US 2011 0195932A1 (19) United States (12) Patent Application Publication (10) Pub. No.: US 2011/0195932 A1 Wynne et al. (43) Pub. Date: Aug. 11, 2011

(54) DRUG COMBINATIONS FOR THE A63L/4045 (2006.01) TREATMENT OF DUCHENNE MUSCULAR C07D 277/66 (2006.01) DYSTROPHY A6II 3/428 (2006.01) C07D 235/04 (2006.01) (76) Inventors: Graham Michael Wynne, A63L/484 (2006.01) Oxfordshire (GB); Stephen Paul C07D 263/58 (2006.01) Wren, Buckinghamshire (GB); A6II 3/423 (2006.01) Peter David Johnson, Oxfordshire C07D 413/02 (2006.01) (GB); Paul Damien Price, Wirral A 6LX 3/5.377 (2006.01) (GB); Olivier De Moor, C07F 9/653 (2006.01) Oxfordshire (GB); Gary Nugent, A 6LX 3/573 (2006.01) Surrey (GB); Richard Storer, Kent A6II 3/58 (2006.01) (GB); Richard Joseph Pye, A6IP 2L/00 (2006.01) Oxfordshire (GB); Colin Richard (52) U.S. Cl...... 514/80; 549/469; 514/469: 548/511: Dorgan, Oxfordshire (GB) 514/415: 548/178: 514/367: 548/310.7: 514/394; 548/221; 514/375; 54.4/137; 514/233.8: 548/113: (21) Appl. No.: 12/600,242 514/171 (22) PCT Filed: Aug. 1, 2008 (57) ABSTRACT (86). PCT No.: PCT/GB2O08/050648 Combinations comprising (or consisting essentially of) one or more compounds of formula (1) with one or more ancillary S371 (c)(1), agents, to processes for preparing the combinations, and to (2), (4) Date: Nov.30, 2010 various therapeutic uses of the combinations. Also provided are pharmaceutical compositions containing the combina (30) Foreign Application Priority Data tions as well as a method of treatment of Duchenne , Becker muscular dystrophy or cachexia using the Aug. 3, 2007 (GB) ...... 0715088.1 combinations. Apr. 21, 2008 (GB) ...... 0807216.7

Publication Classification (1) (51) Int. Cl. A6 IK3I/675 (2006.01) C07D 307/79 (2006.01) A6 IK3I/343 (2006.01) C07D 209/08 (2006.01) Patent Application Publication Aug. 11, 2011 Sheet 1 of 3 US 2011/0195932 A1

Figure 1 Dose dependent luciferase induction

9000 a CPD A A CPD B BOOO

7000 8 R900 5000 4000 s 3OOO 2000 g 1000 7.5 -7.0 -6.5 -6.0 -5.5 -5.0 log Crnpd CPD A CPD B EC50 79756-007 9,5328-007 CPD A - 5-amino-2-(5,6-dimethylbenzodoxazol-2-yl)phenol CPD B-2-(4-(diethylamino)phenyl)-6-methyl-2H-benzod1,2,3triazol-5- amine

Figure 2 An example of TA muscle sections stained with antibody specific for mouse utrophin

MCX- CPD-A 10mg/kg Patent Application Publication Aug. 11, 2011 Sheet 2 of 3 US 2011/0195932 A1

Figure 3 Mice exposed to CPD-A (V2 and V3) showed increased levels of utrophin expression compared to control:

Anti-UTRN

Anti-O-actinin

PD: dis

Patent Application Publication Aug. 11, 2011 Sheet 3 of 3 US 2011/0195932 A1

Figure 4Synergy with prednisone in fatigue reduction test

25O %

200 V

2 1OO : .

Sec. MDX Exer MDX CMPD1 PDN CMPD1+PDN

Resistance to treadmill running, expressed as maximal distance run (in meters) at TO, after 4 (T4) and 5 weeks (T5). Each bar is the meant S.E.M. from 3-7 animals. US 2011/0195932 A1 Aug. 11, 2011

DRUG COMBINATIONS FOR THE cological therapy. Gene- and cell-based therapies offer the TREATMENT OF DUCHENNE MUSCULAR fundamental advantage of obviating the need to separately DYSTROPHY correct secondary defects/pathology (for example, contrac tures), especially if initiated early in the course of the disease. TECHNICAL FIELD Unfortunately, these approaches face a number of technical 0001. This invention relates to combinations comprising hurdles. Immunological responses against Viral vectors, myo (or consisting essentially of) one or more compounds of the blasts and newly synthesized dystrophin have been reported, formula (1) as defined herein with one or more ancillary in addition to toxicity, lack of stable expression and difficulty agents, to processes for preparing the combinations, and to in delivery. various therapeutic uses of the combinations. Also provided 0006 Pharmacological approaches for the treatment of are pharmaceutical compositions containing the combina muscular dystrophy differ from gene- and cell-based tions as well as a method of treatment of Duchenne muscular approaches in not being designed to deliver either the missing dystrophy, Becker muscular dystrophy or cachexia using the gene and/or protein. In general, the pharmacological strate combinations. gies use drugs/molecules in an attempt to improve the phe notype by means such as decreasing inflammation, improving BACKGROUND OF THE INVENTION calcium homeostasis and increasing muscle progenitor pro 0002 Duchenne muscular dystrophy (DMD) is a com liferation or commitment. These strategies offer the advan mon, genetic neuromuscular disease associated with the pro tage that they are easy to deliver systemically and can circum gressive deterioration of muscle function, first described over vent many of the immunological and/or toxicity issues that 150 years ago by the French neurologist, Duchenne de Bou are related to vectors and cell-based therapies. Although logne, after whom the disease is named. DMD has been investigations with corticosteroids and sodium cromoglycate, characterized as an X-linked recessive disorder that affects 1 to reduce inflammation, dantrolene to maintain calcium in 3,500 males caused by mutations in the dystrophin gene. homeostasis and clenbuterol to increase muscle strength, The gene is the largest in the human genome, encompassing have produced promising results none of these potential 2.6 million base pairs of DNA and containing 79 exons. therapies alone has yet been shown to be effective in treating Approximately 60% of dystrophin mutations are large inser DMD. tion or deletions that lead to frameshift errors downstream, 0007 An alternative pharmacological approach is upregu whereas approximately 40% are point mutations or small lation therapy. Upregulation therapy is based on increasing frameshift rearrangements. The vast majority of DMD the expression of alternative genes to replace a defective gene patients lack the dystrophin protein. Becker muscular dystro and is particularly beneficial when an immune response is phy is a much milder form of DMD caused by reduction in the mounted against a previously absent protein. Upregulation of amount, or alteration in the size, of the dystrophin protein. utrophin, an autosomal paralogue of dystrophin has been The high incidence of DMD (1 in 10,000 sperm or eggs) proposed as a potential therapy for DMD (Perkins & Davies, means that genetic screening will never eliminate the disease, Neuromuscul Disord, S1: S78-S89 (2002), Khurana & so an effective therapy is highly desirable. Davies, Nat Rev Drug Discov 2:379-390 (2003)). When utro 0003) A number of natural and engineered animal models phin is overexpressed in transgenic mdX mice it localizes to of DMD exist, and provide a mainstay for preclinical studies the sarcolemma of muscle cells and restores the components (Allamand, V. & Campbell, K. P. Animal models for muscular of the dystrophin-associated protein complex (DAPC), which dystrophy: valuable tools for the development of therapies. prevents the dystrophic development and in turn leads to Hum. Mol. Genet. 9, 2459-2467 (2000).) Although the functional improvement of skeletal muscle. Adenoviral deliv mouse, cat and dog models all have mutations in the DMD ery of utrophin in the dog has been shown to prevent pathol gene and exhibit a biochemical dystrophinopathy similar to ogy. Commencement of increased utrophin expression that seen in humans, they show Surprising and considerable shortly after birth in the mouse model can be effective and no variation interms of their phenotype. Like humans, the canine toxicity is observed when utrophin is ubiquitously expressed, (Golden retriever muscular dystrophy and German short which is promising for the translation of this therapy to haired pointer) models have a severe phenotype; these dogs humans. Upregulation of endogenous utrophin to sufficient typically die of cardiac failure. Dogs offer the best phenocopy levels to decrease pathology might be achieved by the deliv for human disease, and are considered a high benchmark for ery of small diffusible compounds. preclinical Studies. Unfortunately, breeding these animals is expensive and difficult, and the clinical time course can be Ancillary Agents variable among litters. 0004. The mdx mouse is the most widely used model due 0008. A wide variety of ancillary agents find application in to availability, short gestation time, time to mature and rela the combinations of the invention, as described in detail tively low cost (Bulfield, G., Siller, W. G., Wight, P. A. & below. Moore, K. J. X chromosome-linked muscular dystrophy (mdx) in the mouse. Proc. Nail Acad. Sci. USA 81,1189-1192 SUMMARY OF THE INVENTION (1984)). 0005 Since the discovery of the DMD gene about 20 years 0009 We have now found a group of compounds which ago, varying degrees of success in the treatment of DMD have upregulate endogenous utrophin in predictive screens and, been achieved in preclinical animal studies, some of which thus, may be useful in the treatment of DMD. are being followed up in humans. Present therapeutic strate 0010. According to the invention, we provide a combina gies can be broadly divided into three groups: first, gene tion comprising (or consisting essentially of) an ancillary therapy approaches; second, cell therapy; and last, pharma agent and a compound of Formula (I) US 2011/0195932 A1 Aug. 11, 2011

in represents an integer from 0 to 2, in addition, when an adjacent pair of A-A each represent CR, then the A (I) adjacent carbon atoms, together with their substituents may form a ring B. 2-R when X is CRR, R and R7, together with the carbon atom A3.aCO Y to which they are attached may form a ring C, when one of A-A is CR, and R represents COR, R is preferably alkoxy or NRoR, or a pharmaceutically acceptable salt thereof (optionally for in which the therapeutic and/or prophylactic treatment of Duchenne A. A. As and A, which may be the same or different, muscular dystrophy, Becker muscular dystrophy or represent N or CR, cachexia). 0012 Compounds of formula I may exist in tautomeric, X is a divalent group selected from O. S(O), C=W, NR, enantiomeric and diastereomeric forms, all of which are NC(=O)Rs and CRR, included within the scope of the invention. All of the com pounds of formula may be made by conventional methods. W is O, S, NR, Methods of making heteroaromatic ring systems are well known in the art. In particular, methods of synthesis are Y is N or CRs. discussed in Comprehensive Heterocyclic Chemistry, Vol. 1 (Eds.: A R Katritzky, CW Rees), Pergamon Press, Oxford, 00.11 one of R. R. R. R. R. and NRo represents-L-R, 1984 and Comprehensive Heterocyclic Chemistry II: A in which L is a single bond or a linker group, Review of the Literature 1982-1995 The Structure, Reac additionally, R-R, which may be the same or different, tions, Synthesis, and Uses of Heterocyclic Compounds, Alan independently represent hydrogen or a substituent and Rio R. Katritzky (Editor), Charles W. Rees (Editor), E. F. V. represents hydrogen, hydroxyl, alkyl optionally substituted Scriven (Editor), Pergamon Pr, June 1996. Other general by aryl, alkoxy optionally substituted by aryl, aryl, CN, resources which would aid synthesis of the compounds of optionally Substituted alkoxy, optionally Substituted aryloxy, interest include March's Advanced Organic Chemistry: Reactions, Mechanisms, and Structure, Wiley-Interscience: optionally Substitute alkanoyl, optionally Substituted aroyl, 5th edition (Jan. 15, 2001). Of particular relevance are the NO, NRoR, in which Ro and R, which may be the same synthetic methods discussed in WO 2006/044503. Some gen or different, represent hydrogen, optionally substituted alkyl eral methods of synthesis are as follows. or optionally Substituted aryl; additionally, one of Ro and R 0013 Benzoxazoles of formula I or pharmaceutically may represent optionally substituted alkanoyl or optionally acceptable salts thereof may be prepared from compounds of substituted aroyl, formula II.

Scheme 1 N O R1 R9 2N.X when R = CO2H I vi.

Reaction conditions: i. RCOH (or RCOCl), PPA, heat; or RCOCl, dioxane, uwave, then NaOH ii. R'COCl, pryidine, rt iii. TsOH, xylenes iv, RCOH, HATU, pyridine, DMF v. PPA, heat vi. HATU, DMF, PrNet, alkylNH2, rt US 2011/0195932 A1 Aug. 11, 2011

0014 Formation of the benzoxazole I can be carried out in 0018. Another illustrative example of formation of a com a variety of ways, as illustrated above. pound of formula I is shown by steps iv and V. First the amine 0015 For example, reaction of the compound of formula is coupled to an acid using a peptide coupling reagent. Avail able coupling reagents are well known to those skilled in the II with an acyl derivative, such as the acid or the acid chloride, art, and include HBTU, TBTU and HATU. Amide formation and heating in an appropriate solvent and an appropriate in the presence of an appropriate coupling reagent occurs, for temperature in the presence of an acid catalyst, for example example, in DMF in the presence of a nucleophilic catalyst polyphosphoric acid. This is illustrated above as step (i). Such as pyridine. 0016. The reaction may be carried out in an aprotic sol 0019. When R'—COH, this acid may be coupled with an vent, preferably a polar, aprotic solvent, for example tetrahy amine as shown by step (vi). Suitable coupling conditions drofuran, and a temperature of from -10° C. to +150° C. include use of HATU in DMF in the presence of PrNEt, Generally the reaction may be carried on at the reflux tem R''NH at room temperature. perature of the solvent at normal pressure. 0020 Compounds in which the six membered ring is sub 0017 Alternatively, the compound of formula II may first stituted with an amide derivative are of particular interest. be reacted with an excess of an acyl derivative RCOX (where These may be produced from an intermediate amine deriva X is for example Cl), such that acylation takes place on both tive III.

Scheme 2 r OH A-4 NH2 i HN

X-M R9 - ii -- O N XA R2 y - III R17 OH VI Ill. He A-4 NH2 ON W. vii. IV

O N O N \-N Rl5-N R141 V H VIII O VII IX Reaction conditions: i. As for (i); Scheme 1 ii. ROOCl, pyridine (or NEt, DCM); or RCOH, HATU, pyridine, DMF iii. As for (i); Scheme 1 iv. SnCl2, EtOH, heat; or PdC, H2, IMS; or Fe, NH4C1, DMS, water, heat v. RNCO, DCM, rt vi. NaBH(OAc), RCHO, DCE,rt vii. RSOCl, pyridine, DCM, rt oxygen and nitrogen. This can be brought about by, for 0021 Intermediate amine III may be synthesised either by using the method outlined in Scheme 1, step (i) wherein example, reaction in pyridine at room temperature (step ii). R" NH, or alternatively, in a two step process as defined by Ring closure to form the compound of formula II can then steps (iii) and (iv) of scheme 2. Nitro substituted benzoxazole occur in a Subsequent ring closure step in which, for example, derivativeV is produced from nitro substituted phenyl deriva the doubly acylated product is heated in Xylenes in the pres tive IV, also in a method analogous to that illustrated by ence of an acid catalyst Such as a Sulphonic acid (step iii). scheme 1, step 1, and then the nitro-benzoxazole derivative V US 2011/0195932 A1 Aug. 11, 2011 is reduced in a Subsequent step to give intermediate amine III. manipulated in an analogous manner to the primary amine in The skilled person is well aware of suitable methods to reduce scheme 2 steps (ii), (v), (vi) and (vii). a nitro group to give anamine. Selective methods for reducing NO, to NH include Sn/HCl, or H/Pd/C in a suitable solvent, e.g. ethanol at a temperature of from 0 to 80°C. or heating in Scheme 4 the presence of iron, NHCl in industrial methylated spirits/ NH2 Water. 1N i 0022 Intermediate amine III can then be coupled as O.N- --> required. 2 NH2 0023 Amide derivatives of formula VI can be produced by coupling amine III with an acyl derivative. This can be XIII achieved by, for example, reaction of an appropriate acid chloride in either pyridine, or in CHCl (step ii). N 9 II 0024 Sulfonamide derivatives VII can be produced by O.N- A R -> reaction of amine III with an appropriate Sulfonyl chloride in, 2N. for example, CHCl in the presence of pyridine at room XII temperature. 0025 Amine derivatives VIII can be produced by use of an N III appropriate reductive amination strategy. Methods of reduc H.N- A R-> tive amination are well known in the art. They include, for 2NN example, reaction of the amine with an appropriate aldehyde O and sodium triacetoxyborohydride in 1,2-dichloroethane. R17 ( N NH 0026. Urea derivatives of formula IX can be produced, for example, by reaction of amine III with the appropriate isocy N - X- R9 anate, for example, at room temperature in CH2Cl2. 2 N 0027 Bertzothiazoles of formula X or pharmaceutically X NHR iv. w. acceptable salts thereof may be prepared from compounds of Hip- He formula XL ON NO ON NO XIV Scheme 3 R4 4 M C NHR' N --- X- R9 ON NH2 ON NH2 ON N XI XV R4 R4 A N O N iii. 9 ON N R9 ON N l X-R - it -R R17 N N HN N H Reaction conditions: i. RCOH, PPA, heat; or RCOCl, pyridine; then PPA, heat ii. SnCl2. IMS, heat; or PdC, H2, IMS iii. R''COCl, pyridine etc (as per Scheme 1) R17 N O)-r-OX-rN HN N iv. R'NH2, DMSO, base, heat Reaction conditions: v. Sodium dithionite, THF/water; also see (ii) i. RCOCl, pyridine, rt ii. Na2S, S8, DMS, heat iii. Fe, NH4C1, IMS, heat 0030 Benzimidazoles of formula XII can be produced iv, RCOCl, pyridine (or NEt, DCM); or RCOH, HATU, pyridine, DMF according to scheme 4. Reaction of a diaminophenyl deriva tive of formula XIII with an acyl derivative, such as an acid or 0028. The compounds of formula XI can be converted to an acid chloride in an appropriate solvent and at an appropri the corresponding amide by, for example, reaction with the ate temperature in the presence of an acid catalyst, for appropriate acid chloride in pyridine (step (i)), or by using an example polyphosphoric acid, produces a benzimidazole appropriate peptide coupling reagent. Such methods are well derivative of formula XII. This is illustrated above as step (i). known to the person skilled in the art as discussed herein The nitro group may then be reduced and manipulated to above. produce other functionality as discussed hereinabove. 0029. The amide can then be converted to the nitro-ben 0031. Alternatively, benzimidazoles may be produced by Zothiazole of formula XII in a one-pot procedure involving reacting a di-nitro compound of formula XIV, wherein X reaction with NaS, Ss at elevated temperature in industrial represents a leaving group, preferably a halogen Such as chlo methylated spirit. Nitro derivative XII can be reduced as rine or fluorine, with an amine, for example, in DMSO at discussed previously and the resulting primary amine elevated temperature in the presence of a base. Subsequent US 2011/0195932 A1 Aug. 11, 2011

selective reduction of one nitro group using sodium dithionite 0034. For oxazoles of formula XXIII in which X—Br, a in THF/water can then take place to give a diamine of formula Suzuki coupling reaction can then be used to give further XV. Ring closure to formabenzimidazoles, and manipulation derivatives. An example of appropriate conditions are RB of the nitro group can then proceed as illustrated and dis (OH), Pd(PPh), KCO, dioxane?water, pwave, in which a cussed previously. benzoxazole of formula XIX results. The person skilled in the

Scheme 5 Al OH Al OH A. O Al O n n n n Her1. -"11. - || X-R9 st-III X-R9 X 21 NO X 2 NH2 X 2NN 2 HN 2NN XX XXI iv. Br w |

vi. A. A. n O n O Y = C, X=NO X-R X-R RI 2NN RI 2NN XIX XVIII

s O s O S vii. X-Cl X 21 N X 2NN

XXII XXIII A. OH A. 21 N. ix 21 N. O

A| 2 -n - A|| 2 X-R NA4 NH2 NA4 N XVII XVI Reaction conditions: i. Na2S hydrate, MeOH, NH4C1, water; or Na2SO4. EtOH; or SnCl2, EtOH ii. As for (i), Scheme 1; or R9COCl, pyridine; then PPA, heat iii. SnCl2, EtOH, heat iv. RB(OH), Pd(PPh3)4, K2CO3, dioxane?water, uwave v. ROOCl, pyridine, rt vi. EtOC(S)SK, pyridine, heat vii. SOC12; or POCl3 viii. RB(OH)2, Pd(PPh3)4, K2CO3, solvent ix. PPA, RCOH heat

0032 Benzoxazoles of formula XVI can be made by art is familiar with Suzuki coupling reactions and could easily methods analogous to those discussed previously. For manipulate the conditions to produce a wide variety of com example the method illustrated above (ix) involves heating a pounds. compound of formula XVII in an appropriate solvent in the 0035. For oxazoles produced by step (ii) in which presence of acid catalyst and an appropriate acyl derivative eg X=NO, the nitro group can be reduced to the corresponding a carboxylic acid. amine, using any of the methods well known to the person skilled in the art discussed hereinabove. The amine may then 0033 Benzoxazoles of formula XVIII and XIX can be be manipulated using, for example, any of the methods dis synthesised from the appropriate nitro compound of formula cussed in Scheme 2 above, to give, for example, a compound XX. Reduction of the nitro compound XX gives the corre of formula XVIII. sponding amino alcohol XXI (for example using Sn/HCl, or 0036 Alternatively, benzoxazoles of formula XVIII can any of the other appropriate methods well known to the per be made, also from a compound of formula XX, via thiocar son skilled in the art). BenZoxazole formation via reaction of bamate XXII, which is produced by heating a compound of the amino alcohol with an appropriate acyl derivative can then formula XX with EtOC(S)SK in pyridine. The compound of beachieved using any of the methods disclosed hereinabove. formula XXII can be converted to the chloride of formula US 2011/0195932 A1 Aug. 11, 2011

XXIII for example by use of well known reagents such as made by reaction of the appropriate optically active starting SOCl or POCl. A Suzuki coupling using, for example, con materials under conditions which will not cause racemisa ditions illustrated by step viii above gives a benzoxazole of tion. formula XVIII. 0043. Substituents that alkyl may represent include 0037. In the above processes it may be necessary for any methyl, ethyl, butyl, eg sec butyl. Halogen may represent F. functional groups, e.g. hydroxy or amino groups, present in Cl, Brand I, especially C1. 0044) Examples of substituents that R in the compound of the starting materials to be protected, thus it may be necessary formula I may represent include alkyl, alkoxy or aryl, each to remove one or more protective groups to generate the optionally substituted by one or more, preferably one to three compound of formula I. substituents, R, which may be the same or different. 0038 Suitable protecting groups and methods for their 0045. In addition, when L is single bond, R may represent removal are, for example, those described in “Protective thioalkyl optionally substituted by alkyl or optionally substi Groups in Organic Synthesis” by T. Greene and P. G. M. tuted aryl, thioaryl, in which the aryl is optionally substituted, Wutts, John Wiley and Sons Inc., 1991. Hydroxy groups may, optionally substituted aryl, hydroxyl, NO, CN, NRoR, for example, be protected by arylmethyl groups such as phe halogen, SOR, NRSOR, C(=W)R OC(=W) nylmethyl, diphenylmethyl or triphenylmethyl; acyl groups NRR, NRC(=W)R, P(=O)CROR. R. R. R. Such as acetyl, trichloroacetyl or trifluoroacetyl, or as tetrahy R. R. Ris, R. R.17, Rao and Ra, which may be the same dropyranyl derivatives. Suitable amino protecting groups or which may be the same or different, represent hydrogen, include arylmethyl groups such as benzyl. (R.S)-O-phenyl alkyl optionally substituted by optionally substituted aryl, ethyl, diphenylmethyl or triphenylmethyl, and acyl groups optionally substituted aryl, such as acetyl, trichloroacetyl or trifluoroacetyl. Conven 0046 in addition, tional methods of deprotection may be used including hydro 0047 NRoR together with the nitrogen to which they genolysis, acid or base hydrolysis, or photolysis. Arylmethyl are attached may form a ring, groups may, for example, be removed by hydrogenolysis in 0048 R may have the same meaning as NRoR, the presence of a metal catalyst e.g. palladium on charcoal. 0049 when R, represents NRR, that Ro and R. Tetrahydropyranyl groups may be cleaved by hydrolysis which may be the same or different, may represent under acidic conditions. Acyl groups may be removed by hydrogen, COalkyl and CO optionally substituted aryl, hydrolysis with a base Such as Sodium hydroxide or potas 0050 R and R7, which may be the same or different, sium carbonate, or a group such as trichloroacetyl may be may each represent 0051 alkyl substituted by one or more of halogen, removed by reduction with, for example, Zinc and acetic acid. alkoxy optionally substituted aryl or optionally substi 0039. The compounds of formula I, and salts thereof, may tuted aryl, be isolated from their reaction mixtures using conventional techniques. 0.052 optionally substituted aryloxy, 0040 Salts of the compounds of formula I may be formed 0053 aryl or NRoR, by reacting the free acid, or a salt thereof, or the free base, or 0054 and when R or R, represents NRR, one of a salt orderivative thereof, with one or more equivalents of the Ro and R, may additionally represent CO alkyl appropriate base or acid. The reaction may be carried out in a optionally substituted or COaryl optionally substituted, solvent or medium in which the salt is insoluble or in a solvent 0.055 and in addition to the definitions shared with R7, in which the salt is soluble, e.g. ethanol, tetrahydrofuran or R may represent hydroxy. diethyl ether, which may be removed in vacuo, or by freeze 0056. Examples of substituents that RandR, which may drying. The reaction may also be a metathetical process or it be the same or different, may represent include: may be carried out on an ion exchange resin. alkyl optionally Substituted by one or more halogen, alkoxy 0041) Pharmaceutically acceptable salts of the com or optionally Substituted aryl, thioaryl or aryloxyalkoxy pounds of formula I include alkali metal salts, e.g. sodium and optionally substituted by optionally by alkyl or optionally potassium salts; alkaline earth metal salts, e.g. calcium and substituted arylhydroxyl, OC(=W)NRR, optionally magnesium salts; salts of the Group 111 elements, e.g. alu substituted aryl,thioalkyl optionally substituted by alkyl or minium salts; and ammonium salts. Salts with Suitable optionally substituted arylthioaryl, in which the aryl is organic bases, for example, salts with hydroxylamine; lower optionally substituted, NO, CNNRR, halogen, SOR, alkylamines, e.g. methylamine or ethylamine, with Substi NRSOR, C(=W)R. NRC(=W)R7, Rio, R. R. tuted lower alkylamines, e.g. hydroxy Substituted alky R. R. R. R. and R7, which may be the same or differ lamines; or with monocyclic nitrogen heterocyclic com ent, represent hydrogen, alkyl optionally Substituted by pounds, e.g. piperidine or morpholine; and salts with amino optionally substituted aryl, optionally substituted aryl, acids, e.g. with arginine, lysine etc, or an N-alkyl derivative 0057 in addition, thereof, or with an aminosugar, e.g. N-methyl-D-glucamine 0.058 NRoR together with the nitrogen to which they or glucosamine. The non-toxic physiologically acceptable are attached may form a ring, salts are preferred, although other salts are also useful, e.g. in 0059 R may have the same meaning as NRR, isolating or purifying the product. 0060 when R, represents NRR, that Ro and R. 0.042 Diastereoisomers may be separated using conven which may be the same of different, may represent tional techniques, e.g. chromatography or fractional crystal hydrogen, COalkyl and CO optionally substituted aryl, lisation. The various optical isomers may be isolated by sepa 0061 R may represent hydroxy, alkoxy, or NRoR, ration of a racemic or other mixture of the compounds using 0062) R, may represent alkyl substituted by one or conventional, e.g. fractional crystallisation or HPLC, tech more of halogen, alkoxy, optionally Substituted aryl or niques. Alternatively the desired optical isomers may be NRoR US 2011/0195932 A1 Aug. 11, 2011

0063 and when R, represents NRR, that NRR 0080 We particularly prefer compounds in which at least may represent hydrogen, COalkyl and CO optionally one R represents NRC(=W)R7, more particularly the substituted aryl. group NRCOR7. 0064. When L represents a linker group, examples of I0081. We also prefer compounds in which at least one R linker groups that L. may represent include: represents CONRoR. 0065 O, S, (CO)NRs. I0082 For one group of particularly preferred compounds 0.066 alkylene, alkenylene, alkynylene, each of which at least one R represents an amide group NHCOR, wherein may be optionally interrupted by one or more of O, S, R7 is selected from: NRs or one or more C-C single, double or triple 0083) alkyl C-C, bonds, 0084 alkyl C-C substituted by phenyl 0067 a - N N single or double bond, 0085 alkyl C-C substituted by alkoxy C-C, I0086 haloalkyl C-C, 0068 Rs represents hydrogen, alkyl, COR. I0087 perfluoroalkyl C-C, 0069. When L is (CO)NR's n may represent 0, 1 or 2, 0088 phenyl optionally substituted by one or more of when n is 1 or 2, Rs preferably represents hydrogen. halogen, alkyl C-C alkoxy C-C amino, (alkyl 0070 Although the scope for variation of R. R. R. R. C-C)amino, di(alkyl C-C) amino or phenyl, and Rs is large, preferably R. Rs. R. R., and Rs represent I0089 CH:CH phenyl, hydrogen, alkyl or optionally substituted aryl. 0090 naphthyl, pyridinyl, thiophenyl and furanyl. (0071 Preferably Y represents N and X represents O, S or 0091. We prefer compounds in which one or both of R NR. That is preferably the compound according to formula I and R are other than —COOH. is a benzoxazole, a benzthiazole or a benzimidazole. 0092. For another group of particularly preferred com 0072 Although any one of R. R. R. or Rs may represent pounds at least one R represents a group NRCONRoR, -L-R, , in preferred compounds Ro represents -L-Rs. then in which Ro and R, which may be the same or differ 0073 Alkyl may represent any alkyl chain. Alkyl includes ent, are selected from optionally Substituted aryl, alkyl and straight and branched, Saturated and unsaturated alkyl, as COaryl optionally substituted. A particularly preferred group well as cyclic alkyl, such as cyclopropyl, cyclobutyl, cyclo which at least one of R may represent is NHCONFIRs and pentyl, cyclohexyl and cycloheptyl. However, preferably, Rs is selected from phenyl, alkyl C to C and COphenyl when any of the Substituents represents alkyl, alkyl is satu optionally substituted by one or more halogen. rated, linear or branched and has from 1 to 10 carbon atoms, 0093. For another group of particularly preferred com preferably from 1 to 8 carbonatoms and more preferably from pounds at least one R represents alkyl C1 to C6, optionally 1 to 6 carbon atoms. When any of the substituents represents substituted by phenyl or a 5 or 6-membered saturated or alkyl, a particularly preferred group is cycloalkyl, for unsaturated heterocycle containing one to two heteroatoms example cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl selected from N, S and O. and cycloheptyl. 0094 For another group of particularly preferred com 0074 Aryl may represent any aromatic system. Prefer pounds at least one R represents COR and R is alkoxy ably, in the compounds of formula I, aryl is an aromatic C-C, amino, (alkyl C-C)amino or di(alkyl C-C) amino. hydrocarbon or a 5 to 10 membered aromatic heterocycle 0.095 For another group of particularly preferred com containing 1 to 4 hetero atoms selected from an oxygenatom, pounds at least one R represents: a Sulphur atom and a nitrogen atom as a ring constituent 0.096 NO, besides carbon. We prefer heterocycles which contain one or 0097 halogen, two heteroatoms. Aromatic heterocycles that may be men 0.098 amino or (alkyl C-C)amino or di(alkyl C-C) tioned include furan, thiophene, pyrrole, pyridine. amino in which the alkyl C to C is optionally Substi 0075 Particularly preferably, when aryl is an aromatic tuted by phenyl or a 5 or 6 membered saturated or unsat hydrocarbon, aryl represents a 6 to 10 membered monocyclic urated heterocycle, or bicyclic system, for example phenyl or naphthalene. 0099 NHSO-alkyl C-C NHSOphenyl, 0076 Saturated and unsaturated heterocycles that may be 0100 SO, alkyl C-C, mentioned include those containing 4 to 7 ring atoms, pref 0101 phenyl optionally substituted by C to Calkoxy erably 5 or 6 ring atoms, preferably containing one to two C1-C6, heteroatoms selected from N, S and O. Heterocycles that may 0102 a 5-10 membered, saturated or unsaturated, be mentioned include pyrrolidine, piperidine, tetrahydrofu mono- or bi-cyclic heterocycle containing from 1-3 het ran, piperazine and morpholine. N-containing heterocycles eroatoms selected from N, S and O. are particularly preferred, eg when NRoR forms a hetero 0103) There is also wide scope for variation of the group cyclic ring. R. Preferably R represents aryland is optionally substituted 0077. As detailed above, when an adjacent pair of Al-A by one to three substituents, R, which may be the same or each represent CR, the adjacent carbon atoms, together with different. Particularly preferably, R is a 5-10 membered aro their Substituents may form a ring ring B. Also, when X is matic mono- or bi-cyclic system, especially a hydrocarbon CRR, R and R-7, together with the carbon to which they are 5-10 membered aromatic mono- or bi-cyclic system, for attached may form a ring C. Preferably ring Band/or ring C is example benzene or naphthalene. Alternatively, the 5-10 a saturated or unsaturated 3 to 10 membered carbocylic or membered aromatic mono- or bi-cyclic system, may be a heterocyclic ring. heterocyclic system containing up to three heteroatoms 0078 Particularly preferably ring B is benzene ring. selected from N, O and S, for example a thiophene, furan, 0079 Particularly preferably ring C is a 3-10 membered pyridine or pyrrole. saturated or unsaturated carbocylic ring. 0104 Preferably the substituent(s) R is/are selected from: US 2011/0195932 A1 Aug. 11, 2011

0105 alkyl C-C optionally substituted by thiophenyl may represent optionally Substituted alkanoyl or optionally or phenoxy, each optionally Substituted by halogen, Substituted aroyl, in represents an integer from 0 to 2. 0106 alkoxy C-C, 0.130 R represents alkyl, alkoxy or aryl, each option 0107 phenyl, ally substituted by one to three substitutents, R, which (0.108 thioalkyl C-C, may be the same or different 0109 thiophenyl, optionally substituted by halogen, I0131 R and R, which may be the same or different, 10110 NO, represent: 0111 CN 0132) alkyl optionally substituted by one or more halo 0112 NRR, in which Ro and R, which may be the gen, alkoxy or optionally Substituted aryl, thioaryl or same or different represent hydrogen, alkyl C-C, or aryloxy, together with the nitrogen to which they are attached 0.133 alkoxy optionally substituted by optionally by form a 5 to 7 membered ring which may contain one or alkyl or optionally substituted aryl, more additional heteroatoms selected from N, O and S, 0134 hydroxyl, 0113 halogen 0135) OC(=W)NRR 0.136 optionally substituted aryl, I0114 SOR, in which R' represents a 5 to 7 mem 0.137 thioalkyl optionally substituted by alkyl or bered ring which may contain one or more additional optionally substituted aryl, heteroatoms selected from N, O and S 0.138 thioaryl, in which the aryl is optionally substi 0115 NHCOR, in which R, represents tuted, 0116 alkyl C-C optionally substituted by: 0139 NO, 0.117 phenyl or halogen, or 0140. CN, 0118 phenyl optionally substituted by alkoxy 0.141 NRoR, C-C carboxy or halogen, 0.142 halogen, 0119 or 0143 SOR, 0120 a 5 or 6 membered saturated or unsaturated heterocycle, 0144) NRSOR, I0121 phenyl or a 5 or 6 membered saturated or unsat urated heterocycle optionally Substituted by halogen, 0147 Rio, R. R. R. R. Rs. R and R17, which alkoxy C to C. carboxy or a group SONRoR, may be the same or different, represent hydrogen, alkyl 0122 Particularly preferably when R represents optionally substituted by optionally substituted aryl, NRR, NRoR represents N-pyrrole, N-piperidine, optionally substituted aryl, N'(C-C) alkyl N piperazine or N-morpholine. 0.148 in addition, 0123 Preferably the linker group L represents: 0.149 NRoR together with the nitrogen to which they 0.124 - NH.NH are attached may form a ring, 0.125 CH-CH 0150 R may have the same meaning as NRoR, (0.126 - NCOR in which R" represents phenyl or a 5 0151 when R, represents NRR, that NR1OR'' or 6 membered saturated or unsaturated heterocycle may represent hydrogen, COalkyl and CO optionally optionally substituted by halogen, alkoxy C1 to C6, substituted aryl, carboxy. 0152 R may represent hydroxy, alkoxy, or NRR, 0127 A-A may represent N or CR. Consequently, the 0.153 R, may represent alkyl substituted by one or benzoxazole six membered ring may contain 1, 2, 3 or 4 more of halogen, alkoxy, optionally Substituted aryl or nitrogenatoms. Embodiments of the invention exist in which NRoR, two of A-A represent nitrogen, one of A-A represents and when R, represents NRoR, that NRoR may repre nitrogen and in which all of A-A represents CR. sent hydrogen, COalkyl and CO optionally substituted aryl, 0128. In a particularly preferred group of compounds: and in addition, A. A. As and A, which may be the same or different, when an adjacent pair of A-A each represent CR, then the represent N or CR. adjacent carbon atoms, together with their substituents may X is a divalent group selected from O. S(O), C=W, NR, form a ring B. NC(=O)R and CRR, when X is CRR, R and R7, together with the carbon atom to which they are attached may form a ring C, or a pharma W is O, S, NR, ceutically acceptable salt thereof, optionally for the therapeu tic and/or prophylactic treatment of Duchenne muscular dys Y is N or CRs, trophy, Becker muscular dystrophy or cachexia. 0154 We also provide a method for the treatment or pro 0129 one of R. R. R. Rs. Ro and NRo represents-L-R, phylaxis of Duchenne muscular dystrophy, Becker muscular in which L is a single bond or a linker group, dystrophy or cachexia in a patient in need thereof, comprising additionally, R-R, which may be the same or different, independently represent hydrogen or a substituent and Ro administering to the patient an effective amount of a combi represents hydrogen, hydroxyl, alkyl optionally substituted nation of the invention. by aryl, alkoxy optionally substituted by aryl, aryl, CN, optionally Substituted alkoxy, optionally Substituted aryloxy, General Preferences and Definitions optionally Substitute alkanoyl, optionally Substituted aroyl, 0155 The combinations of the invention may produce a NO, NRoR, in which Ro and R, which may be the same therapeutically efficacious effect relative to the therapeutic or different, represent hydrogen, optionally substituted alkyl effect of the individual compounds when administered sepa or optionally Substituted aryl; additionally, one of Ro and R rately. US 2011/0195932 A1 Aug. 11, 2011

0156 The term “efficacious” includes advantageous which the two or more compounds/agents are associated. The effects such as additivity, synergism, reduced side effects, terms “combined and “combining in this context are to be reduced toxicity, increased time to disease progression, interpreted accordingly. increased time of Survival, sensitization or resensitization of 0163 The association of the two or more compounds/ one agent to another, or improved response rate. Advanta agents in a combination may be physical or non-physical. geously, an efficacious effect may allow for lower doses of Examples of physically associated combined compounds/ each or either component to be administered to a patient, agents include: thereby decreasing the toxicity of chemotherapy, whilst pro 0.164 compositions (e.g. unitary formulations) com ducing and/or maintaining the same therapeutic effect. prising the two or more compounds/agents in admixture (for example within the same unit dose); 0157 A“synergistic effect in the present context refers to 0.165 compositions comprising material in which the a therapeutic effect produced by the combination which is two or more compounds/agents are chemically/physico larger than the sum of the therapeutic effects of the compo chemically linked (for example by crosslinking, nents of the combination when presented individually. molecular agglomeration or binding to a common 0158. An “additive' effect in the present context refers to vehicle moiety); a therapeutic effect produced by the combination which is 0166 compositions comprising material in which the larger than the therapeutic effect of any of the components of two or more compounds/agents are chemically/physico the combination when presented individually. chemically co-packaged (for example, disposed on or 0159. A “pharmaceutical composition is a solid or liquid within lipid vesicles, particles (e.g. micro- or nanopar composition in a form, concentration and level of purity Suit ticles) or emulsion droplets): able for administration to a patient (e.g. a human or animal 0.167 pharmaceutical kits, pharmaceutical packs or patient) upon which administration it can elicit the desired patient packs in which the two or more compounds/ physiological changes. Pharmaceutical compositions are agents are co-packaged or co-presented (e.g. as part of typically sterile and/or non-pyrogenic. The term non-pyro an array of unit doses); 0168 Examples of non-physically associated combined genic as applied to the pharmaceutical compositions of the compounds/agents include invention defines compositions which do not elicit undesir 0.169 material (e.g. a non-unitary formulation) com able inflammatory responses when administered to a patient. prising at least one of the two or more compounds/agents (0160. As used herein, the terms mobilizing agent and together with instructions for the extemporaneous asso mobilization are terms of art referring to agents and treat ciation of the at least one compound/agent to form a ments which serve to promote the migration of CD34', stem, physical association of the two or more compounds/ progenitor and/or precursor cells from the marrow to the agents; peripheral blood (for a review, see e.g. Cottler-Fox et al. 0170 material (e.g. a non-unitary formulation) com (2003)Stem cell mobilization Hematology. 419-437). Current prising at least one of the two or more compounds/agents standard agents for mobilization Suitable for use according to together with instructions for combination therapy with the invention include G-CSF (FilgrastimTM, Amgen), GM the two or more compounds/agents; CSF (SargramostimTM, Berlex, Richmond, Calif.) and eryth 0171 material comprising at least one of the two or ropoietin (which has some mobilizing activity w.r.t. CD34" more compounds/agents together with instructions for cells). Alternative agents include stem cell factor (SCF) administration to a patient population in which the other (which is particularly effective when used in combination (s) of the two or more compounds/agents have been (or with G-CSF) and various derivatives of G-CSF (Peg are being) administered; filgrastimTM Amgen) and erythropoietin (Darbopoietin R, 0172 material comprising at least one of the two or Amgen). The latter agents benefit from extended half-lives more compounds/agents in an amount or in a form and so increase the temporal window available for collection. which is specifically adapted for use in combination AMD3100 (AnorMedTM, Vancouver, Canada), which is a with the other(s) of the two or more compounds/agents. reversible inhibitor of the binding of stromal derived factor 0173 As used herein, the term “combination therapy” is (SDF-1a) to its cognate receptor CXCR4, is currently in intended to define therapies which comprise the use of a clinical trials as a mobilizing agent. Other agents include combination of two or more compounds/agents (as defined docetaxel (see e.g. Prince et al. (2000) Bone Marrow Trans above). Thus, references to “combination therapy”, “combi plantation 26: 483-487). nations” and the use of compounds/agents “in combination' 0161 The term “upregulation of utrophin' as used herein in this application may refer to compounds/agents that are includes elevated expression or over-expression of utrophin, administered as part of the same overall treatment regimen. including gene amplification (i.e. multiple gene copies) and As such, the poSology of each of the two or more compounds/ increased expression by a transcriptional effect, and hyper agents may differ: each may be administered at the same time activity and activation of utrophin, including activation by or at different times. It will therefore be appreciated that the mutations. The term “utrophin upregulating agent' is to be compounds/agents of the combination may be administered interpreted accordingly. Thus, upregulation of utrophin cov sequentially (e.g. before or after) or simultaneously, either in ers increasing utrophin activity at the level of the encoding the same pharmaceutical formulation (i.e. together), or in DNA as well as the transcriptional, translational or post different pharmaceutical formulations (i.e. separately). translational level. Preferred compounds of formula (I) are Simultaneously in the same formulation is as a unitary for utrophin upregulators (as disclosed herein). mulation whereas simultaneously in different pharmaceutical 0162. As used herein, the term “combination', as applied formulations is non-unitary. The posologies of each of the two to two or more compounds and/or agents (also referred to or more compounds/agents in a combination therapy may herein as the components), is intended to define material in also differ with respect to the route of administration. US 2011/0195932 A1 Aug. 11, 2011

0.174 As used herein, the term “pharmaceutical kit' transgenic animals. The term “antibody' also includes multi defines an array of one or more unit doses of a pharmaceutical mericantibodies and higher-order complexes of proteins (e.g. composition together with dosing means (e.g. measuring heterodimericantibodies). device) and/or delivery means (e.g. inhaler or Syringe), optionally all contained within common outer packaging. In Ancillary Agents for Use According to the Invention pharmaceutical kits comprising a combination of two or more 0180. Any of a wide variety of ancillary agents may be compounds/agents, the individual compounds/agents may used in the combinations of the invention. Preferably, the unitary or non-unitary formulations. The unit dose(s) may be ancillary agents for use in the combinations of the invention contained within a blister pack. The pharmaceutical kit may as described herein are selected from the following classes: optionally further comprise instructions for use. 1. Antiinflammatory agents; 0.175. As used herein, the term “pharmaceutical pack” 2. Protease inhibitors; defines an array of one or more unit doses of a pharmaceutical 3. Myostatin antagonists; composition, optionally contained within common outer 4. Cytokines and mobilizing agents; packaging. In pharmaceutical packs comprising a combina tion of two or more compounds/agents, the individual com 5. Corticosteroids; pounds/agents may unitary or non-unitary formulations. The 0181 6. Anabolic steroids; unit dose(s) may be contained within a blister pack. The 7. TGF-B antagonists; pharmaceutical pack may optionally further comprise 8. Antioxidants and mitochondrial Supporting agents; instructions for use. 9. Dystrophin expression enhancing agents; 0176). As used herein, the term “patient pack” defines a 10. Gene replacement/repair agents; package, prescribed to a patient, which contains pharmaceu 11. Cell-based compositions: tical compositions for the whole course of treatment. Patient packs usually contain one or more blister pack(s). Patient 12. Creatine; packs have an advantage over traditional prescriptions, where 0182 13. anti-osteoporotic agents: a pharmacist divides a patient's Supply of a pharmaceutical 14. auxiliary utrophin upregulating agents; from a bulk Supply, in that the patient always has access to the 15. cQMP signalling modulators; and package insert contained in the patient pack, normally miss 16. a combination of two or more of the foregoing classes. ing in patient prescriptions. The inclusion of a package insert 0183. A reference to a particular ancillary agent herein is has been shown to improve patient compliance with the phy intended to include ionic, salt, Solvate, isomers, tautomers, sician's instructions. N-oxides, ester, prodrugs, isotopes and protected forms 0177. The combinations of the invention may produce a thereof (preferably the salts or tautomers or isomers or N-ox therapeutically efficacious effect relative to the therapeutic ides or solvates thereof, and more preferably, the salts or effect of the individual compounds/agents when adminis tered separately. tautomers or N-oxides or solvates thereof). 0.178 The term “ancillary agent” as used herein may 1. Antinflammatory Agents define a compound/agent which yields an efficacious combi nation (as herein defined) when combined with a compound 0.184 Muscles affected by DMD show signs of inflamma of the formula (1) as defined herein. The ancillary agent may tion, including an abundance of macrophages. Thus, a wide therefore act as an adjunct to the compound of the formula (1) range of antiinflammatory agents can be used in the treatment as defined herein, or may otherwise contribute to the efficacy of muscular dystrophies, as discussed below. of the combination (for example, by producing a synergistic or additive effect or improving the response rate, as herein 1.1 Beta2-Adrenergic Receptor Agonists defined). 0185. In one embodiment of the invention, the ancillary (0179. As used herein, the term “antibody” defines whole agent is a beta2-adrenergic receptor agonist (e.g. albuterol). antibodies (including polyclonal antibodies and monoclonal 0186. Definitions and technical background: The term antibodies (Mabs)). The term is also used herein to refer to beta2-adrenergic receptor agonist is used herein to define a antibody fragments, including F(ab), F(ab'), F(ab')2, Fv, Fcs class of drugs which act on the 32-adrenergic receptor, and single chain antibodies (and combinations thereof), thereby causing Smooth muscle relaxation resulting in dila which may be produced by recombinant DNA techniques or tion of bronchial passages, vasodilation in muscle and liver, by enzymatic or chemical cleavage of intact antibodies. The relaxation of uterine muscle and release of insulin. A pre term “antibody' is also used herein to cover bispecific or ferred beta2-adrenergic receptoragonist for use according to bifunctional antibodies which are synthetic hybrid antibodies the invention is albuterol, an immunosuppressant drug that is having two different heavy/light chain pairs and two different widely used in inhalant form for asthmatics. Albuterol is binding sites. Bispecific antibodies can be produced by a thought to slow disease progression by Suppressing the infil variety of methods including fusion of hybridomas or linking tration of macrophages and other immune cells that contrib of Fab' fragments. Also covered by the term “antibody' are ute to inflammatory tissue loss. Albuterol also appears to have chimaeric antibodies (antibodies having a human constant Some anabolic effects and promotes the growth of muscle antibody immunoglobulin domain coupled to one or more tissue. Albuterol may also suppress protein degradation (pos non-human variable antibody immunoglobulin domain, or sibly via calpain inhibition). fragments thereof). Such chimaeric antibodies therefore 1.2 nNOS Stimulators include “humanized antibodies. Also covered by the term 0187. The loss of dystrophin leads to breaks in the mem “antibody” are minibodies (see WO94/09817), single chain brane, and destabilizes neuronal nitric oxide synthase Fv-Fc fusions and human antibodies antibodies produced by (nNOS), a protein which normally generates nitric oxide US 2011/0195932 A1 Aug. 11, 2011

(NO). It is thought that at least part of the muscle degeneration adjusting the dose up to 10 mg/kg or treating as often as every observed in DMD patients may result from the reduced pro 4 weeks. Humira is marketed in both preloaded 0.8 ml duction of muscle membrane-associated neuronal nitric Syringes and also in preloaded pen devices, both injected oxide synthase. This reduction may lead to impaired regula Subcutaneously, typically by the patient at home. Etanercept tion of the vasoconstrictor response and eventual muscle can be administered at a dose of 25 mg (twice weekly) or 50 damage. mg (once weekly). 1.3 Nuclear Factor Kappa-B Inhibitors 1.5 Ciclosporin 0188 A preferred class of antiinflammatory agent suitable 0193 In one embodiment of the invention, the antinflam for use in the combinations of the invention are Nuclear matory agent is ciclosporin. Ciclosporin A, the main form of Factor Kappa-B (NF-kB) inhibitors. NE-kB is a major tran the drug, is a cyclic nonribosomal peptide of 11 amino acids Scription factor modulating the cellular immune, inflamma produced by the fungus Tolypocladium inflatum. Ciclosporin tory and proliferative responses. NF-kB functions inactivated is thought to bind to the cytosolic protein cyclophilin (immu macrophages to promote inflammation and muscle necrosis nophilin) of immunocompetent lymphocytes (especially and in skeletal muscle fibers to limit regeneration through the T-lymphocytes). This complex of ciclosporin and cyclophy inhibition of muscle progenitor cells. The activation of this lin inhibits calcineurin, which under normal circumstances is factor in DMD contributes to diseases pathology. Thus, NF responsible for activating the transcription of interleukin-2. It kB plays an important role in the progression of muscular also inhibits lymphokine production and interleukin release dystrophy and the IKK/NF-B signaling pathway is a potential and therefore leads to a reduced function of effector T-cells. It therapeutic target for the treatment of DMD. Inhibitors of does not affect cytostatic activity. It has also an effect on NF-kB (for example, IRFI 042, a vitamin E analogue) ame mitochondria, preventing the mitochondrial PT pore from liorate muscle function, decrease serum CKlevel and muscle opening, thus inhibiting cytochrome c release (a potent apo necrosis and enhance muscle regeneration. Furthermore, spe ptotic stimulation factor). Ciclosporin may be administered at cific inhibition of NF-kB/IKK-mediated signalling has simi a dose of 1-10 mg/kg/day. lar benefits. 2. Protease Inhibitors 1.4 TNF-C. Antagonists 0194 Proteins in skeletal muscle are degraded by at least (0189 TNFC. is one of the key cytokines that triggers and three different proteolytic pathways: (a) lysosomal proteases Sustains the inflammation response. In one embodiment of the (e.g. the cathepsins); (b) non-lysosomal Ca"-dependent pro invention, the ancillary agent is a TNF-C. antagonist (e.g. teases (e.g. calpain); and (c) non-lysosomal ATP-ubiquitin infliximab). dependent proteases (e.g. the multicatalytic protease complex (0190. Preferences and specific embodiments: Preferred or proteasome). Several lines of evidence have Suggested that TNF-C. antagonists for use according to the invention include enhanced activation of proteolytic degradation pathways infliximab (RemicadeTM), a chimeric monoclonal antibody underlies the pathogenesis of muscular dystrophy. Thus, pro comprising murine VK and VH domains and human constant tease inhibitors can be used in the treatment of muscular Fc domains. The drug blocks the action of TNFC. by binding dystrophies, as discussed below. to it and preventing it from signaling the receptors for TNFC. 0.195 Preferred protease inhibitors for use according to on the surface of cells. Another preferred TNF-C. antagonists the invention may specifically target one of the three degrad for use according to the invention is adalimumab (HumiraTM). tion pathways described above. Particularly preferred are Adalimumab is a fully human monoclonal antibody. Another protease inhibitors which target the non-lysosomal Ca"-de preferred TNF-C. antagonists for use according to the inven pendent pathway (calpain inhibitors) or the non-lysosomal tion is etanercept (EnbrelTM) Etanercept is a dimeric fusion ATP-ubiquitin-dependent pathway (proteasome inhibitors), protein comprising soluble human TNF receptor linked to an as described below: Fc portion of an IgG1. It is a large molecule that binds to and so blocks the action of TNFC. Etanercept mimics the inhibi 2.1 Calpain Inhibitors tory effects of naturally occurring soluble TNF receptors, but 0196. In one embodiment of the invention, the ancillary as a fusion protein it has a greatly extended half-life in the agent is a calpain inhibitor. bloodstream and therefore a more profound and long-lasting 0197) Definitions and technical background: The term inhibitory effect. Enbrel is marketed as a lyophylized powder “calpain inhibitor is used herein to define any agent capable in 25 mg vials which must be reconstituted with a diluent and of inhibiting the activity of calpain. Calpain is a ubiquitous then injected Subcutaneously, typically by the patient at calcium-dependent cysteine protease which cleaves many home. cytoskeletal and myelin proteins. Calpains belong to a family 0191 Another preferred TNF-C. antagonist for use accord of Ca" activated intracellular proteases whose activity is ing to the invention is pentoxifylline (TrentalTM), chemical accelerated when abnormal amounts of Ca" enter the cell by name 1-(5-oxohexyl)-3,7-dimethylxanthine. The usual dos virtue of increased membrane permeability as a result of age in controlled-release tablet form is one tablet (400 mg) Some traumatic or ischemic event and/or a genetic defect. three times a day with meals. Calpain is one of a relatively small family of cysteine pro 0.192 PoSology: Remicade is administered by intravenous teases, which are active in promoting programmed cell death, infusion, typically at 2-month intervals. The recommended or apoptosis. It has been implicated in the initiation of both dose is 3 mg/kg given as an intravenous infusion followed necrotic and apoptotic cell death. When calpain is abnormally with additional similar doses at 2 and 6 weeks after the first up regulated, the accelerated degradation process breaks infusion then every 8 weeks thereafter. For patients who have down cells and tissues faster than they can be restored, result an incomplete response, consideration may be given to ing in several serious neuromuscular and neurodegenerative US 2011/0195932 A1 Aug. 11, 2011

diseases. Calpain has been implicated in the accelerated tis Solvate, isomers, tautomers, N-oxides, ester, prodrugs, iso Sue breakdown associated with muscular dystrophies (includ topes and protected forms thereof (preferably the salts or ing DMD). The trigger which activates calpain is Ca' ions tautomers or isomers or N-oxides or solvates thereof, and leaking into cells, where the levels are generally very low. The more preferably, the salts or tautomers or N-oxides or solvates dystrophin gene is involved in maintaining membrane integ thereof), as described above. rity, and when it is mutated, the membrane is more permeable to calcium ions. Thus, the inhibition of calpain activity in the 0202 Preferences and specific embodiments: There are muscles of DMD patients can preserve muscle integrity and several classes of proteasome Inhibitors suitable for usein the prfevent or slow muscle deterioration. combinations of the invention, including peptide aldehydes 0198 Preferences and specific embodiments: Calpain (such as MG-132) and the dipeptidylboronic acid bortezimib inhibitors for use according to the invention preferably com (VelcadeTM; formerly known as PS-341) which is a more prise a calpain inhibiting moiety linked to (or associated with) specific inhibitor of the proteasome. Thus, preferred protea a carrier (which acts to facilitate targeting of the calpain some inhibitors for use in accordance with the invention inhibiting moiety to muscle tissue). The targeting moiety may include bortezimib (I(1R)-3-methyl-1-(2S)-1-oxo-3-phe be chemically linked to the calpain inhibiting moiety, or may nyl-2-(pyrazinylcarbonyl)aminopropylaminobutyl-bo be physically associated therewith (a liposome carrier). Pre ronic acid). Bortezimib is commercially available for ferred targeting moieties include carnitine or aminocarnitine. example from Millennium Pharmaceuticals Inc under the The calpain inhibiting moiety may be leupeptin. Particularly trade name Velcade, or may be prepared for example as preferred may be Ceptor's MyodurTM. Other such calpain described in PCT patent specification No. WO96/13266, or inhibitors are described in WO2005124563 (the contents of by processes analogous thereto. Bortezimib specifically which are incorporated herein by reference). Other suitable interacts with a key amino acid, namely threonine, within the calpain inhibitors are the O-ketocarbonyl calpain inhibitors catalytic site of the proteasome. Another preferred protea disclosed in WO 2004/078.908 (the contents of which are some inhibitor for use in the combinations of the invention is incorporated herein by reference). Of the calpain inhibitors the cell-permeable proteasomal inhibitor CBZ-leucyl-leucyl described in WO 2004/078908, preferred may be those which leucinal (MG-132) (as described in Bonuccelli et al. (2003) target both calpain and the proteasome. Am J Pathol. October; 163(4): 1663-1675, the content of 0199 The calpain inhibitors for use according to the which relating to this compound is incorporated herein by invention may be chimaeric compounds or combinations in reference). Other inhibitors include those structurally related which the calpain inhibiting moiety is associated (e.g. com to MG-132, including MG-115 (CBZ-leucyl-leucyl-norvali bined with, co-administered with or covalently linked) to a nal) and ALLN (N-acetyl-leucyl-leucyl-norleucinal) (as also ROS inhibitor. Such agents combine relief of oxidative stress described in Bonuccelli et al. (2003) Am J Pathol. October; with a reduction in calpain-mediated muscle tissue break 163(4): 1663-1675, the content of which relating to this com down. Suitable dual action calpain/ROS inhibitors are pound is incorporated herein by reference). described for example in WOO1/32654, WO2007/045761, 0203 Posology: The proteasome inhibitor (such as borte WO2005/056551 and WO 2002/40016 (the contents of which Zimib) can be administered in a dosage such as 100 to 200 are incorporated herein by reference). mg/m. These dosages may be administered for example 0200. Other suitable calpain inhibitors can be identified once, twice or more per course of treatment, which may be using commercially available assay kits (e.g. the calpain repeated for example every 7, 14, 21 or 28 days. MG-132 can activity kit based on a fluorogenic Substrate from Oncogene be administered at a dose of 10 ug/kg/day. Research Products, San Diego, Calif.). This assay measures the ability of calpain to digest the synthetic substrate Suc 3. Myostatin Antagonists LLVY-AMC: free AMC can be measured fluorometrically at 0204 Definitions and technical background: Another an excitation of 360-380 nm and an emission of 440-460 nm. class of adjunctive agents Suitable for use in the combinations of the invention are myostatin antagonists. Myostatin, also 2.2 Proteasome Inhibitors known as growth/differentiation factor 8 (GDF-8) is a trans 0201 Definitions and technical background: Another forming growth factor-fi (TGF-3) family member involved in class of adjunctive agents Suitable for use in the combinations the regulation of skeletal muscle mass. Most members of the of the invention are proteasome inhibitors. Proteasomes con TGF-B-GDF family are widely expressed and are pleiotropic: trol the half-life of many short-lived biological processes. At however, myostatin is primarily expressed in skeletal muscle the plasma membrane of skeletal muscle fibers, dystrophin tissue where it negatively controls skeletal muscle growth. associates with a multimeric protein complex, termed the Myostatin is synthesized as an inactive preproprotein which dystrophin-glycoprotein complex (DGC). Protein members is activated by proteolyic cleavage. The precurser protein is of this complex are normally absent or greatly reduced in cleaved to produce an approximately 109 amino acid COOH dystrophin-deficient skeletal muscle fibers and inhibition of terminal protein which, in the form of a homodimer of about the proteasomal degradation pathway rescues the expression 25kDa, is the mature, active form. The mature dimer appears and Subcellular localization of dystrophin-associated pro to circulate in the blood as an inactive latent complex bound teins. Thus, proteasome inhibitors have recently been identi to the propeptide. As used herein the term “myostatin antago fied as potential therapeutics for the treatment of DMD (see nist' defines a class of agents which inhibit or block at least Bonuccelli et al. (2003) Am J Pathol. October; 163(4): 1663 one activity of myostatin, or alternatively, blocks or reduces 1675). The term “proteasome inhibitor as used herein refers the expression of myostatin or its receptor (for example, by to compounds which directly or indirectly perturb, disrupt, interference with the binding of myostatin to its receptor block, modulate or inhibit the action of proteasomes (large and/or blocking signal transduction resulting from the bind protein complexes that are involved in the turnover of other ing of myostatin to its receptor). Such agents therefore cellular proteins). The term also embraces the ionic, salt, include agents which bind to myostatin itselforto its receptor. US 2011/0195932 A1 Aug. 11, 2011

0205 Preferences and specific embodiments: Myostatin US2004/0223966 and WO2006/012.627, both of which are antagonists for use according to the invention include anti incorporated herein by reference). bodies to GDF-8; antibodies to GDF-8 receptors; soluble 0208. Other preferred myostatin antagonists based on the GDF-8 receptors and fragments thereof (e.g. the ActRIIB myostatin receptors are ALK-5 and/or ALK-7 inhibitors (see fusion polypeptides as described in U.S. Pat. No. 10/689,677, for example WO2006025988 and WO2005084699, the dis including soluble ActRIIB receptors in which ActRIIB is closure of which is incorporated herein by reference). As a joined to the Fc portion of an immunoglobulin); GDF-8 TGF-B cytokine, myostatin signals through a family of single propeptide and modified forms thereof (e.g. as described in transmembrane serine/threonine kinase receptors. These WO 02/068650 or U.S. Pat. No. 10/071,499, including forms receptors can be divided in two classes, the type I or activin in which GDF-8 propeptide is joined to the Fc portion of an like kinase (ALK) receptors and type II receptors. The ALK immunoglobulin and/or form in which GDF-8 is mutated at receptors are distinguished from the Type II receptors in that an aspartate (asp) residue, e.g., asp-99 in murine GDF-8 the ALK receptors (a) lack the serine/threonine rich intracel propeptide and asp-100 inhuman GDF-8 propeptide); a small lular tail, (b) possess serine/threonine kinase domains that are molecule inhibitor of GDF-8; follistatin (e.g. as described in very homologous between Type I receptors, and (c) share a U.S. Pat. No. 6,004,937) or follistatin-domain-containing common sequence motif called the GS domain, consisting of proteins (e.g. GASP-1 or other proteins as described in U.S. a region rich in glycine and serine residues. The GS domain is Pat. No. 10/369,736 and U.S. Pat. No. 10/369,738); and at the amino terminal end of the intracellular kinase domain modulators of metalloprotease activity that affect GDF-8 and is believed to be critical for activation by the Type II activation, as described in U.S. Pat. No. 10/662,438. receptor. Several studies have shown that TGF-B signaling 0206 Preferred myostatin antagonists include myostatin requires both the ALK (Type I) and Type II receptors. Spe antibodies which bind to and inhibit or neutralize myostatin cifically, the Type II receptor phosphorylates the GS domain (including the myostatin proprotein and/or mature protein, in of the Type 1 receptor for TGF-beta ALK5, in the presence monomeric or dimeric form). Myostatin antibodies are pref of TGF-beta. The ALK5, in turn, phosphorylates the cyto erably mammalian or non-mammalian derived antibodies, for plasmic proteins Smad3 and Smad3 at two carboxy terminal example an IgNAR antibody derived from sharks, or huma serines. Generally, it is believed that in many species, the nised antibodies (or comprise a functional fragment derived Type II receptors regulate cell proliferation and the Type I from antibodie. Such antibodies are described, for example, receptors regulate matrix production. Various ALK5 receptor in US 2004/0142383, US 2003/1038422, WO 2005/0944.46 inhibitors have been described (see, for example, U.S. Pat. and WO 2006/116269 (the content of which is incorporated No. 6,465,493, US2003/0149277, US2003/0166633, herein by reference). Myostatin antibodies also include those US20040063745, and US2004/0039198, the disclosure of which bind to the myostatin proprotein and prevent cleavage which is incoprorated herein by reference). Thus, the myo into the mature active form. A particularly preferred myosta statin antagonists for use according to the invention may tin antibody for use in the combinations of the invention is comprise the myostatin binding domain of an ALK5 and/or Wyeth's (MYO-029). MYO-029 is a recombi ALK7 receptor. nant human antibody which binds to and inhibits the activity 0209. Other preferred myostatin antagonists include of myostatin. Other preferred antibody antagonists include soluble ligandantagonists which compete with myostatin for the antibodies described in U.S. Pat. No. 6,096,506 and U.S. binding to myostatin receptors. The term "soluble ligand Pat. No. 6,468,535 (incorporated herein by reference). In antagonist’ here refers to soluble peptides, polypeptides or some embodiments, the GDF-8 inhibitor is a monoclonal peptidomimetics capable of non-productively binding the antibody or a fragment thereof that blocks GDF-8 binding to myostatin receptor(s) (e.g. the activin type HB receptor (Ac its receptor. Other illustrative embodiments include murine tRHA)) and thereby competitively blocking myostatin-recep monoclonal antibody JA-16 (as described in US2003/ tor signal transduction. Soluble ligand antagonists include 0138422 (ATCC Deposit No. PTA-4236); humanized deriva variants of myostatin, also referred to as “myostatin ana tives thereof and fully human monoclonal anti-GDF-8 anti logues' that have homology with but not the activity of myo bodies (e.g., Myo-29. Myo-28 and Myo-22, ATCC Deposit statin. Such analogues include truncates (such an N- or C-ter Nos. PTA-4741, PTA-4740, and PTA-4739, respectively, or minal truncations, Substitutions, deletions, and other derivatives thereof) as described in US2004/0142382 and alterations in the amino acid sequence. Such as variants hav incorporated herein by reference. ing non-amino acid Substitutions). 0207. Other preferred myostatin antagonists include 0210. Other preferred myostatin antagonists further soluble receptors which bind to myostatin and inhibit at least include polynucleotide antagonists. These antagonists one activity thereof. The term “soluble receptor here include antisense or sense oligonucleotides comprising a includes truncated versions or fragments of the myostatin single-stranded polynucleotide sequence (either RNA or receptor which specifically bind myostatin thereby blocking DNA) capable of binding to target mRNA (sense) or DNA or inhibiting myostatin signal transduction. Truncated ver (antisense) sequences. Antisense or sense oligonucleotides sions of the myostatin receptor, for example, include the for use according to the invention comprise fragments of the naturally-occurring soluble domains, as well as variations targeted polynucleotide sequence encoding myostatin or its elaborated by proteolysis of the N- or C-termini. The soluble receptor, transcription factors, or other polynucleotides domain includes all or part of the extracellular domain of the involved in the expression of myostatin or its receptor. Such a receptor, either alone or attached to additional peptides or fragment generally comprises at least about 14 nucleotides, other moieties. Since myostatin binds activin receptors (in typically from about 14 to about 30 nucleotides. Antisense or cluding activin type IEE receptor (ActRHB) and activin type sense oligonucleotides further comprise oligonucleotides HA receptor (ActRHA), activin receptors can form the basis having modified Sugar-phosphodiester backbones (or other of Soluble receptor antagonists. Soluble receptor fusion pro sugar linkages, such as those described in WO91/06629) and teins can also be used, including soluble receptor Fc (see wherein such Sugar linkages are resistant to endogenous US 2011/0195932 A1 Aug. 11, 2011

nucleases. Such oligonucleotides with resistant Sugar link 0213 Preferences and specific embodiments: Preferred ages are stable in Vivo but retain sequence specificity to be cytokines for use according to the invention include IGF-1 able to bind to target nucleotide sequences. Other examples of and IGF-2. Approximately 99% of IGF-1 inhealthy individu sense orantisense oligonucleotides include those oligonucle als circulates in the blood stream bound to IGFBP-3 where it otides which are covalently linked to organic moieties. Such forms a large ternary 150 kD complex after association with as those described in WO 90/10448, and other moieties that acid-labile subunit protein (ALS). The ternary complex is increases affinity of the oligonucleotide for a target nucleic restricted to the circulation by the capillary endothelium and acid sequence. Such as poly-(L)-lysine and morpholinos. Fur thus serves as a circulatory reservoir of IGF-1. Thus, for ther still, intercalating agents, such as ellipticine, and alky therapeutic applications according to the invention IGF-1 is lating agents or metal complexes may be attached to sense or preferably administered in the form of a complex. For antisense oligonucleotides to modify binding specificities of example, a preferred cytokine for use in the combinations of the antisense or sense oligonucleotide for the target nucle the invention is IPLEXTM (recombinant protein complex of otide sequence. Thus, RNA interference (RNAi) produced by insulin-like growth factor-I (IGF-1) and its most abundant the introduction of specific small interfering RNA (siRNA), binding protein, insulin-like growth factor binding protein-3 may also be used to inhibit or eliminate the activity of myo (IGFBP-3)). Another suitable cytokine is G-CSF (or other statin. mobilizing agents as herein defined, e.g. GM-CSF), which 0211 Particularly preferred myostatin antagonists include can Support muscle regeneration by mobilizing stem cells but are not limited to follistatin, the myostatin prodomain, from the marrow. Other preferred cytokines include IGF-1 growth and differentiation factor 11 (GDF-11) prodomain, derivatives (IGF-1E peptides) as described in prodomain fusion proteins, antagonistic antibodies that bind WO2006056885 (the content of which is incorporated herein to myostatin, antagonistic antibodies or antibody fragments by reference) which have the appropriate subsets of the func that bind to the activin type IEB receptor, soluble activin type tion of the full-length IGF-1 and, in particular, its regenerative IHB receptor, soluble activin type IEB receptor fusion pro capacity. Thus, in a preferred embodiment the combinations teins, soluble myostatin analogs (soluble ligands), oligo of the invention comprise the IGF-I Ea peptide (i.e. the 35 nucleotides, Small molecules, peptidomimetics, and myosta amino acid C terminal peptide translated from part of exons 4 tin binding agents. disclose anti-myostatin antibodies. Other and 5 of the IGF-I gene as part of the IGF-I propeptide and preferred antagonists include the peptide immunogens which is cleaved off during post-translational processing) described in U.S. Pat. No. 6,369,201 and WO 01/05820 (in and/or the IGF-I Eb peptide (i.e. the 41 amino acid C terminal corporated herein by reference) and myostatin multimers and peptide translated from parts of exons 4, 5 and 6 of the IGF-I immunoconjugates capable of eliciting an immune response gene as part of the IGF-I propeptide and which is cleaved off and thereby blocking myostatin activity. Other preferred during post-translational processing). antagonists include the protein inhibitors of myostatin 0214) Posology: IPLEXTM can be administered via subcu described in WO02/085306 (and incorporated herein by ref taneous injection at an initial dose of 0.5 mg/kg, to be erence), which include the truncated Activin type II receptor, increased into the therapeutic dose range of 1 to 2 mg/kg, the myostatin pro-domain, and follistatin. Other myostatin given once daily. IPLEXTM can be given in the morning or in inhibitors include those released into culture from cells over the evening but should be administered at approximately the expressing myostatin (see WO00/43781), dominant nega same time every day. In order to establish tolerability to tives of myostatin (see WO 01/53350) including the Pied IPLEXTM, glucose monitoring should be considered at treat montese allele, and mature myostatin peptides having a ment initiation or when a dose has been increased. If frequent C-terminal truncationata position either at or between amino symptoms of hypoglycemia or severe hypoglycemia occur, acid positions 335 to 375. The small peptides described in preprandial glucose monitoring should continue. Glucose US2004/0181033 (incorporated herein by reference) which monitoring is also advised for patients with recent occur comprise the amino acid sequence WMCPP, are also suitable rences of asymptomatic or symptomatic hypoglycemia. If for use in the combinations of the invention. evidence of hypoglycemia is present at the time of dosing, the dose should be withheld. 4. Cytokines and Mobilizing Agents 0215 Dosage can be titrated up to a maximum of 2 mg/kg 0212 Definitions and technical background: Another daily based on measurement of IGF-1 levels obtained 8-18 class of adjunctive agents Suitable for use in the combinations hours after the previous dose. Dosage should be adjusted of the invention are cytokines, and in particular anabolic downward in the event of adverse effects (including hypogly cytokines and insulin-like growth factors (such as IGF-1 or cemia) and/or IGF-1 levels that are greater than or equal to 3 IGF-2). The anabolic effect of IGF-1 on muscle is very well standard deviations above the normal reference range for established. In muscular dystrophies, a progressive reduction IGF-1. in the proliferative capacity of satellite cells occurs and this loss of proliferative capacity may be ameliorated by treatment 5. Corticosteroids with IGF-1. Thus, IGF-1 (and other members of this class of 0216. In one embodiment of the invention, the ancillary cytokine) may help to slow the progress of the dystrophino agent is a corticosteroid. pathies by enhancing activation of dormant satellite cells. 0217 Definition and biological activities: The term “cor Insulin-like growth factors (IGFs) are members of the highly ticosteroid as used herein refers to any of several steroid diverse insulin gene family that includes insulin, IGF-1, IGF hormones secreted by the cortex of the adrenal glands and 11, relaxin, prothoraciotropic hormone (PTTH), and mollus which are involved in one or more of the following physi can insulin-related peptide. The IGFs are circulating, mito ological processes: stress response, immune response and genic peptide hormones that have an important role in regulation of inflammation, carbohydrate metabolism, pro stimulating growth, differentiation, metabolism and regen tein catabolism and blood electrolyte levels. The term also eration both in vitro and in vivo. includes synthetic analogues which share the aforementioned US 2011/0195932 A1 Aug. 11, 2011 properties. Corticosteroids include glucocorticoids and min compared with prednisone, though biological equivalence eralocorticoids. Glucocorticoids control carbohydrate, fat between deflazacort and prednisone also depends on the spe and protein metabolism and are anti-inflammatory. Mineralo cific actions under examination. Corticosterods (including corticoids control electrolyte and water levels, mainly by delazacort and prednisone) are usually taken orally but can be promoting Sodium retention in the kidney. Some corticoster delivered by intramuscular injection. oids have dual glucocorticoid and mineralocorticoid activi ties. For example, prednisone (see below) and its derivatives 6. Anabolic Steroids have some mineralocorticoid action in addition to a glucocor 0221. In one embodiment of the invention, the ancillary ticoid effect. The precise cellular mechanism(s) by which agent is an anabolic steroid. corticosteroids produce antidystrophic effects are not yet 0222 Definition and biological activities: The term “ana known. A multifactorial mechanism is likely and the effects bolic steroid’ as used herein refers to any of several steroid of corticosteroids probably involve a reduction of inflamma hormones related to the male hormone testosterone and Syn tion, Suppression of the immune system, improvement in thetic analogues thereof. Such steroids are may also be calcium homeostasis, upregulation of the expression of com referred to as “anabolic-androgenic steroids' or 'AAS'. Ana pensatory proteins and an increase in myoblast proliferation. bolic steroids increase protein synthesis within cells, promot 0218. Problems: The use of corticosteroids is associated ing anabolism (especially in muscles). The precise cellular with side effects which vary from person to person and on the mechanism(s) by which anabolic steroids produce antidyS dosage of the regime used, but they can be severe. The most trophic effects are not yet known, but it seems that their common side effects are weight gain and mood changes. anabolic effects in muscles effectively compensates for Weight gain (and attendant changes in muscle activity and muscle loss. Oxandrolone has been shown to have anabolic use) can abrogate Some of the benefits of treatment. Long effects on DMD muscle as well as decreasing muscle degen term use may lead to growth suppression, cataracts, eration and so easing the demands for muscle regeneration. osteoporosis and muscle atrophy (affecting the same proxi By conserving regenerative capacity, anabolic steroids Such mal muscles affected in DMD and BMD). These side effects as Oxandrolone may prolong muscle function. may limit the long-term effectiveness of corticosteroid 0223 Problems: The use of anabolic steroids is associated therapy. Other side effects include hypertension, diabetes, with severe side effects. The most common side effects are skin atrophy, poor wound healing and immunosuppression. liver and kidney damage, sterility, stunting of growth and Deflazacort was evaluated in the hope that it would have severe mood Swings. Anabolic steroids also also tend to be fewer side effects than prednisone. androgenizing and can promote growth of beard and body 0219 Preferences and Specific embodiments: Preferred hair, maturation of genitalia and development of acne. With are glucocorticoids (or corticosteroids having dual glucocor drawal can lead to rapid and severe deterioration in muscle ticoid/minerlocorticoid activity). Synthetic corticosteroids mass and function. are preferred. In one embodiment, the corticosteroid is pred 0224 Preferences and Specific embodiments: Preferred nisone (prodrug) or prednisolone (liver metabolite of pred are synthetic anabolic steroids such as Oxandrolone (Anavar), nisone and active drug). In another embodiment, the corticos norethandrolone and methandrostenolone (Dianabol). Oxan teroid is deflazacort. Deflazacort is an oxazoline analogue of drolone (an oral synthetic analog of testosterone) may be prednisone. Other synthetic corticosteroids suitable for use in particularly preferred because in addition to its anabolic prop the combinations of the invention include one or more corti erties it also blocks the binding of cortisol to glucocorticoid costeroids selected from: alclometasone, amcinonide, receptors on muscle, thus preventing muscle breakdown. beclomethasone (including beclomethasone dipropionate), Otheranabolic steroids suitable foruse in the combinations of betamethasone, budesonide, ciclesonide, clobetasol, clobeta the invention include one or more anabolic steroids selected Sone, clocortolone, cloprednol, cortivaZol, deoxycorticoster from: DHEA, DHT methenolone, oxymetholone, quin one, desonide, desoximetasone, dexamethasone, diflorasone, bolone, stanozolol, ethylestrenol, nandrolone (Deca Durabo diflucortolone, difluprednate, fluclorolone, fludrocortisone, lin), oxabolone cipionate, boldenone undecylenate (Equi fludroxycortide, flumetasone, flunisolide fluocinolone poise), stanozolol (Winstrol), oxymetholone (Anadrol-50), acetonide, fluocinonide, fluocortin, fluocortolone, fluo fluoxymesterone (Halotestin), trenbolone (Fina), meth rometholone, fluperolone, fluprednidene, fluticaSone, formo enolone enanthate (Primobolan), 4-chlordehydromethyltest cortal, halcinonide, halometasone, hydrocortisone ace osterone (Turinabol), mesterolone (Proviron), mibolerone ponate, hydrocortisone buteprate, hydrocortisone butyrate, (Cheque props), tetrahydrogestrinone and testosterone (or loteprednol, medrysone, meprednisone, methylprednisolone, combinations and/or derivatives (e.g. pharmaceutically methylprednisolone aceponate, mometaSone furoate, acceptable salts) of one or more of the foregoing). paramethasone, prednicarbate, prednylidene, rimexolone, 0225 Posology: Anabolic steroids may be administered as tiXocortol, triamcinolone and ulobetasol (or combinations orally in the form of pills, by injection or via skin patches. and/or derivatives (e.g. pharmaceutically acceptable salts) of Oral administration is most convenient, but since the steroid one or more of the foregoing). Suitable endogenous corticos must be chemically modified so that the liver cannot break it teroids for use in the combinations of the invention include down before it reaches the blood stream these formulations include one or more corticosteroids selected from aldoster can cause liver damage in high doses. Injectable steroids are one, cortisone, hydrocortisonefcortisol and desoxycortone typically administered intramuscularly. Transdermal patches (or combinations and/or derivatives (e.g. pharmaceutically can be sued to deliver a steady dose through the skin and into acceptable salts) of one or more of the foregoing). the bloodstream. Oxandrolone may be administered orally at 0220 PoSology: Prednisone may be administered daily in a daily dosage of 0.1 mg/kg. dosages ranging from 0.3 to 1.5 mg/kg (typically 0.7 mg/kg). 7. TGF-B antagonists Some patienmts respond better to 22.5 mg/kg every other 0226 Definitions and technical background: Transform day. Deflazacort has an estimated dosage equivalency of 1:1.3 ing growth factor beta (TGF-3) promotes fibrosis in response US 2011/0195932 A1 Aug. 11, 2011

to muscle tissue damage associated with DMD that can con (including EGCG and other polyphenols) may improve MD tribute to disease pathology. In one embodiment of the inven prognosis by reducing this oxidative stress. Feeding studies tion, the ancillary agent is a TGF-B antagonist. with mdx mice have shown a protective effect of EGCG 0227. The term TGF-B antagonist is used herein to refer to against the first massive wave of necrosis. It also stimulated compounds which directly or indirectly perturb, disrupt, muscle adaptation toward a stronger and more resistant phe block, modulate or inhibit the action of TGF-B. The term also embraces the ionic, salt, Solvate, isomers, tautomers, N-ox notype. The effective dosage corresponds to about seven cups ides, ester, prodrugs, isotopes and protected forms thereof of brewed green tea per day in humans (preferably the salts or tautomers or isomers or N-oxides or 0232 Coenzyme Q10 (CoQ10; also called ubiquitin) is a solvates thereof, and more preferably, the salts or tautomers powerful antioxidant and mitochondrial respiratory chain or N-oxides or solvates thereof). cofactor. It possesses membrane-stabilizing properties and is 0228 Preferences and specific embodiments: Preferred capable of penetrating cell membranes and mitochondria. TGF-B antagonists for use according to the invention include Dosages of 100 mg CoQ10 daily for three months have been anti-TGF-3 antibodies, tamoxifen, losartan and pirfenidone. Pirfenodone is an orally active synthetic antifibrotic agent shown to be beneficial in human trials, though higher dosages structurally similar to pyridine 2,4-dicarboxylate. Pirfeni are likely to yield better results. done inhibits fibroblast, epidermal, platelet-derived, and 0233. Idebenone is a synthetic analog of Coenzyme Q10 TGF-B-1 growth factors and also inhibits DNA synthesis and and is thought to perform the same functions as CoQ10 with the production of mRNA for collagen types I and Ill, resulting out the risk of auto-oxidation. Like CoQ10, idebenone can in a reduction in radiation-induced fibrosis. Losartan is an therefore contribute to maintaining correct electron balance, angiotensin II receptor antagonist drug used mainly to treat which is necessary for the production of cellular energy. hypertension currently marketed by Merck & Co. under the Since muscle cells are particularly energy-demanding, ide trade name CozaarTM. However, losartan also downregulates the expression of (TGF-B types I and II receptors. Tamoxifen benone and CoQ10 can preserve mitochondrial function and is an orally active selective estrogen receptor modulator protect cells from oxidative stress. (SERM) which is used in the treatment of breast cancer and is 0234 Glutamine is an important energy source and acute currently the world's largest selling drug for this indication. oral glutamine administration appears to have a protein-spar Tamoxifen is sold under the trade names NolvadexTM, Istu ing effect. Arginine (and other pharmacological activators of balTM and ValodexTM. Tamoxifen may be administered at a the NO pathway) may enhance the production of utrophin in dose of 10-100 mg per day (e.g. 20-40 mg/day). MDX mice. The increase is likely to be mediated by arginine fueled production of nitric oxide (NO), which plays an impor 8. Antioxidants and Mitochondrial Supporting Agents tant role in blood vessel function and is generally lower in 0229. In Duchenne Muscular Dystrophy (DMD), the people with MD. Studies with MDX mice have also shown cytoskeletal protein dystrophin is absent leading to numerous that a combination of arginine and deflazacort may be more cellular dysfunctions that culminate in muscle cell necrosis. beneficial than deflazacort alone. Subsequently, an inflammatory response develops in the necrotic muscle tissue, resulting in increased oxidative stress, 0235. Other antioxidants suitable for use according to the responsible for further tissue damage. In the mdx dystrophic invention are the chimaeric compounds or combinations in mouse, both inflammation and oxidative stress have been which the a ROS inhibitor is associated (e.g. combined with, identified as aggravating factors for the course of the disease. co-administered with or covalently linked) to calpain inhib 0230 GTE and EGCG also display unexpected pro myo iting moiety. Such agents combine relief of oxidative stress genic properties. Primary cultures of skeletal muscle cells with a reduction in calpain-mediated muscle tissue break were established from both normal and dystrophic mice and down. Suitable dual action calpain/ROS inhibitors are treated with GTE and EGCG for 1-7 days. As judged by in situ described for example in WOO1/32654, WO2007/045761, staining of myosin heavy chains (My HC), we found that GTE WO2005/056551 and WO 2002/40016 (the contents of which and EGCG concentration-dependently stimulated the rate of are incorporated herein by reference). formation of myotubes within the first 2-4 days of applica tion. The amount of myotubes reached similar level with both agents compared to control thereafter. Western-blot analysis 9. Dystrophin Expression Enhancing Agents was performed on myotube cultures treated for 7 days. GTE 9.1 Read-Through Agents and EGCG promoted the expression of several muscle-spe cific proteins, such as dystrophin (in control cultures), sarco 0236. A subset of DMD patients (around 15%) have a meric alpha actinin, and MyHC, while myogenin was nonsense mutation that produces a premature stop signal in unchanged. By contrast, the expression of desmin was down their RNA, resulting in abnormal truncation of protein trans regulated and redistributed to Z discs. Our results suggest that green tea polyphenols display pro myogenic properties by lation. In one embodiment of the invention, the ancillary acting directly on skeletal muscle cells. These findings Sug agent is an agent which promotes readthrough of premature gest a beneficial action for muscle regeneration and strength stop codons (“read-through agent'), thereby bypassing the ening in dystrophic condition. premature stop codon and restoring the expression of full 0231 Green tea polyphenols, such as epigallocatechin length, functional dystrophin. gallate (EGCG), are known to be powerful antioxidants. 0237 Suitable read-through agents for use according to Because inflammation is involved in the degradation of the invention are 1,2,4-oxadiazole compounds as described in muscle tissue in MD, oxidative stress is believed to play a role U.S. Pat. No. 6,992,096 (which is incorporated herein by in this process. Thus, green tea and its active constituents reference): US 2011/0195932 A1 Aug. 11, 2011 17

may be adeno-associated virus (AAV) vector-mediated gene therapy, preferably using the microdystrophin gene. Highly R2 l-O abbreviated microdystrophin cDNAs have been developed 3 X Z for adeno-associated virus (AAV)-mediated DMD gene R N therapy. Among these, a C-terminal-truncated AR4-R23/AC microgene (AR4/AC) is a very promising therapeutic candi date gene. R4 R5 0243 Targeted correction of mutations in the genome holds great promise for the repair/treatment of disease caus () ing mutations either on their own applied directly to the affected tissue, or in combination with other techniques such (2) indicates text missing or illegible when filed as stem cell transplantation. Various DNA or RNA/DNA based Corrective Nucleic Acid (CNA) molecules such as 0238. One such compound is 3-5-(2-fluoro-phenyl)-I.2. chimeraplasts, single stranded oligonucleotides, triplex 4-oxadiazol-3-yl)-benzoic acid. A preferred readthrough forming oligonucleotides and SFHR have been used to agent is PTC 124. PTC 124 is a 284-Dalton 1,2,4-oxadiazole change specific mutant loci. MyoDyS(R) is comprised of plas that promotes ribosomal readthrough of premature stop mid DNA encoding the full-length human dystrophin gene. codons in mRNA. Thus, the combinations of the invention Mirus Pathway IVTM delivery technology is used to admin may comprise 1,2,4-oxadiazole benzoic acid compounds (in ister the plNA to a patient's limb skeletal muscles. cluding 3-5-(2-fluoro-phenyl)-I,2,4-oxadiazol-3-yl)-ben Zoic acid) (see e.g. WO2006110483, the content of which is 11. Cell-Based Therapies incorporated herein by reference). 0244. In one embodiment of the invention, the ancillary 0239 PTC 124, 3-5-(2-fluoro-phenyl)-1,2,4-oxadiazol agent is a myogenic cell or tissue composition. Various types 3-yl)-benzoic acid or a pharmaceutically acceptable salt, Sol of myogenic cell have been shown to have potential in the vate or hydrate thereof can be administered in single or treatment of DMD, including stem cells from umbilical cord, divided (e.g., three times daily) doses between 0.1 mg/kg and mesenchymal stem cells and muscle-derived stem cells. 500 mg/kg, 1 mg/kg and 250 mg/kg, 1 mg/kg and 150 mg/kg, 1 mg/kg and 100 mg/kg, 1 mg/kg and 50 mg/kg, 1 mg/kg and 12. Creatine 25 mg/kg, 1 mg/kg and 10 mg/kg or 2 mg/kg and 10 mg/kg to a patent in need thereof. In a particular embodiment, the 0245. Definition and biological activities: Creatine is an 3-5-(2-fluoro-phenyl)-1,2,4-oxadiazol-3-yl)-benzoic acid energy precursor that is naturally produced by the body. Cre atine kinase (CK) phosphorylates creatine for later donation or a pharmaceutically acceptable salt, Solvate or hydrate to contractile muscle filaments: phosphocreatine enters thereof is administered in a dose of about 4 mg/kg, about 7 muscle cells and promotes protein synthesis while reducing mg/kg, about 8 mg/kg, about 10 mg/kg, about 14 mg/kg or protein breakdown. In healthy individuals, creatine has been about 20 mg/kg. shown to enhance endurance and increase energy levels by 0240. Other readthrough agents for use according to the preventing depletion of adenosine triphosphate. Among MD invention include aminoglycoside antibiotics, including gen patients, studies have Suggested that Supplemental creatine tamicin. Particularly preferred may be aminoglycosides that can improve muscle performance and strength, decrease contain a 6' hydroxyl group (e.g. paromomycin), which may fatigue, and slightly improve bone mineral density. be effective at lower doses and may display less toxicity than 0246 Problems: High doses of creatine can cause kidney compounds such as gentamicin. damage and requires cohydration. Behavioral changes have 9.2 Exon Skipping been recorded. 0247 Posology: Creatine can be administered as a pow 0241 Most cases of Duchenne muscular dystrophy dered nutritional supplement. In recent trials with DMD (DMD) are caused by dystrophin gene mutations that disrupt patients, slight increases in muscle strength on administration the mRNA reading frame. In some cases, forced exclusion of low levels (1 to 10 g/day) of creatine monohydrate have (skipping) of a single exon can restore the reading frame, been recorded. Intermittent administration (involving a break giving rise to a shorter, but still functional dystrophin protein of one to several weeks) may mitigate side effects whilst (so called quasi-dystrophin). Antisense oligonucleotides providing the same benefits as constant use. Dosages in the (AONs) designed to cause exon skipping can target abroader region of 100 mg/kg/day are well-tolerated and have been range of mutations than can compounds that cause cells to found to decrease bone degradation and increase strength and ignore premature stop codons by induce cells to leave out fat-free mass. Benefits have been reported for the co-admin sections of genetic instructions that contain mistakes and join istration of creatine with conjugated linoleic acid (alpha together the Surrounding, correct instructions. However, lipoic acid), hydroxyl-beta-methylbutyrate and prednisolone. since AONs are not self-renewed, they cannot achieve long term correction. To overcome this limitation, antisense 13. Anti-Osteoporotic Agents sequences can be introduced into Small nuclear RNAS (Sn 0248 Combined therapy to inhibit bone resorption, pre RNA) and vectorized in AAV and lentiviral vectors. vent osteoporosis, reduce skeletal fracture, enhance the heal 10. Gene Replacement/Repair Agents ing of bone fractures, stimulate bone formation and increase bone mineral density can be effectuated by combinations 0242. In one embodiment of the invention, the ancillary comprising various anti-osteoporotic agents. Preferred are agent is a nucleic acid construct adapted to replace or repair bisphosphonates including alendronate, tiludronate, dim non-functional endogenous genetic material. Gene therapy ethyl-APD, risedronate, etidronate, YM-175, clodronate, US 2011/0195932 A1 Aug. 11, 2011

pamidronate and BM-210995 (ibandronate). Others include pharmaceutically acceptable salt thereof, as defined above, in oestrogen agonist/antagonists. The term oestrogen agonist/ admixture with a pharmaceutically acceptable diluent or car antagonists refers to compounds which bind with the estrogen 1. receptor, inhibit bone turnover and prevent bone loss. In par 0255 We also provide a process for the production of such ticular, oestrogen agonists are herein defined as chemical a pharmaceutical composition which comprises mixing the compounds capable of binding to the estrogen receptor sites ingredients. Examples of pharmaceutical formulations which in mammalian tissue, and mimicking the actions of estrogen may be used, and Suitable diluents or carriers, are as follows: in one or more tissue. Exemplary oestrogen agonist/antago for intravenous injection or infusion-purified water or saline Solution; for inhalation compositions—coarse lactose; for nists include droloxifene and associated compounds (see U.S. tablets, capsules and dragees-microcrystalline cellulose, cal Pat. No. 5,047.431), tamoxifen and associated compounds cium phosphate, diatomaceous earth, a Sugar Such as lactose, (see U.S. Pat. No. 4,536,516), 4-hydroxytamoxifen (see U.S. dextrose or mannitol, talc, Stearic acid, starch, Sodium bicar Pat. No. 4,623,660), raloxifene and associated compounds bonate and/or gelatin; for Suppositories—natural or hardened (see U.S. Pat. No. 4,418,068 and idoxifene and associated oils or waxes. compounds (see U.S. Pat. No. 4,839,155). 0256 When the compound is to be used in aqueous solu tion, e.g. for infusion, it may be necessary to incorporate other 14. Auxiliary Utrophin Upregulating Agents excipients. In particular there may be mentioned chelating or sequestering agents, antioxidants, tonicity adjusting agents, 0249. In addition to the compounds of formula (I) as pH-modifying agents and buffering agents. defined herein, the combinations of the present invention may 0257 Solutions containing a compound of formula I may, include one or more auxiliary utrophin upregulating agents. if desired, be evaporated, e.g. by freeze drying or spray dry Such auxiliary utrophin upregulating agents are compounds ing, to give a solid composition, which may be reconstituted that upregulate (i.e. increase the expression or activity of prior to use. utrophin) and which do not conform to the structure of for 0258 When not in solution, the compound of formula I mula (I) as defined herein (or the ionic, salt, Solvate, isomers, preferably is in a form having a mass median diameter of from tautomers, N-oxides, ester, prodrugs, isotopes and protected 0.01 to 10 um. The compositions may also contain suitable forms thereof). The auxiliary utrophin upregulating agents preserving, stabilising and wetting agents, solubilisers, e.g. a for use in the combinations of the invention preferably water-soluble cellulose polymer Such as hydroxypropyl upregulate utrophin via a mechanism that is different from methylcellulose, or a water-soluble glycol such as propylene that of the compounds of formula (I) described herein. glycol, Sweetening and colouring agents and flavourings. 15. cQMP signalling Modulators Where appropriate, the compositions may be formulated in (0250. It has recently been shown (Khairallah et al. (2008) Sustained release form. PNAS 105(19): 7028-7033) that enhancement of cQMP sig 0259. The content of compound formula I in a pharmaceu naling by administration of the phosphodiesterase 5 (PDE5) tical composition is generally about 0.01-about 99.9 wt %, inhibitor sildenafil prevents deterioration of myocardial con preferably about 0.1-about 50 wt %, relative to the entire tractile performance in mdx hearts. preparation. 0251 Thus, c0MP signaling enhancers, including in par 0260 The dose of the compound of formula I is deter ticular selective PDE5 inhibitors (including for example mined in consideration of age, body weight, general health sildenafil, tadalafil. Vardenafil. udenafil and avanafil) may be condition, diet, administration time, administration method, used in combination with the compounds of the invention to clearance rate, combination of drugs, the level of disease for treat DMD or BMD. Such combinations find particular appli which the patient is under treatment then, and other factors. cation in the treatment of dystrophic cardiopmyopathies and 0261) While the dose varies depending on the target dis may be used to prevent or delay the onset of dystrophin ease, condition, Subject of administration, administration related cardiomyopathies as the clinical course of DMD/ method and the like, for oral administration as a therapeutic BMD progresses. agent for the treatment of Duchenne muscular dystrophy in a 0252. Thus, the invention contemplates combinations of patient Suffering from Such a disease is from 0.01 mg-10g, the compounds of the invention with cGMP signaling preferably 0.1-100 mg, is preferably administered in a single enhancers, including in particular selective PDE5 inhibitors. dose or in 2 or 3 portions per day. Preferred combinations are comprise a compound of the EXAMPLES invention and a PDE5 inhibitor selected from sildenafil, tad 0262 The potential activity of the compounds of formula alafil. Vardenafil. udenafil and avanafil. Particularly preferred I for use in the treatment of DMD may be demonstrated in the is a combination comprising a compound of the invention and following predictive assay and screens. sildenafil. The compound of the invention for use in the afor mentioned combinations is preferably compound number 1. Luciferase Reporter Assay (Murine H2K Cells) 390 of Table 1 being 5-(ethylsulfonyl)-2-(naphthalen-2-yl) 0263. The cell line used for the screen is an immortalized benzodoxazole. mdx mouse H2K cell line that has been stably transfected with a plasmid containing OO5 kb fragment of the Utrophin A Formulation and Posology promoter including the first untranslated exon linked to a luciferase reporter gene. Under conditions of low temperature 0253) The compounds of formula I for use in the treatment and interferon containing media, the cells remain as myo of DMD will generally be administered in the form of a blasts. These are plated into 96 well plates and cultured in the pharmaceutical composition. presence of compound for three days. The level of luciferase 0254 Thus, according to a further aspect of the invention is then determined by cell lysis and reading of the light output there is provided a pharmaceutical composition including from the expressed luciferase gene utilising a plate luminom preferably less than 80% w/w, more preferably less than 50% eter. Example of pharmacological dose response of com w/w, e.g. 0.1 to 20%, of a compound of formula I, or a pounds in the assay is shown in FIG. 1. US 2011/0195932 A1 Aug. 11, 2011

2. mdx Mouse (0273 Interferon (1276905 Roche) Add fresh 10 ul/50 mls medium 0264. Data obtained from the ADMET data was priori tised and the compounds with the best in vitro luciferase Luciferase Assay for 96 Well Plates activity and reasonable ADMET data were prioritised for (0274 The H2K/mdx/Utro A reporter cell line cells were testing in the mdx proof of concept study where the outcome plated out into 96 well plates (Falcon 353296, white opaque) was to identify whether any of the compounds had the ability at a density of approximately 5000 cells/well in 190 ul normal to increase the levels of utrophin protein in dystrophin defi growth medium. The plates were then incubated at 33°C. in cient muscle when compared to vehicle only dosed control the presence of 10% CO for 24 hrs. Compounds were dosed animals. by adding 10 ul of diluted compound to each well giving a 0265. There were two animals injected with up to 50 final concentration of 10 uM. The plates were then incubated mg/kg (e.g. 10 mg/kg) of compound administered ip daily for for a further 48 hrs. Cells were then lysed in situ following the 28 days plus age matched controls. Muscle samples were manufacture's protocols (Promega Steady-Glo Luciferase taken and processed for sectioning (to identify increases in Assay System (E2520). Then counted for 10 seconds using a sarcolemmal staining of utrophin) and Western blotting (to plate luminometer (Victorl 420). identify overall increases in utrophin levels). Compound Storage 0266 FIG. 2 shows an example of TA muscle sections stained with antibody specific for mouse utrophin. Compari 0275 Compounds for screening were stored at -20°C. as son to the mdx muscle only injected with vehicle shows an 10 mM stocks in 100% DMSO until required. increase in the amount of sarcolemmal bound utrophin. Injection of mdx Mice with Compounds Muscles from the above treated mice were also excised and 0276 Mdx from a breeding colony were selected for test processed for Western blotting and stained with specific anti ing. Mice were injected daily with either vehicle or 10 mg/kg bodies (see FIG. 3). Again using muscle dosed with CPD-A of compound using the intreperitoneal route (ip). Mice were shows a significant increase in the overall levels of utrophin weighed and compounds diluted in 5% DMSO, 0.1% tween present in both the TA leg muscle and the diaphragm. Both in PBS. mice exposed to CPD-A (V2 and V3) showed increased levels 0277 Mice were sacrificed by cervical dislocation at of utrophin expression compared to control. Positive upregu desired time points, and muscles excised for analysis lation data from the first 28 day study were then repeated in a further two mouse 28 day study. A total of three different Muscle Analysis compounds have shown in duplicate the ability to increase the Immunohistochemistry level of utrophin expression in themdx mouse when delivered daily by ip for 28 days. This data demonstrates the ability of 0278 Tissues for sectioning were dissected, immersed in the compound when delivered ip causes a significant increase OCT (Bright Cryo-M-Bed) and frozen on liquid nitrogen in the levels of utrophin found in the mdx muscle and there cooled isopentane. Unfixed 8 LM cryosections were cut on a fore gives us the confidence that this approach will ameliorate Bright Cryostat, and stored at -80° C. the disease as all the published data to date demonstrates that 0279. In readiness for staining, sections were blocked in any increase of utrophin levels over three fold has significant 5% foetal calf serum in PBS for 30 mins. The primary anti functional effects on dystrophin deficient muscle. bodies were diluted in blocking reagent and incubated on H2K/mdx/Utro A reporter Cell Line Maintenance sections for 1.5 hrs in a humid chamber then washed three 0267. The H2K/mdx/Utro A reporter cell line was pas times for 5 mins in PBS. Secondary antibodies also diluted in saged twice a week until 530% confluent. The cells were blocking reagent, were incubated for 1 hr in the dark in a grown at 33°C. in the presence of 10% CO. To remove the humid chamber. Finally sections were washed three times 5 myoblasts for platting, they were incubated with Trypsin mins in PBS and coverslip Mounted with hydromount. Slides EDTA until the monolayer started to detach. were analysed using a Leica fluorescent microscope. Results Growth Medium 0280 Biological activity as assessed using the luciferase 0268 DMEM Gibco 41966 reporter assay in murine H2K cells, and is classified as fol 0269) 20% FCS lows: + Up to 200% relative to control 0270 1% Pen/Strep ++ Between 201% and 300% relative to control (0271 1% glutamine +++ Between 301% and 400% relative to control (0272 10 mls Chick Embryo Extract ++++ Above 4.01% relative to control

TABLE 1. Compounds made by methods described herein Example Chemical Name Activity 1 N-(2-(4-(dimethylamino)phenyl)benzodoxazol-5-yl)isonicotinamide -- 2 N-(2-(4-fluorophenyl)benzodoxazol-5-yl)furan-2-carboxamide -- 3 2-((4-chlorophenoxy)methyl)-1-methyl-1H-benzodimidazole ------4 2-(4-methoxyphenoxy)methyl)-1H-benzodimidazole ---- 5 phenyl(2-phenyl-1H-benzodimidazol-6-yl)methanone -- 6 N-(2-phenylbenzodoxazol-5-yl)nicotinamide -- 7 3-phenyl-N-(2-phenylbenzodoxazol-5-yl)propanamide --

US 2011/0195932 A1 Aug. 11, 2011 30

TABLE 2-continued Compounds made by analogues methods to those described herein, or by literature methods known or adapted by the persons skilled in the art. 459 Chemical Name Activity

671 N-(benzodthiazol-2-yl)-5-bromo-2-chlorobenzamide ---- 672 N-(4-(6-methylbenzodoxazol-2-yl)phenyl)thiophene-2-carboxamide ---- 673 3-amino-N-(2-fluorophenyl)-6,7-dihydro-5H-cyclopentaethieno2,3- ---- bipyridine-2-carboxamide 674 (3-(benzofuran-2-yl)-1-phenyl-1H-pyrazol-4-yl)methanol ---- 675 2-(4-methoxyphenyl)-5-methylbenzodoxazole ---- 676 4-ethoxy-N-((1-methyl-1H-benzodimidazol-2-yl)methyl)aniline ---- 677 N-(2-chloro-5-(5-methylbenzodoxazol-2-yl)phenyl)furan-2- ---- carboxamide 678 N-(4-methyl-6,7-dihydro-5H-cyclopentadpyrimidin-2- ---- yl)benzodoxazol-2-amine 679 N-(1H-benzodimidazol-2-yl)-3-chlorobenzamide ---- 680 N-(4-(benzodthiazol-2-yl)phenyl)tetrahydrofuran-2-carboxamide ---- 681 1-(2-(benzodoxazol-2-ylamino)-4,6-dimethylpyrimidin-5-yl)ethanone ---- 682 N-(4-methylpyrimidin-2-yl)benzodoxazol-2-amine ---- 683 N-(6-(N,N-dimethylsulfamoyl)benzodthiazol-2-yl)thiophene-2- ---- carboxamide 684 5-bromo-N-(4-hydroxy-3-(5-methylbenzodoxazol-2- ---- yl)phenyl)nicotinamide 685 2-(1H-1,2,4-triazol-3-ylthio)-N-(4-(benzodthiazol-2- ---- yl)phenyl)acetamide 686 5-(5-methoxybenzodoxazol-2-yl)-2-methylaniline ---- 687 N-(6-(N-methylsulfamoyl)benzodthiazol-2-yl)thiophene-2- ---- carboxamide 688 2-chloro-N-(4-(5-methylbenzodthiazol-2-yl)phenyl)-5-(4H-1,2,4- ---- triazol-4-yl)benzamide 689 N-(2-fluorophenyl)-2-(1H-indol-3-yl)-2-oxoacetamide ---- 670 N-(2,3-dihydrobenzob.14dioxin-6-yl)-2-(1H-indol-3-yl)-2- ---- oxoacetamide

Experimental When peak multiplicities are reported, the following abbre (0281 HPLC-UV-MS was performed on a Gilson 321 viations are used S (singlet), d (doublet), t (triplet), m (mul HPLC with detection performed by a Gilson 170 DAD and a tiplet), br(broadened), dd (doublet of doublets), dt (doublet of Finnigan AQA mass spectrometer operating in electrospray triplets), td (triplet of doublets). Coupling constants, when ionisation mode. The HPLC column used is a Phenomenex given, are reported in Hertz (Hz). Column chromatography Gemini C18 150x4.6 mm. Preparative HPLC was performed was performed either by flash chromatography (40-65 um on a Gilson 321 with detection performed by a Gilson 170 silica gel) or using an automated purification system (SP1TM DAD. Fractions were collected using a Gilson 215 fraction Purification System from Biotage(R). Reactions in the micro collector. The preparative HPLC column used is a Phenom wave were done in an Initiator 8TM (Biotage). The abbrevia enex Gemini C18 150x10 mm and the mobile phase is aceto tions used are DMSO (dimethylsulfoxide), HATU (O-(7-aza nitrile/water. benzotriazol-1-yl)-N.N.N',N'-tetramethyluronium 0282 'H NMR spectra were recorded on a Bruker instru hexafluorophosphate), HCl (hydrochloric acid), MgSO ment operating at 300 MHz. NMR spectra were obtained as (magnesium sulfate), NaOH (sodium hydroxide), NaCO CDC1 solutions (reported in ppm), using chloroform as the (sodium carbonate), NaHCO (sodium bicarbonate), STAB reference standard (7.25 ppm) or DMSO-D (2.50 ppm). (sodium triacetoxyborohydride), THF (tetrahydrofuran).

21 OH R9 R- -- o={ N NH2 X X = OH or Cl PPA, 110° C.-180° C. 3-16 h X = OH (Method 1A)

Microwave, dioxane, 210°C., 15 min X = Cl (Method 1B)

US 2011/0195932 A1 Aug. 11, 2011 40

-continued afNO l,R 21y- l,R 1y- H3C - afN-O R1SN-1a V O)-I X-R, S1, N-EH 's-NN X-R, O1 N-HN || X-R,- S--N I X-R)- CH3 3 WIb VII VI DX

Method 2A (Compounds Ib) cooling, the solution was poured into ice/water and neutralize 2-p-Tolvlb d 1-5-ami with Saturated aqueous NaHCO. The aqueous layer was then p olylbenzodoxazol-5-amine extracted twice with ethyl acetate (500 mL). The combined 0426 To 5-nitro-2-p-tolylbenzodoxazole (4.8 g. 18.90 organic layers were dried over anhydrous MgSO and evapo mmol) in ethyl acetate/acetic acid (250 ml/1 mL) was added rated to afford 188 mg (65%) of the title compound (LCMS palladium on carbon (480 mg). The reaction vessel was RT=6.76 min, MH 225.1) purged three times with nitrogen, followed by hydrogen three times, and then left stirring under hydrogen for 16 h. The 0433 H NMR (DMSO): 8.04 (2H, d, J 8.2 Hz), 7.41 (2H, reaction vessel was finally purged three times with nitrogen, d, J 8.0 Hz), 7.07 (1H, t, J8.0 Hz), 6.85 (1H, dd, J8.00.8 Hz), before filtration on a pad of Celite(R), which was washed with 6.55 (1H, dd, J 8.00.8 Hz), 5.67 (2H, s), 2.41 (3H, s) ethyl acetate. The organic Solution was washed with Saturated 0434. The compound below was prepared following the aqueous NaCO, followed by brine. The combined organic same general method. layers were dried over anhydrous MgSO and evaporated to afford 2.5 g (60%) of the title compound. 2-Phenyloxazolo 5,4-bipyridin-6-amine 0427 H NMR (DMSO): 8.02 (2H, d, J 8.2 Hz), 7.39 (3H, d.J8.5 Hz), 6.86 (1H, d, J2.0Hz), 6.65 (1H, dd, J 8.72.2 Hz), 0435 LCMS RT=5.41 min, MH 212.1; H NMR 5.09 (2H, s), 2.40 (3H, s) (DMSO): 8.19-8.15 (2H, m), 7.73 (1H, d, J24 Hz), 7.63-7.58 (3H, m), 7.32 (1H, d, J 2.4 Hz), 5.38 (2H, s) Method 2B (Compounds Ib) 0428. As Method 2A, except ethanol was used instead of Method 2E (Compounds Ib) ethyl acetate/acetic acid. After evaporation of the solvents, the material was taken up in 2M HCl, the resulting precipitate 2-p-Tolylbenzodoxazol-6-amine was discarded, and the solution was basified with 2N NaOH to afford the title compound as a precipitate. 0436 To 6-nitro-2-p-tolylbenzodoxazole (2.1 g, 8.27 mmol) in ethanol: water 2:1 V/v (60 mL) at 70° C. was added 2-Phenylbenzodoxazol-6-amine iron powder (2.14 g., 38.3 mmol) and ammonium chloride (819 mg, 15.3 mmol). The suspension was stirred at reflux for 0429 LCMS RT=5.93 min, MH 211.1; H NMR 16 h. After cooling, the solution was filtered through a pad of (DMSO): 8.10-8.07 (2H, m), 7.58-7.54 (3H, m), 7.42 (1H, d, CeliteR) and washed with ethanol. After evaporation of the J8.4 Hz), 6.83 (1H, d, J 1.9 Hz), 6.65 (1H, dd, J 8.52.0 Hz), Solvent, the aqueous layer was extracted twice with ethyl 5.46 (2H, s) acetate. The combined organic layers were dried over anhy Method 2C (Compounds Ib) drous MgSO and evaporated. The resulting solid was puri fied by column chromatography eluting with ethyl acetate: 2-(4-(Trifluoromethoxy)phenyl)benzodoxazol-5-amine hexanes 1:3 V/v to afford 70 mg (4%) of the title compound 0430. To 5-nitro-2-(4-(trifluoromethoxy)phenyl)benzod (LCMS RT=6.12 min, MH 223.1) oxazole (850 mg, 2.62 mmol) in ethanol (20 mL) was added 0437. HNMR (DMSO): 7.97 (2H, d, J8.1 Hz), 7.40-7.36 ammonium formate (827 mg, 13.1 mmol) and palladium on (3H, m), 6.82 (1H, d, J 1.9 Hz), 6.64 (1H, dd, J 8.5 2.0 Hz), carbon (85 mg). The mixture was stirred at room temperature 5.41 (2H, s), 2.39 (3H, s) for 20 min, then filtrated through a pad of Celite R, and 0438 All compounds below were prepared following the washed with ethyl acetate. The organic Solution was washed same general method. with water, followed by brine. The combined organic layers were dried over anhydrous MgSO and evaporated to afford 2-Phenethylbenzodoxazol-5-amine 434 mg (56%) of the title compound (LCMS RT=6.51 min, MH' 294.9) 0431 'HNMR (DMSO): 8.25 (2H, d, J 8.9 Hz), 7.62-7.56 0439 LCMS RT=5.82 min, MH 238.9; H NMR (2H, m), 7.44 (1H, d, J 8.7 Hz), 6.89 (1H, d, J 2.0 Hz), 6.70 (DMSO): 7.29-7.16 (6H, m), 6.76 (1H, d, J 1.9 Hz), 6.57 (1H, (1H, dd, J 8.72.3 Hz), 5.16 (2H, s) dd, J 8.62.2 Hz), 4.98 (2H, s), 3.19-3.06 (4H, m) Method 2D (Compounds Ib) 2-(Benzod1.3dioxol-5-yl)benzodoxazol-5-amine 2-p-Tolylbenzodoxazol-4-amine 0440 LCMS RT=5.77 min, MH 2549; H NMR 0432 To 4-nitro-2-p-tolylbenzodoxazole (330 mg, 1.30 (DMSO): 7.69 (1H, dd, J 8.2 1.7 Hz), 7.58 (1H, d, J 1.7 Hz), mmol) in ethanol (20 mL) was added tin (II) chloride (1.23g. 7.37 (1H, d, J 8.6 Hz), 7.11 (1H, d, J 8.2 Hz), 6.84 (1H, d, J 2.0 6.5 mmol). The suspension was stirred at 70° C. for 16 h. After Hz), 6.64 (1H, dd, J 8.8 2.2 Hz), 6.16 (2H, s), 5.07 (2H, s)

US 2011/0195932 A1 Aug. 11, 2011

N-(2-(4-Chlorophenyl)benzodoxazol-5-yl)-3-meth Hz), 7.55 (1H, dd, J 8.82.1 Hz), 7.46-7.41 (3H, m), 3.60(1H, oxypropanamide q, J 7.0 Hz), 1.41 (3H, d, J 7.0 Hz) 0635 LCMS RT=6.32 min, MH 331.1; H NMR Method 3G (Compounds II) (DMSO): 10.17 (1H, s), 8.22-8.17 (3H, m), 7.74-7.68 (3H, 0644 As Method 3C, except instead of the acid chloride, m), 7.54 (1H, dd, J 8.92.1 Hz), 3.65 (2H, t, J 6.1 Hz), 3.26 the corresponding anhydride was used (3H, s), 2.59 (2H, t, J 6.1 Hz) 2.2.2-Trifluoro-N-(2-p-tolylbenzodoxazol-5-yl) N-(2-(4-Chlorophenyl)benzodoxazol-5-yl)-3,3,3- acetamide trifluoropropanamide 0645 LCMS RT=6.93 min, MH 321.0: "H NMR 0636 LCMS RT=6.72 min, MH 354.7; H NMR (DMSO): 11.46 (1H, br), 8.22 (2H, d, J 8.6 Hz), 8.15 (1H, d, (DMSO): 10.52 (1H, s), 8.21 (2H, d, J 8.7 Hz), 8.14 (1H, d, J J 1.9 Hz), 7.85 (1H, d, J 8.8 Hz), 7.73-7.68 (3H, m) 2.0 Hz), 7.78 (1H, d, J 8.7 Hz), 7.70 (2H, d, J 8.7 Hz), 7.52 (1H, dd, J 8.82.1 Hz), 3.56 (2H, q, J 11.1 Hz) N-(2-(4-Chlorophenyl)benzodoxazol-5-yl)-2.2.2- trifluoroacetamide N-(2-(3,4-Dichlorophenyl)benzodoxazol-5-yl)-3,3, 0646 LCMS RT=7.12 min, WI340.8: "H NMR 3-trifluoropropanamide (DMSO): 11.43 (1H, br), 8.12-8.09 (3H, m), 7.82(1H, d, J8.8 0637 LCMS RT=7.41 min, MH 388.8: "H NMR Hz), 7.67 (1H, dd, J 8.92.1 Hz), 7.45 (2H, d, J 8.0 Hz) (DMSO): 10.54 (1H, s), 8.36 (1H, d, J 2.0 Hz), 8.17-8.14 (2H, m), 7.90 (1H, d, J8.4 Hz), 7.80 (1H, d.J8.8 Hz), 7.54 (1H, dd, Method 3H (Compounds II) J8.92.1 Hz), 3.56 (2H, q, J 11.1 Hz) 3-Morpholino-N-(2-phenylbenzodoxazol-5-yl)propanamide N-(2-(2,3-Dichlorophenyl)benzodoxazol-5-yl)-3,3, 0647. To 2-phenylbenzodoxazol-5-amine (75 mg, 0.36 3-trifluoropropanamide mmol) and methyl 3-morpholinopropanoate (63 uL, 0.39 mmol) in toluene (2.5 mL) was added a 2M solution of 0638 LCMS RT=6.76 min, MH 388.9; H NMR trimethylaluminum in toluene (0.22 mL, 0.43 mmol). The (DMSO): 10.55 (1H, s), 8.20 (1H, d, J 1.9 Hz), 8.12 (1H, dd, resulting solution was heated twice for 5 min at 160° C. in the J 7.9 1.6 Hz), 7.94 (1H, dd, J 8.1 1.6 Hz), 7.83 (1H, d, J 8.8 microwave. After cooling, sodium bicarbonate solution was Hz), 7.64-7.55 (2H, m), 3.58 (2H, q, J 11.1 Hz) added and the aqueous layer was extracted with ethyl acetate. 0639. The compounds below were obtained by Method The organic layer was then washed with brine and the com 3F, using the appropriate Boc-amino acid. Coupling was fol bined organic layers were dried over anhydrous MgSO. lowed by deprotection of Boc group using 4M HCl in dioxane After evaporation, the impurities were removed by filtering for 20 min at room temperature. through on a plug of silica eluting with ethyl acetate/hexanes 50:50 V/v and the desired product was obtained by elution with methanol to afford 17.5 mg (14%) of the title compound (S)-N-(2-(4-Chlorophenyl)benzodoxazol-5-yl) (LCMS RT=5.54 min, MH 352.0) pyrrolidine-2-carboxamide 0648 H NMR (DMSO): 10.25 (1H, s), 8.22-8.15 (3H, 0640 LCMS RT=4.54 min, MH 342.0; H NMR m), 7.73 (1H, d, J 8.8 Hz), 7.65-7.59 (3H, m), 7.52(1H, dd, J (DMSO): 10.15 (1H, s), 8.23 (1H, d, J 1.7 Hz), 8.19 (2H, d, J 8.8 2.0 Hz), 3.60-3.57 (4H, m), 2.68-2.64 (2H, m), 2.44-2.41 8.7 Hz), 7.74-7.63 (4H, m), 3.75-3.69 (1H, m), 2.91 (2H, t, J (4H, m) 6.5 Hz), 2.12-2.00 (1H, m), 1.90-1.60 (3H, m) Method 4 (Compounds III) (S)-N-(2-(4-Chlorophenyl)benzodoxazol-5-yl)-2- (methylamino)propanamide N-(2-Phenylbenzodoxazol-5-yl)propane-1-sulfonamide 0649. To a solution of 2-phenylbenzodoxazol-5-amine 0641 LCMS RT=446 min, MH 330.1; H NMR (100 mg 0.48 mmol) in dichloromethane (2 mL) at room (DMSO): 8.20-8.10 (3H, m), 7.67 (2H, d, J8.8 Hz), 7.58-7.47 temperature was added pyridine (83 uL, 0.95 mmol) followed (2H, m), 3.00 (1H, q, J 6.8 Hz), 2.24 (3H, s), 1.16 (3H, d, J 6.8 by propane-1-sulfonyl chloride (61 uL, 0.52 mmol). The Hz) resulting Solution was then stirred at room temperature for 16 0642. The compound below was obtained by Method 3F, h. Dichloromethane was added and the organic layer was using the appropriate Fmoc-amino acid. Coupling was fol washed with Saturated aqueous copper Sulfate solution. The lowed by deprotection of the Fmoc group using piperidine/ combined organic layers were dried over anhydrous MgSO DMF 20:80 VfV. and evaporated. The resulting insoluble solid was washed with saturated aqueous NaHCO to afford 37.2 mg (25%) of (S)-2-Amino-N-(2-(4-chlorophenyl)benzodoxazol the title compound (LCMS RT=6.25 min, MH 317.0) 5-yl)propanamide 0650 "H NMR (DMSO): 9.89 (1H, br), 8.21-8.18 (2H, m), 7.77 (1H, d, J 8.8 Hz), 7.66-7.59 (4H, m), 7.28 (1H, dd, J 0643 LCMS RT=446 min, MH 316.1; H NMR 8.82.1 Hz), 3.10-3.04 (2H, m), 1.77-1.64 (2H, m), 0.94 (3H, (CDC1): 9.56 (1H, s), 8.11 (2H, d, J 8.7 Hz), 7.98 (1H, d, J 1.9 t, J 7.5 Hz)

US 2011/0195932 A1 Aug. 11, 2011 d, J 2.1 Hz), 6.71 (1H, dd, J 8.82.3 Hz), 5.70 (1H, t, J5.7 Hz), 0676) "H NMR (CDC1): 8.13 (2H, d, J8.6Hz), 7.55-7.52 2.86 (2H, t, J 6.3 Hz), 2.40 (3H, s), 1.95-1.81 (1H, m), 0.97 (2H, m), 7.46 (2H, d, J 8.6 Hz), 7.13 (1H, dd, J 8.3 2.0 Hz), (6H, d, J 6.7 Hz) 3.26 (3H, s), 2.04 (2H, q, J 7.6 Hz), 1.00 (3H, t, J 7.6 Hz) Method 7 (Compounds V1b) N-Benzyl-2-p-tolylbenzodoxazol-5-amine N-(2-(4-Chlorophenyl)benzodoxazol-5-yl)-N- 0669 LCMS RT=7.95 min, MH 315.1; H NMR methyisobutyramide (DMSO): 8.00 (2H, d, J 8.1 Hz), 7.46-7.21 (8H, m), 6.77-6.73 0677 LCMS RT=7.44 min, MH 329.1; H NMR (2H, m), 6.37 (1H, t, J 6.4 Hz), 4.32 (2H, d, J 6.0 Hz), 2.40 (DMSO): 8.22 (2H, d, J 8.7 Hz), 7.91-7.87 (2H, m), 7.72 (2H, (3H, s) d, J 8.6 Hz), 7.43 (1H, d, J 8.3 Hz), 3.20 (3H, s), 2.46-2.42 (1H, m), 0.93 (6H, d, J 6.6 Hz) 2-(4-Chlorophenyl)-N,N-diisobutylbenzodoxazol 2-(4-Chlorophenyl)-N-isobutyl-N-methylbenzod 5-amine Oxazol-5-amine 0670 LCMS RT=17.03 min, MH 357.1; H NMR 06781 LCMS RT=10.59 min, MH 315.1; H NMR (DMSO): 8.26 (2H, d, J 8.5 Hz), 7.80 (2H, d, J 8.6 Hz), 7.67 (DMSO): 8.15 (2H, d, J 8.6 Hz), 7.67 (2H, d, J 8.6 Hz), 7.56 (1H, d, J 9.0 Hz), 7.09 (1H, d, J2.3 Hz), 6.96 (1H, dd, J9.12.4 (1H, d, J9.0Hz), 6.96 (1H, d, J2.5 Hz), 6.84 (1H, dd, J9.12.5 HZ), 3.32 (4H, d, J 7.2 Hz), 2.20-2.10 (2H, m), 1.02 (12H, d, Hz), 3.17 (2H, d, J 7.3 Hz), 2.97 (3H, s), 2.13-197 (1H, m), J 6.6 Hz) 0.90 (6H, d, J 6.7 Hz) Method 8 (Compounds VII) Method 6 (Compounds V) N-(2-(4-Chlorophenyl)benzodoxazol-5-yl)-2- 1-Phenyl-3-(2-phenylbenzodoxazol-5-yl)urea methylpropanethioamide 0679 To N-(2-(4-chlorophenyl)benzodoxazol-5-yl) 0671 To 2-phenylbenzodoxazol-5-amine (75 mg, 0.36 isobutyramide (50 mg, 0.16 mmol) in toluene (2 mL) at 110° mmol) in dichloromethane (2 mL) at room temperature was C. was added Lawesson's reagent (35 mg, 0.09 mmol). The added phenyl isocyanate (43 uL, 0.39 mmol). The solution resulting solution was heated at 110°C. for 7h. After cooling, was stirred at room temperature for 16 h. The resulting pre the solution was diluted with water and extracted with ethyl cipitate was filtered off and washed with dichloromethane to acetate. The combined organic layers were washed with afford 99.9 mg (85%) of the title compound (LCMSRT=6.45 brine, then dried over anhydrous MgSO and evaporated. The min, MH 330.1) resulting solid was purified by column chromatography elut 0672 H NMR (DMSO): 8.85 (1H, s), 8.71 (1H, s), 8.22 ing with ethyl acetate/hexanes 20:80 V/v to afford 13.8 mg 8.19 (2H, m), 8.00 (1H, d, J 2.0 Hz), 7.71 (1H, d, J 8.9 Hz), (26%) of the title compound (LCMS RT=7.36 min, MH 331. 7.65-7.60 (3H, m), 7.50-7.47 (2H, m), 7.40 (1H, dd, J 8.82.1 2) Hz), 7.30 (2H, t, J 8.4 Hz), 7.02-6.95 (1H, m) 0680 "H NMR (DMSO): 11.59 (1H, s), 8.37 (1H, d, J 2.0 0673. The compound below was prepared following the Hz), 8.22 (2H, d, J 8.8 Hz), 7.83 (1H, d, J 8.7 Hz), 7.73-7.65 same general method. (3H, m), 3.18-3.09 (1H, m), 1.25 (6H, d, J 6.7 Hz) Method 9 (Compound VIII) 1-Isopropyl-3-(2-phenylbenzodoxazol-5-yl)urea 2-(4-Chlorophenyl)benzodoxazole-5-diazonium 0674) LCMS RT=5.94 min, MH 296.0; H NMR tetrafluoroboric acid salt (DMSO): 8.47 (1H, s), 8.20-8.16 (2H, m), 7.93 (1H, d, J 1.9 0681 To a solution of 2-(4-chlorophenyl)benzodoxazol Hz), 7.64-7.59 (4H, m), 7.30 (1H, dd, J 8.82.1 Hz), 6.03 (1H, 5-amine (500 mg, 2.04 mmol) in water (3 mL) and tetrafluo d, J 7.5 Hz), 3.85-3.74 (1H, m), 1.12 (6H, d, J 6.5 Hz) roboric acid (50% in water, 2 mL) at 0° C. was added a solution of sodium nitrite (140 mg, 2.04 mmol) in water (2 mL), dropwise over 5 min. The resulting mixture was stirred Method 7 (Compounds VI) at 0°C. for 15 min, and then at room temperature for 1 h. The N-(2-(4-Chlorophenyl)benzodoxazol-5-yl)-N- solid was then filtered off, washed with dilute aqueous tet methylpropionamide rafluoroboric acid solution and methanol to afford 370 mg (53%) of the title compound, which was used directly without 0675 To sodium hydride (15 mg, 0.37 mmol) under nitro characterization. gen at 0°C. was slowly added a solution of N-(2-(4-chlo Method 10 (Compound IX) rophenyl)benzodoxazol-5-yl)propionamide (100 mg, 0.33 mmol) in dimethylformamide (10 mL). After 10 min at 0°C., S-2-(4-Chlorophenyl)benzodoxazol-5-yl ethanethioate methyl iodide (56 uL, 0.37 mmol) was added, and the solution 0682 To a stirred solution of potassium thioacetate (130 was left warming up to room temperature for 16 h. The mg, 1.13 mmol) in DMSO (2.8 mL) at room temperature was mixture was then diluted with ethyl acetate, and then washed added dropwise a solution of 2-(4-chlorophenyl)benzodox three times with water. The combined organic layers were azole-5-diazonium tetrafluoroboric acid salt (370 mg, 1.08 dried over anhydrous MgSO and evaporated. The resulting mmol) in DMSO (1.4 mL). After 15 min, the mixture was Solid was purified by column chromatography eluting with heated at 70° C. for 1 h. After cooling, the mixture was diluted ethyl acetate/hexanes 50:50 v/v to afford 36.4 mg (35%) of with water, and then extracted several times with ethyl the title compound (LCMS RT=7.00 min, MH"315.0) acetate. The combined organic layers were washed with

US 2011/0195932 A1 Aug. 11, 2011

2-(3',4'-Dichlorophenyl)-5-(1'-hydroxyethyl)-ben Zoxazole 0742 LCMS RT=7.18 min, MH 308.1; H NMR (DMSO): 8.36 (1H, d, J 2.0 Hz), 8.16 (1H, dd, J 8.5 2.0 Hz), 7.90 (1H, d, J 8.4 Hz), 7.78-7.77 (1H, m), 7.74 (1H, d, J 8.4 Hz), 7.47 (1H, dd, J 8.6 1.7 Hz), 5.32 (1H, d, J 4.3 Hz), 4.93-4.84 (1H, m), 1.39 (3H, d, J 6.4 Hz)

OH

ON

Pyridine, 120° C.,6h then rt, 16 h

RR2NH, THF, MW, RSnBus, 150° C., 15 min O O CI-P'sa P's O S-e- X-NR,R, O NX-R- Pd(PPh3)4Dioxane, - Method 21 N Method 16 ON 100° C., 16 h O2N ON H R N XXI Method 22 XX XV R = NO2 Method XVI R = NH. A 17a or 17b

1-(bromomethyl)-4-chlorobenzene NEt3, chloroform, 60° C., 2h Method 18 OX 7-O- XVII R=NO ) Iron, NHCl, ethanol/water XVII R = NH2 90° C., 4h Method 19

RCOCl, pyridine 16 h, rt Method 20 uC2 XIX

5-Nitrobenzodoxazole-2(3H)-thione water and aqueous hydrochloric acid was added. The result ing precipitate was collected by filtration, washed with dilute 0743. In accordance with well known procedures, (see aqueous hydrochloric acid, followed by water and then dried Batista-Parra, A et at Heterocycles, 2003, 60, 1367), a sus pension of 2-amino-4-nitrophenol (1.54 g. 10 mmol) and in the vacuum oven to afford 3.3 g (84%) of the title com potassium O-ethyl carbonodithioate (1.68 g, 10.5 mmol) in pound. dry pyridine (10 mL) was stirred at 120° C. for 6 h, and then 0744 H NMR (DMSO): 8.18 (1H, dd, J 8.92.4 Hz), 7.94 at room temperature for 16 h. The solution was poured into (1H, dd, J 2.40.4 Hz), 7.73 (1H, dd, J 8.9 0.4 Hz) US 2011/0195932 A1 Aug. 11, 2011 62

Method 16 (Compound XV) (0752 H NMR (DMSO): 8.52(1H, d, J 2.3 Hz), 8.27(1H, dd, J 8.92.3 Hz), 7.92(1H, d, J9.0Hz), 7.58 (2H, d, J8.5 Hz), 2-Morpholino-5-nitrobenzodoxazole 7.42 (2H, d, J 8.5 Hz), 4.67 (2H, s) 0745 5-nitrobenzodoxazole-2(3H)-thione (98.1 mg 0.5 mmol) and morpholine (66 uL, 0.75 mmol) in tetrahydrofu Method 19 (Compound XVIII) ran (3 mL) were heated at 150° C. for 15 min in the micro 2-(4-Chlorobenzylthio)benzodoxazol-5-amine wave. After cooling, the mixture was poured into water and extracted with ethyl acetate. The organic layer was washed 0753. To a suspension of 2-(4-chlorobenzylthio)-5-ni with brine, dried over anhydrous MgSO and absorbed on trobenzodoxazole (220 mg. 0.70 mmol) in ethanol/water (5 silica. Purification by column chromatography, eluting with mL/5 mL) at 90° C. was added ammonium chloride (75 mg. ethyl acetate/hexanes 25:75 v/v affords 115 mg (92%) of the 1.4 mmol), followed by iron powder (192 mg, 3.44 mmol). title compound. The resulting mixture was stirred for 4 h at 90° C. After cooling, ethyl acetate was added and the Solution was passed 0746 H NMR (DMSO): 8.10 (1H, d, J 2.3 Hz), 8.00 (1H, through a pad of Celite(R). The organic layer was then washed dd, J 8.8 2.4 Hz), 7.66 (1H, d, J 8.8 Hz), 3.76-3.72 (4H, m), with brine, dried over anhydrous MgSO and evaporated to 3.67-3.64 (4H, m) afford 190 mg (94%) of the title compound (LCMS RT=6.54 min, MH 290.9) Method 17a (Compound XVI) (0754 H NMR (DMSO): 7.52(2H, d, J 8.7 Hz), 7.40 (2H, 2-Morpholinobenzodoxazol-5-amine d, J 8.5 Hz), 7.26 (1H, d, J8.7 Hz), 6.74 (1H, d, J 1.9 Hz), 6.54 (1H, dd, J 8.72.2 Hz), 5.06 (2H, s), 4.55 (2H, s) 0747 A solution of 2-morpholino-5-nitrobenzodox azole (130 mg, 0.52 mmol) in ethanol/water 1:1 V/v (10 mL) Method 20 (Compounds XIX) was treated with sodium dithionite (182 mg, 1.04 mmol) at room temperature and then refluxed for 16 h. After cooling, N-(2-(4-Chlorobenzylthio)benzodoxazol-5-yl)acetamide the mixture was diluted with water and extracted with ethyl 0755. To a solution of 2-(4-chlorobenzylthio)benzodox acetate. The organic layer was dried over anhydrous MgSO azol-5-amine (87 mg, 0.30 mmol) in dry pyridine (3 mL) at and evaporated to afford 35 mg (30%) of the title compound. room temperature was added acetyl chloride (21 uL, 0.30 0748 H NMR (DMSO): 7.03 (1H, d, J 8.5 Hz), 6.50 (1H, mmol). The resulting solution was stirred at room tempera d, J 2.1 Hz), 6.25 (1H, dd, J 8.4 2.2 Hz), 4.80 (2H, s), 3.71 ture for 16 h. Water was then added, the precipitate was 3.68 (4H, m), 3.53-3.50 (4H, m) collected by filtration, washed with diluted aqueous hydro chloric acid and then with water. Trituration with diethyl ether Method 17b (Compound XVI) afforded 40 mg (40%) of the title compound (LCMSRT=6.40 min, MH 333.1) N-2-(4-Chlorobenzyl)benzodoxazole-2,5-diamine (0756 H NMR (DMSO): 10.08 (1H, s), 8.01 (1H, d, J 1.8 0749. To a suspension of N-(4-chlorobenzyl)-5-ni Hz), 7.58-7.52 (3H, m), 743-7.35 (3H, m), 4.60 (2H, s), 2.06 trobenzodoxazol-2-amine (150 mg. 0.50 mmol) in ethanol/ (3H, s) water 1:1 V/v (10 mL) at 90° C. was added ammonium chlo 0757. The compound below was prepared following the ride (53 mg, 1.0 mmol), followed by ironpowder (140 mg, 2.5 same general method. mmol). The resulting mixture was stirred for 4 h at 90° C. After cooling, ethyl acetate was added and the solution was N-(2-(4-Chlorobenzylthio)benzodoxazol-5-yl) passed through a pad of Celite(R). The organic layer was then isobutyramide washed with brine, dried over anhydrous MgSO and evapo 0758 LCMS RT=7.00 min, MH 361.1; H NMR rated. The resulting Solid was purified by column chromatog (DMSO): 9.96 (1H, s), 8.04 (1H, d, J 1.8 Hz), 7.58-7.52 (3H, raphy eluting with ethyl acetate/hexanes 50:50 V/v to afford m), 7.43-7.39 (3H, m), 4.60 (2H, s), 2.65-2.60 (1H, m), 1.12 60 mg (44%) of the title compound (LCMS RT=5.60 min, (6H, d, J 6.8 Hz) MH 274.0) (0750 "H NMR (DMSO): 8.20(1H, t, J 6.2 Hz), 7.42-7.36 Method 21 (Compound XX) (4H, m), 6.97 (1H, d, J 8.4 Hz), 6.45 (1H, d, J 2.1 Hz), 6.20 (1H, dd, J 8.4 2.2 Hz), 4.72 (2H, s), 4.46 (2H, d, J 6.2 Hz) 2-Chloro-5-nitrobenzodoxazole (0759. To a solution of 5-nitrobenzodoxazole-2(3H)- Method 18 (Compound XVII) thione (2.52 g, 12.86 mmol) in phosphorous oxychloride (21 2-(4-Chlorobenzylthio)-5-nitrobenzodoxazole mL) was added phosphorous pentachloride (2.68 g, 12.86 mmol) in one portion. The mixture was then heated to 100° C. 0751. To a suspension of 5-nitrobenzodoxazole-2(3H)- for 2.5 h. After cooling, the excess of phosphorous oxychlo thione (196.2 mg, 1.0 mmol) in chloroform (10 mL) was ride was removed in vacuo and the resulting mixture was used added triethylamine (278 uL., 2 mmol) followed by 1-(bro crude without characterisation. momethyl)-4-chlorobenzene (226 mg, 1.1 mmol). The reac tion was stirred at 60°C. for 2 h. After cooling, the reaction Method 22 (Compound XXI) was diluted with ethyl acetate, washed with dilute aqueous 5-Nitro-2-(thiophen-2-yl)benzodoxazole hydrochloric Solution and brine. The combined organic layers were dried over anhydrous MgSO and evaporated. The 0760. A mixture of 2-chloro-5-nitrobenzodoxazole (404 resulting solid was purified by column chromatography elut mg, 2.04 mmol), 2-(tributylstannyl)-thiophene (648 uL 2.04 ing with ethyl acetate/hexanes 1:10 V/v to afford 250 mg mmol) and tetrakis(triphenylphosphine)palladium (0) (40.8 (78%) of the title compound (LCMS RT-7.64 min) mg) in dioxane (12.2 mL) was heated at 100° C. for 16 hunder US 2011/0195932 A1 Aug. 11, 2011 nitrogen. Ethyl acetate was added, the organic layer was 2-(6-Nitro-1H-benzodimidazol-2-yl)phenol washed with water, dried over anhydrous MgSO and evapo 0766 LCMS RT=6.48 min, MH 256.0; H NMR rated. The resulting Solid was purified by column chromatog (DMSO): 13.50 (1H, br), 12.40 (1H, br), 8.56 (1H, s), 8.20 raphy eluting with ethyl acetate/hexanes 10:90 V/V, and then 8.14 (2H, m), 7.84 (1H, d, J 8.8 Hz), 7.48-743 (1H, m), purified by reverse phase HPLC to afford 3 mg (2%) of the 7.12-7.04 (2H, m) title product (LCMS RT=6.95 min) 0761 'H NMR (DMSO): 8.54 (1H, d, J 2.2 Hz), 8.27 (1H, 2-(4-Chlorophenyl)-6-nitro-1H-benzodimidazole dd, J9.02.3 Hz), 8.04 (1H, dd, J5.41.2 Hz), 7.93 (1H, d, J 9.0 0767 LCMS RT=6.24 min, MH 273.9; H NMR Hz), 7.69 (1H, dd, J 3.7 1.2 Hz), 7.29 (1H, dd, J 5.43.7 Hz) (DMSO): 13.80 (1H, br), 8.54 (1H, d, J 2.1 Hz), 8.29 (2H, d, J8.6 Hz), 8.21 (1H, dd, J 8.92.2 Hz), 7.84 (1H, d, J 8.9 Hz), 7.75 (2H, d, J 8.5 Hz) NH2 R PPA, 130° C., 16 h 21 9 (Method 23) Method 24 (Compound XXIIb) R-H + O. ={ 2-(5-Amino-1H-benzodimidazol-2-yl)phenol N NH2 OH H 0768 To 2-(5-nitro-1H-benzodimidazol-2-yl)phenol 21 N (90 mg, 0.35 mmol) in ethyl acetate/waterfacetic acid 1:1:0. 01 V/v/v (10 mL) was added palladium on carbon (15 mg). R-H X- Ro The reaction vessel was purged three times with nitrogen, N / followed by three times with hydrogen, and then left stirring XXII under hydrogen for 2 h. The reaction vessel was finally XXIIa R = NO2 ) (Method 24) purged three times with nitrogen, before filtration on a pad of XXIIb R = NH2 Celite R, which was washed with ethyl acetate. The organic layer was washed with saturated aqueous NaHCO. The com bined organic layers were dried over anhydrous MgSO and so16 h (Method pyridine, 25) rt, evaporated. The resulting solid was purified by column chro matography eluting with ethyl acetate/hexanes 2:1 V/v to 21 afford 60 mg (76%) of the title compound (LCMS RT=5.39 min, MH" 226.1) N 0769 H NMR (DMSO): 13.28 (1H, br), 12.63 (1H, br), 7.72 (1H, d, J 8.4 Hz), 7.33-7.28 (2H, m), 6.99-6.94 (2H, m), o- N 6.71-6.58 (2H, m), 5.12 (2H, s) XXIII Method 25 (Compounds XXIII) N-(2-p-Tolyl-1H-benzodimidazol-5-yl)butyramide Method 23 (Compounds XXII) 0770. To a solution of 2-p-tolyl-1H-benzodimidazol-5- amine (150 mg. 0.67 mmol) in pyridine (10 mL) at room 5-Amino-2-(5,6-dimethyl-1H-benzodimidazol-2-yl)phenol temperature was added butyryl chloride (77 uL, 0.74 mmol). The resulting mixture was stirred at room temperature for 16 0762. To polyphosphoric acid at 130°C. were added 4.5- h. Ethyl acetate was added and the organic layer was washed dimethylbenzene-1,2-diamine (500 mg, 3.67 mmol) and twice with Saturated aqueous copper Sulfate, followed by 4-amino-2-hydroxybenzoic acid (562 mg, 3.67 mmol), and Sodium bicarbonate and brine. The combined organic layers the resulting mixture was then heated to 130° C. for 16 h. The were dried over anhydrous MgSO and evaporated. The Solution was then poured into water and the resulting precipi resulting solid was purified by column chromatography elut tate was dissolved in ethyl acetate and washed with NaCO. ing with ethyl acetate/hexanes 50:50 V/v to afford 56 mg The aqueous layer was separated and extracted twice with (28%) of the title compound (LCMS RT=6.96 min, MH" ethyl acetate. The combined organic layers were dried over 294.0) anhydrous MgSO and evaporated. The resulting Solid was (0771 'H NMR (DMSO): 12.68 (1H, br), 9.87 (1H, s), purified by column chromatography eluting with ethyl 8.08-8.00 (3H, m), 7.52-7.20 (4H, m), 2.38 (3H, s), 2.31 (2H, acetate/hexanes 50:50 v/v to afford 200 mg (28%) of the title t, J 7.3 Hz), 1.70- 1.58 (2H, m), 0.94 (3H, t, J 7.4 Hz) compound (LCMS RT=6.14 min, MH" 254.1) 0772 All compounds below were prepared following the 0763 H NMR (DMSO): 13.07 (1H, s), 12.40 (1H, s), same general method. 7.61 (1H, d, J8.3 Hz), 7.35 (1H, s), 7.24 (1H, s), 6.19 (1H, dd, N-(2-p-Tolyl-1H-benzodimidazol-5-yl)isobutyra J8.52.2 Hz), 6.12(1H, d, J 2.1 Hz), 5.59 (2H, s), 2.32 (3H, s), mide 2.30 (3H, s) 0773) LCMS RT=5.43 min, MH 294.1; H NMR 0764 All compounds below were prepared following the (DMSO): 12.67 (1H, br), 9.91 (1H, s), 8.08-8.01 (3H, m), same general method. 7.50-728 (4H, m), 4.03 (1H, q, J 7.2 Hz), 2.38 (3H, s), 1.13 (6H, d, J 6.8 Hz) 2-(3-Methyl-4-nitrophenyl)-1H-benzodimidazole N-(2-(4-Chlorophenyl)-1H-benzodimidazol-5-yl) 0765 LCMS RT=5.87 min, MH 254.1; H NMR butyramide (DMSO): 13.20 (1H, br), 9.31 (1H, s), 8.21-8.19 (2H, m), 0774 LCMS RT=5.54 min, MH 314.0; H NMR 7.67-7.64 (2H, m), 7.28-7.25 (2H, m), 2.65 (3H, s) (DMSO): 12.85 (1H, br), 9.90 (1H, s), 8.14 (3H, d, J 8.6 Hz), US 2011/0195932 A1 Aug. 11, 2011 64

7.62 (2H, d, J 8.7 Hz), 7.53 (1H, br), 7.25 (1H, br), 2.32 (2H, washed with 2M aqueous hydrochloric acid and then brine. t, J 7.1 Hz), 1.71-1.58 (2H, m), 0.94 (3H, t, J 7.5 Hz) The combined organic layers were dried over anhydrous MgSO and evaporated to afford 1.1 g (33%) of the title N-(2-Phenyl-1H-benzodimidazol-5-yl)isobutyramide compound. 0775 LCMS RT=5.32 min, MH 280.0; H NMR (0779) 'H NMR (DMSO): 7.61-7.55 (2H, m), 7.47 (1H, d, (DMSO): 12.80 (1H, br), 9.85 (1H, s), 8.16-8.09 (3H, m), J 8.2 Hz), 7.31 (3H, s) 7.58-7.47 (4H, m), 7.28 (1H, d, J 8.1 Hz), 2.68-2.60 (1H, m), 1.13 (6H, d, J 6.9 Hz) Method 27 (Compound XXV) N-(2-Phenyl-1H-benzodimidazol-5-yl)butyramide 2-(2-Chlorophenyl)-5-nitrobenzodthiazole 0776 LCMS RT=5.32 min, MH 280.0; H NMR (DMSO): 12.77 (1H, br), 9.90 (1H, s), 8.15-8.12 (3H, m), 0780 2-Amino-4-nitrobenzenethiol (315 mg, 1.85 7.58-7.45 (4H, m), 7.26 (1H, br), 2.31 (2H, t, J 7.1 Hz), mmol), 2-chlorobenzoic acid (290 mg, 1.85 mmol) and 1.71-1.58 (2H, m), 0.94 (3H, t, J 7.5 Hz) Eaton's reagent (5 mL) were combined and heated in the microwave at 130° C. for 10 min. After cooling, the mixture N-(2-(4-Chlorophenyl)-1H-benzodimidazol-5-yl) was poured into water and basified with 5M aqueous sodium isobutyramide hydroxide to give a precipitate, which was filtered off and 0777 LCMS RT=5.71 min, MH 314.0; H NMR dried to afford 530 mg (98%) of the title compound. (DMSO): 12.89 (1H, br), 9.86 (1H, s), 8.18-8.10 (3H, m), 0781 'H NMR (DMSO): 8.93 (1H, d, J 2.2 Hz), 8.53 (1H, 7.62 (2H, d, J 8.8 Hz), 7.52(1H, d, J 8.8 Hz), 7.29 (1H, d, J8.3 d, J 8.9 Hz), 8.37 (1H, dd, J8.92.2 Hz), 8.29 (1H, dd, J 7.41.9 Hz), 2.66-2.60 (1H, m), 1.13 (6H, d, J 6.9 Hz) Hz), 7.78-7.75 (1H, m), 7.70-7.60 (2H, m)

C C -- 1. ON NH O Ro ON

Pyridine, rt, 16h Na2S-9H2O, NaHCO3, H2O Method 28 microwave, 5 min, 125°C. Method 26

C SH

ON NH ON NH2 1. Ro XXVI XXIV

Na2S-9H2O, S OH Ethanol, 85°C., 3 h Method 29 1. R9 Eaton's reagent, microwave, 10 min, 130°C., Method 27 O S -- R * Ethanol/Water (2:1 v/v), Pyridine, rt, 16 h-48 h. X-R, 1 h, 80° C. Method 31 oss N Method 30 H XXVIII

Method 26 (Compound XXIV) Method 28 (Compound XXVI) 2-Amino-4-nitrobenzenethiol N-(2-Chloro-5-nitrophenyl)-4-methylbenzamide 0778 2-Fluoro-5-nitroaniline (1 g, 6.41 mmol), sodium 0782. To 2-chloro-5-nitroaniline (2 g, 11.59 mmol) in sulfide nonahydrate (1.7g, 7.05 mmol), sodium bicarbonate pyridine (5 mL) at room temperature was added 4-methyl (600 mg, 7.05 mmol) and water (15 mL) were combined and benzoyl chloride (1.6 mL, 12.17 mmol), followed by pyridine heated in the microwave at 125° C. for 5 min. After cooling, (5 mL). The mixture was then stirred at room temperature for dichloromethane was added, and the organic layer was 16 h. Ethyl acetate was then added to the solution to give a US 2011/0195932 A1 Aug. 11, 2011

precipitate, which was filtered off and washed twice with (1H, d, J 8.6 Hz), 7.64-7.55 (2H, m), 7.19 (1H, d, J 1.7 Hz), ethyl acetate, and then hexanes. The resulting Solid was then 6.82 (1H, dd, J 8.62.2 Hz), 5.37 (2H, s) washed with aqueous sodium bicarbonate, 1M aqueous Sodium hydroxide, water and hexanes to afford (1.4g, 42%) 2-(3,4-Dichlorophenyl)benzodthiazol-5-amine of the title compound. 0783 H NMR (DMSO): 10.26 (1H, s), 8.58 (1H, d, J 2.8 0793 LCMS RT=7.49 min, MH 294.9; H NMR Hz), 8.11 (1H, dd, J 8.9 2.8 Hz), 7.93 (2H, d, J 8.2 Hz), 7.87 (DMSO): 8.24 (1H, d, J 2.1 Hz), 8.00 (1H, dd, J 8.42.1 Hz), (1H, d, J 8.9 Hz), 7.38 (2H, d, J 8.0 Hz), 2.41 (3H, s) 7.82(1H, d, J 8.4 Hz), 7.76 (1H, d, J 8.6 Hz), 7.20(1H, d, J 1.9 Method 29 (Compound XXV) Hz), 6.84 (1H, dd, J 8.62.2 Hz), 5.41 (2H, s) 5-Nitro-2-p-tolylbenzodthiazole 2-(2,3-Dichlorophenyl)benzodthiazol-5-amine 0784 Sodium sulfide nonahydrate (875 mg, 3.78 mmol) and sulfur (120 mg, 3.78 mmol) were heated until molten. 0794. LCMS RT=7.00 min, MH 294.9; H NMR The water was driven off with nitrogen to give a solid. The (DMSO): 8.08 (1H, dd, J 7.9 1.6 Hz), 7.84 (1H, dd, J 8.01.6 obtained solid was added in portions to N-(2-chloro-5-nitro Hz), 7.78 (1H, d, J 8.6 Hz), 7.55 (1H, t, J 8.0 Hz), 7.22(1H, d, phenyl)-4-methylbenzamide (1 g, 3.44 mmol) in ethanol (20 J 2.0 Hz), 6.87 (1H, dd, J 8.62.2 Hz), 5.38 (2H, s) mL) at 85°C. The solution was stirred at 85°C. for 3 h. After cooling, 2M aqueous HCl was added, and the solution was Method 31 (Compounds XXVIII) extracted three times with ethyl acetate. The combined N-(2-p-Tolylbenzodthiazol-5-yl)butyramide organic layers were dried over anhydrous MgSO and evapo rated. The resulting Solid was purified by column chromatog 0795 To a solution of 2-p-tolylbenzodthiazol-5-amine raphy eluting with ethyl acetate/hexanes 10:90 V/v to afford (110 mg, 0.46 mmol) in pyridine (3 mL) at room temperature 400 mg (43%) of the title compound. was added butyrylchloride (53 uL, 0.50 mmol). The resulting 0785 H NMR (DMSO): 8.81 (1H, d, J 2.2 Hz), 8.45 (1H, mixture was stirred at room temperature for 2 days. Ethyl d.J8.8 Hz), 8.29 (1H, dd, J8.82.3 Hz), 8.06 (2H, d, J 8.2 Hz), acetate was added and the organic layer was washed with 7.44 (2H, d, J 8.0 Hz), 2.42 (3H, s) saturated aqueous copper Sulfate, followed by aqueous sodium bicarbonate and finally with brine. The combined Method 30 (Compounds XXVII) organic layers were dried over anhydrous MgSO and evapo 2-p-Tolylbenzodthiazol-5-amine rated. The resulting Solid was purified by column chromatog 0786 5-Nitro-2-p-tolylbenzodthiazole (400 mg, 1.48 raphy eluting with ethyl acetate/hexanes 50:50 V/v to afford mmol) was suspended in ethanol/water (8 mL/4 mL) and 25 mg (18%) of the title compound (LCMS RT=7.10 min, heated at 80° C. Ammonium chloride (160 mg, 2.96 mmol) MH311.0) and iron powder (414 mg, 7.40 mmol) were added to the suspension, and the mixture was left stirring at 80°C. for 75 0796 H NMR (DMSO): 10.12 (1H, s), 8.43 (1H, d, J 1.8 min. After cooling, the solution was filtrated through a pad of Hz), 8.02-7.96 (3H, m), 7.58 (1H, dd, J 8.62.0 Hz), 7.38 (2H, CeliteR) and the pad washed with ethanol. Water was added to d, J8.0Hz), 2.40 (3H, s), 2.35 (2H, t, J 7.5 Hz), 1.72-1.60(2H, the filtrate, ethanol was evaporated and the remaining aque m), 0.95 (3H, t, J 7.4 Hz) ous layer was extracted with ethyl acetate. The combined 0797 All compounds below were prepared following the organic layers were dried over anhydrous MgSO and evapo same general method. rated to afford 220 mg (62%) of the title compound (LCMS RT=6.51 min, MH 241.0) N-(2-p-Tolylbenzodthiazol-5-yl)isobutyramide 0787 "H NMR (DMSO): 7.91 (2H, d, J 8.1 Hz), 7.68 (1H, 0798 LCMS RT=7.06 min, MH 311.0: "H NMR d, J8.6Hz), 7.35 (2H, d, J8.0Hz), 7.15 (1H, d, J 2.0 Hz), 6.77 (DMSO): 10.08 (1H, s), 8.44 (1H, d, J 1.3 Hz), 8.03-7.96 (3H, (1H, dd, J 8.6 2.0 Hz), 5.32 (2H, s), 2.39 (3H, s) m), 7.60 (1H, dd, J 8.7 1.6 Hz), 7.38 (2H, d, J 8.0 Hz), 0788 All compounds below were prepared following the 2.69-2.60 (1H, m), 2.40 (3H, s), 1.15 (6H, d, J 6.8 Hz) same general method. 2-Phenylbenzodthiazol-5-amine N-(2-Phenylbenzodthiazol-5-yl)isobutyramide 0789 LCMS RT=6.11 min, MH 227.1; H NMR 0799 LCMS RT=6.56 min, MH 297.0; H NMR (DMSO): 8.04-8.01 (2H, m), 7.71 (1H, d, J8.5 Hz), 7.56-7.53 (DMSO): 10.07 (1H, s), 8.47 (1H, d, J 1.8 Hz), 8.11-8.07 (2H, (3H, m), 7.18 (1H, d, J 2.1 Hz), 6.79 (1H, dd, J 8.62.3 Hz), m), 8.04 (1H, d, J 8.6 Hz), 7.64-7.56 (4H, m), 2.70-2.61 (1H, 5.33 (2H, s) m), 1.15 (6H, d, J 6.8 Hz) 2-(4-Chlorophenyl)benzodthiazol-5-amine N-(2-(4-Chlorophenyl)benzodthiazol-5-yl)isobu 0790 LCMS RT=6.72 min, MH 260.7; H NMR tyramide (DMSO): 8.04 (2H, d, J 8.7 Hz), 7.72 (1H, d, J 8.6 Hz), 7.61 (2H, d, J 8.7 Hz), 7.17 (1H, d, J20Hz), 6.80 (1H, dd, J8.62.2 0800 LCMS RT=7.42 min, MH 331.0; H NMR Hz), 5.35 (2H, s) (DMSO): 10.08 (1H, s), 8.47 (1H, d, J 1.9 Hz), 8.10 (2H, d, J 2-(2-Chlorophenyl)benzodthiazol-5-amine 8.6 Hz), 8.05 (1H, d, J 8.7 Hz), 7.67-7.61 (3H, m), 2.70-2.60 (1H, m), 1.15 (6H, d, J 6.8 Hz) 0791) LCMS RT=6.49 min, MH 260.8: "H NMR (DMSO): 8.19-8.16 (1H, m), 7.76 (1H, d, J8.6Hz), 7.69-7.66 N-(2-(2-Chlorophenyl)benzodthiazol-5-yl)isobu (1H, m), 7.56-7.52 (2H, m), 7.22(1H, d, J 2.0 Hz), 6.85 (1H, tyramide dd, J 8.6 2.1 Hz), 5.37 (2H, s) 0801 LCMS RT=6.99 min, MH 330.9; H NMR 2-(3-Chlorophenyl)benzodthiazol-5-amine (DMSO): 10.12(1H, s), 8.55 (1H, d, J 1.9 Hz), 8.25-8.22(1H, 0792 LCMS RT=6.79 min, MH 260.8: "H NMR m), 8.10 (1H, d, J 8.8 Hz), 7.74-7.55 (4H, m), 2.72-2.63 (1H, (DMSO): 8.05-8.04 (1H, m), 7.96 (1H, dt, J 7.01.7 Hz), 7.74 m), 1.17 (6H, d, J 6.8 Hz) US 2011/0195932 A1 Aug. 11, 2011 66

N-(2-(3-Chlorophenyl)benzodthiazol-5-yl)isobu mmol). The resulting solution was cooled to 10-15° C. and tyramide 4-chlorobenzoyl chloride (1.14 mL, 9.00 mmol) was added over 5 min. The mixture was stirred at room temperature for 0802 LCMS RT=7.34 min, MH 330.9; H NMR 90 min. The precipitate was filtered off, washed with dichlo (DMSO): 10.11 (1H, s), 8.50 (1H, d, J 1.7 Hz), 8.13-8.03 (3H, romethane, 1 Maqueous sodium hydroxide solution and 1M m), 7.69-7.60 (3H, m), 2.71-2.62 (1H, m), 1.17 (6H, d, J 6.8 aqueous hydrochloric acid solution to afford 2.12 g (93%) of Hz) the title compound. N-(2-(3,4-Dichlorophenyl)benzodthiazol-5-yl) 0806 H NMR (DMSO): 10.31 (1H, s), 7.98 (2H, d, J 8.7 isobutyramide Hz), 7.73 (2H, d, J 8.8 Hz), 7.61 (2H, d, J 8.8 Hz), 7.28 (2H, 0803 LCMS RT=8.21 min, MH 364.7; H NMR d, J 8.8 Hz), 2.47 (3H, s) (DMSO): 10.11 (1H, s), 8.52-8.50 (1H, m), 8.30 (1H, d, J 2.1 Hz), 8.10-8.04 (2H, m), 7.85 (1H, d, J8.4 Hz), 7.69-7.64 (1H, Method 33 (Compound XXX) m), 2.71-2.64 (1H, m), 1.17 (6H, d, J 6.8 Hz) 4-Chloro-N-(4-(methylthio)phenyl)benzothioamide N-(2-(2,3-Dichlorophenyl)benzodthiazol-5-yl) isobutyramide 0807. A suspension of 4-chloro-N-(4-(methylthio)phe nyl)benzamide (1 g, 3.60 mmol) and Lawesson’s reagent (875 0804 LCMS RT-7.62 min, MH 364.9; "H NMR mg, 2.16 mmol) in toluene (25 mL) was heated to 110°C. for (DMSO): 10.12(1H, s), 8.55 (1H, d, J 1.7 Hz), 8.14-8.10 (2H, 16 h. After cooling, toluene was removed in vacuo and the m), 7.88 (1H, dd, J 8.01.4 Hz), 7.67 (1H, dd, J 8.8 2.0 Hz), resulting solid was purified by column chromatography elut 7.58 (1H, t, J 8.0 Hz), 2.70-2.61 (1H, m), 1.15 (6H, d, J 6.8 ing using a gradient (hexanes to ethyl acetate/hexanes 30:70 Hz) V/v) to afford 503 mg (48%) of the title compound. (0808 LCMS RT=6.98 min, MH' 294.1; H NMR S (DMSO): 11.80 (1H, s), 7.85 (2H, d, J 8.6 Hz), 7.78 (2H, d, J 1. 8.7 Hz), 7.54 (2H, d, J 8.6 Hz), 7.33 (2H, d, J 8.7 Hz) --

NH2 Method 34 (Compound XXXI) C Pyridine, DCM 2-(4-Chlorophenyl)-6-(methylthio)benzodthiazole -e- rt, 90 min O Method 32 0809. To a solution of potassium hexacyanoferrate(III) (670 mg, 2.04 mmol) in water (5 mL) at 90° C. was added dropwise over 5 minutes a solution of 4-chloro-N-(4-(meth C ylthio)phenyl)benzothioamide (150 mg, 0.51 mmol) in etha 1S O Lawesson's, nol (2 mL) and 3M aqueous sodium hydroxide Solution (1.4 Toluene mL, 4.08 mmol). The resulting mixture was heated at 90° C. for 30 minutes. After cooling, the precipitate formed was Ol N o 110°Method C., 1633 h filtered off and washed with water to give a yellow solid. The yellow solid was purified by column chromatography eluting C using a gradient (hexanes to ethylacetate/hexanes 5:95 V/v) to XXIX afford 100 mg (67%) of the title compound (LCMS RT=9.37 1. S min, MH 292.2) K3Fe(CN)6, 08.10 "H NMR (DMSO); 8.11-8.06 (3H, m), 7.98 (1H, d, Ethanol N -as J 8.6 Hz), 7.65 (2H, d, J 8.7 Hz), 7.45 (1H, dd, J 8.61.9 Hz), H NaOH, 90° C., 2.58 (3H, s) 30 min C Method 34 Method 35 (Compound XXXII) XXX S S 1 mCPBA, DCM 2-(4-Chlorophenyl)-6-(methylsulfonyl)benzodthiazole C -ss M rt, 3 h 0811. To a solution of 2-(4-chlorophenyl)-6-(methylthio) N Method 35 benzodthiazole (240 mg, 0.82 mmol) in dichloromethane XXXI (20 mL) was added 3-chloroperoxybenzoic acid (77% in MeOS S water, 710 mg. 4.11 mmol) over 5 min. The resulting mixture was stirred at room temperature for 3 h. 1 Maqueous sodium N hydroxide solution was added carefully, and the mixture was XXXII then stirred for 5 min. The organic layer was then washed with 1M aqueous sodium hydroxide solution, dried over anhy drous MgSO and evaporated. The resulting Solid was recrys tallised from hot ethyl acetate to afford 125 mg (47%) of the Method 32 (Compound XXIX) title compound (LCMS RT=6.81 min, MH 324.0) 4-Chloro-N-(4-(methylthio)phenyl)benzamide 0812 H NMR (CDC1): 8.61 (1H, dd, J 1.8 0.4 Hz), 8.26 0805. To 4-(methylthio)aniline (1 mL, 8.19 mmol) in (1H, dd, J 8.6 0.5 Hz), 8.14-8.09 (3H, m), 7.57 (2H, d, J 8.6 dichloromethane (20 mL) was added pyridine (2 mL. 24.6 Hz), 3.19 (3H, s) US 2011/0195932 A1 Aug. 11, 2011 67

-continued Y-H21 S. B(OH)2 Br R N N Pd(PPh3)4, dioxane?water R Iron, NH4Cl C MW, 160° C., 15 min XXXIV R1 = ...) Ethanol/Water, N Method 36 XXXV R = NH 70° C., 2h

Method 38

RCOCI Pyridine, rt, 16 h Method 39

R N

O4. N N H V R XXXVI Method 36 (Compounds XXXIII) 2-(4-Chlorophenyl)-5-phenyl-1H-indole Method 37 (Compound XXXIV) 0813 To a suspension of 5-bromo-2-(4-chlorophenyl)- 1-Methyl-6-nitro-1H-indole 1H-indole (200 mg. 0.65 mmol) in dioxane?water 4:1 V/v (5 0817 To a solution of 6-nitro-1H-indole (100 mg. 0.62 mL) was added phenylboronic acid (87 mg, 0.72 mmol) and mmol) and 18-Crown-6 (180 mg. 0.68 mmol) in anhydrous a few milligrams of tetrakis(triphenylphosphine)palladium tetrahydrofuran (2 mL) at room temperature was slowly (0). The resulting Suspension was heated in the microwave at added potassium tert-butoxide (76 mg, 0.68 mmol) followed by methyl iodide (42 uL, 0.68 mmol). The solution was stirred 160° C. for 15 min. After cooling, the reaction was poured at room temperature for 30 min. Tetrahydrofuran was into water to give a precipitate, which was filtered off and removed in vacuo. Ethyl acetate was added, and the organic washed with water. The resulting solid was purified by col layer was washed with water and then brine. The combined umn chromatography eluting using a gradient (hexanes to organic layers were dried over anhydrous MgSO and evapo ethyl acetate/hexanes 30:70 V/v), followed by a recrystallisa rated to afford 91 mg (84%) of the title compound. tion from hot ethyl acetate to afford 21 mg (11%) of the title 0818 H NMR (CDC1): 8.16 (1H, d, J 1.8 Hz), 7.85 (1H, compound (LCMS RT=8.54 min, MH 304.1) dd, J 8.71.9 Hz), 7.49 (1H, d, J 8.7 Hz), 7.19 (1H, d, J3.1 Hz), 0814 H NMR (DMSO): 11.68 (1H, s), 7.91 (2H, d, J 8.6 6.43 (1H, dd, J 3.10.9 Hz), 3.74 (3H, s) Hz), 7.81 (1H, d, J 1.1 Hz), 7.70-7.66 (2H, m), 7.55 (2H, d, J 8.6 Hz), 7.50-741 (4H, m), 7.33-7.28 (1H, m), 7.01 (1H, d, J Method 38 (Compound XXXV) 1.2 Hz) 1-Methyl-1H-indol-6-amine 0815. The compound below was prepared following the 0819 1-methyl-6-nitro-1H-indole (90 mg 0.51 mmol), same general method. ammonium chloride (55 mg, 1.02 mmol) and iron powder (143 mg, 2.55 mmol) were suspended in ethanol/water (2 N-(4-(2-(4-Chlorophenyl)-1H-indol-5-yl)phenyl) ml 11 mL) and heated at 70° C. for 2 h. After cooling, the solution was filtrated through a pad of Celite(R), which was acetamide washed with ethanol. Ethyl acetate was added to the filtrate and the organic layer was washed with water twice. The 0816 LCMS RT=6.69 min, MH 361.0; H NMR combined organic layers were dried over anhydrous MgSO (DMSO): 11.64 (1H, s), 9.98 (1H, s), 7.91 (2H, d, J 8.6 Hz), and evaporated to afford 30 mg (37%) of the title compound 7.77 (1H, d, J 1.0 Hz), 7.68-7.60 (4H, m), 7.54 (2H, d, J 8.6 0820 "H NMR (DMSO): 7.17 (1H, d, J 8.4 Hz), 6.93 (1H, Hz), 7.46 (1H, d, J 8.3 Hz), 7.41 (1H, dd, J 8.5 1.6 Hz), 6.99 d, J 3.1 Hz), 6.51-6.49 (1H, m), 6.41 (1H, dd, J 8.3 1.9 Hz), (1H, d, J 1.5 Hz), 2.07 (3H, s) 6.16 (1H, d, J 3.1 Hz), 4.76 (2H, s), 3.60 (3H, s) Method 39 (Compounds XXXVI) KO'Bu, RX, 18-Crown-6 N-(1H-indol-6-yl)isobutyramide N THF, rt, 30 min 0821. To a solution of 1-methyl-1H-indol-6-amine (45 Method 37 mg, 0.31 mmol) in pyridine (2 mL) at room temperature was ON N added isobutyryl chloride (35 uL, 0.34 mmol). The resulting mixture was stirred at room temperature for 16 h. Ethyl acetate was added and the organic layer was washed three US 2011/0195932 A1 Aug. 11, 2011

times with brine. The combined organic layers were dried over anhydrous MgSO and evaporated to afford 24.3 mg -continued (36%) of the title compound (LCMS RT=5.73 min, MH" 217.2) R O N ( ) 0822 H NMR (DMSO): 9.78 (1H, s), 7.96 (1H, m), 7.44 O4. N N (1H, d, J8.4 Hz), 7.24 (1H, d, J3.1 Hz), 7.08 (1H, dd, J8.41.7 H V Hz), 6.35 (1H, dd, J 3.00.7 Hz), 3.73 (3H, s), 2.68-2.61 (1H, R m), 1.13 (6H, d, J 6.9 Hz) XLII 0823. The compound below was prepared following the XLIIb, R= Me same general method. N-(1-Benzyl-1H-indol-6-yl)isobutyramide Method 40 (Compound XXXVII) 0824 LCMS RT=6.36 min, MH 293.2; H NMR N-(2-Hydroxy-5-nitrophenyl)butyramide (DMSO): 9.73 (1H, s), 7.90 (1H, m), 7.45 (1H, d, J 8.5 Hz), 0825 To a solution of 2-amino-4-nitrophenol (10g, 64.9 7.41 (1H, d, J 3.1 Hz), 7.34-7.24 (3H, m), 7.14-7.10 (3H, m), mmol) in dichloromethane (250 mL) under nitrogen at 0°C. 6.42 (1H, d, J 3.2 Hz), 5.35 (2H, s), 1.08 (6H, d, J 6.8 Hz) was added pyridine (10.5 mL, 129.9 mmol) followed by butyrylchloride (7.05 mL, 68.2 mmol) over a period of 5 min. After 30 min at 0°C., the solution was left warming up to OH room temperature for 2 days. The organic layer was washed butyryl chloride, pyridine, DCM with aqueous copper Sulfate Solution and brine. Insoluble 2 days, rt material from the aqueous layer was filtered off and washed ON NH2 Method 40 with water to afford 4.95 g (34%) of the title compound. OH 0826 HNMR (DMSO): 11.64 (1H, br),9.37 (1H, s), 8.95 O NaH, TfG), MeCN -e- (1H, d, J 2.8 Hz), 7.89 (1H, dd, J 8.92.8 Hz), 7.02(1H, d, J 8.9 3 h, rt Hz), 2.43 (2H, t, J 7.4 Hz), 1.67-1.55 (2H, m), 0.92 (3H, t, J ON N Method 41 7.5 Hz) Method 41 (Compound XXXVIII) XXXVII OTf 2-Butyramido-4-nitrophenyl trifluoromethanesulfonate O 0827. To a solution of sodium hydride (220 mg, 5.58 Phenyl acetylene, Pd(PPh3)4 mmol) in dry acetonitrile (40 mL) at 0°C. under nitrogen was ON N CuI, BuNI, NEt3, MeCN added a solution of N-(2-hydroxy-5-nitrophenyl)butyramide -e- 2 h, rt (1 g, 4.46 mmol) in dry acetonitrile (90 mL). The solution was Method 42 then stirred at 0° C. for 30 min. Trifluoromethanesulfonic XXXVIII anhydride (825 uL, 4.90 mmol) was added dropwise at 0°C. over a period of 10 min. After 3 h at 0°C., the solution was stirred at room temperature for 3 h. Water was added, and the 'BuOK, NMP aqueous layer was extracted with ethyl acetate. The organic -- 2 6h, 70° C. layer was then washed with dilute aqueous hydrochloric acid, aqueous Sodium bicarbonate and brine. The combined O O Method 43 organic layers were dried over anhydrous MgSO and evapo rated. The resulting oil was purified by column chromatog ON raphy eluting using a gradient (hexanes to ethyl acetate/hex anes 25:75 V/v) to afford 860 mg (54%) of the title compound. 0828 H NMR (CDC1): 9.37 (1H, d, J 2.8 Hz), 8.09 (1H, XXXIX dd, J9.12.8 Hz), 7.53 (2H, d, J9.1 Hz), 2.50 (2H, t, J 7.6 Hz), Iron, NH4C1, 1.91-1.78 (2H, m), 1.09 (3H, t, J 7.5 Hz) N Ethanol Water 70° C., 2h Method 42 (Compound XXXIX) ON N Method 45 N-(5-Nitro-2-(phenylethynyl)phenyl)butyramide * KO'Bu, Mel, 18-Crown-6 0829. To a solution of 2-butyramido-4-nitrophenyltrifluo XLaR = H ) THF, rt, 30 min romethanesulfonate (860 mg, 2.42 mmol) in dry acetonitrile XLb R = Me Method 44 (30 mL) under nitrogen was added tetrabutylammonium N RICOCl, pyridine iodide (1.34g, 3.62 mmol), tetrakis(triphenylphosphine)pal He ladium(0) (280 mg, 0.24 mmol) and copper iodide (140 mg. rt, 48 h. 0.72 mmol). Triethylamine (6 mL) was then added, followed HN N Method 46 by phenyl acetylene (530 ul, 4.83 mmol). The resulting solu R tion was stirred at room temperature for 2 h. Ammonium XLI chloride was then added to quench the reaction, and the XLIb, R= Me aqueous layer was extracted with ethyl acetate. The organic layer was washed with brine. The combined organic layers US 2011/0195932 A1 Aug. 11, 2011 69 were dried over anhydrous MgSO and evaporated. The 0836) "H NMR (DMSO): 10.88 (1H, s), 7.76-772 (2H, resulting solid was purified by column chromatography elut m), 7.39 (2H, t, J 7.9 Hz), 7.23-7.16 (2H, m), 6.67 (1H, dd, J ing using a gradient (hexanes to ethyl acetate) to afford 580 2.00.7 Hz), 6.59-6.57 (1H, m), 6.39 (1H, dd, J 8.4 2.0 Hz), mg (78%) of the title compound. 4.82 (2H, s) 0830 "H NMR (DMSO): 9.81 (1H, s), 8.74 (1H, d, J 2.3 Hz), 8.01 (1H, dd, J 8.6 2.4 Hz), 7.83 (1H, d, J 8.6 Hz), Method 46 (Compounds XLII) 7.70-7.67 (2H, m), 7.52-749 (3H, m), 1.73-1.61 (2H, m), N-(2-Phenyl-1H-indol-6-yl)isobutyramide 0.96 (3H, t, J 7.5 Hz) 0837 To a solution of 2-phenyl-1H-indol-6-amine (54 mg, 0.26 mmol) in pyridine (2 mL) at room temperature was Method 43 (Compound XLa) added isobutyryl chloride (30 uL, 0.29 mmol). The resulting mixture was stirred at room temperature for 2 days. When 6-Nitro-2-phenyl-1H-indole water was added, a precipitate was formed. This solid was recrystallised from hot ethyl acetate to afford 15 mg (21%) of 0831. To a solution of N-(5-nitro-2-(phenylethynyl)phe the title compound (LCMS RTh-6.27 min, MH 279.0) nyl)butyramide (580 mg, 1.88 mmol) in 1-Methyl-2-pyrroli 0838 H NMR (DMSO): 11.40 (1H, s), 9.74 (1H, s), 8.02 dinone (20 mL) under nitrogen was added potassium tert (1H, s), 7.82 (2H, d, J 7.5 Hz), 747-7.40 (3H, m), 7.28 (1H, butoxide (243 mg, 2.16 mmol). The resulting solution was t, J 7.3 Hz), 7.07 (1H, dd, J 8.5 1.6 Hz), 6.83 (1H, d, J 1.1 Hz), heated at 70° C. for 6 h, and then left at room temperature for 2.67-2.60 (1H, m), 1.13 (6H, d, J 6.7 Hz) 16 h. Water was added and the aqueous layer was extracted 0839. The compound below was prepared following the several times with ethyl acetate. The combined organic layers same general method. were washed 10 times with water, 3 times with brine, dried over anhydrous MgSO and evaporated. The resulting mate N-(1-Methyl-2-phenyl-1H-indol-6-yl)isobutyramide rial was purified by column chromatography eluting with 0840 LCMS RT=6.66 min, MH 293.2; H NMR ethylacetate/hexanes 15:85 V/v to afford 175 mg (39%) of the (DMSO): 9.83 (1H, s), 8.02 (1H, s), 7.61-7.39 (6H, m), 7.13 title compound. (1H, dd, 18.5 1.7 Hz), 6.50 (1H, d, J 0.5 Hz), 3.69 (3H, s), 0832 'H NMR (DMSO): 12.35 (1H, s), 8.30 (1H, d, J 2.1 2.69-2.60 (1H, m), 1.13 (6H, d, J 6.8 Hz) Hz), 7.97-7.90 (3H, m), 7.73 (1H, d, J 8.9 Hz), 7.57-7.52(2H, m), 7.47-7.42 (1H, m), 7.17 (1H, dd, J 2.00.8 Hz) OH Method 44 (Compound XLb) 1-Methyl-6-nitro-2-phenyl-1H-indole ON I Prolinol, Pd/C, CuI, PPh3 0833. To a solution of 6-nitro-2-phenyl-1H-indole (106 Water, 80°C., 3 h mg, 0.44 mmol) and 18-Crown-6 (130 mg, 0.49 mmol) in R Method 47 anhydrous tetrahydrofuran (2 mL) at room temperature was added potassium tert-butoxide (55 mg, 0.49 mmol) followed by methyl iodide (31 uL, 0.49 mmol). The solution was stirred ON O) ()- at room temperature for 30 min. Tetrahydrofuran was XLIIIa R=NO Fe, NH4C1, EtOHWater 2:1 removed in vacuo. Ethyl acetate was added, and the organic XLIIIb R = NH ) viv, 80°C., 4h layer was washed with water and then brine. The combined Method 48 organic layers were dried over anhydrous MgSO and evapo rated to afford 110 mg (98%) of the title compound. RICOCl, pyridine, RICOH, DIPEA, DCM, 0834 H NMR (DMSO): 8.60 (1H, d, J 2.1 Hz), 8.03 (1H, rt, 16 h HATU dd, J 8.8 2.1 Hz), 7.82 (1H, d, J 8.7 Hz), 7.74-7.71 (2H, m), Method 49 rt, 48 h. 7.67-7.57 (3H, m), 6.87 (1H, d, J 0.8 Hz), 3.95 (3H, s) Method 50 O R Method 45 (Compound XLI) 4. / ( ) R O N 2-Phenyl-1H-indol-6-amine H XLIV 0835 6-Nitro-2-phenyl-1H-indole (175 mg, 0.73 mmol), ammonium chloride (80 mg, 1.47 mmol) and iron powder (205 mg, 3.68 mmol) were suspended in ethanol/water (4 mL/2 mL) and heated at 70° C. for 2 h. After cooling, the Method 47 (Compound XLIIIa) solution was filtrated through a pad of Celite(R), which was 5-Nitro-2-phenylbenzofuran washed with ethanol. The organic layer was evaporated into vacuo to obtain a solid, which was purified by column chro 0841. A solution of 2-iodo-4-nitrophenol (500 mg, 1.89 matography eluting using a gradient (ethyl acetate/hexanes mmol), prolinol (573 mg, 5.66 mmol), palladium on carbon 10:90 v/v to ethyl acetate/hexanes 50:50 v/v) to afford 54 mg (60 mg, 0.06 mmol), triphenylphosphine (59.4 mg., 0.226 (35%) of the title compound. mmol) and copper iodide (22 mg 0.113 mmol) in water (6 US 2011/0195932 A1 Aug. 11, 2011 70 mL) was stirred for 1 hat room temperature. Ethynylbenzene 0850. The mixture was then stirred at room temperature (482 mg, 4.72 mmol) was slowly added, and the resulting for 10 min. 2-phenylbenzofuran-5-amine (234 mg, 1.12 mixture was heated at 80° C. for 3 h. After cooling, ethyl mmol) was then added and the resulting mixture was stirred at acetate was added, and the mixture was passed through a pad room temperature for 48 h. Ethyl acetate was added and the of Celite(R). The filtrate was washed with water; the combined organic layer was washed once with Saturated aqueous water. organic layers were dried over anhydrous MgSO and evapo The combined organic layers were dried over anhydrous rated. The resulting material was purified by column chroma MgSO and evaporated. The resulting solid was purified by tography eluting with ethyl acetate/hexanes 1:40 V/v to afford column chromatography eluting using a gradient (ethyl 134 mg (30%) of the title compound. acetate/hexanes 1:3 V/v to ethyl acetate/hexanes 1:1 V/v) fol 0842 'H NMR (DMSO): 8.62(1H, d, J24 Hz), 8.23 (1H, lowed by trituration in ethyl acetate to afford 99.3 mg (28%) dd, J 9.1 2.5 Hz), 8.00-7.96 (2H, m), 7.89 (1H, d, J 9.0 Hz), of the title compound (LCMS RT=6.62 min) 7.66 (1H, d, J 0.4 Hz), 7.60-746 (3H, m) 0851 H NMR (DMSO): 10.37 (1H, s), 8.01 (1H, d, J 2.0 Hz), 7.92 (2H, dd, J 7.5 1.5 Hz), 7.61 (1H, d, J 8.8 Hz), Method 48 (Compound XLIIIb) 7.55-7.41 (4H, m), 7.38 (1H, dd, J 8.9 2.2 Hz), 3.53 (2H, q, J 11.2 Hz) 2-Phenylbenzofuran-5-amine 0852. The compounds listed in Table 2, can be prepared by 0843. To 5-Nitro-2-phenylbenzofuran (250 mg, 1.04 analogues methods to those described above, or by literature mmol) in ethanol/water 2:1 V/v (12 mL) at 80°C. was added methods known or adapted by the persons skilled in the art. ammonium chloride (112 mg, 2.09 mmol) and iron powder Synergistic Effect of Combinations with Prednisone (292 mg, 5.23 mmol). The resulting mixture was heated at 80° 0853. The effect of combinations of a compound of for C. for 4 h. After cooling, the solution was filtrated through a mula (1) with a corticosteroid (prednisolone) was examined pad of Celite R, which was washed with ethanol. The organic by measuring fatigue induced by a forced exercise regime in layer was evaporated into vacuo to obtain a Solid, which was mdx mice. then taken up in ethyl acetate and washed with water. The 0854 Four to five week-old male mdx mice were forced to combined organic layers were dried over anhydrous MgSO exercise (30 min running on horizontal treadmill at 12 m/min and evaporated to afford 211 mg (96%) of the title compound. twice a week on the same day and at the same time) over a five 0844 H NMR (DMSO): 7.87-7.83 (2H, m), 7.50-7.44 week treatment course. Each treatment group included 6 (2H, mi, 7.40-7.34 (1H, m), 7.28 (1H, d, J 8.7 Hz), 7.20 (1H, 1CC. d, J0.7 Hz), 6.74 (1H, d, J2.2 Hz), 6.60 (1H, dd, J8.72.3 Hz), 0855. At the end of the treatment course, exhaustion was 4.88 (2H, s) induced by running on a horizontal treadmill at 5 m/min for 5 min, with incremental speed increases of 1 m/min each Method 49 (Compounds XLIV) minute until exhaustion. The total distance run was then mea Sured. N-(2-Phenylbenzofuran-5-yl)isobutyramide 0856 Treatment included dosing (ip) with 50 mg/kg 5-(ethylsulfonyl)-2-(naphthalen-2-yl)benzodoxazole daily 0845 To a solution of 2-phenylbenzofuran-5-amine (210 for 5 weeks, alone and in combination with prednisolone. mg, 1.00 mmol) in pyridine (5 mL) at room temperature was Sedentary and vehicle only groups acted as negative controls. added isobutyrylchloride (120 uL. 1.10 mmol). The resulting 0857. The results are shown in FIG. 4. They show that mixture was stirred at room temperature for 16 h. Ethyl prednisolone (PDN) and 5-(ethylsulfonyl)-2-(naphthalen-2- acetate was added and the organic layer was washed with saturated aqueous copper Sulfate Solution followed by Satu yl)benzodoxazole (CMPD1) can act in synergy to reduce rated aqueous potassium carbonate solution. The combined exercise-induced fatigue in the mdx mouse. organic layers were dried over anhydrous MgSO and evapo rated. The resulting material was purified by column chroma EQUIVALENTS tography eluting using a gradient (ethyl acetate/hexanes 1:3 0858. The foregoing examples are presented for the pur V/v to ethyl acetate/hexanes 1:2 v/v) to afford 134 mg (48%) pose of illustrating the invention and should not be construed of the title compound (LCMS RT=6.81 min, MH 280.1) as imposing any limitation on the Scope of the invention. It 0846 H NMR (DMSO): 9.87 (1H, s), 8.05 (1H, s), 7.91 will readily be apparent that numerous modifications and (2H, d, J 7.4 Hz), 7.58-748 (3H, m), 7.45-7.38 (3H, m), alterations may be made to the specific embodiments of the 2.66-2.54 (1H, m), 1.13 (6H, d, J 6.8 Hz) invention described above and illustrated in the examples 0847 The compound below was prepared following the without departing from the principles underlying the inven same general method. tion. All such modifications and alterations are intended to be embraced by this application. 2-(4'-Chlorophenyl)-5-isobutyramido-benzofuran 1. A combination comprising an ancillary agent and a 0848 LCMS RT=7.41 min, MH 314.2; H NMR compound of Formula (1): (DMSO): 9.88 (1H, s), 8.06 (1H, d, J 1.9 Hz), 7.92 (2H, d, J 8.7 Hz), 7.59-7.53 (3H, m), 7.49 (1H, d, J 0.8 Hz), 7.43 (1H, dd, J9.02.2 Hz), 2.66-2.56 (1H, m), 1.13 (6H, d, J 6.8 Hz) (1) Method 50 (Compound XLIV) 2-Phenyl-5-(3',3',3'-trifluoro propanamido)benzofuran 0849. To 3,3,3-trifluoropropanoic acid (136 mg, 1.06 mmol) in dry dichloromethane (10 mL) was added HATU (46.8 mg, 1.23 mmol) and diisopropylethylamine (580 uL. 3.35 mmol). US 2011/0195932 A1 Aug. 11, 2011 71

in which optionally substituted aryloxy, A. A. As and A, which may be the same or different, each aryl or NRoR; or represent N or CR; when R or R7 represents NRoR, one of Ro and R. X is a divalent group selected from O, S(O)C—W, NR may additionally represent optionally substituted NC(=O)Rs and CRR, COalkyl or optionally substituted COaryl; and W is O, S, or NR: R may additionally represent hydroxy. Y is N or CRs: 4. The combination according to claim 2, in which the one of R. Rs. Re, Rs. Ro and NRo represents -L-R, in Substituent(s) on R is R and R and R, which may be the which L is a single bond or a linker group; same or different, each represent: additionally, R, R-R, which may be the same or differ alkyl optionally Substituted by one or more halogen, ent, each independently represent hydrogen or a Sub alkoxy or optionally Substituted aryl, thioaryl oraryloxy, stituent; alkoxy optionally substituted by optionally by alkyl or Ro represents hydrogen, hydroxyl, alkyl optionally Sub optionally substituted aryl, stituted by aryl, alkoxy optionally substituted by aryl, hydroxyl, aryl, CN, optionally substituted alkoxy, optionally sub stituted aryloxy, optionally Substitute alkanoyl, option aryl, ally substituted aroyl, NO, or NRoR, in which Rio thioalkyl optionally substituted by alkyl or optionally sub and R, which may be the same or different, each rep stituted aryl, resent hydrogen, optionally Substituted alkyl or option thioaryl, in which the aryl is optionally substituted, ally Substituted aryl; or one of Ro and R is represent NO, optionally substituted alkanoyl or optionally substituted CN, aroyl, and the other of Ro and R is hydrogen, option NRoR, ally substituted alkyl or optionally substituted aryl. halogen, in represents an integer from 0 to 2: SOR, when an adjacent pair of A-A each represent CR, the NRSOR, adjacent carbon atoms, together with their Substituents may form a ring B; and when X is CRR 7, RandR 7, together with the carbonatom P(=O)CRR, wherein to which they are attached may form a ring C; or a Rio, R1, R12, R1s. R14, R1s. R16, R17, Rao and R41, which pharmaceutically acceptable salt thereof. may be the same or different, each represent hydrogen, 2. The combination according to claim 1, wherein R rep alkyl optionally substituted by optionally substituted resents alkyl, alkoxy or aryl, each optionally substituted by aryl, or optionally substituted aryl; one to three substituents, which may be the same or different. in addition, 3. The combination according to claim 1, wherein L is NRoR together with the nitrogen to which they are single bond; and R represents: attached may form a ring; thioalkyl optionally substituted by alkyl or optionally sub R may have the same meaning as NRoR, stituted aryl, when R, represents NRR, one of Ro and R may thioaryl, in which the aryl is optionally substituted, represent hydrogen, COalkyl or optionally substituted optionally substituted aryl, COaryl; hydroxyl, R may represent hydroxy, alkoxy, or NRoR, NO, R7 may represent alkyl optionally substituted by one or CN, more of halogen, alkoxy, optionally Substituted aryl or NRoR, NRoR; or halogen, when R, represents NRR, one of Ro and R may SOR, represent hydrogen, COalkyl or optionally substituted NRSOR, COaryl. OC(=W)NRR, or 5. The combination of according to claim 1, in which NRC(=W)R; wherein L represents a linker group which is: Rio R, R2, R. R. Rs. RandR 17, which may be the O, S, (CO)NRs, whereinn represents an integer from 0 to same or different, each represent hydrogen, alkyl option 2. ally substituted by optionally substituted aryl, or option alkylene, alkenylene, or alkynylene, each of which may be ally substituted aryl; optionally interrupted by one or more of O, S, or NRs. in addition, or one or more C-C single, double or triple bonds, or NRoR together with the nitrogen to which they are a —N N— single or double bond; attached may form a ring; Rs represents hydrogen, alkyl, or COR: R may have the same meaning as NRoR, R. R. R. R., and Rs each independently represent hydro when R7 represents NRoR, one of Ro and R may gen, alkyl or optionally Substituted aryl; represent hydrogen, COalkyl or optionally substituted Y represents N: COaryl; X represents O, S or NR; and RandR, which may be the same or different, may each R represents -L-R represent 6-8. (canceled) alkyl optionally Substituted by one or more of halogen, 9. The combination according to claim 1 in which alkoxy optionally substituted aryl or optionally substi when any of the substituents represents alkyl, the alkyl is tuted aryl, Saturated and has from 1 to 10 carbon atoms; and US 2011/0195932 A1 Aug. 11, 2011 72

when any of the Substituents represents aryl, the aryl is an 29. The combination according to claim 26, in which the aromatic hydrocarbon or a 5- to 10-membered aromatic aromatic ring is benzene, naphthalene, thiophene, furan, pyri heterocycle containing from 1 to 4 hetero atoms selected dine or pyrrole. from O, S and N. 30-31. (canceled) 10. (canceled) 32. The combination according to claim 2, in which the 11. The combination according to claim 9, in which the aryl Substituent(s) on R is R., and R independently: is phenyl, naphthalene, furan, thiophene, pyrrole or pyridine. alkyl C-C optionally Substituted by thienyl or phenoxy, 12. (canceled) each optionally Substituted by halogen, 13. The combination according to claim 1, in which ring B alkoxy C-C, or ring C is a saturated or unsaturated 3- to 10-membered phenyl, carbocyclic or heterocyclic ring. thioalkyl C-C, 14. The combination according to claim 13, in which ring thienyl, optionally Substituted by halogen, B is a benzene ring, and ring C is a 3- to 10-membered NO, saturated or unsaturated carbocyclic ring. CN, 15. (canceled) NRR, in which Ro and R, which may be the same or 16. The combination according to claim 1, in which at least different, each represent hydrogen, or alkyl C-C, or one R represents: together with the nitrogen to which they are attached NRC(=W)R; form a 5 to 7-membered ring which may contain one or NRC(=O)R7; more additional heteroatoms selected from N, O and S, CONRR: halogen, NHCOR, wherein R, is: SOR2, in which R2 represents a 5- to 7-membered ring C-C alkyl optionally substituted with one or more which may contain one or more additional heteroatoms halo, phenyl or C-C alkoxy, selected from N, O and S, or phenyl optionally substituted with one or more of halo, NHCOR7, in which R, represents C-C alkyl, alkyl C-C optionally substituted by: C-C alkoxy, amino, (C-C alkyl)amino, di(C-C, phenyl or halogen, or alkyl)amino, or phenyl, CH:CH phenyl. phenyl optionally Substituted by alkoxy C-C car naphthyl, pyridinyl, thienyl, or furanyl: boxy, or halogen, or NRCONRR, wherein Ro and R, which may be the a 5 or 6 membered saturated or unsaturated hetero same or different, are each optionally Substituted aryl, cycle, or alkyl, or optionally substituted COaryl; phenyl or a 5 or 6 membered saturated or unsaturated NHCONHRs, wherein Rs is phenyl, C-C alkyl, or heterocycle optionally substituted by halogen, alkoxy COphenyl optionally substituted with one or more halo: C to C, carboxy or SO2NRoR. C-C alkyl optionally substituted with phenyl or a 4- to 33. The combination according to claim 32 in which 7-membered saturated or unsaturated heterocycle con NRoR represents N-pyrrole, N-piperidine, N'-(C-C, taining one to two heteroatoms selected from N, S and alkyl)-N-piperazine or N-morpholine. O; 34. The combination according to claim 5 in which L COR, wherein R is C-C alkoxy, amino, (C-C alkyl) represents: amino, or di(C-C alkyl)amino; NH.NH , NO; CH-CH, or halogen; NCOR in which R represents phenyl or a 5 or 6 mem amino, (C-C alkyl)amino, or di(C-C alkyl)amino, bered saturated or unsaturated heterocycle optionally wherein the C-C alkyl is optionally substituted with Substituted by halogen, alkoxy C to C, or carboxy. phenyl or a 5- or 6-membered saturated or unsaturated 35. The combination according to claim 1 in which two of heterocycle; A-A represent nitrogen; or one of A-A represents nitro NHSO(C-C alkyl); gen; or all of A-A represents CR. NHSO-phenyl: 36-37. (canceled) SO(C-C alkyl); 38. The combination of claim 1 wherein the compound of phenyl optionally Substituted with one or more C-C, formula (1) is: alkoxy; or N-(2-(4-(dimethylamino)phenyl)benzodoxazol-5-yl) 5- to 10-membered saturated or unsaturated mono- or bi isonicotinamide; cyclic heterocycle containing from 1 to 3 heteroatoms N-(2-(4-fluorophenyl)benzodoxazol-5-yl)furan-2-car selected from N, S and O. boxamide: 17-19. (canceled) 2-((4-chlorophenoxy)methyl)-1-methyl-1H-benzodimi 20. The combination according to claim 1 in which one or dazole; both of R and R is other than —COOH. 2-((4-methoxyphenoxy)methyl)-1H-benzodimidazole; 21-25. (canceled) phenyl(2-phenyl-1H-benzodimidazol-6-yl)methanone; 26. The combination according to claim 1, in which R is a N-(2-phenylbenzodoxazol-5-yl)nicotinamide: 5- to 10-membered aromatic mono- or bi-cyclic hydrocarbon 3-phenyl-N-(2-phenylbenzodoxazol-5-yl)propanamide: ring or heterocyclic ring optionally Substituted by one to three N-(2-phenylbenzodoxazol-5-yl)acetamide: substituents, which may be the same or different, wherein the N-(2-phenylbenzodoxazol-5-yl)propionamide: heterocyclic ring contains one to three heteroatoms selected N-(2-phenylbenzodoxazol-5-yl)butyramide: from N, O and S. N-(2-phenylbenzodoxazol-5-yl)pentanamide: 27-28. (canceled) N-(2-phenylbenzodoxazol-5-yl)isobutyramide:

US 2011/0195932 A1 Aug. 11, 2011 78

2-(Naphthalen-2-yl)-5-(trifluoromethoxy)benzodox the two or more compounds together with instructions for azole; administration to a patient population in which the other(s) of 2-(Naphthalen-2-yl)benzodoxazole-5-carboxylic acid; the two or more compounds have been or are being) admin 2-(Naphthalen-2-yl)benzodoxazole; istered; (d) at least one of the two or more compounds in an 5-tert-Butyl-2-(naphthalen-2-yl)benzodoxazole; amount or in a form which is specifically adapted for use in 5,6-Difluoro-2-(naphthalen-2-yl)benzodoxazole; combination with the other(s) of the two or more compounds. 1-(2-(3",4"-Dichlorophenyl)benzodoxazol-5-yl)etha 43. A pharmaceutical pack, kit or patient pack comprising the combination of claim 1. none. N-(4-(Benzodoxazol-2-yl)phenyl)isobutyramide: 44-46. (canceled) Methyl 2-(4-chlorophenyl)benzodoxazol-5-yl(ethyl) 47. A method for the treatment or prophylaxis of Duchenne phosphinate; muscular dystrophy, Becker muscular dystrophy or cachexia, 2-(3',4'-Dichlorophenyl)-5-(1'-hydroxyethyl)-benzox comprising administering the combination of claim 1. 48. (canceled) azole; 49. The combination of claim 1 wherein the ancillary agent 2-(4-Chlorophenyl)-6-methylbenzodoxazole; or is selected from: (a) an anti-inflammatory agent; (b) a pro 5-Methyl-2-(naphthalen-2-yl)benzodoxazole. tease inhibitor; (c) a myostatin antagonist; (d) a cytokine or 39. The combination of claim 1 wherein the ancillary agent mobilizing agent; (e)a corticosteroid, (f) an anabolic steroid; and the compound of formula (1) are physically associated; or (g) a TGF-3 antagonist: (h) an antioxidant or mitochondrial the ancillary agent and the compound of formula (1) are Supporting agent; (i) a dystrophin expression enhancing non-physically associated; or the ancillary agent and the com agent; (ii) a gene replacement/repair agent; (k) a cell-based pound of formula (1) are composition; (1) creatine; (m) an anti-osteoporotic agent; (n) (a) in admixture; an auxiliary utrophin upregulating agent; (o) a coMP signal (b) chemically/physicochemically linked; ing modulator; and (p) a combination of two or more of the (c) chemically/physicochemically co-packaged; or foregoing classes (a) to (o). (d) unmixed but co-packaged or co-presented. 50. The combination of claim 1 wherein the compound of 40-41. (canceled) formula (I) is 5-(ethylsulfonyl)-2-(naphthalen-2-yl)benzod 42. The combination of claim 39 wherein the combination oxazole. comprises: (a) at least one of the two or more compounds 51. The invention combination of claim 50 wherein the together with instructions for the extemporaneous association ancillary agent is a corticosteroid, prednisone, prednisolone, of the at least one compound to form a physical association of or deflazacort. the two or more compounds; or (b) at least one of the two or 52-53. (canceled) more compounds together with instructions for combination therapy with the two or more compounds; (c) at least one of