You are on page 1of 6

PROGRESS

T R A N S L AT I O N A L G E N E T I C S
delivery to all muscle, which constitutes

Therapy for Duchenne muscular ~35% of the body mass, is a considerable


hurdle. Many therapies work well in the mdx

dystrophy: renewed optimism from


mouse — the frequently used animal model
of the disease, which has a stop codon in
exon 23 of the dystrophin gene — and the
genetic approaches golden retriever muscular dystrophy dog
models of the disease. In these models, point
mutations result in the absence of dystro‑
Rebecca J. Fairclough, Matthew J. Wood and Kay E. Davies phin, but efficacy will need to be improved
in patients to lead to significant disease
Abstract | Duchenne muscular dystrophy (DMD) is a devastating progressive
modification.
disease for which there is currently no effective treatment except palliative Just as the identification of the dystro‑
therapy. There are several promising genetic approaches, including viral delivery phin gene was one of the first successes of
of the missing dystrophin gene, read-through of translation stop codons, exon positional cloning, the pioneering genetic
skipping to restore the reading frame and increased expression of the approaches now entering the clinic for DMD
(TABLE 1) are also applicable to other diseases.
compensatory utrophin gene. The lessons learned from these approaches will be
applicable to many other disorders.
Viral gene therapy
Direct replacement of the missing pro‑
Duchenne muscular dystrophy (DMD) is a DMD. In‑frame deletions result in a milder tein can be effected by viral gene therapy,
relentless, X‑linked recessive muscle-wasting form of the disease, Becker muscular dys‑ although this strategy has had a cheq‑
disease with one of the highest known rates trophy (BMD), in which patients express a uered history when applied to other dis‑
of new mutations, meaning that many cases truncated, partially functional dystrophin. orders because of deaths in clinical trials
in the clinic have no previous family his‑ Symptoms of BMD range from loss of ambu‑ and immune reactions to viral proteins6.
tory 1. The gene responsible for the disease lation in the late teens to manifestation only However, this approach is now coming of
encodes the large cytoskeletal structural as a cardiomyopathy in patients who live age as more is understood about the viral
protein dystrophin, which is absent or pre‑ into their 80s. genome and the relationship with its host.
sent at very low levels in patients with DMD. No effective treatment exists for DMD. Adeno-associated virus (AAV) vectors in
Dystrophin is essential for muscle mem‑ Although there are numerous pharmaco‑ particular are providing spectacular results
brane stability as it provides an important logical strategies that delay symptoms by for eye disorders7, and expression of the
link between the dystrophin-associated pro‑ tackling the secondary effects of DMD, transgene has been reported in a haemophilia
tein complex (DAPC) at the muscle mem‑ many are only partially effective because B patient 10 years after gene transfer into
brane and the actin cytoskeleton2 (FIG. 1). they treat just one aspect of the pathogenesis, muscle8. In DMD, the challenge has been the
Boys affected by DMD are usually diagnosed and they may be toxic in the longer term4. large size of the mRNA (14 kb) and the need
at 3–5 years and go into a wheelchair at Thus there has been great interest in devel‑ to target all muscles. Dystrophin mini- or
about 12 years of age. Abnormal cardiac oping genetic approaches to treat the disease micro-genes (~6 kb) have been designed on
function observed by magnetic resonance that tackle the primary defect. This article the basis of the mutant form of the gene car‑
imaging (MRI) or echocardiography is usu‑ summarizes recent progress with these ried in a mildly affected patient with BMD.
ally seen in early teens but may be observed genetic approaches to therapy, including This form has a large in‑frame rod domain
as early as age 8. Patients usually die in their both the ‘traditional’ gene therapy approach deletion and fits into AAV vectors even when
20s from respiratory or heart failure, and of direct replacement of the protein, and muscle-specific promoters are incorporated9.
a minority of patients also show mental newer approaches involving the manipula‑ These mini-genes have been optimized to
impairment. tion of the cellular machinery at the level include motifs such as neuronal nitric oxide
The full-length dystrophin gene (FIG. 2Aa) of gene transcription, mRNA processing or synthase (nNOS) binding sites10.
is predominantly expressed in skeletal translation. The challenge is that any effec‑ Although in mice no cellular immune
and cardiac muscle with smaller amounts tive therapy will need to restore dystrophin responses against either the capsid proteins
expressed in the brain. Sixty-five per cent of in both skeletal and heart muscle, and the or transgene products have been observed
the mutations are deletions of the whole or treatment would need to be life-long. Studies using these mini-genes, studies in larger ani‑
a part of the gene. There are two hotspots of patients with X‑linked dilated cardio‑ mals and humans have resulted in variable
for deletions within the gene sequence3. myopathy show that dystrophin levels as immunological outcomes. In a recent clini‑
Deletions that leave the mRNA out‑of‑frame low as 30% in skeletal muscle are sufficient cal trial using AAV delivery to the biceps of
abolish protein production and lead to to prevent muscle weakness5. Nevertheless, a mini-dystrophin gene under the control

NATURE REVIEWS | GENETICS VOLUME 14 | JUNE 2013 | 373

© 2013 Macmillan Publishers Limited. All rights reserved


PROGRESS

α2 Laminin 2 of dystrophin protein was included as an


exploratory endpoint, no correlation has
β1 γ1 been reported between the 6MWT results
and the level of dystrophin expression.
Extracellular matrix
Ataluren was generally well tolerated. In the
Sarcoglycan αDG Dystroglycans
complex United States, all boys who participated in
the Phase IIb study have been in an exten‑
Sarcolemma δ γ β α βDG sion trial, and these are about to start in
Sarcospan other countries as well. It will be interesting
β1 Syntrophins to determine whether there is a cumulative
Cytoplasm β2 effect of treatment, such that more signifi‑
nNOS binding sites α
α-Dystrobrevin
cant clinical improvement will be seen at the
end of these extended trials.
Ataluren has demonstrated proof of prin‑
Actin filaments Dystrophin
ciple of reading through stop codons as an
approach to therapy, although the efficacy
needs to be improved to achieve higher lev‑
els of dystrophin and improved clinical out‑
comes. Other compounds that show greater
Figure 1 | The dystrophin-associated protein complex.  DystrophinNature acts asReviews
an important link
| Genetics efficacy are being reported but have yet to
between the internal cytoskeleton and the extracellular matrix. Neuronal nitric oxide synthase be tested in patient trials19. This approach
(nNOS) binds to α-syntrophin but also has a binding site in repeat 17 of the rod domain of dystrophin is being applied to other genetic disorders,
(see FIG. 2A for details of dystrophin domains). αDG, α-dystroglycan; βDG, β-dystroglycan. such as cystic fibrosis and spinal muscular
atrophy (SMA), as well as for the treatment
of adenomatous polyposis coli-associated
of the cytomegalovirus (CMV) promoter, events. In future trials, transient immuno‑ colorectal cancers, where it would be
an immune response directed against the suppression may prove to be highly effective applicable to 24% of cases20.
transgene product was observed11. However, for the systemic delivery of viral vectors,
a trial delivering an α‑sarcoglycan transgene as has been shown for the dog model of Exon skipping
to patients with limb girdle muscular dys‑ DMD14. The use of utrophin (FIG. 2Ad) rather Approximately 83% of all DMD mutations3,
trophy (LGMD) did not evoke an immune than dystrophin mini-genes would avoid the including most out‑of‑frame deletions, are
response. This may have been because the immune response to dystrophin. However, amenable to correction by the skipping of
transgene was under the control of a muscle- even if the immune response can be mini‑ specific exons21 (FIG. 2B). In this correction
specific promoter 12. In the DMD trial, it was mized, delivery to all muscles in the body strategy, RNaseH‑independent antisense
also proposed that rare dystrophin-positive remains an important challenge with viral oligonucleotides (AONs) hybridize to
revertant fibres, which may have resulted therapy for DMD. complementary sequences in or adjacent
from a mutation at a second site or from to the target exon, modulating precursor
alternative splicing, could express an immu‑ Termination codon read-through mRNA (pre-mRNA) splicing and achieving
nogenic epitope never previously seen by the Premature termination codon mutations reading frame restoration with the expres‑
patient 13. These data emphasize the impor‑ occur in ~15% of patients with DMD (see sion of a BMD-like truncated dystrophin
tance of pre-screening patients for immune Leiden Muscular Dystrophy Pages), and protein. Proof of principle has been estab‑
responses to dystrophin before admission therefore these boys could potentially be lished in Phase I and Phase II clinical trials
into trials. In addition, more needs to be treated by drugs that promote read-through. led by groups in the Netherlands (Leiden
understood regarding why certain patients Aminoglycosides restore protein transla‑ University Medical Centre in collaboration
have T cells that are activated in response to tion in cell assays, but many of these have with Prosensa–GlaxoSmithKline) and the
dystrophin epitopes. long-term toxic effects15. PTC Therapeutics United Kingdom (MDEX Consortium in
Little is known about how long dystro‑ have screened for drugs that promote read- collaboration with Sarepta Therapeutics),
phin expression will be sustained after a sin‑ through of stop codons in DMD cell lines targeting DMD exon 51, an approach
gle administration in humans, and repeated and the mdx mouse16. Ataluren (FIG. 2Ac) is that is applicable to ~13% of patients. The
treatment may be necessary. Clinical trials the first drug in this class, but initial human 2′-O-methyl-phosphorothioate (2′OMe) and
in patients with LGMD have highlighted trials have been disappointing. Dystrophin morpholino phosphorodiamidate oligonu‑
the need to be vigilant about pre-existing expression appeared to be increased in most cleotide (PMO) AONs used in these trials
immunity to the AAV capsid, although it is patients but no clear quantitative analyses were well-tolerated and restored dystrophin
possible to use different AAV serotypes in were presented for this Phase IIa clinical protein to variable degrees (up to 55%
successive treatments. study 17. The clinical endpoint for these trials dystrophin-positive fibres in one patient)
Although more work is needed on vector was the six-minute walk test (6MWT)17,18, when delivered via intramuscular 22,23 and
design and the use of muscle-specific pro‑ which is a standardized test of ambulation. systemic routes24,25. Further clinical trials are
moters to minimize any immune response, Data from the 48‑week Phase IIb study in progress to establish a long-term adminis‑
the local delivery trials have been very showed that treated patients walked on tration regimen, to evaluate clinical efficacy
informative and show that viral therapy average 30 metres further than patients on in pivotal studies and to extend this to other
is generally safe with no reported adverse placebo. However, although the evaluation DMD exons.

374 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/genetics

© 2013 Macmillan Publishers Limited. All rights reserved


PROGRESS

A Rod domain
a Full-length dystrophin Actin binding H1 R R R H2 R R R R R R R R R R R R R R R R H3 R R R R R H4 CRD CTD
1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 16 17 18 19 20 21 22 23 24

b ∆17–48 Actin binding H1 R R R R R R R R H4 CRD


H3 20 CTD
1 2 3 21 22 23 24
(BMD mutation)

∆D3990 Actin binding H1 R R R R R H4 CRD


1 2 H3 22 23 24
(Mini-dystrophin)

PTC
Rod domain
c Dystrophin-containing
nonsense mutation Actin binding H1 H2 H3 H4 CRD CTD

– Ataluren Shortened non-functional protein


mRNA
+ Ataluren

Rod domain
d Utrophin Actin binding H1 H2 H3 H4 CRD CTD

B U1 U2 SF2
snRNP snRNP U2 ASF
AF
a DMD 49 51 52
ESE ESS

U1 U2
snRNP AON snRNP U2
AF
b DMD and AON 49 51 52

49 52

Figure 2 | Dystrophin, utrophin and genetic approaches to therapy.  synthase (nNOS) to the sarcolemma, as is the case in some patients with
Aa | Wild-type dystrophin. Full-length dystrophin comprises an amino‑ Nature
BMD. B | The principles of antisense oligonucleotide Reviews | Genetics
(AON)-mediated skip-
terminal actin-binding domain, four hinge domains (H1–H4) and a rod ping of dystrophin exon 51. Ba | The Duchenne muscular dystrophy (DMD)
domain consisting of 24 spectrin-like repeats (R1–R24), within which lie locus in a patient with a deletion of exon 50. As a result of the deletion,
a second actin-binding domain, a cysteine-rich domain (CRD) and a exons 49 and 51 are out‑of‑frame, which leads to an unstable precursor
carboxy‑terminal domain (CTD). Ab | Mini-genes used in gene therapy. A mRNA (pre-mRNA) transcript and a lack of dystrophin protein. Also shown
deletion of dystrophin exons 17–48 was identified in a mildly affected in this region of the locus are some of the key cis- and trans-acting ele-
patient with Becker’s muscular dystrophy (BMD) and was shown to correct ments that regulate the splicing of the dystrophin pre-mRNA, including
95% of the dystrophic pathology in the Mdx mouse. A clinical trial has been U1 and U2 small nuclear RNAs (snRNAs), which define exon–intron bound-
carried out using mini-dystrophin ΔD3990, which consists of a truncated aries and also exon internal sequences, such as exonic splicing enhancers
protein that encodes the N‑terminal domain, the CRD and the rod domain, (ESEs) and exonic splicing silencers (ESSs) that promote and inhibit exon
but with a reduced number of spectrin repeats (namely, R1, R2, R22, R23 inclusion during pre-mRNA splicing, respectively. Bb | An AON-targeting
and R24) and three hinges (namely, H1, H3 and H4). Omitting the CTD was exon internal sequences within exon 51 adjacent to putative ESE sites may
not found to be crucial for function. Ac | Nonsense suppression. Ataluren inhibit the association of splicing regulatory proteins (for example, of the
allows read-through of premature termination codons (PTC) to restore SF2 and ASF protein families) at this recognition site and therefore pro-
production of a full-length functional dystrophin protein. Ad | Utrophin mote the skipping of this exon during pre-mRNA splicing. Skipping of exon
lacks sequence corresponding to spectrin-like repeats 15 and 19 of dystro- 51 is able to restore a viable mRNA reading frame as axons 49 and 52 are
phin and binds actin only through the N‑terminal domain. Utrophin and in‑frame exons, and therefore a truncated but semi-functional dystrophin
some of the dystrophin mini-genes will not localize neuronal nitric oxide isoform is generated. snRNP, small nuclear ribonucleoprotein.

NATURE REVIEWS | GENETICS VOLUME 14 | JUNE 2013 | 375

© 2013 Macmillan Publishers Limited. All rights reserved


PROGRESS

Table 1 | Clinical trials using genetic therapies for Duchenne’s muscular dystrophy
Drug name Description Company Delivery Results to date Current Clinical trial number Refs
route stage and/or URL*
Viral gene therapy
Biostrophin rAAV2.5‑CMV Asklepios IM Failed to establish long-term Phase I http://www.askbio. 11
mini-dystrophin Biopharmaceutical (biceps) dystrophin expression; (completed) com
(d3990) immune response against
transgene in 4 out of 6 patients
Termination codon read-through
Ataluren Nonsense PTC Therapeutics Oral Slowed loss of walking ability Phase III NCT01557400; http:// 17,
suppression in patients with DMD or BMD www.ptcbio.com 49
(n = 174) at the lower of two
doses tested
Exon skipping
Eteplirsen PMO morpholino Sarepta IV Well-tolerated and restored Phase II NCT00844597; 22,
(AVI‑4568) targeting exon 51 Therapeutics dystrophin expression in http://www. 24
7 out of 19 patients in a sareptatherapeutics.
dose-dependent manner com
(<20% normal levels)
GSK2402968 2′OMePS AON Prosensa– SC Restored dystrophin Phase III NCT01480245; 23,
(PRO051); targeting exon 51 GlaxoSmithKline expression in 10 out of 12 http://www.gsk.com; 25
Drisapersen patients (<20% normal levels) http://www.prosensa.
eu
PRO044 2′OMePS AON Prosensa SC or IV Study ongoing Phase I/IIa NCT01037309 50
targeting exon 44
Increasing levels of utrophin
SMT‑C1100 Utrophin Summit PLC Oral Safe, well-tolerated, achieved Phase I http://www. 42
modulator plasma levels shown to (completed) summitplc.com
increase utrophin in DMD
patient cells in vitro
*Where possible, the clinical trial identification number is given where the trial is ongoing or where data have not yet been published. 2′OMePS, 2′O‑methyl-
phosphorothioate; AAV, adeno-associated virus; AON, antisense oligonucleotide. BMD, Becker muscular dystrophy; CMV, cytomegalovirus; DMD, Duchenne
muscular dystrophy; IM, intramuscular; IV, intravenous; PMO, phosphorodiamidate morpholino oligomer; SC, subcutaneous.

Although progress is promising, the factors might improve outcome in certain implies that high-frequency mutations will
translation of exon-skipping therapies is cases. From initial clinical trials, it appears be addressed first and that some patients
complex, and caution in the interpretation that some patients might be ‘high responders’, with DMD who harbour lower-frequency
of current clinical trial data is warranted. although whether this has a genetic or mutations might remain untreated. These
First, the efficacy of exon-skipping AONs is stochastic basis is unknown24. Some muta‑ problems could be partially solved with
limited by poor cellular uptake, resulting in tions might be better targets for therapy as the advent of successful multi-exon skip‑
low and variable levels of dystrophin protein some DMD exons appear easier to skip27. ping, which has recently been demonstrated
restoration in skeletal muscle and little or Finally, prolonged treatment might improve in principle28. An additional issue is that
no restoration in heart muscle. A potential clinical outcome as may be the case with although first-generation AONs have
solution is the development of second- read-through of stop codons. acceptable safety profiles, long-term safety
generation AONs with improved skeletal However, further important caveats exist is currently unknown, and this is crucial
and cardiac muscle penetration26. Second, regarding the exon-skipping approach. when treatment is essentially life-long. More
the variable nature of the disease course and These include, first, that the restored generally, the lack of validated biomarkers to
of the current clinical outcome measures dystrophin protein is truncated and semi- monitor AON efficacy continues to hinder
necessitates that clinical trials are appropri‑ functional, and therefore at best the clinical DMD drug development, although
ately powered and double-blind and that the outcome is conversion to the corresponding candidate serum protein and microRNA
AON drug is evaluated against a placebo or BMD phenotype. Second, therapy is highly biomarkers have now been identified29–31.
a reference DMD natural history data set. personalized and mutation-specific, neces‑ Overall, although results targeting exon
These are demanding requirements for a sitating the clinical development of multiple 51 are promising, major hurdles in the clini‑
rare disease but are nonetheless important. AONs. Moreover, this mutation-specific cal development of exon-skipping AONs
The most likely outcome with first- requirement has direct cost and regula‑ remain that are likely to limit the effective‑
generation exon-skipping AONs is limited tory implications, given that each AON in ness of first-generation compounds. To
clinical efficacy. This constitutes progress, development is regarded by the US Food maximize the likelihood of success, efforts
but the level of efficacy would probably and Drug Administration (FDA) and other should focus on validating appropriate bio‑
fall short of what is required for a disease- regulatory agencies as a new drug. Third, markers, understanding factors related to
modifying therapy. However, a number of the fact that therapy is highly personalized individual responses to exon-skipping AON

376 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/genetics

© 2013 Macmillan Publishers Limited. All rights reserved


PROGRESS

compounds, advancing second-generation Utrophin is more highly expressed in slow medicine), and advances in technology have
AONs and establishing an accelerated regu‑ fibres, and therefore the promotion of the made many of the problems with therapeutic
latory process. Although success to date is slow oxidative myogenic programme may approaches more tractable. Many challenges
partial, the progress already made is having be beneficial. This has been demonstrated remain, however, not least for the develop‑
an impact on the development of AON- through the activation of the calcineurin– ment of reliable endpoints for clinical trials
based therapies for related neuromuscular NFAT (nuclear factor of activated T cells) in view of the variability in the 6MWT. The
disorders, including SMA and myotonic pathway 44 and by increasing peroxisome problem of delivery to all muscles of the
dystrophy 21. Beyond such diseases, the use of proliferator-activated receptor‑γ coactivator body will also need to be resolved. However,
AONs to modulate pre-mRNA splicing has 1α (PGC1α)45. However, PGC1α regulates there is little doubt that although a cure
also been successfully extended to cancer, in the neuromuscular junction programme and remains some way off, treatments are cur‑
which disrupted alternative splicing com‑ therefore may be affecting other pathways rently entering trials that have the potential
monly occurs and in which redirecting splic‑ in addition to increasing utrophin levels. for providing a significant clinical impact on
ing of, for example, signal transducer and Other pharmacological activators of oxida‑ the quality of life of patients. In turn, these
activator of transcription 3 (STAT3) can lead tive metabolism may also be helpful, and approaches are being applied to many other
to a favourable anti-oncogenic outcome32. some are in clinical trials for other disorders, genetic disorders. The age of genomic medi‑
such as diabetes4. Summit plc has recently cine is moving forwards with rapid speed.
Increasing levels of utrophin reported positive data from a Phase I trial of Rebecca J. Fairclough, Matthew J. Wood and Kay
The dystrophin-related protein utrophin SMT C1100, which is a drug derived from E. Davies are at the Department of Physiology,
shows sequence and structural similarity a high-throughput transcription screen and Anatomy and Genetics, University of Oxford, Parks
to dystrophin and can functionally com‑ which increases utrophin RNA and protein Road, Oxford OX1 3PT, UK.

pensate for the lack of dystrophin in the up to twofold in some muscles42. This is now Rebeccan J. Fairclough and Kay E. Davies are also at
mdx mouse4,33,34. Utrophin is restricted to proceeding to Phase II. It is worth noting the MRC Functional Genomics Unit, MRC Functional
Genomics Unit, Department of Physiology, Anatomy
neuromuscular and myotendinous junc‑ that, in the wild-type mouse, very high levels
and Genetics, Parks Road, Oxford OX1 3PT, UK.
tions in adult muscle, but during embryonic of utrophin are observed in the kidneys and
development and in patients with DMD, it lungs, suggesting that high levels are possible Correspondence to K.E.D. 
e-mail: kay.davies@dpag.ox.ac.uk
is localized at the sarcolemma when dystro‑ in adult tissue.
doi:10.1038/nrg3460
phin is absent or present only at low levels. Increasing utrophin should be effective in
Published online 23 April 2013
Studies in animal models have provided all patients with DMD, irrespective of their
compelling evidence that utrophin func‑ dystrophin mutation and will circumvent 1. Emery, A. E. H. Muscular Dystrophy (The Facts)
(Oxford Univ. Press, 2008).
tions in these scenarios directly to protect immunological challenges faced by 2. Cohn, R. D. & Campbell, K. P. Molecular basis of
the muscle against dystrophic degeneration. dystrophin-based therapies13. Although none muscular dystrophies. Muscle Nerve 23, 1456–1471
(2000).
However, utrophin does not anchor nNOS to of the utrophin-based approaches developed 3. Aartsma-Rus, A. et al. Theoretic applicability of
the sarcolemma, which is a requirement so far has achieved the two- to fourfold antisense-mediated exon skipping for Duchenne
muscular dystrophy mutations. Hum. Mutat. 30,
to regulate blood flow to the muscle and to increase that has been set as the benchmark 293–299 (2009).
ensure that all of its metabolic needs are for preclinical rescue46, the continued clinical 4. Fairclough, R. J., Perkins, K. J. & Davies, K. E.
Pharmacologically targeting the primary defect and
met. This issue may also be encountered in evaluation of SMT C1100 may provide the downstream pathology in Duchenne muscular
strategies that are based on the skipping of first insight into therapeutic levels required dystrophy. Curr. Gene Ther. 12, 206–244 (2012).
5. Neri, M. et al. Dystrophin levels as low as 30% are
particular exons and for some mini-genes. in patients. A recent study has reported in sufficient to avoid muscular dystrophy in the human.
The implications of this are uncertain, given the mdx mouse that as little as 4% of wild- Neuromuscul. Disord. 17, 913–918 (2007).
6. Cavazzana-Calvo, M. & Fischer, A. Gene therapy for
that there may be compensatory pathways type dystrophin levels may be sufficient severe combined immunodeficiency: are we there yet?
for the lack of nNOS and that several mildly for significant survival and motor function J. Clin. Invest. 117, 1456–1465 (2007).
7. Vandenberghe, L. H. & Auricchio, A. Novel adeno-
affected patients with BMD have large dele‑ improvement, emphasizing the need to take associated viral vectors for retinal gene therapy. Gene
tions that also lack nNOS binding sites9,35,36. drugs such as SMT C1100 into patients for Ther. 19, 162–168 (2012).
8. Buchlis, G. et al. Factor IX expression in skeletal
In the mdx mouse, an increase in utrophin evaluation47. The change of expression of a muscle of a severe hemophilia B patient 10 years after
protein in all tissues, which is sufficient related or similar protein was first used for AAV-mediated gene transfer. Blood 119, 3038–3041
(2012).
for complete rescue, does not cause the treatment of β‑thalassaemia, in which 9. Pichavant, C. et al. Current status of pharmaceutical
toxicity 37. Encouragingly, small differences increased levels of fetal haemoglobin were and genetic therapeutic approaches to treat DMD.
Mol. Ther. 19, 830–840 (2011).
in utrophin protein levels between patients used to compensate for the lack of the adult 10. Lai, Y. et al. Dystrophins carrying spectrin-like repeats
with DMD positively correlate with the age β-globin gene48. Such an approach could be 16 and 17 anchor nNOS to the sarcolemma and
enhance exercise performance in a mouse model of
of confinement to a wheelchair, demon‑ applicable to many other disorders. muscular dystrophy. J. Clin. Invest. 119, 624–635
strating that even small increases might be (2009).
11. Mendell, J. R. et al. Gene therapy for muscular
beneficial38. Conclusion dystrophy: lessons learned and path forward.
Utrophin levels can be increased by Five years ago, effective treatment for DMD Neurosci. Lett. 527, 90–99 (2012).
12. Mendell, J. R. et al. Sustained α-sarcoglycan
direct delivery of the protein39, by stabi‑ seemed an impossible dream, and few phar‑ gene expression after gene transfer in limb-girdle
lization of the protein or RNA40,41 or by maceutical companies were interested in muscular dystrophy, type 2D. Ann. Neurol. 68,
629–638 (2010).
transcriptional upregulation42. Agents such investing in therapy for a fairly rare orphan 13. Mendell, J. R. et al. Dystrophin immunity in
as biglycan that stabilize the DAPC hold disease. However, this has all changed, as Duchenne’s muscular dystrophy. N. Engl. J. Med. 363,
1429–1437 (2010).
therapeutic promise, and clinical trials DMD is now seen as a model for the devel‑ 14. Wang, Z. et al. Successful regional delivery and
are planned43, but whether this approach opment of treatment on the basis of the long-term expression of a dystrophin gene in
canine muscular dystrophy: a preclinical model for
will result in sufficient utrophin levels for specific mutation or mutations that are pre‑ human therapies. Mol. Ther. 20, 1501–1507
therapeutic benefit remains unknown. sent in an individual (that is, personalized (2012).

NATURE REVIEWS | GENETICS VOLUME 14 | JUNE 2013 | 377

© 2013 Macmillan Publishers Limited. All rights reserved


PROGRESS

15. Bidou, L., Allamand, V., Rousset, J. P. & Namy, O. Sense 30. Roberts, T. C. et al. Expression analysis in multiple 44. Angus, L. M. et al. Calcineurin-NFAT signaling,
from nonsense: therapies for premature stop codon muscle groups and serum reveals complexity in the together with GABP and peroxisome PGC‑1α, drives
diseases. Trends Mol. Med. 18, 679–688 (2012). microRNA transcriptome of the mdx mouse with utrophin gene expression at the neuromuscular
16. Welch, E. M. et al. PTC124 targets genetic disorders implications for therapy. Mol. Ther. Nucleic Acids 1, junction. Am. J. Physiol. Cell Physiol. 289,
caused by nonsense mutations. Nature 447, 87–91 e39 (2012). C908–C917 (2005).
(2007). 31. Nadarajah, V. D. et al. Serum matrix 45. Handschin, C. et al. PGC‑1α regulates the
17. Finkel, R. S. Read-through strategies for suppression metalloproteinase‑9 (MMP‑9) as a biomarker neuromuscular junction program and ameliorates
of nonsense mutations in Duchenne/Becker muscular for monitoring disease progression in Duchenne Duchenne muscular dystrophy. Genes Dev. 21,
dystrophy: aminoglycosides and ataluren (PTC124). muscular dystrophy (DMD). Neuromuscul. Disord. 21, 770–783 (2007).
J. Child Neurol. 25, 1158–1164 (2010). 569–578 (2011). 46. Tinsley, J. et al. Expression of full-length utrophin
18. McDonald, C. M. et al. The 6‑minute walk test as a 32. Zammarchi, F. et al. Antitumorigenic potential of prevents muscular dystrophy in mdx mice. Nature
new outcome measure in Duchenne muscular STAT3 alternative splicing modulation. Proc. Natl Med. 4, 1441–1444 (1998).
dystrophy. Muscle Nerve 41, 500–510 (2010). Acad. Sci. USA 108, 17779–17784 (2011). 47. van Putten, M. et al. Low dystrophin levels increase
19. Kayali, R. et al. Read-through compound 13 restores 33. Khurana, T. S. & Davies, K. E. Pharmacological muscle survival and improve muscle pathology
dystrophin expression and improves muscle function strategies for muscular dystrophy. Nature Rev. Drug and function in dystrophin/utrophin double-knock-
in the mdx mouse model for Duchenne muscular Discov. 2, 379–390 (2003). out mice. FASEB J. 4 Mar 2013 (doi:10.1096/
dystrophy. Hum. Mol. Genet. 21, 4007–4020 (2012). 34. Miura, P. & Jasmin, B. J. Utrophin upregulation for fj.12‑224170).
20. Bordeira-Carrico, R., Pego, A. P., Santos, M. & treating Duchenne or Becker muscular dystrophy: 48. Gambari, R. & Fibach, E. Medicinal chemistry
Oliveira, C. Cancer syndromes and therapy by stop- how close are we? Trends Mol. Med. 12, 122–129 of fetal hemoglobin inducers for treatment of
codon readthrough. Trends Mol. Med. 18, 667–678 (2006). β-thalassemia. Curr. Med. Chem. 14, 199–212
(2012). 35. Li, D. et al. Sarcolemmal nNOS anchoring reveals a (2007).
21. Muntoni, F. & Wood, M. J. Targeting RNA to treat qualitative difference between dystrophin and 49. Hirawat, S. et al. Safety, tolerability, and
neuromuscular disease. Nature Rev. Drug Discov. 10, utrophin. J. Cell Sci. 123, 2008–2013 (2010). pharmacokinetics of PTC124, a nonaminoglycoside
621–637 (2011). 36. Ramachandran, J. et al. Nitric oxide signaling pathway nonsense mutation suppressor, following single- and
22. Kinali, M. et al. Local restoration of dystrophin in Duchenne muscular dystrophy mice: upregulation of multiple-dose administration to healthy male
expression with the morpholino oligomer AVI‑4658 in l‑arginine transporters. Biochem. J. (2012). and female adult volunteers. J. Clin. Pharmacol. 47,
Duchenne muscular dystrophy: a single-blind, placebo- 37. Fisher, R. et al. Non-toxic ubiquitous over-expression 430–444 (2007).
controlled, dose-escalation, proof‑of‑concept study. of utrophin in the mdx mouse. Neuromuscul. Disord. 50. Heemskerk, H. A. et al. In vivo comparison of
Lancet Neurol. 8, 918–928 (2009). 11, 713–721 (2001). 2′‑O‑methyl phosphorothioate and morpholino
23. van Deutekom, J. C. et al. Local dystrophin restoration 38. Kleopa, K. A., Drousiotou, A., Mavrikiou, E., antisense oligonucleotides for Duchenne muscular
with antisense oligonucleotide PRO051. N. Engl. Ormiston, A. & Kyriakides, T. Naturally occurring dystrophy exon skipping. J. Gene Med. 11, 257–266
J. Med. 357, 2677–2686 (2007). utrophin correlates with disease severity in Duchenne (2009).
24. Cirak, S. et al. Exon skipping and dystrophin restoration muscular dystrophy. Hum. Mol. Genet. 15,
in patients with Duchenne muscular dystrophy after 1623–1628 (2006). Acknowledgements
systemic phosphorodiamidate morpholino oligomer 39. Sonnemann, K. J. et al. Functional substitution by The authors are very grateful to the UK Medical Research
treatment: an open-label, phase 2, dose-escalation TAT-utrophin in dystrophin-deficient mice. PLoS Med. Council, the Wellcome Trust, the UK Muscular Dystrophy
study. Lancet 378, 595–605 (2011). 6, e1000083 (2009). Campaign, the US Muscular Dystrophy Association, Action
25. Goemans, N. M. et al. Systemic administration of 40. Chakkalakal, J. V., Miura, P., Belanger, G., Duchenne, the Association Française Contre les Myopathies
PRO051 in Duchenne’s muscular dystrophy. N. Engl. Michel, R. N. & Jasmin, B. J. Modulation of utrophin A and the International Consortium on Exon Skipping for
J. Med. 364, 1513–1522 (2011). mRNA stability in fast versus slow muscles via an support.
26. Betts, C. A., Hammond, S. M., Yin, H. F. & Wood, M. J. AU‑rich element and calcineurin signaling. Nucleic
Optimizing tissue-specific antisense oligonucleotide- Acids Res. 36, 826–838 (2008). Competing interests statement
peptide conjugates. Methods Mol. Biol. 867, 41. Moorwood, C., Soni, N., Patel, G., Wilton, S. D. & The authors declare competing financial interests: see Web
415–435 (2012). Khurana, T. S. A cell-based high-throughput screening version for details.
27. Mitrpant, C. et al. Rational design of antisense assay for posttranscriptional utrophin upregulation.
oligomers to induce dystrophin exon skipping. J. Biomol. Screen. 18, 400–406 (2012).
Mol. Ther. 17, 1418–1426 (2009). 42. Tinsley, J. M. et al. Daily treatment with SMTC1100, FURTHER INFORMATION
28. Aoki, Y. et al. Bodywide skipping of exons 45–55 in a novel small molecule utrophin upregulator, Matthew Wood’s homepage: http://www.dpag.ox.ac.uk/
dystrophic mdx52 mice by systemic antisense delivery. dramatically reduces the dystrophic symptoms in the academic_staff/matthew_wood
Proc. Natl Acad. Sci. USA 109, 13763–13768 (2012). mdx mouse. PLoS ONE 6, e19189 (2011). MRC Functional Genomics Unit homepage: http://www.
29. Cacchiarelli, D. et al. miRNAs as serum biomarkers for 43. Amenta, A. R. et al. Biglycan recruits utrophin to the mrcfgu.ox.ac.uk
Duchenne muscular dystrophy. EMBO Mol. Med. 3, sarcolemma and counters dystrophic pathology in mdx ALL LINKS ARE ACTIVE IN THE ONLINE PDF
258–265 (2011). mice. Proc. Natl Acad. Sci. USA 108, 762–767 (2011).

378 | JUNE 2013 | VOLUME 14 www.nature.com/reviews/genetics

© 2013 Macmillan Publishers Limited. All rights reserved

You might also like