol, 1.05 eq.), the product from Step 1 (38 mg, 0.19 mmol) and HATU (81 mg, 0.21 mmol, 1 .2 eq.) in EtOAc (2.0 ml_). The mixture was stirred for 18 hours before being partitioned between EtOAc and a saturated solution of NaHC0 3 . The layers were separated and the aqueous layer extracted with EtOAc (3X). The combined organic layers were dried (MgS0 4 ), filtered and concentrated under reduced pressure. The product was isolated as a colorless solid by flash chromatography on silica gel eluting with an increasing proportion of EtOAc in hexanes.

1 H NMR (DMSO-d6): δ 7.84 (0.45H), 7.70 (0.55H), 7.54-7.26 (10H), 7.23-7.18 (1 H), 7.02- 6.94 (1.55H), 6.90-6.86 (0.45H), 4.86 (1.1 H), 4.79 (0.9H), 4.58 (0.9H), 4.54 (1 .1 H). MS ESI: 464.2 [M + Na] + .

Example 7: Effect of disruption of CUG-rich RNA foci on expression of splicing factors and mRNA splicing

A primary consequence of RNA toxicity in DM1 is dysfunction of two classes of splicing regulators: the CUG binding protein 1 (CUGBP1/CELF1 ) and the muscleblind protein 1 (MBNL1 ). MBNL1 recognizes YGCY motifs in RNA and binds with high affinity to CUG repeats (Goers et al. 2010). MBNL1 has been shown to co-localize in vivo with the nuclear foci of CUG-triplet repeat transcripts (Fardei et al. 2001 ), suggesting that the sequestration of this factor in the nucleus may lead to DM1 pathogenesis. Expanded CUG-repeat RNA is believed to increase CUGBP1 levels and activity in DM1 cells via activation of protein kinase C (PKC), which phosphorylates and stabilizes CUGBP1 protein levels (Kuyumcu-Martinez et al. 2007). CUGBP1 was found to bind CUG-repeat RNA but does not co-localize in vivo with the nuclear DMPK expanded-repeat foci (Fardei et al. 2001 ). It was next tested whether treatment of DM1 patient myoblasts with compounds 15 and 32 and representative analogues thereof could correct the increased CUGBP1 protein levels observed in these cells (FIG. 7). Western Blot analyses show that DM1 cells have a level of CUGBP1 about 2.5-times higher than WT cells (lane 1 compared to lanes 3 and 4). The myoblast cells were treated for 7 days at a 20μΜ concentration of the compounds or DMSO. As a control, it was verified that the treatment of WT cells with compound 15 does not change the levels of CUGBP1 (lane 2 compared to lane 1 ). Treatment of the DM1 myoblasts with compound 15 reduces CUGBP1 levels by 85% (lane 7 compared to lanes 3 and 4). Compounds 32 and 43 also reduce CUGBP1 levels by 30% and 45%, respectively (lanes 8 and 9 compared to lanes 3 and 4). Thus, the drug candidates have the capacity to correct the stabilization of the CUGBP1 alternative splicing factor in DM1 myoblasts. The abnormal increase in CUGBP1 levels has been linked to the severe muscle wasting which debilitates DM1 patients in advanced stages of the disease (Orengo et al. 2008).

The effect of treatment with candidate drugs 15 and 32 on the alternative splicing of specific mRNA, such as SERCA1 exon 22, TNNT2 exon 5, and MBNL2 exon 7, which are all mis- spliced to re-express a foetal or neonatal form in DM1 cells, was tested. Patient myoblasts were treated for 7 days with a 20μΜ concentration of candidate drugs. Total RNA was extracted, and used in RT-PCR reactions to quantify the splicing of the endogenous mRNA. SERCA1 (ATP2A1 ) is a sarcoplasmic/ER Ca 2+ -ATPase expressed in skeletal muscle. As shown in FIG. 8B, SERCA1 exon 22 inclusion changes from 59% in the WT cells, to less than 2% in the DM1 myoblasts. Treatment with compound 15 restores inclusion of exon 22 to 17% of total transcripts. The SERCA1 exon 22+ variant is the adult form, and the SERCA1 exon 22- is the neonatal form (Kimura et al. 2005). Mis-splicing of SERCA1 is thought to be involved in muscle wasting in DM1 . The data depicted in FIG. 8C shows that treatment with candidate compound 15 reduces the inclusion of foetal TNNT2 exon 5 from 90% to 76% of total transcripts. Mis-splicing of this exon 5 of cardiac Troponin T (TNNT2/cTNT) causes cardiac conduction defects in DM1 (Ho et al. 2005, Warf et al. 2009). MBNL2 is a muscleblind splicing regulator that is ubiquitously expressed. In FIG. 8D, it is shown that treatment with compound 15 reduces the inclusion of exon 7 from 40% to 25%. Mis- splicing of MBNL2 is thought to result in the mis-splicing of several transcripts in the brain in DM1 (Charizanis et al. 2012).

Compounds 15 and 32, and some of their analogues, were tested for the correction of

MBNL2 exon 7 mis-splicing. The data depicted in FIG. 9 shows that in DM1 cells which expressed 60% of total transcripts with the foetal form (exon 7+), treatment with the compounds reduces inclusion of exon 7 to about 45% of total transcripts. Overall, these results show that the compounds can correct some of the molecular defects in DM1 patient cells.

Example 8: Further testing of compounds in the alternative splicing assay

DM1 patient myoblasts were treated for 7 days with a 20μΜ concentration of each compound. Total RNA was extracted, and used in RT-PCR reactions to quantify the alternative splicing of exon 7 of MBNL2 and MBNL1 mRNAs, and exon 22 of Sercal mRNA, which are all mis- spliced to re-express a foetal or neonatal form in DM1 cells (Charizanis et al. 2012, Kimura et al. 2005). With the exception of analogue 31 (which exhibits cellular toxicity), all the analogues partially corrected the mis-splicing of MBNL2 and MBNL1 mRNA (FIG. 10). However, beside compound 15, most of these analogues had little effect on the mis-splicing of exon 22 of Sercal mRNA. The other analogues of compounds 15 and 32 (FIG. 6C) were tested in splicing assays to measure alternative splicing correction of MBNL2, MBNL1 and Sercal pre-mRNAs in DM1. Mis- splicing of exon 5 of cardiac Troponin T (TNNT2/cTNT), which causes cardiac conduction defects in DM1 (Ho et al. 2005, Warf et al. 2009), was also tested. Analogues which corrected splicing of at least 3 control pre-mRNAs above 5% were considered active. As shown in FIG. 1 1 , analogues 102, 196, and 214 were able to correct at least 3 mis-splicing of the pre-mRNAs tested. Depending on the mRNA tested, analogue 196 (which is a "chimera" of compounds 15 and 32) is either better or comparable to the original compound 15, since it significantly corrects mis-splicing of all the pre- mRNAs tested.

Example 9: Dose-response, specificity and toxicity of the compounds

To determine if the identified compounds act as true pharmacological inhibitors, a dose- response assay was performed with the alternative splicing assay using compound 32. Normal (M908) and patient (DM1 ) myoblasts were treated for 7 days with compound 32 at concentrations varying from 5 to 60 μΜ. Total RNA was extracted, and used in RT-PCR reactions to quantify the alternative splicing of the endogenous MBNL2 and MBNL1 mRNAs. As shown in FIGs. 12A and 12B, a clear dose-response effect can be observed on the splicing of MBNL2 (FIG. 12A) and MBNL1 (FIG. 12B) mRNAs upon treatment of DM1 myoblasts, as the splicing pattern of both mRNAs shift toward the pattern observed in normal myoblasts as the concentration of compound 32 increases.

To eliminate the possibility that compounds 15 and 32 act as general regulators of splicing, the specificity of the two families of compounds was tested on the inclusion of exon 2 of the hnRNPA2/B1 pre-mRNA, which is not mis-spliced in DM1 patients (Philips et al., 1998). Normal (M908) and patient (DM1 ) myoblasts were treated for 7 days with a 20μΜ concentration of the compounds. Total RNA was extracted, and used in RT-PCR reactions to quantify the splicing of the endogenous mRNA. As shown in FIG. 13, no significant change in exon 2 exclusion of the hnRNPA2/B1 pre-mRNA was observed between control (DMSO treated or inactive analogues) and cells treated with active compounds, for either normal or DM1 myoblasts. These results show that the compounds do not affect general splicing of pre-mRNAs, but act specifically on pre-mRNA mis- spliced in DM1 .

The cellular toxicity of compounds 15 and 32, and their analogs was also measured using a validated cytotoxicity assay. Normal or DM1 myoblasts were treated with 20μΜ of compound or DMSO. The ratio of viable cells was measured with a High Content Screening microscope in live cells treated with Hoescht 33342 (labels all cells) and propidium iodide (labels dead cells). The total number of cells was counted and the number of dead cells was subtracted to obtain the number of viable cells, which is presented as a ratio (drug-treated versus DMSO-treated cells). The percentage of dead cells was also measured using propidium iodide. As shown in FIG. 10, while some compounds like 15 and 31 show some toxicity, other compounds like 32 and 46 had little toxicity on the cells at this concentration.

The cellular toxicity of the compounds was also measured in a dose-response assay. In this assay, normal (M908) and patient (DM1 ) myoblasts were treated for 7 days with compound 32 or 196 at concentrations varying from 5 to 60 μΜ. Cell number and cell death was quantified by High Content Screening microscope, as specified above. For both compounds, little cytotoxicity was observed at concentration of 20μΜ and below (FIGs. 14A-D). The main phenotype observed was a cytostatic effect at higher concentrations, as cell death remains below 10% at concentrations lower than 60 μΜ. These results also show that some of the novel analogues were less toxic than the original compounds 15 and 32. Indeed, the ratio of viable cells at 20μΜ of compound 15 is 0,22 (FIG. 10), while for compound 196, an analogue of compound 15, it is above 1 at the same concentration (FIG. 14C).

Although the present invention has been described hereinabove by way of specific embodiments thereof, it can be modified, without departing from the nature of the subject invention as defined in the appended claims. In the claims, the word "comprising" is used as an open-ended term, substantially equivalent to the phrase "including, but not limited to". The singular forms "a", "an" and "the" include corresponding plural references unless the context clearly dictates otherwise.

REFERENCES

1. Brook, J. D., McCurrach, M. E., Harley, H. G., Buckler, A. J., Church, D., Aburatani, H., Hunter, K., Stanton, V. P., Thirion, J. P., Hudson, T. et al. (1992). Molecular basis of myotonic dystrophy: expansion of a trinucleotide (CTG) repeat at the 3' end of a transcript encoding a protein kinase family member. Cell 68, 799-808.

2. Buxton, J., Shelbourne, P., Davies, J., Jones, C, Van Tongeren, T., Aslanidis, C, de Jong, P., Jansen, G., Anvret, M., Riley, B. et al. (1992). Detection of an unstable fragment of DNA specific to individuals with myotonic dystrophy. Nature 355, 547-548.

3. Charizanis K., Lee K.Y., Batra R., Goodwin M., Zhang C, Yuan Y., Shiue L, Cline M., Scotti M.M., Xia G. et al. (2012). Muscleblind-like 2-mediated alternative splicing in the developing brain and dysregulation in Myotonic Dystrophy. Neuron 75: 437-50.

4. Daughters, R. S., Tuttle, D. L, Gao, W., Ikeda, Y., Moseley, M. L, Ebner, T. J., Swanson, M. S. and Ranum, L. P. W. (2009). RNA gain-of-function in spinocerebellar ataxia type 8. PLoS Genet. 5, e1000600. 5. Davis, B. M., McCurrach, M. E., Taneja, K. L, Singer, R. H., and Housman, D. E. (1997) Expansion of a CUG trinucleotide repeat in the 3' untranslated region of myotonic dystrophy protein kinase transcripts results in nuclear retention of transcripts PNAS 94: 7388-7393.

6. Fardaei, M., Larkin, K., Brook, J.D., and Hamshere, M.G. (2001 ). In vivo co-localisation of MBNL protein with DMPK expanded-repeat transcripts. Nucleic Acids Research. 29, 2766- 2771.

7. Fu, Y. H., Pizzuti, A., Fenwick, R.G. Jr., King, J., Rajnarayan, S., Dunne, P.W., Dubel, J., Nasser, G.A., Ashizawa, T., de Jong, P., et al. (1992) An unstable triplet repeat in a gene related to myotonic muscular dystrophy. Science 255:1256-1258.

8. Goers, E.S., Purcell, J., Voelker, R.B., Gates, D.P., and Berglund, J.A. (2010). MBNL1 binds GC motifs embedded in pyrimidines to regulate alternative splicing . Nucleic Acids Research 38,

2467-2484.

9. Ho T.H., Bundman D., Armstrong D.L., Cooper T.A. (2005). Transgenic mice expressing CUG- BP1 reproduce splicing mis-regulation observed in myotonic dystrophy. Hum. Mol. Genet. 14:1539-47. 10. Kimura T., Nakamori M., Lueck J.D., Pouliquin P., Aoike F., Fujimura H., Dirksen R.T., Takahashi M.P., Dulhunty A.F., Sakoda S. (2005). Altered mRNA splicing of the skeletal muscle ryanodine receptor and sarcoplasmic/endoplasmic reticulum Ca2+-ATPase in myotonic dystrophy type 1 . Hum. Mol. Genet. 14 :2189-2200. 1 1 . Kuyumcu-Martinez, N. M., Wang, G.-S., and Cooper, T. A. (2007) Increased Steady-State Levels of CUGBP1 in Myotonic Dystrophy 1 Are Due to PKC-Mediated Hyperphosphorylation. Molecular Cell 28, 68-78.

12. Liquori, CL et ai, (2001 ). Myotonic dystrophy type 2 caused by a CCTG expansion in intron 1 of ZNF9. Science 293, 864-867. 13. Mahadevan, M., Tsilfidis, C, Sabourin, L., Shutler, G., Amemiya, C, Jansen, G., Neville, C, Narang, M., Barcelo, J., O'Hoy, K. et al. (1992). Myotonic dystrophy mutation: an unstable CTG repeat in the 3 untranslated region of the gene. Science 255, 1253-1255.

14. Ranum, L. P. W. and Cooper, T. A. (2006). RNA-mediated neuromuscular disorders. Annu.

Rev. Neurosci. 29, 259-277 15. Rudnicki, D. D., Holmes, S. E., Lin, M. W., Thornton, C. A., Ross, C. A. and Margolis, R. L.

(2007). Huntington's disease-like 2 is associated with CUG repeat-containing RNA foci. Ann. Neurol. 61 , 272-282.

16. Tassone, F., Iwahashi, C. and Hagerman, P. J. (2004). FMR1 RNA within the intranuclear inclusions of fragile X-associated tremor/ataxia syndrome (FXTAS). RNA Biol. 1 , 103-105 17. Warf MB, Diegel JV, von Hippel PH., Berglund JA. (2009). The protein factors MBNL1 and U2AF65 bind alternative RNA structures to regulate splicing. Proc Natl Acad Sci USA. Jun 9; 106(23):9203-8.

18. Wheeler, T. M., Thornton, C.A. (2007) Myotonic dystrophy: RNA-mediated muscle disease.

Curr. Opin. Neurol. 20, 572-576. 19. Wheeler, T. M. (2008) Myotonic Dystrophy: Therapeutic Strategies for the Future.

Neurotherapeutics 5, 592-600.

20. Philips, Anne V., Timchenko, Lubov T., and Cooper, Thomas A., Disruption of Splicing Regulated by a CUG-Binding Protein in Myotonic Dystrophy. Science 280 (5364), 737 (1998).