Review Article Antisense oligonucleotides for the treatment of cardiomyopathy in Duchenne muscular dystrophy

Page created by Dwight Sanders
 
CONTINUE READING
Review Article Antisense oligonucleotides for the treatment of cardiomyopathy in Duchenne muscular dystrophy
Am J Transl Res 2019;11(3):1202-1218
www.ajtr.org /ISSN:1943-8141/AJTR0085739

Review Article
Antisense oligonucleotides for the treatment of
cardiomyopathy in Duchenne muscular dystrophy
Quynh Nguyen1, Toshifumi Yokota1,2
1
 Department of Medical Genetics, Faculty of Medicine and Dentistry, University of Alberta, 8812-112 St.,
Edmonton, AB T6G 2H7, Canada; 2The Friends of Garret Cumming Research and Muscular Dystrophy Canada HM
Toupin Neurological Science Research Chair, 8812-112 St., Edmonton, AB T6G 2H7, Canada
Received September 19, 2018; Accepted January 2, 2019; Epub March 15, 2019; Published March 30, 2019

Abstract: Duchenne muscular dystrophy (DMD) is an X-linked recessive fatal neuromuscular disorder characterized
by progressive muscle degeneration which affects one in 3500-5000 males born worldwide. DMD is caused by
loss-of-function mutations in the dystrophin (DMD) gene encoding for dystrophin, a cytoskeletal protein that sup-
ports the structural integrity of myofibers during cycles of muscle contraction and relaxation. DMD patients do not
only experience skeletal muscle deterioration but also severe cardiomyopathy, which is recognized as the current
leading cause of death for the disease. Among the therapies being developed, exon skipping using antisense oligo-
nucleotides (AOs) is one of the most promising approaches. AOs effectively restore dystrophin expression in skeletal
muscles; however, they are highly inefficient in the heart due to endosomal entrapment. Improving skeletal muscle
function without restoring dystrophin expression in cardiac tissue may exacerbate cardiomyopathy due to increased
voluntary activity. This review consolidates the preclinical antisense approaches to improve dystrophin restoration,
with a special focus on the heart.

Keywords: Duchenne muscular dystrophy, dystrophin, cardiomyopathy, antisense therapy, morpholinos, drug de-
livery

Introduction                                                  damage, patients with DMD have markedly ele-
                                                              vated serum levels of creatine kinase (CK), even
Duchenne muscular dystrophy (DMD) is an                       at birth [11]. Death usually occurs in their 20s
X-linked recessive disorder characterized by                  due to respiratory or cardiac complications
progressive muscle degeneration [1]. The prev-                [11-14].
alence of DMD is estimated to be 1 in 3500-
5000 live male births, making it the most com-                DMD is caused by mutations in the DMD gene,
mon lethal neuromuscular disorder [2, 3]. Pa-                 which encodes a membrane-associated protein
tients with DMD generally stay asymptomatic at                called dystrophin. Dystrophin has 4 domains:
birth although they could show signs of delayed               an actin-binding N-terminal domain, a rod do-
gross motor development compared to their                     main consisting of 24 spectrin-like repeat
peers [4-6]. The disease progresses rapidly,                  motifs for structural flexibility, a cysteine-rich
with muscle weakness and wasting observed                     domain for facilitating protein-protein interac-
first in the proximal muscles then extending to               tion, and a C-terminal domain for binding sarco-
the more distal muscles [7]. Affected boys                    lemmal proteins [15, 16]. As a member of the
often lose ambulation by the age of 12 and                    dystrophin-glycoprotein complex (DGC), dystro-
experience multiple organ system dysfunction                  phin functions to link the actin cytoskeleton of
such as neuromuscular scoliosis, joint contrac-               muscle cells to the extracellular matrix, provid-
ture, osteoporosis, restrictive lung disease,                 ing mechanical support and membrane stabili-
obstructive sleep apnea, cardiomyopathy, and                  zation during muscle contraction and relaxation
psychological problems [8-10]. The age of                     cycles [15, 17-20]. In the absence of dystrophin,
onset and the rate of decline are highly variable             muscle fibers experience mechanical stress
among patients. Due to continuous muscle                      and become susceptible to tearing and frag-
Antisense therapy for DMD cardiomyopathy

mentation, resulting in degeneration [21, 22].      has been pushed into the late 20s [12, 13].
Other studies have suggested that dystrophin        Since respiratory dysfunction is now better
also serves non-mechanical roles [23].              managed, cardiomyopathy is presently the lea-
                                                    ding cause of death among DMD patients.
The DMD gene is located on the short arm of         Cardiomyopathy is estimated to present in 25%
the X chromosome (Xp21.3-p21.2) spanning            of patients at the age of 6, and 59% of patients
2.4 Mb with 79 exons which produces a 14 kb         at the age of 10 [27, 46]. The majority of
transcript [24-26]. The DMD gene has 3 main         patients will have developed cardiomyopathy
promoters that produce full-length dystrophin       by 18 years of age. Recognition of signs and
(M, B, and P), with other promoters responsible     symptoms of cardiac dysfunction can be chal-
for the transcription of different, smaller iso-    lenging since DMD patients are usually wheel-
forms [27, 28]. The M promoter produces the         chair-bound and do not perform increased car-
Dp427m isoform which is expressed in cardiac        diac workload [47]. The correlation between a
and skeletal muscle, the B promoter produces        patient’s genotype and the severity of their car-
the Dp427c isoform which is expressed in neu-       diac phenotype remains uncertain [48-52].
rons of the cortex and the Cornu Ammonis (CA)
region of the hippocampus, and the P promoter       The earliest clinical signs of cardiomyopathy
produces the Dp427p isoform which is ex-            are either decreased systolic function or sinus
pressed in the Purkinje cells of the brain. Other   tachycardia, with the latter commonly seen in
shorter isoforms include Dp260 expressed in         teenage DMD patients [53, 54]. Sinus tachycar-
the retina, Dp140 expressed in the kidney and       dia results in elevated heart rate, which in-
brain, Dp116 expressed in the peripheral            creases the workload of the already deteriorat-
nerves and Dp71 which is ubiquitously ex-           ing dystrophic myocardium [55-57]. Arrhyth-
pressed [29-33].                                    mias are a common cardiac involvement in
                                                    patients with DMD and are significantly corre-
DMD is considered one of the largest genes in
                                                    lated with decreased heart function. App-
the human genome. Furthermore, it is located
                                                    roximately 44% of DMD patients show signs of
in a genomic region with high rates of recombi-
                                                    arrhythmias [58]. Supraventricular tachycardia
nation [34-36]. Due to its length and location,
                                                    (SVT) and ventricular tachycardia (VT) are
DMD is highly susceptible to mutation. Ap-
                                                    observed in 10% of individuals with DMD [58].
proximately 60% of DMD cases are due to dele-
                                                    Although DMD cardiomyopathy is routinely
tions of one or more exons, ~6% due to duplica-
tions, and the rest due to small insertions/dele-   described as dilated cardiomyopathy (DCM),
tions or point mutations [11, 16, 37, 38]. These    patients with decreased systolic or diastolic
mutations usually disrupt the reading frame or      function do not necessarily show ventricular
introduce a premature stop codon, both of           dilation [49, 59, 60]. Additionally, myocardial
which lead to the absence of dystrophin. There      fibrosis usually precedes left ventricular dys-
is no significant correlation between mutation      function (LVD) [61, 62]. The age of onset and
size and disease severity [39-41]. Most muta-       the rate of progression are variable for LVD.
tions that maintain the DMD reading frame pro-      However, patients with onset of LVD before 18
duce truncated yet partly functional dystrophin,    years of age were found to have a significantly
and often result in a milder form of the disease    shorter lifespan compared to those who had
known as Becker muscular dystrophy (BMD)            LVD after 18 years old [48]. Other features of
[42-44]. Although the skeletal muscle symp-         DMD cardiomyopathy include abnormal cardiac
toms are less severe, the majority of BMD           conduction (as caused by, for example, vacuole
patients also develop cardiomyopathy, and it is     degeneration in the Purkinje fibers), congestive
the leading cause of death in patients with         heart failure and sudden cardiac arrest [27, 28,
BMD [45].                                           47, 63-66].

Cardiomyopathy in DMD patients                      Myocardial issues in DMD patients are unavoid-
                                                    able since dystrophin serves the same func-
The life expectancy for patients with DMD used      tions in cardiomyocytes as in skeletal muscle
to be less than 20 years of age just two decades    cells [67]. In the absence of dystrophin, cardio-
ago. Due to advancements in ventilator support      myocytes experience increased structural vul-
and spinal surgery, the average age of mortality    nerability and are susceptible to membra-

1203                                                       Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

Figure 1. Antisense-induced exon skipping in DMD. Deletion of exon 49 and 50 in the DMD gene creates a frame-
shift mutation which results in a premature stop codon and no dystrophin produced. In the case shown, AOs can
specifically bind to exon 51 and interfere with the splicing machinery to exclude exon 51 from the mature mRNA,
thereby restoring the reading frame and producing truncated but partly functional dystrophin.

ne instability and tearing [22]. Cardiomyocyte              other interventions such as ventricular assist
remodeling occurs secondary to myocardial                   devices (VADs) or cardiac transplantation [9,
wasting, which leads to ventricular enlarge-                47, 82-92]. Treatment with corticosteroids has
ment and fibrosis [68]. As previously men-                  been shown to improve cardiac function. DMD
tioned, affected hearts do not always show                  patients who received steroids such as pred-
signs of ventricular dilation. Another conse-               nisolone/prednisone or deflazacort had signifi-
quence of myocardial tearing and fragmenta-                 cantly delayed onset of cardiomyopathy, less
tion is the dysregulation of ion influx/efflux in           ventricular dilation and systolic dysfunction
the heart [69, 70]. Notably, damages to the                 than those who did not. However, patients
myocardial membrane disrupt Ca2+ homeosta-                  using corticosteroids can experience some
sis. Since Ca2+ acts as a second messenger in               long-term complications such as weight gain,
many cellular pathways, Ca2+ dysregulation re-              bone fractures, cataracts, etc. [9, 10, 93-96],
sults in multiple downstream abnormalities                  which limits their use for therapy. ACEIs and
within myocardial tissue. One significant conse-            ARBs have proven effective in reducing left ven-
quence of Ca2+ dysregulation includes increa-               tricular hypertrophy and fibrosis. Furthermore,
sed reactive oxygen species (ROS) production                initiation of beta-adrenergic receptor blocker
and mitochondrial dysfunction. Elevated Ca2+                treatment along with ACEIs has been demon-
levels within cardiomyocytes activate the mito-             strated to improve left ventricular systolic func-
chondria to produce ROS which leads to apop-                tion in DMD patients [88, 91, 97]. However,
tosis and further muscle wasting [69-79]. An                most of these studies have their inherent limi-
important second messenger affected by Ca2+                 tations, which makes it challenging for data
dysregulation is nitric oxide (NO) which has                interpretation as well as treatment application
roles in blood flow and vasoregulation [80, 81].            in clinical settings.
Increased intracellular Ca2+ concentrations
also enhance the risk of Ca-dependent arrhyth-              Antisense oligonucleotides for the treatment
mias and cellular Ca2+ overload in DMD patients             of DMD
[69].
                                                            At present, there is no cure for DMD. Current
Cardiomyopathy in DMD patients is currently                 treatments are mostly palliative, aiming at alle-
not curative. Therapies commonly used include:              viating the symptoms of the disease [11, 14].
corticosteroids, angiotensin-converting enzyme              Several promising approaches have been in-
inhibitors (ACEIs), angiotensin II receptor block-          vestigated such as utrophin up-regulation, viral
ers (ARBs), beta-adrenergic receptor blockers,              gene therapy, cell-based therapy, antisense oli-
mineralocorticoid receptor antagonists and                  gonucleotides (AOs) for exon skipping and,

1204                                                               Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

Figure 2. Chemical structures commonly used in antisense therapy. (A) RNA, (B) 2’-OMe-phosphorothioate, (C) Tricy-
clo-DNA, (D) Phosphorodiamidate morpholino oligomers. X or Y can be a peptide; Y can be an octaguanidine moiety
(E) as seen in Vivo-morpholino.

most recently, CRISPR-Cas9-mediated gene                     sistance to nuclease degradation, but also
editing [69]. In this review, we will discuss the            enhance their pharmacological properties. Al-
use of AOs in the treatment of DMD with a spe-               terations at the 2’ position of the ribose sugar
cific focus on treating the heart.                           have yielded a class of AOs with improved safe-
                                                             ty and efficacy profiles. 2’-O-methyl (2’-OMe)
AOs are short, synthetic nucleic acid sequenc-               and 2’-O-methoxyethyl (2’-MOE) alkyl substitu-
es which bind to complementary target mRNA                   ents are the most studied members of this
sequences and lead to either endonuclease-                   group. In addition, the use of a phosphorothio-
mediated transcript knockdown or splice mod-                 ate backbone which contains a sulfur in a non-
ulation [98, 99]. AO-mediated exon skipping                  bridging location on the phosphate backbone
can correct the reading frame by removing an                 significantly improves AO nuclease resistance
out-of-frame exon or exons from the DMD pre-                 and binding affinity to serum proteins [98, 107-
mRNA, producing a truncated but partly func-                 109]. Antisense chemistries that contain both
tional dystrophin protein [100-102] (Figure 1).              a phosphorothioate backbone and a 2’-alkyl
The first generation of AOs has the unmodified               substituent, such as 2’-OMe-phosphorothioate
phosphoribose backbone, making them highly                   (2’OMePS) AOs, have shown several favorable
susceptible to degradation by nucleases [103,                properties compared to their unmodified coun-
104] (Figure 2). Due to their short half-life, first         terparts [105, 110-113]. The phosphodiester
generation AOs rarely achieve sufficient intra-              backbone can also be replaced with a polyam-
cellular concentrations to have a therapeutic                ide structure made up of repeating N-(2-
effect [105, 106]. Second and third generation               aminoethyl) glycine units, which has given rise
AOs contain chemically modified structures.                  to another class of AOs termed peptide nucleic
These modifications not only increase AO re-                 acids (PNAs) [114]. This new class of AO can

1205                                                                 Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

sterically block splicing factors, inhibit ribo-     and multi-vesicular bodies (MVBs), and lyso-
some recruitment and bind non-coding RNAs,           somes. Most AOs are initially delivered to early
thereby further expanding therapeutic potential      endosomes. Subsequently, they can be traf-
[69].                                                ficked to lysosomes for degradation or seques-
                                                     tered in late endosomes/MVBs. Antisense dru-
Among these next-generation chemistries, ph-         gs trapped within endomembrane compart-
osphorodiamidate morpholino oligomers (PM-           ments are pharmacologically inactive as they
Os) represent the most advanced use of anti-         remain separated from their target sites by
sense therapy for DMD. PMOs have the deoxy-          membrane barriers [106, 129]. Nevertheless,
ribose/ribose moiety replaced by a morpholine        a small portion of AOs escapes endosomes and
ring, and the charged phosphodiester inter-          only then can they reach their targets and
subunit linkage replaced by an uncharged             become pharmacologically active. The concept
phosphorodiamidate linkage, making PMOs              of endosomal escape has been widely accept-
nuclease-resistant and charge-neutral [115,          ed as the most important barrier to the effec-
116]. One of the challenges with using nucleic       tive use of antisense drugs [127, 130]. Im-
acid-based molecules as therapeutics is their        portantly for developing heart-effective AOs,
stability and potential toxicity in cells. The       the research found that AOs (particularly PMOs)
advantage of being charge-neutral, as in the         tend to be trapped in endosomes in cardiomyo-
case of PMOs, is that this imparts an even           cytes more than in skeletal muscle cells [131].
greater resistance to nucleases, which typically     Since cardiomyopathy is the leading cause of
target charged molecules. Additionally, due to       death in DMD patients, researchers have
the lack of charge, PMOs are safer since they        looked into different approaches to enhance
are unlikely to activate Toll-like receptors, a      delivery of AOs to cardiac tissues. The next sec-
class of receptors involved in producing innate      tion will discuss some of the most promising
immune responses against pathogenic materi-          approaches to improving AO efficacy for treat-
al [117]. In fact, a PMO targeting DMD exon 51,      ing the heart.
called eteplirsen or Exondys 51 (Sarepta
Therapeutics, Cambridge, MA, USA), was condi-        Strategies to improve the efficacy of antisense
tionally approved by the U.S. Food and Drug          drugs for the treatment of cardiomyopathy in
Administration (FDA) in 2016, and several            DMD patients
PMOs targeting other DMD exons, including
golodirsen or SRP-4053 (Sarepta Therapeutics)        Tricyclo-DNA
and NS-065/NCNP-01 (NS Pharma, Paramus,
NJ, USA) are currently undergoing clinical trials    In the late nineties, a novel class of AOs known
[118-121].                                           as tricyclo-DNAs (tcDNAs) was shown to display
                                                     unique pharmacological properties. tcDNA is a
Although AOs effectively rescue dystrophin           conformationally-constrained DNA analog that
expression in skeletal muscles, they are highly      deviates from the natural DNA structure by the
inefficient in cardiac muscles [122, 123].           presence of an ethylene bridge connecting C3’
Failure to restore dystrophin expression in the      and C5’ [132-134]. tcDNA shows enhanced
heart in the presence of rescued dystrophin in       RNA affinity and nuclease resistance, as well
skeletal muscles can exacerbate cardiac issues       as the inability to elicit RNase H activity [135-
due to increased patient voluntary activity          138]. Furthermore, tcDNAs can spontaneously
[124, 125]. The major barrier to effective use of    form defined nanoparticles which potentia-
AOs in therapy is the delivery of antisense drugs    lly improves delivery and cellular uptake com-
to their intracellular targets. Upon reaching the    pared to other antisense chemistries [139-
cell surface, AOs are internalized by endocyto-      141].
sis. A variety of cell-surface receptors have
been suggested to bind AOs and facilitate their      In a study by Goyenvalle and colleagues, a
entry into the cell including integrins, scavenger   mouse model carrying a nonsense mutation in
receptors and Toll-like receptors [126-128].         exon 23 of the Dmd gene (hereafter referred to
Regardless of the endocytosis route taken, AOs       as mdx mice) was injected intravenously with a
entering a cell will encounter an intricate net-     15 nucleotide long (15-mer) tcDNA-AO that tar-
work of membrane compartments that include           gets the skipping of Dmd exon 23 [139].
early and recycled endosomes, late endosomes         Treatment with 200 mg/kg/week of the drug

1206                                                        Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

for 12 weeks rescued up to 40% of wild type          experiments are mentioned in this paper, but a
(WT) dystrophin expression in the heart.             selected few will be discussed for each
Additionally, tcDNA-treated mice showed signif-      approach.
icantly higher levels of dystrophin rescue, espe-
cially in the diaphragm and the heart, com-          The (RXR)4 peptide (with R standing for arginine
pared to those treated with PMO and 2’-OMe           and X standing for aminohexanoic acid) was the
AOs at an equimolar dosage. Similar results          first arginine-rich CPP tested in the mdx model
were observed in a dystrophin/utrophin double        of DMD [146]. In a study by Yin and colleagues,
knockout mouse model where treatment with            (RXR)4 peptides were conjugated to PMOs tar-
weekly intravenous (IV) injections of 200 mg/        geting Dmd exon 23. A single IV injection at 25
kg of tcDNA over a period of 5-20 weeks              mg/kg via tail vein resulted in ~50% exon 23
restored 53% of WT dystrophin levels in the          skipping in the heart of treated mice. A wide-
heart. Treatment with tcDNA also ameliorated         spread, uniform distribution of dystrophin-posi-
cardiac systolic function as demonstrated by         tive fibers was also observed in cardiac tissue
improved ventricular ejection fractions and          as shown by immunostaining. Western blot
shortening fractions in echocardiography.            analysis demonstrated dystrophin restoration
                                                     at levels between 10 and 20% of that found in
One of the advantages of tcDNA is its increased      the WT mouse heart. Even at lower doses
RNA binding affinity. Due to this feature, shorter   administered (weekly injections at 6 mg/kg for
tcDNAs can be designed, reducing the mass of         3 weeks), dystrophin restoration was still
AOs administered and potential AO-induced            observed in cardiac tissue as shown by immu-
toxicity while retaining their therapeutic effect.   nostaining and Western blotting.
To investigate the efficacy of shorter tcDNAs,
mdx mice were injected intravenously with a          Subsequently, another arginine-rich peptide,
13-mer tcDNA targeting Dmd exon 23 at 200            named the B-peptide, demonstrated high lev-
mg/kg/week for 12 weeks. The 13-mer tcDNA            els of exon skipping in various muscles includ-
induced particularly high levels of exon skip-       ing the heart. The B-peptide contains arginine,
ping and restored dystrophin in the heart as         aminohexanoic acid and beta alanine (abbrevi-
shown by RT-PCR, Western blotting and immu-          ated B) in an (RXRRBR)2 sequence [147-149]. A
nostaining. The safety profile of the treatment      single IV injection at 30 mg/kg of B-peptide-
also showed that tcDNAs were well-tolerated,         conjugated PMO (PPMO) restored 94% of dys-
with only minimal renal toxicity and liver inflam-   trophin expression in cardiac muscle fibers of
mation observed [138]. Although tcDNAs have          treated mdx mice by immunohistochemistry,
the potential to rescue dystrophin in cardiac tis-   50% of WT dystrophin levels by Western blot,
sue, treatment with these AOs requires high          and 63% exon 23 skipping by RT-PCR. Repeated
dosage and multiple dose administrations.            treatment for 3 months at the same dose at
                                                     biweekly intervals further improved dystrophin
Peptides                                             restoration in cardiac muscle. Skipped tran-
                                                     script levels increased to 72%. Immunohis-
Among the various delivery systems studied,          tochemistry and Western blot demonstrated
cell-penetrating peptides (CPPs) have gained         dystrophin rescue at levels comparable to that
the most considerable interest. CPPs are com-        in WT hearts. Dystrophin restoration was also
posed of cationic and/or amphipathic amino           observed in the muscles of the atria and large
acids that are capable of delivering a wide          vessels such as the aorta or vena cava and the
range of nucleic acid-based molecules both in        pulmonary arteries. PPMO treatment signifi-
vitro and in vivo [142-144]. These peptides          cantly improved cardiac function and prevent-
either possess natural translocating properties      ed cardiac failure under dobutamine stress
or were engineered by combining different pro-       [150]. A PPMO cocktail was also shown to
tein domains to have these properties [145].         restore dystrophin expression in the myocardi-
Many studies have investigated peptide-conju-        um and cardiac Purkinje fibers, and improve
gated AOs for the potential treatment of DMD.        cardiac conduction abnormalities in a canine
This review will discuss some of the most prom-      model of DMD [151].
ising approaches, namely: arginine-rich pep-
tides, B peptides, PNA/PMO internalization           Another promising class of CPPs is the Pips.
peptides (Pips), and phage peptides. Not all         Pips are a novel series of transduction peptides

1207                                                        Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

developed from mutagenesis and functional            also evaluated and no significant changes were
studies of a derivative of Penetratin, with the      detected for liver and kidney damage. This
six arginine residues added to the N terminus        result is encouraging since the phage peptides
of the CPP (R6Pen) [152]. Penetratin is origi-       could be used as an alternative to arginine-rich
nally derived from the homeobox peptide of           peptides, which have shown toxicity in higher
Drosophila Antennapedia. The CPPs in the Pip         animals [156-158].
series are characterized by a central hydropho-
bic motif anchored on each side by arginine-         Octaguanidine morpholino
rich sequences similar to that of the B-peptide
which contains arginine, aminohexanoic acid,         In an effort to improve the efficacy of AO sys-
and beta-alanine residues. Among the highly          temic delivery and to reduce the immunogenic-
efficient members of this series are Pip5 and        ity of cell penetrating peptides, dendrimeric
Pip6, both of which have demonstrated high           octaguanidine moieties have been conjugated
levels of dystrophin restoration in various mus-     to PMOs to form a new class of AOs named
cles including the heart [153, 154]. A single IV     Vivo-Morpholinos or vPMOs [150, 159].
injection of 25 mg/kg of Pip5e-PMO (a member
                                                     The guanidine groups in vPMOs work similarly
of the Pip5 family) induced almost complete
                                                     to the arginine-rich CPPs found conjugated to
exon skipping in cardiac tissue where full length
                                                     PMOs. As such, vPMOs have been shown to
uncorrected transcript was barely detectable.        induce high levels of dystrophin expression es-
Immunohistochemistry data revealed more              pecially in the heart [149]. Additionally, the
than 90% of dystrophin-positive fibers in cardi-     combination of a synthetic scaffold and multi-
ac muscle. Dystrophin restoration was further        ple unnatural side chains makes vPMOs unlike-
confirmed by Western blot which revealed more        ly to elicit an immune response, thus allowing
than 50% of WT dystrophin levels in the heart        for multiple administrations to maintain ade-
[153]. Modifications to the central hydrophobic      quate levels of dystrophin in the body [160].
core of the Pip5 peptide gave rise to a novel
derivative of the Pip series, consecutively          vPMOs have been shown to rescue cardiac dys-
named Pip6. When directly compared to Pip5e-         trophin expression in vivo [161]. A single IV
PMO treatment, a member of the Pip6 family           injection of a vPMO targeting mouse Dmd exon
exhibited significantly higher dystrophin resto-     23 (Vivo-ME23) at 6 mg/kg resulted in a strong
ration in the heart of treated mice at very low      dystrophin signal in a significant number of car-
doses (12.5 mg/kg). The Pip6 series have also        diomyocytes in mdx mice. Repeated treatment
been demonstrated to prevent exercise-               at the same dose for five times biweekly res-
induced cardiomyopathy following Pip-PMO             cued dystrophin expression in more than 40%
treatment in dystrophic mice [154].                  of cardiomyocytes as observed by immunos-
                                                     taining. Western blot showed dystrophin rescue
While the above-mentioned CPPs work well for         to approximately 10% of normal levels in cardi-
neutral AOs like PMOs, it is challenging to con-     ac muscles. vPMOs have also been shown to
jugate positively charged peptides to negative-      restore dystrophin expression in a canine mo-
ly-charged AOs such as 2’OMePS due to the            del of DMD, however, no systemic injection was
presence of strong electrostatic interactions. In    performed so whether vPMOs can rescue dys-
an effort to find uncharged CPPs, Jirka and          trophin in canine cardiac tissues remains to be
colleagues screened a phage display peptide          determined [162]. Regardless, these encourag-
library and identified a 7-mer peptide (P4) that     ing results represent an important step forward
significantly enhanced AO uptake in cardiac tis-     in our effort to overcome the challenges associ-
sue [155]. Dystrophic mdx mice were injected         ated with the delivery of AOs to the heart.
subcutaneously with 200 mg/kg/week doses
of 2’OMePS-P4 conjugate targeting mouse              Ultrasound and microbubbles
Dmd exon 23 for 6 weeks. Treatment with pep-
tide-conjugated AOs resulted in enhanced exon        Another promising approach for improving AO
skipping efficacy with a significant difference in   delivery to cardiac tissue is the use of ultra-
cardiac tissue compared to naked AOs, possi-         sound in combination with contrast-enhancing
bly due to improved uptake in cardiac muscle.        microbubbles [163]. Microbubbles are used to
The safety profile of the peptide conjugate was      carry the drugs to an area of interest, then

1208                                                        Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

ultrasound is applied to burst the microbub-          ZM2, have been shown to restore dystrophin
bles, resulting in tissue-specific delivery of the    expression in the heart of mdx mice [172, 173].
therapeutic materials [164-167]. Ultrasound
exposure improves the efficacy of intracellular       T1 nanoparticles are a type of cationic core-
delivery due to a phenomenon known as sono-           shell nanospheres, made up of a core of poly-
poration or cellular sonification. This effect cre-   methylmethacrylate (PMMA) and surrounded
ates transient pores in the cellular plasma           by a shell of cationic groups, which facilitates
membrane by a process known as inertial cavi-         the binding of charged AOs. In a study by
tation, thus allowing bioactive materials to          Rimessi and colleagues, T1 nanoparticles were
enter the cells. Inertial cavitation is enhanced      demonstrated to deliver 2’OMePS AOs and
by using microbubbles of contrast agents [164,        induce dystrophin rescue in body-wide striated
                                                      muscles. AOs targeting the boundary sequenc-
167-170]. AOs can be mixed with or incorporat-
                                                      es of exon and intron 23 of mouse Dmd (M23D)
ed into microbubbles in a number of ways such
                                                      were conjugated with T1 nanoparticles, and
as binding to the microbubble shell or using
                                                      delivered by weekly intraperitoneal (IP) injec-
site-specific ligands. The combination of ultra-
                                                      tions at 2.7 mg/kg to mdx mice [172]. T1
sound and microbubbles not only improves
                                                      nanoparticles showed a wide distribution in the
intracellular delivery but also reduces the
                                                      body, including the heart. At one week post-
amount of drugs used systemically thereby le-
                                                      injection, immunohistochemical analysis of
ssening the potential toxicity and side effects       treated mice showed the presence of dystro-
of treatment.                                         phin-positive cardiomyocytes in various areas
                                                      of the heart. Dystrophin was however undetect-
In order to investigate the microbubble proper-
                                                      able in cardiac tissues 6 weeks after the last
ties that are important for influencing drug
                                                      injection. Total and skipped dystrophin tran-
delivery efficacy in vivo, PMOs targeting Dmd
                                                      script levels in the hearts of treated mice were
exon 23 of the mdx mouse were injected sys-
                                                      also significantly increased with 80% and 16%
temically at 16 mg/kg via the tail vein [171].
                                                      more transcripts, respectively. Although dystro-
PMOs were incorporated into three different
                                                      phin was undetected in the heart by Western
commercially available microbubbles: Sona-
                                                      blot, the results were positive considering a sig-
zoid, Optison, and SonoVue. Ultrasound was            nificantly reduced dose compared to previous
then applied to the heart for two minutes after       studies. From a safety aspect, T1 nanoparticles
injections. Treated hearts were collected 24          may cause adverse effects due to their slow
hours or one-week post injection. Of the three        biodegradability, indicating a possibility of
microbubbles tested, Sonazoid and Optison             accumulative toxicity for long-term treatments
induced significant amounts of dystrophin-pos-        [172]. Also, T1 nanoparticles can form small
itive fibers in cardiac tissues. Treatments with      aggregates and are therefore not suitable for IV
PMOs only (no ultrasound or microbubbles)             injections.
were ineffective in terms of restoring dystro-
phin in cardiomyocytes. Microbubbles were             To improve the efficacy of T1 nanoparticles, a
also found to increase AO delivery to the heart       more efficient class of nanoparticles termed
in a dose-dependent manner. Histological anal-        ZM2 was designed [173]. ZM2 nanoparticles
ysis showed no signs of tissue damage or              are composed of a PMMA core and a random
observable morphological differences between          copolymer shell consisting of units derived
samples treated with ultrasound and microbub-         from N-isopropylacrylamide+ (NIPAM) and reac-
bles and non-treated controls. Additionally,          tive methacrylate-bearing cationic groups. mdx
microbubble stability was determined to be the        mice were injected IP weekly for 7 weeks at 7.5
major factor affecting the efficiency of thera-       mg/kg with M23D AOs loaded onto ZM2
peutic drug delivery.                                 nanoparticles. Immunohistochemical findings
                                                      showed 3% by manual count and 6% by semi-
Nanoparticles                                         automated count of dystrophin-positive signal
                                                      in the heart. Dystrophin was also detectable,
In order to improve AO stability and enable the       though faintly, in the heart of ZM2-M23D treat-
use of lower AOs concentrations in vivo, AO           ed mice via Western blot. However, dystrophin
delivery using nanoparticles was developed            was not detectable in the heart at 3 months
and demonstrated to be another promising              post-injection as demonstrated in a follow-up
approach. Two classes of nanoparticles, T1 and        study.

1209                                                         Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

Polymers                                                Conclusion

Different series of polymers were investigated          To summarize, with the exception of tricyclo-
for their ability to improve the delivery of AOs in     DNAs, naked AOs are generally ineffective in
vitro and in vivo. The first class is a series of low   treating the heart. Conjugation to CPPs, espe-
molecular weight polyethylenimine (LPEI) conju-         cially arginine-rich peptides and the Pip series,
gated pluronic copolymers (PCMs) [174]. Am-             appears to be a promising approach to improve
phiphilic pluronic, poly (ethylene oxide)-b-poly        AO uptake and dystrophin restoration in cardi-
(propylene oxide)-b-poly (ethylene oxide) (PEO-         ac tissue. However, the practicality of CPPs in
PPO-PEO triblock copolymer) has been used               the clinical setting remains to be determined
widely as a pharmaceutical adjuvant and a free          largely due to concerns over our complex
adjuvant for gene delivery. Depending on the            immune system. Phage peptides and octagua-
molecular weight, hydrophilic-lipophilic balance        nidine moieties were developed in an effort to
(HLB), and other characteristics of the pluron-         reduce the immunogenicity of arginine-rich
ics, PCMs can have different AO delivery effi-          peptides, however, they have been less suc-
ciencies and toxicities. In a study by Wang and         cessful in rescuing dystrophin in systemic deliv-
colleagues, mdx mice were injected intrave-             ery of antisense drugs to the heart. Other non-
nously with 0.4 mg of PCMs and 2 mg of PMOs             covalent conjugation approaches such as the
targeting the boundary sequences of exon and            use of nanoparticles, polymers, or ultrasound
intron 23 of mouse Dmd gene (PMOE23), or 2              and microbubbles are promising strategies to
mg of PMOE23 only as the control. Two weeks             enhance the efficacy of AOs in cardiac tissue,
after injection, dystrophin was not detectable          especially for charged antisense chemistries.
in cardiac muscle of control mice, while immu-          With a growing number of AO-based approach-
nohistochemistry showed membrane-localized              es being investigated, antisense therapy holds
dystrophin in more than 5% of cardiomyocytes            the potential to improve the treatments for
in some areas of the heart from treated mice.           DMD patients, with special focus on cardiac
Treatment also demonstrated no detectable               aspects of the disease.
toxicity in muscles, liver, and kidneys after sys-
temic administration.                                   Tricyclo-DNA clearly appears as a promising
                                                        chemistry for the antisense therapy of DMD-
Another class of amphiphilic polymers, termed           associated cardiomyopathy due to its unique
Z polymers, is constructed from Tween 85 and            pharmacological properties and high therapeu-
LPEI [175]. Based on preliminary results, two Z         tic potential. Besides their ability to improve
polymers Z7 and Z8 were selected for systemic           delivery and uptake into cardiac tissue, tcDNAs
injection. Dystrophic mdx mice were treated by          can also cross the blood-brain barrier at low
IV injection with 1 mg Z7/1 mg PMO complex, or          levels after systemic administration [139].
0.5 mg Z8/1 mg PMO conjugate, or 1 mg PMO               Along with other distinctive properties, this out-
as a control. Dystrophin expression was not             standing feature opens up more therapeutic
detected in the PMO-only group, however,                options for other neuromuscular and neurode-
Z8-PMO treated mice demonstrated mem-                   generative disorders such as spinal muscular
brane-localized dystrophin-positive fibers in           atrophy and Huntington’s disease [135].
some areas of the heart. No adverse effects             Although the safety profile of tcDNAs has been
were observed in systemic delivery under the            evaluated, observed toxicities were ascribed
experimental conditions. The mechanism/s by             more to the phosphorothioate linkages rather
which amphiphilic polymers improve gene                 than the tcDNA chemistry itself [138]. More
transfer is/are not fully understood. Their hydro-      extensive toxicological studies are recommend-
phobic interaction with cell membranes could            ed to assess the full safety profile of this prom-
reduce membrane viscosity and facilitate the            ising chemistry. Given its positive performance
entry of AOs [173]. Amphiphilic polymers could          in the preclinical phase, the evaluation of tcD-
also prevent nonspecific binding of AOs to              NAs in human clinical trials seems to be the
charged extracellular components, thus                  most logical next step. Similar to other chemis-
increasing the effective concentrations of ther-        tries, the outcome essentially depends on how
apeutic drugs.                                          well tcDNAs will be tolerated in patients.

1210                                                           Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

An active body of research on DMD has signifi-          [3]    Mendell JR, Shilling C, Leslie ND, Flanigan KM,
cantly broadened our knowledge on the pathol-                  Al-Dahhak R, Gastier-Foster J, Kneile K, Dunn
ogy as well as potential therapeutic treatments                DM, Duval B, Aoyagi A, Hamil C, Mahmoud M,
                                                               Roush K, Bird L, Rankin C, Lilly H, Street N,
for the disease. To this day, DMD is still an
                                                               Chandrasekar R, Weiss RB. Evidence-based
invariably fatal disease that affects the whole                path to newborn screening for Duchenne mus-
body. It is, therefore, essential to focus on treat-           cular dystrophy. Ann Neurol 2012; 71: 304-
ments for individual systems such as the heart.                313.
Advances in the understanding of antisense              [4]    Moser H. Duchenne muscular dystrophy:
therapy have provided a strong foundation for                  pathogenetic aspects and genetic prevention.
the transition of AOs drugs from the lab bench                 Hum Genet 1984; 66: 17-40.
to the clinic. However, delivery and uptake in          [5]    Duchenne. The pathology of paralysis with
                                                               muscular degeneration (paralysie myosclero-
cardiac tissue remain the key limiting factor in
                                                               tique), or paralysis with apparent hypertrophy.
the bioavailability of AO-based therapies for the              Br Med J 1867; 2: 541-542.
treatment of DMD. Perhaps more basic                    [6]    Hoffman EF, Kunkel LM. Dystrophin abnormali-
research into understanding the underlying                     ties in Duchenne/becker muscular review dys-
mechanism of these processes is needed to                      trophy genes. Cell 1989; 2: 1019-1029.
overcome this major obstacle. Up to this point,         [7]    Mercuri E, Muntoni F. Muscular dystrophies.
clinical trials have focused mostly on the effi-               Lancet 2013; 381: 845-860.
cacy of AOs treatments on skeletal muscle,              [8]    Tsuda T. Clinical manifestations and overall
without much emphasis on the heart. Given                      management strategies for Duchenne muscu-
                                                               lar dystrophy. Methods Mol Biol 2018; 1687:
how cardiomyopathy is currently the leading
                                                               19-28.
cause of death in patients with DMD, an                 [9]    Bushby K, Finkel R, Birnkrant DJ, Case LE, Cle-
increased focus on treatments for the heart is                 mens PR, Cripe L, Kaul A, Kinnett K, McDonald
very much needed. In 2017, a clinical trial                    C, Pandya S, Poysky J, Shapiro F, Tomezsko J,
investigating the efficacy of a peptide-conjugat-              Constantin C. Diagnosis and management of
ed version of eteplirsen was initiated. The trial              Duchenne muscular dystrophy, part 1: diagno-
is expected to be completed in January 2019.                   sis, and pharmacological and psychosocial
With the promising results shown by peptide                    management. Lancet Neurol 2010; 9: 77-93.
                                                        [10]   Strehle EM, Straub V. Recent advances in the
conjugated AOs in pre-clinical studies, it would
                                                               management of Duchenne muscular dystro-
be exciting to see the therapeutic outcome of                  phy. Arch Dis Child 2015; 100: 1173-1177.
the trial, particularly the cardiac aspects of it.      [11]   Manzur AY, Kinali M, Muntoni F. Update on the
Hopefully, these efforts will facilitate the prog-             management of Duchenne muscular dystro-
ress of therapeutic development so that one                    phy. Arch Dis Child 2008; 93: 986-990.
day DMD will no longer be as devastating a dis-         [12]   Eagle M, Baudouin SV, Chandler C, Giddings
ease as it is now.                                             DR, Bullock R, Bushby K. Survival in Duchenne
                                                               muscular dystrophy: improvements in life ex-
Disclosure of conflict of interest                             pectancy since 1967 and the impact of home
                                                               nocturnal ventilation. Neuromuscul Disord
None.                                                          2002; 12: 926-929.
                                                        [13]   Passamano L, Taglia A, Palladino A, Viggiano E,
Address correspondence to: Dr. Toshifumi Yokota,               D’Ambrosio P, Scutifero M, Cecio MR, Torre V,
                                                               De Luca F, Picillo E, Paciello O, Piluso G, Nigro
Department of Medical Genetics, Faculty of
                                                               G, Politano L. Improvement of survival in Duch-
Medicine and Dentistry, University of Alberta, 8812-           enne muscular dystrophy: retrospective analy-
112 St., Edmonton, AB T6G 2H7, Canada. Tel:                    sis of 835 patients. Acta Myol 2012; 31: 121-
+1-780-492-1102; E-mail: toshifum@ualberta.ca                  125.
                                                        [14]   Mah JK. Current and emerging treatment strat-
References                                                     egies for Duchenne muscular dystrophy. Neu-
                                                               ropsychiatr Dis Treat 2016; 12: 1795-1807.
[1]   Nakamura A, Takeda S. Mammalian models of         [15]   Ervasti JM. Dystrophin, its interactions with
      Duchenne muscular dystrophy: pathological                other proteins, and implications for muscular
      characteristics and therapeutic applications. J          dystrophy. Biochim Biophys Acta 2007; 1772:
      Biomed Biotechnol 2011; 2011: 184393.                    108-117.
[2]   Emery AE. Population frequencies of inherited     [16]   van Deutekom JC, van Ommen GJ. Advances in
      neuromuscular diseases-A world survey. Neu-              Duchenne muscular dystrophy gene therapy.
      romuscul Disord 1991; 1: 19-29.                          Nat Rev Genet 2003; 4: 774-783.

1211                                                            Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

[17] Campbell KP. Three muscular dystrophies: loss     [33] Pillers DA, Bulman DE, Weleber RG, Siges-
     of cytoskeleton-extracellular matrix linkage.          mund DA, Musarella MA, Powell BR, Murphey
     Cell 1995; 80: 675-679.                                WH, Westall C, Panton C, Becker LE, Worton
[18] Whitmore C, Morgan J. What do mouse models             RG, Ray PN. Dystrophin expression in the hu-
     of muscular dystrophy tell us about the DAPC           man retina is required for normal function as
     and its components? Int J Exp Pathol 2014;             defined by electroretinography. Nat Genet
     95: 365-377.                                           1993; 4: 82-86.
[19] Ervasti JM, Campbell KP. A role for the dystro-   [34] Takeshima Y, Yagi M, Okizuka Y, Awano H,
     phin-glycoprotein complex as a transmem-               Zhang Z, Yamauchi Y, Nishio H, Matsuo M. Mu-
     brane linker between laminin and actin. J Cell         tation spectrum of the dystrophin gene in 442
     Biol 1993; 122: 809-823.                               Duchenne/becker muscular dystrophy cases
[20] Ervasti JM, Campbell KP. Membrane organiza-            from one Japanese referral center. J Hum Gen-
     tion of the dystrophin-glycoprotein complex.           et 2010; 55: 379-388.
     Cell 1991; 66: 1121-1131.                         [35] Buzin CH, Feng J, Yan J, Scaringe W, Liu Q, Den
[21] Lapidos KA, Kakkar R, McNally EM. The dystro-          Dunnen J, Mendell JR, Sommer SS. Mutation
     phin glycoprotein complex: signaling strength          rates in the dystrophin gene: a hotspot of mu-
     and integrity for the sarcolemma. Circ Res             tation at a CpG dinucleotide. Hum Mutat 2005;
     2004; 94: 1023-1031.                                   25: 177-188.
[22] Petrof BJ, Shrager JB, Stedman HH, Kelly AM,      [36] Nachman MW, Crowell SL. Contrasting evolu-
     Sweeney HL. Dystrophin protects the sarco-             tionary histories of two introns of the Duch-
     lemma from stresses developed during mus-              enne muscular dystrophy gene, Dmd, in hu-
     cle contraction. Proc Natl Acad Sci U S A 1993;        mans. Genetics 2000; 155: 1855-1864.
     90: 3710-3714.                                    [37] Yokota T, Pistilli E, Duddy W, Nagaraju K. Poten-
[23] Nichols B, Takeda S, Yokota T. Nonmechanical           tial of oligonucleotide-mediated exon-skipping
     roles of dystrophin and associated proteins in         therapy for Duchenne muscular dystrophy. Ex-
     exercise, neuromuscular junctions, and brains.         pert Opin Biol Ther 2007; 7: 831-842.
     Brain Sci 2015; 5: 275-98.                        [38] White S, Kalf M, Liu Q, Villerius M, Engelsma D,
[24] Hoffman EP, Brown RH, Kunkel LM. Dystro-               Kriek M, Vollebregt E, Bakker B, van Ommen
     phin: the protein product of the Duchenne              GJ, Breuning MH, den Dunnen JT. Comprehen-
     muscular dystrophy locus. Cell 1987; 51: 919-          sive detection of genomic duplications and de-
     928.                                                   letions in the DMD gene, by use of multiplex
[25] Koenig M, Hoffman EP, Bertelson CJ, Monaco             amplifiable probe hybridization. Am J Hum
     AP, Feener C, Kunkel LM. Complete cloning of           Genet 2002; 71: 365-374.
     the Duchenne muscular dystrophy (DMD)             [39] Ferlini A, Neri M, Gualandi F. The medical ge-
     cDNA and preliminary genomic organization of           netics of dystrophinopathies: molecular genet-
     the DMD gene in normal and affected individu-          ic diagnosis and its impact on clinical practice.
     als. Cell 1987; 50: 509-517.                           Neuromuscul Disord 2013; 23: 4-14.
[26] Roberts RG, Coffey AJ, Bobrow M, Bentley DR.      [40] Flanigan KM, Dunn DM, Von Niederhausern A,
     Exon structure of the human dystrophin gene.           Soltanzadeh P, Gappmaier E, Howard MT,
     Genomics 1993; 16: 536-538.                            Sampson JB, Mendell JR, Wall C, King WM,
[27] Tsuda T, Fitzgerald K. Dystrophic cardiomyopa-         Pestronk A, Florence JM, Connolly AM,
     thy: complex pathobiological processes to gen-         Mathews KD, Stephan CM, Laubenthal KS,
     erate clinical phenotype. J Cardiovasc Dev Dis         Wong BL, Morehart PJ, Meyer A, Finkel RS,
     2017; 4: 14.                                           Bonnemann CG, Medne L, Day JW, Dalton JC,
[28] Kamdar F, Garry DJ. Dystrophin-deficient car-          Margolis MK, Hinton VJ, Weiss RB, Bromberg
     diomyopathy. J Am Coll Cardiol 2016; 67:               M, Swoboda K, Kerr L, Gurvich O, Tuohy T, Tay-
     2533-2546.                                             lor L, Zhao L, Hart K, Moural C, Hak K, Duval B,
[29] Ahn AH, Kunkel LM. The structural and func-            Hamil C, Mahmoud M, Aoyagi A, Viollet L, Gai-
     tional diversity of dystrophin. Nat Genet 1993;        ley S, Lopate G, Golumbek P, Schierbecker J,
     3: 283-291.                                            Malkus B, Siener C, Baldwin K, Glanzman AM,
[30] Mehler MF. Brain dystrophin, neurogenetics             Flickinger J, Naughton CE. Mutational spec-
     and mental retardation. Brain Res Rev 2000;            trum of DMD mutations in dystrophinopathy
     32: 277-307.                                           patients: application of modern diagnostic
[31] Chelly J, Hamard G, Koulakoff A, Kaplan JC,            techniques to a large cohort. Hum Mutat
     Kahn A, Berwald-Netter Y. Dystrophin gene              2009; 30: 1657-1666.
     transcribed from different promoters in neuro-    [41] Tuffery-Giraud S, Béroud C, Leturcq F, Yaou R
     nal and glial cells. Nature 1990; 344: 64-65.          Ben, Hamroun D, Michel-Calemard L, Moizard
[32] Lidov HG, Selig S, Kunkel LM. Dp140: a novel           MP, Bernard R, Cossée M, Boisseau P, Blayau
     140 kDa CNS transcript from the dystrophin             M, Creveaux I, Guiochon-Mantel A, De Martin-
     locus. Hum Mol Genet 1995; 4: 329-335.                 ville B, Philippe C, Monnier N, Bieth E, Van Kien

1212                                                           Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

       PK, Desmet FO, Humbertclaude V, Kaplan JC,         [51] Ashwath ML, Jacobs IB, Crowe CA, Ashwath
       Chelly J, Claustres M. Genotype-phenotype               RC, Super DM, Bahler RC. Left ventricular dys-
       analysis in 2,405 patients with a dystrophinop-         function in Duchenne muscular dystrophy and
       athy using the UMD-DMD database: a model of             genotype. Am J Cardiol 2014; 114: 284-289.
       nationwide knowledgebase. Hum Mutat 2009;          [52] Magri F, Govoni A, D’Angelo MG, Del Bo R, Ghe-
       30: 934-945.                                            zzi S, Sandra G, Turconi AC, Sciacco M, Ciscato
[42]   Hoffman EP, Kunkel LM, Angelini C, Clarke A,            P, Bordoni A, Tedeschi S, Fortunato F, Lucchini
       Johnson M, Harris JB. Improved diagnosis of             V, Bonato S, Lamperti C, Coviello D, Torrente Y,
       Becker muscular dystrophy by dystrophin test-           Corti S, Moggio M, Bresolin N, Comi G Pietro.
       ing. Neurology 1989; 39: 1011-1017.                     Genotype and phenotype characterization in a
[43]   Arahata K, Hoffman EP, Kunkel LM, Ishiura S,            large dystrophinopathic cohort with extended
       Tsukahara T, Ishihara T, Sunohara N, Nonaka I,          follow-up. J Neurol 2011; 258: 1610-1623.
       Ozawa E, Sugita H. Dystrophin diagnosis: com-      [53] Spurney CF. Cardiomyopathy of Duchenne
       parison of dystrophin abnormalities by immu-            muscular dystrophy: current understanding
       nofluorescence and immunoblot analyses.                 and future directions. Muscle Nerve 2011; 44:
       Proc Natl Acad Sci U S A 1989; 86: 7154-7158.           8-19.
[44]   Neri M, Torelli S, Brown S, Ugo I, Sabatelli P,    [54] Thomas TO, Morgan TM, Burnette WB,
       Merlini L, Spitali P, Rimessi P, Gualandi F,            Markham LW. Correlation of heart rate and
       Sewry C, Ferlini A, Muntoni F. Dystrophin levels        cardiac dysfunction in Duchenne muscular
       as low as 30% are sufficient to avoid muscular          dystrophy. Pediatr Cardiol 2012; 33: 1175-
       dystrophy in the human. Neuromuscul Disord              1179.
       2007; 17: 913-918.                                 [55] Miller G, D’Orsogna L, O’Shea JP. Autonomic
[45]   Melacini P, Fanin M, Danieli GA, Villanova C,           function and the sinus tachycardia of Duch-
       Martinello F, Miorin M, Freda MP, Miorelli M,           enne muscular dystrophy. Brain Dev 1989; 11:
                                                               247-250.
       Mostacciuolo ML, Fasoli G, Angelini C, Dalla
                                                          [56] Lanza GA, Russo A Dello, Giglio V, De Luca L,
       Volta S. Myocardial involvement is very fre-
                                                               Messano L, Santini C, Ricci E, Damiani A, Fu-
       quent among patients affected with subclinical
                                                               magalli G, De Martino G, Mangiola F, Bellocci F.
       Becker’s muscular dystrophy. Circulation
                                                               Impairment of cardiac autonomic function in
       1996; 94: 3168-3175.
                                                               patients with Duchenne muscular dystrophy:
[46]   Nigro G, Comi LI, Politano L, Bain RJ. The inci-
                                                               relationship to myocardial and respiratory
       dence and evolution of cardiomyopathy in
                                                               function. Am Heart J 2001; 141: 808-812.
       Duchenne muscular dystrophy. Int J Cardiol
                                                          [57] Kedem J, Sonn J, Scheinowitz M, Weiss HR. Re-
       1990; 26: 271-277.
                                                               lationship between local oxygen consumption
[47]   McNally EM, Kaltman JR, Benson D, Canter CE,
                                                               and local and external cardiac work: effect of
       Cripe LH, Duan D, Finder JD, Hoffman EP,                tachycardia. Cardiovasc Res 1989; 23: 1043-
       Judge DP, Kertesz N, Kinnett K, Kirsch R,               1052.
       Metzger JM, Pearson GD, Rafael-Fortney JA,         [58] Chiang DY, Allen HD, Kim JJ, Valdes SO, Wang
       Raman SV, Spurney CF, Targum SL, Wagner                 Y, Pignatelli RH, Lotze TE, Miyake CY. Relation
       KR, Markham LW. Contemporary cardiac is-                of cardiac dysfunction to rhythm abnormalities
       sues in Duchenne muscular dystrophy. Work-              in patients with Duchenne or becker muscular
       ing group of the national heart, lung, and blood        dystrophies. Am J Cardiol 2016; 117: 1349-
       institute in collaboration with parent project          1354.
       muscular dystrophy. Circulation 2015; 131:         [59] Ramaciotti C, Heistein LC, Coursey M, Lemler
       1590-1598.                                              MS, Eapen RS, Iannaccone ST, Scott WA. Left
[48]   Wang M, Birnkrant DJ, Super DM, Jacobs IB,              ventricular function and response to enalapril
       Bahler RC. Progressive left ventricular dysfunc-        in patients with Duchenne muscular dystrophy
       tion and long-term outcomes in patients with            during the second decade of life. Am J Cardiol
       Duchenne muscular dystrophy receiving car-              2006; 98: 825-827.
       diopulmonary therapies. Open Hear 2018; 5:         [60] Su JA, Ramos-Platt L, Menteer JD. Left ventric-
       e000783.                                                ular tonic contraction as a novel biomarker of
[49]   Jefferies JL, Eidem BW, Belmont JW, Craigen             cardiomyopathy in Duchenne muscular dystro-
       WJ, Ware SM, Fernbach SD, Neish SR, Smith               phy. Pediatr Cardiol 2016; 37: 678-685.
       EOB, Towbin JA. Genetic predictors and remod-      [61] Silva MC, Meira ZM, Gurgel Giannetti J, da Sil-
       eling of dilated cardiomyopathy in muscular             va MM, Campos AF, Barbosa Mde M, Starling
       dystrophy. Circulation 2005; 112: 2799-2804.            Filho GM, Ferreira Rde A, Zatz M, Rochitte CE.
[50]   Nigro G, Politano L, Nigro V, Petretta VR, Comi         Myocardial delayed enhancement by magnetic
       LI. Mutation of dystrophin gene and cardiomy-           resonance imaging in patients with muscular
       opathy. Neuromuscul Disord 1994; 4: 371-                dystrophy. J Am Coll Cardiol 2007; 49: 1874-
       379.                                                    1879.

1213                                                              Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

[62] Silva MC, Magalhães TA, Meira ZM, Rassi CH,                tion in Duchenne muscular dystrophy and the
     Andrade AC, Gutierrez PS, Azevedo CF, Gurgel-              role of second messengers. Med Res Rev
     Giannetti J, Vainzof M, Zatz M, Kalil-Filho R,             2013; 33: 1174-1213.
     Rochitte CE. Myocardial fibrosis progression in     [74]   Berridge MJ, Lipp P, Bootman MD. The versatil-
     Duchenne and becker muscular dystrophy: a                  ity and universality of calcium signalling. Nat
     randomized clinical trial. JAMA Cardiol 2017;              Rev Mol Cell Biol 2000; 1: 11-21.
     2: 190-199.                                         [75]   Vallejo-Illarramendi A, Toral-Ojeda I, Aldanondo
[63] Mavrogeni SI, Markousis-Mavrogenis G, Pa-                  G, López de Munain A. Dysregulation of calci-
     pavasiliou A, Papadopoulos G, Kolovou G.                   um homeostasis in muscular dystrophies. Ex-
     Cardiac involvement in Duchenne muscular                   pert Rev Mol Med 2014; 16: e16.
     dystrophy and related dystrophinopathies.           [76]   Zorov DB, Juhaszova M, Sollott SJ. Mitochon-
     Methods Mol Biol 2018; 1687: 31-42.                        drial reactive oxygen species (ROS) and ROS-
[64] Urasawa N, Wada MR, Machida N, Yuasa K,                    induced ROS release. Physiol Rev 2014; 94:
     Shimatsu Y, Wakao Y, Yuasa S, Sano T, Nonaka               909-950.
     I, Nakamura A, Takeda S. Selective vacuolar         [77]   Gorski PA, Ceholski DK, Hajjar RJ. Altered myo-
     degeneration in dystrophin-deficient canine                cardial calcium cycling and energetics in heart
     Purkinje fibers despite preservation of dystro-            failure - a rational approach for disease treat-
     phin-associated proteins with overexpression               ment. Cell Metab 2015; 21: 183-194.
     of Dp71. Circulation 2008; 117: 2437-2448.          [78]   Fong P, Turner PR, Denetclaw WF, Steinhardt
[65] Nomura H, Hizawa K. Histopathological study                RA. Increased activity of calcium leak channels
     of the conduction system of the heart in Duch-             in myotubes of Duchenne human and mdx
     enne progressive muscular dystrophy. Acta                  mouse origin. Science 1990; 250: 673-676.
     Pathol Jpn 1982; 32: 1027-33.                       [79]   Franco A, Lansman JB. Calcium entry through
[66] Yanagisawa A, Miyagawa M, Yotsukura M,                     stretch-inactivated ion channels in mdx myo-
     Tsuya T, Shirato C, Ishihara T, Aoyagi T, Ishika-          tubes. Nature 1990; 344: 670-673.
     wa K. The prevalence and prognostic signifi-        [80]   Reid MB. Role of nitric oxide in skeletal mus-
     cance of arrhythmias in Duchenne type mus-                 cle: synthesis, distribution and functional im-
     cular dystrophy. Am Heart J 1992; 124:                     portance. Acta Physiol Scand 1998; 162: 401-
     1244-50.                                                   409.
[67] Wallace GQ, McNally EM. Mechanisms of mus-          [81]   Stamler JS, Meissner G. Physiology of nitric ox-
     cle degeneration, regeneration, and repair in              ide in skeletal muscle. Physiol Rev 2001; 81:
     the muscular dystrophies. Annu Rev Physiol                 209-237.
     2009; 71: 37-57.                                    [82]   Raman SV, Hor KN, Mazur W, Halnon NJ, Kissel
[68] Konstam MA, Kramer DG, Patel AR, Maron MS,                 JT, He X, Tran T, Smart S, McCarthy B, Taylor
     Udelson JE. Left ventricular remodeling in                 MD, Jefferies JL, Rafael-Fortney JA, Lowe J,
     heart failure: current concepts in clinical sig-           Roble SL, Cripe LH. Eplerenone for early car-
     nificance and assessment. JACC Cardiovasc                  diomyopathy in Duchenne muscular dystrophy:
     Imaging 2011; 4: 98-108.                                   a randomised, double-blind, placebo-con-
[69] van Westering T, Betts C, Wood M. Current un-              trolled trial. Lancet Neurol 2015; 14: 153-161.
     derstanding of molecular pathology and treat-       [83]   Mendell JR, Moxley RT, Griggs RC, Brooke MH,
     ment of cardiomyopathy in Duchenne muscu-                  Fenichel GM, Miller JP, King W, Signore L, Pan-
     lar dystrophy. Molecules 2015; 20: 8823-8855.              dya S, Florence J, Schierbecker J, Robison J,
[70] Shirokova N, Niggli E. Cardiac phenotype of                Kaiser K, Mandel S, Arfken C, Gilder B. Ran-
     Duchenne muscular dystrophy: insights from                 domized, double-blind six-month trial of pred-
     cellular studies. J Mol Cell Cardiol 2013; 58:             nisone in Duchenne’s muscular dystrophy. N
     217-224.                                                   Engl J Med 1989; 320: 1592-1597.
[71] Fanchaouy M, Polakova E, Jung C, Ogrodnik J,        [84]   Dec GW. Steroid therapy effectively delays
     Shirokova N, Niggli E. Pathways of abnormal                Duchenne’s cardiomyopathy. J Am Coll Cardiol
     stress-induced Ca2+ influx into dystrophic mdx             2013; 61: 955-956.
     cardiomyocytes. Cell Calcium 2009; 46: 114-         [85]   Wu RS, Gupta S, Brown RN, Yancy CW, Wald
     121.                                                       JW, Kaiser P, Kirklin NM, Patel PC, Markham
[72] Nakayama H, Chen X, Baines CP, Klevitsky R,                DW, Drazner MH, Garry DJ, Mammen PP. Clini-
     Zhang X, Zhang H, Jaleel N, Chua BHL, Hewett               cal outcomes after cardiac transplantation in
     TE, Robbins J, Houser SR, Molkentin JD. Ca2+-              muscular dystrophy patients. J Heart Lung
     and mitochondrial-dependent cardiomyocyte                  Transplant 2010; 29: 432-438.
     necrosis as a primary mediator of heart failure.    [86]   Ryan TD, Jefferies JL, Sawnani H, Wong BL,
     J Clin Invest 2007; 117: 2431-2444.                        Gardner A, Del Corral M, Lorts A, Morales DL.
[73] Mosqueira M, Zeiger U, Förderer M, Brinkmeier              Implantation of the HeartMate II and Heart-
     H, Fink RH. Cardiac and respiratory dysfunc-               Ware left ventricular assist devices in patients

1214                                                             Am J Transl Res 2019;11(3):1202-1218
Antisense therapy for DMD cardiomyopathy

       with Duchenne muscular dystrophy: lessons                  prednisone therapy in Duchenne muscular
       learned from the first applications. ASAIO J               dystrophy. Neurology 1991; 41: 1874-1877.
       2014; 60: 246-248.                                   [97] McMurray JJ, Östergren J, Swedberg K, Grang-
[87]   Stoller D, Araj F, Amin A, Fitzsimmons C, Mor-             er CB, Held P, Michelson EL, Olofsson B, Yusuf
       lend R, Thibodeau JT, Ramaciotti C, Drazner                S, Pfeffer MA. Effects of candesartan in pa-
       MH, Meyer DM, Mammen PPA. Implantation of                  tients with chronic heart failure and reduced
       a left ventricular assist device to provide long-          left-ventricular systolic function taking angio-
       term support for end-stage Duchenne muscu-                 tensin-converting-enzyme inhibitors: the cha-
       lar dystrophy-associated cardiomyopathy. ESC               rm-added trial. Lancet 2003; 362: 767-771.
       Hear Fail 2017; 4: 379-383.                          [98] Wu H, Lima WF, Zhang H, Fan A, Sun H, Crooke
[88]   Duboc D, Meune C, Pierre B, Wahbi K, Eymard                ST. Determination of the role of the human
       B, Toutain A, Berard C, Vaksmann G, Weber S,               RNase H1 in the pharmacology of dna-like an-
       Bécane HM. Perindopril preventive treatment                tisense drugs. J Biol Chem 2004; 279: 17181-
       on mortality in Duchenne muscular dystrophy:               9.
       10 years’ follow-up. Am Heart J 2007; 154:           [99] Lee J, Yokota T. Antisense therapy in neurology.
       596-602.                                                   J Pers Med 2013; 3: 144-176.
[89]   Viollet L, Thrush PT, Flanigan KM, Mendell JR,       [100] Kole R, Krieg AM. Exon skipping therapy for
       Allen HD. Effects of angiotensin-converting en-            Duchenne muscular dystrophy. Adv Drug Deliv
       zyme inhibitors and/or beta blockers on the                Rev 2015; 87: 104-107.
       cardiomyopathy in Duchenne muscular dystro-          [101] Guncay A, Yokota T. Antisense oligonucleotide
       phy. Am J Cardiol 2012; 110: 98-102.                       drugs for Duchenne muscular dystrophy: how
[90]   Allen HD, Flanigan KM, Thrush PT, Dvorchik I,              far have we come and what does the future
       Yin H, Canter C, Connolly AM, Parrish M, Mc-               hold? Future Med Chem 2015; 7: 1631-1635.
       Donald CM, Braunlin E, Colan SD, Day J, Dar-         [102] Nguyen Q, Yokota T. Immortalized muscle cell
                                                                  model to test the exon skipping efficacy for
       ras B, Mendell JR. A randomized, double-blind
                                                                  Duchenne muscular dystrophy. J Pers Med
       trial of lisinopril and losartan for the treatment
                                                                  2017; 7: E13.
       of cardiomyopathy in Duchenne muscular dys-
                                                            [103] Dias N, Stein CA. Antisense oligonucleotides:
       trophy. PLoS Curr 2013; 5.
                                                                  basic concepts and mechanisms. Mol Cancer
[91]   Kajimoto H, Ishigaki K, Okumura K, Tomimatsu
                                                                  Ther 2002; 1: 347-355.
       H, Nakazawa M, Saito K, Osawa M, Nakanishi
                                                            [104] Eder PS, DeVine RJ, Dagle JM, Walder JA. Sub-
       T. Beta-blocker therapy for cardiac dysfunction
                                                                  strate specificity and kinetics of degradation of
       in patients with muscular dystrophy. Circ J
                                                                  antisense oligonucleotides by a 3’ exonucle-
       2006; 70: 991-994.
                                                                  ase in plasma. Antisense Res Dev 1991; 1:
[92]   Matsumura T, Tamura T, Kuru S, Kikuchi Y,
                                                                  141-151.
       Kawai M. Carvedilol can prevent cardiac events       [105] Geary RS, Norris D, Yu R, Bennett CF. Pharma-
       in Duchenne muscular dystrophy. Intern Med                 cokinetics, biodistribution and cell uptake of
       2010; 49: 1357-1363.                                       antisense oligonucleotides. Adv Drug Deliv Rev
[93]   Schram G, Fournier A, Leduc H, Dahdah N,                   2015; 87: 46-51.
       Therien J, Vanasse M, Khairy P. All-cause mor-       [106] Juliano RL, Carver K. Cellular uptake and intra-
       tality and cardiovascular outcomes with pro-               cellular trafficking of oligonucleotides. Adv
       phylactic steroid therapy in Duchenne muscu-               Drug Deliv Rev 2015; 87: 35-45.
       lar dystrophy. J Am Coll Cardiol 2013; 61:           [107] De Clercq E, Eckstein E, Merigan TC. Interferon
       948-954.                                                   induction increased through chemical modifi-
[94]   Buyse GM, Goemans N, Van Den Hauwe M,                      cation of a synthetic polyribonucleotide. Sci-
       Meier T. Effects of glucocorticoids and ideben-            ence 1969; 165: 1137-1139.
       one on respiratory function in patients with         [108] Rifai A, Brysch W, Fadden K, Clark J, Schlingen-
       Duchenne muscular dystrophy. Pediatr Pulm-                 siepen KH. Clearance kinetics, biodistribution,
       onol 2013; 48: 912-920.                                    and organ saturability of phosphorothioate oli-
[95]   Tandon A, Villa CR, Hor KN, Jefferies JL, Gao Z,           godeoxynucleotides in mice. Am J Pathol
       Towbin JA, Wong BL, Mazur W, Fleck RJ, Sticka              1996; 149: 717-725.
       JJ, Benson DW, Taylor MD. Myocardial fibrosis        [109] Watanabe TA, Geary RS, Levin AA. Plasma pro-
       burden predicts left ventricular ejection frac-            tein binding of an antisense oligonucleotide
       tion and is associated with age and steroid                targeting human ICAM-1 (ISIS 2302). Oligonu-
       treatment duration in Duchenne muscular dys-               cleotides 2006; 16: 169-180.
       trophy. J Am Heart Assoc 2015; 4: e001338.           [110] Freier S, Altmann KH. The ups and downs of
[96]   Fenichel GM, Florence JM, Pestronk A, Mendell              nucleic acid duplex stability: structure-stability
       JR, Moxley RT, Griggs RC, Brooke MH, Miller JP,            studies on chemically-modified DNA: RNA du-
       Robison J, King W, Signore L, Pandya S, Schier-            plexes. Nucleic Acids Res 1997; 25: 4429-
       becker J, Wilson B. Long-term benefit from                 4443.

1215                                                                Am J Transl Res 2019;11(3):1202-1218
You can also read