You are on page 1of 14

JOURNAL OF THE AMERICAN COLLEGE OF CARDIOLOGY VOL. 67, NO.

21, 2016

ª 2016 BY THE AMERICAN COLLEGE OF CARDIOLOGY FOUNDATION ISSN 0735-1097/$36.00

PUBLISHED BY ELSEVIER http://dx.doi.org/10.1016/j.jacc.2016.02.081

REVIEW TOPIC OF THE WEEK

Dystrophin-Deficient Cardiomyopathy
Forum Kamdar, MD, Daniel J. Garry, MD, PHD

ABSTRACT

Dystrophinopathies are a group of distinct neuromuscular diseases that result from mutations in the structural
cytoskeletal Dystrophin gene. Dystrophinopathies include Duchenne muscular dystrophy (DMD) and Becker muscular
dystrophy (BMD), X-linked dilated cardiomyopathy, as well as DMD and BMD female carriers. The primary presenting
symptom in most dystrophinopathies is skeletal muscle weakness. However, cardiac muscle is also a subtype of striated
muscle and is similarly affected in many of the muscular dystrophies. Cardiomyopathies associated with dystrophin-
opathies are an increasingly recognized manifestation of these neuromuscular disorders and contribute significantly to
their morbidity and mortality. Recent studies suggest that these patient populations would benefit from cardiovascular
therapies, annual cardiovascular imaging studies, and close follow-up with cardiovascular specialists. Moreover, patients
with DMD and BMD who develop end-stage heart failure may benefit from the use of advanced therapies. This review
focuses on the pathophysiology, cardiac involvement, and treatment of cardiomyopathy in the dystrophic patient.
(J Am Coll Cardiol 2016;67:2533–46) © 2016 by the American College of Cardiology Foundation.

DYSTROPHINOPATHIES: the development of diagnostic and genetic testing, as


A HISTORICAL PERSPECTIVE well as the establishment of disease models that have
enhanced our understanding of DMD. Although a
The earliest detailed descriptions of muscular dys- number of questions remain regarding the muscular
trophies were by Meryon (1) in England in 1852 and by dystrophies, the rich history of discovery has signifi-
Duchenne (2) in France in 1868, identifying a disease cantly accelerated our knowledge of DMD in the last
that affected young boys with severe muscular 30-year period.
weakness and an abnormal increase in calf size.
Duchenne (2) observed fatty-fibrous replacement of DYSTROPHINOPATHIES: INHERITANCE
skeletal muscle in muscle biopsies obtained from
these boys; for this contribution, the disease was DMD is the most common form of muscular dys-
named after him. In 1886, Gowers (3) described how a trophy, affecting 1 in every 5,000 boys born in the
child affected with muscular dystrophy used his arms United States (Table 1) (6–8). DMD is a result of
to rise from the ground. Although Duchenne an inherited or spontaneous mutation of the DMD
muscular dystrophy (DMD) had been known as a gene. DMD is a 2.5-Mb gene located on chromosome
clinical entity since the 1860s, its cause was not Xp21.1 (5,9), and is the largest known gene,
elucidated until over 100 years later. In a landmark harboring 79 exons and encoding a 14-kb transcript
discovery, Louis Kunkel’s laboratory (1986) identified (Figure 1). The full-length dystrophin gene has 3
the DMD gene responsible for causing DMD and, a promoters: the M promoter produces the Dp427m
year later, demonstrated that mutations resulted isoform, expressed in skeletal and cardiac muscle;
Listen to this manuscript’s in the absence of the 427-kDa rod-like protein dys- the B promoter produces Dp427c, expressed in
audio summary by
trophin (4,5). These major advancements allowed for the brain; and the P promoter produces Dp427p,
JACC Editor-in-Chief
Dr. Valentin Fuster.

From the Cardiovascular Division, Lillehei Heart Institute, University of Minnesota, Minneapolis, Minnesota. Both authors have
reported that they have no relationships relevant to the contents of this paper to disclose.

Manuscript received December 14, 2015; revised manuscript received February 16, 2016, accepted February 23, 2016.
2534 Kamdar and Garry JACC VOL. 67, NO. 21, 2016

DMD and BMD Cardiomyopathy MAY 31, 2016:2533–46

ABBREVIATIONS expressed in the Purkinje cells in the brain function or absence of 1 or several of these DGC pro-
AND ACRONYMS (Figure 1) (10). teins leads to plasma membrane fragility. DMD and
An out-of-frame mutation of 1 or several of Becker muscular dystrophy (BMD) arise from muta-
ACEI = angiotensin-converting
enzyme inhibitor
the 79 exons in the full-length Dystrophin tions in dystrophin, and other forms of muscular dys-
gene results in an absence of a functional trophies arise from mutations in DGC components. In
BMD = Becker muscular
dystrophy dystrophin protein, causing the DMD pheno- skeletal muscle, the DGC is located at regular intervals
CMR = cardiac magnetic type. DMD is inherited in an X-linked reces- in structures known as costameres, whereas in cardiac
resonance imaging sive manner, where a female carrier with 1 X muscle, the DGC is not located in discrete costameres
CRISPR = clustered regularly- chromosome carrying the DMD mutation has and may have a unique composition (15,16).
interspaced short palindromic a 50% chance of passing on the mutated X
repeat
chromosome to her son. Female carriers can
PATHOGENESIS AND PHYSICAL FINDINGS
DGC = dystroglycan complex
also have symptoms on the basis of their
DMD = Duchenne muscular Skeletal muscle and heart lacking functional dystro-
X-inactivation. Although the majority of DMD
dystrophy phin are mechanically weak, and contraction of the
mutations are inherited, spontaneous muta-
ECG = electrocardiogram cell (skeletal myocytes and cardiac myocytes) leads
tions account for 30% of DMD cases (11).
ICD = implantable to membrane damage (17–19) (Central Illustration).
cardioverter-defibrillator DYSTROPHIN AND DYSTROGLYCAN Loss of membrane integrity results in a cascade of
LVAD = left ventricular COMPLEX ORGANIZATION increased calcium influx into the cell and eventual
assist device
cell death. Clinically, dystrophin loss manifests as
LVEF = left ventricular ejection Dystrophin, a rod-shaped cytoplasmic pro- progressive muscle weakness (6). Symptoms are first
fraction
tein, connects the dystroglycan complex noted in early childhood and include calf pseudohy-
(DGC) to the intracellular contractile apparatus and pertrophy, an inability to stand without using the
extracellular matrix (ECM) of the cell (Figure 2) (12). arms for assistance (Gower maneuver), toe walking,
Dystrophin is similar to spectrin and consists of 2 and difficulty keeping up with peers. As the disease
ends separated by long, flexible, rod-like regions. The progresses, patients’ skeletal muscle becomes
N-terminus binds actin and the C-terminus binds to increasingly weak, causing atrophy and contractures,
glycoproteins in the sarcolemma. The role of dystro- which subsequently results in the loss of ambulation
phin is to stabilize the plasma membrane by trans- in their teens. Due to ongoing muscle damage, pa-
mitting forces generated by the sarcomeric tients with DMD have markedly elevated serum levels
contraction to the ECM (Figure 2). of the muscle protein creatine kinase (CK), which may
The DGC is a multimeric complex composed of be 10 to over 100 the normal limit, and an elevated
glycated integral membrane proteins and peripheral CK level is a diagnostic sign (20). The diagnosis of
proteins that form a structural link between the DMD can be confirmed by muscle biopsy demon-
filamentous (F)-actin cytoskeleton and the ECM in strating the absence of dystrophin (Figure 3) and
both cardiac and skeletal muscle (13). The DGC using genetic testing for dystrophin mutations.
is comprised of cytoskeletal proteins, dystrophin,
syntrophins, dystroglycans, sarcoglycans, DGC- SURVIVAL
associated proteins, neuronal nitric oxide synthase,
and dystrobrevin (14). The fully nucleated DGC pro- Historically, this progressive muscle weakness resul-
vides mechanical support to the skeletal or cardiac ted in loss of ambulation by 10 to 12 years of age and
plasma membrane during contraction, and loss of death during the second decade of life, primarily due
to respiratory failure. However, with the advent of
nocturnal ventilation, spinal stabilization surgery,
T A B L E 1 DMD Versus BMD
and steroid treatment, the life expectancy of boys
DMD BMD with DMD has increased to the late twenties to early
Dystrophin protein Absent Partially functional thirties (21) (Figure 4). Although respiratory failure
Incidence 1:5,000 male births 1:19,000 was previously the major cause of death, cardiomy-
Mean age at onset, yrs 3–5 12
opathy has now emerged as the leading cause of
Mean age of becoming w12 w27
nonambulatory, yrs death in patients with DMD (22).
Mean life expectancy, yrs Mid to late 20s 40s
Onset of cardiomyopathy, yrs 16–18 Variable; cardiomyopathy
NONCARDIAC TREATMENTS
may precede skeletal
symptoms
Advances in management, namely corticosteroid
BMD ¼ Becker muscular dystrophy; DMD ¼ Duchenne muscular dystrophy.
treatments, spinal stabilization, and improved pul-
monary support, have significantly improved the life
JACC VOL. 67, NO. 21, 2016 Kamdar and Garry 2535
MAY 31, 2016:2533–46 DMD and BMD Cardiomyopathy

F I G U R E 1 Dystrophin Gene and Protein

A B/Dp427

M/Dp427
R/Dp260 B,K/Dp140 S/Dp116 G/Dp71
Gene P/Dp427
500 kb 1000 kb 1500 kb 2500kb

Exons N terminus 1 2 1 2 3 4 4 5 5 6 6 7 7
0 0 0 0 5 0 6 0 3 0 9
Dystrobrevin binding site
Actin nNOS Dystroglycan Syntrophin binding site
binding binding Binding site
domain
B H H H 2 H
Structural NH2 1 COOH
1 2 3 4 4
Dy Sy
str n
og d tro
lyc ystro phin
Rod Domain an bre
vin

(A) The 79 exons of the full-length dystrophin gene with promoters for the alternatively spliced isoforms indicated by red arrows and boxes
specific for each promoter. Full-length dystrophin promoters include: M, muscle promoter; B, brain promoter; and P, Purkinje promoter. Other,
shorter dystrophin isoforms include: Dp260, expressed in the retina (R); Dp140, expressed in kidney (K) and brain (B); Dp116, expressed in
Schwann cells (S); and Dp71, which is ubiquitously expressed. (B) Schematic of the protein structure of dystrophin, including the actin-binding
site at the N-terminus and the DGC protein-binding site at the C-terminus. The rod domain contains 24 spectrin-like repeats with 4 hinge
regions. DGC ¼ dystroglycan complex.

expectancy of boys/men with DMD to the late Additionally, nutrition, exercise therapy, and psy-
twenties and early thirties (21). Glucocorticoids are chosocial support are critical in the interdisciplinary
one of the mainstays of treatment for DMD and have care of patients with DMD (27).
been shown to improve muscle strength, function,
and pulmonary function (23). Glucocorticoids are CARDIAC INVOLVEMENT IN DMD
currently recommended in patients with DMD who
are 5 years of age or older who are not gaining or have The reduction of respiratory-related deaths due to
a decline in motor skills (24). Current glucocorticoid nocturnal ventilation and spinal stabilization has
protocols that have been tested in randomized clin- contributed to the increase of DMD cardiomyopathy
ical trials include: daily prednisone 0.75 mg/kg/day; due to the increased survival and advanced age of
intermittent prednisone 0.75 mg/kg/day, 10 days on patients with DMD (21,31). The incidence of cardio-
and 10 days off; and daily deflazacort 0.9 mg/kg/day myopathy in DMD increases with age. Although it is
(25). Deflazacort is another steroid that is used in the estimated that 25% of boys have cardiomyopathy at
DMD patient population and may have a more limited 6 years of age and 59% by 10 years of age, cardiac
side effect profile compared with prednisone (26). involvement is nearly ubiquitous in older patients
Scoliosis and kyphosis, common, progressive with DMD, as more than 90% of young men over
sequelae of DMD, subsequently result in decreased 18 years of age demonstrate evidence of cardiac
pulmonary function and discomfort in these patients; dysfunction (22). Dilated cardiomyopathy typically
however, implementing spinal stabilization surgery has an onset in the midteen years and progressively
using Harrington rods has improved survival, com- remodels and contributes to the demise of patients
fort, and pulmonary function (27). Respiratory com- with DMD (22,32) (Figure 5). Distinct dystrophin
plications occur frequently in patients with DMD as mutations have been correlated to an increased
losing respiratory muscle strength leading to incidence of cardiomyopathy and possible response
decreased ventilation, and they are at increased risk to treatment (33). Recognition of heart failure
for pneumonia, atelectasis, and respiratory failure symptoms in patients with DMD can be challenging
(28). The majority of deaths in end-stage DMD pre- due to physical inactivity and other respiratory
viously occurred as a result of respiratory failure and complaints that can obscure the diagnosis (34).
infections; however, the introduction of noninvasive Currently, clinical guidelines recommend the initial
nocturnal mechanical ventilation has improved sur- cardiac screening at the time of diagnosis of DMD,
vival in patients with DMD and may improve every 2 years until 10 years of age, and then yearly
cardiac function through afterload reduction (29,30). thereafter (24).
2536 Kamdar and Garry JACC VOL. 67, NO. 21, 2016

DMD and BMD Cardiomyopathy MAY 31, 2016:2533–46

F I G U R E 2 Schematic Diagram of the DGC

ECM
Laminin
α-dystroglycan

Extracellular
Sarcoglycans β-dystroglycan
Sarcolemma

Syntrophin
nNOS Dystrobrevin

Dystrophin COOH

Intracellular

Actin cytoskeleton
NH3

The DGC spans the sarcolemma and links the cytoplasmic actin cytoskeleton to the ECM via dystrophin. Transmembrane components of the DGC
include sarcoglycans, beta-dystroglycan, and extracellular alpha-dystroglycan, which binds laminin. Cytoplasmic components include dystrophin
(or utrophin), which binds dystrobrevin, syntrophin, and nitric oxide synthase. DGC ¼ dystroglycan complex; ECM ¼ extracellular matrix.

Most patients with DMD will have abnormal elec- patients with DMD (46). Dalmaz et al. (47) demon-
trocardiographic tracings. The classical pattern dem- strated that urinary catecholamines increased in
onstrates tall R waves and increased R/S amplitude in patients with DMD around 10 years of age, which
lead V 1 , Q waves in the left precordial leads, right axis corresponded temporally to heart rate elevation and
deviation, or complete right bundle branch block cardiac involvement. In patients with DMD cardio-
(Figure 6) (35–37). These electrocardiographic (ECG) myopathy, atrial arrhythmias, including atrial fibril-
findings correlate with the pathological studies that lation, atrial flutter, and atrial tachycardias, can also
have demonstrated a predilection for fibrosis in the occur. Ventricular tachycardia, premature ventricular
basal posterior wall of the heart in patients with complexes, and other conduction abnormalities have
muscular dystrophies and may reflect a reduction in been noted, and patients with DMD with left ven-
electrical activity in the inferobasal ventricular wall tricular ejection fraction (LVEF) <35% have a signifi-
(38). ECG findings are believed to precede echocar- cantly higher burden of ventricular tachycardia
diographic findings of cardiomyopathy; however, no (48,49). In addition to low LVEF, increased QT
correlation between ECG findings and the presence of dispersion has been identified as a risk factor for
cardiomyopathy has yet been established (35,39). The ventricular arrhythmias in DMD; however, there was
use of serum biomarkers, such as cardiac troponin or no prognostic value of signal-averaged ECG (49,50).
B-type natriuretic peptides (BNPs), has not been well The role of implantable cardioverter-defibrillators
established for DMD screening and will need further (ICDs) is well-established for primary prevention of
examination in the future. sudden cardiac death in patients with an LVEF <35%;
Arrhythmias are a common cardiac involvement in however, the use of ICDs has not been well estab-
patients with muscular dystrophies. Sinus tachy- lished in patients with DMD cardiomyopathy (51). The
cardia is a common finding in patients with DMD guidelines discourage the use of ICDs for a patient
(40–42). Dystrophic patients have elevated heart with a life expectancy <1 year, and ICDs may not have
rates, even when compared with patients with other been utilized in the past due to the previous high
muscular dystrophies or deconditioned patients (42). mortality in patients with DMD. The recent use of
Pathological examination of the heart in dystrophic neurohormonal therapy may also improve LVEF to
patients has demonstrated fibrosis of the conduction reduce the risk of sudden cardiac death. We would
system, in addition to the myocardium (43), which recommend ICD implantation candidacy for patients
may, in part, explain the autonomic dysfunction with DMD with LVEF <35%; however, an individual-
in the DMD population (44,45). Additionally, sinus ized discussion with patients and families is neces-
tachycardia may correlate with cardiac dysfunction in sary, given the course of disease progression.
JACC VOL. 67, NO. 21, 2016 Kamdar and Garry 2537
MAY 31, 2016:2533–46 DMD and BMD Cardiomyopathy

CENTRAL ILLUSTRATION Duchenne Muscular Dystrophy: Pathogenesis and Therapies

DUCHENNE MUSCULAR DYSTROPHY PATHOGENESIS THERAPIES

Disease typically affects boys Inherited or Current cardiac treatments


(1 in every 5,000 boys born in the U.S.) spontaneous mutation
in the DMD gene • Steroid treatment (glucocorticoids)
• Neurohormonal therapy: Angiotensin
converting enzyme inhibitors (ACEIs) and
Possible learning angiotensin receptor blockers (ARBs)
difficulties Absence of dystrophin—
the anchor that connects
Scoliosis Dystrophin the extracellular matrix Current non-cardiac treatments
and membrane proteins • Noninvasive nocturnal ventilation
Cardiomyopathy to the cell cytoskeleton
• Nutrition, exercise therapy,
Weak and psychological support
respiratory • Spinal stabilization
muscles Contraction of muscle
cell leads to cell
membrane damage Emerging treatments
Muscle loss
and weakness • Implantable cardioverter defibrillation
leading to • Beta-blockers
Increased calcium
loss of
influx into damaged • Aldosterone inhibitors
ambulation
cell leads to cell death • Exon skipping
• Dystrophin gene replacement
Progressive • Gene editing mediated with CRISPR/Cas9
muscle weakness • Advanced heart failure therapies

Kamdar, F. et al. J Am Coll Cardiol. 2016;67(21):2533–46.

ACEI ¼ angiotensin-converting enzyme inhibitor; ARB ¼ angiotensin receptor blocker; CRISPR/Cas9 ¼ clustered regularly interspaced short palindromic
repeats/CRISPR-associated systems; DMD ¼ Duchenne muscular dystrophy.

CARDIAC IMAGING IN DMD function (54–56). Silva et al. (54) performed gadolin-
ium contrast-enhanced CMR on 10 patients with
Cardiac imaging can be challenging in patients with dystrophinopathies (8 patients with DMD and 2 with
end-stage DMD due to scoliosis, ventilation abnor- BMD) and were the first to demonstrate late gado-
malities, and contractures. Current, commonly used linium enhancement (LGE) by CMR in the dystrophic
imaging modalities include echocardiography and heart. They further demonstrated that LGE was pre-
cardiac magnetic resonance (CMR). Echocardiography sent by echocardiography, even with normal left
in DMD cardiomyopathy has demonstrated regional ventricular function (54). Puchalski et al. (57) subse-
wall motion abnormalities in the posterior basal wall, quently performed a study of 74 patients with DMD,
left ventricular dilation, and overall reduced systolic where the majority of patients had LGE in the post-
function (52). Current guidelines recommend an erobasal region of the left ventricle in a subepicardial
echocardiogram at the time of diagnosis or by 6 years distribution (Figure 7). This pattern of LGE in the
of age, with repeat echocardiograms every 1 to 2 years basal inferior and inferolateral walls is consistent
until 10 years of age. After 10 years of age, it is rec- with the pathological findings of fibrosis in the infe-
ommended that patients have an annual echocar- rior basal wall (38,58) (Figure 8). Myocardial fibrosis,
diogram to assess left ventricular function (27). as assessed by LGE in DMD, has been demonstrated to
Although echocardiography is easily accessible, rela- increase with age and correlates with a decline in
tively quick, and a cost-effective imaging modality LVEF (59,60). Thus, CMR may provide earlier detec-
to utilize, it can be technically challenging in tion of cardiovascular involvement in DMD, allow for
patients with DMD due to chest wall deformities, accurate and reproducible quantification of left ven-
scoliosis, and respiratory dysfunction, thus limiting tricular function and size, and promote initiation of
the diagnostic yield (53). CMR has been used to image earlier cardioprotective treatment. CMR has many
patients with DMD and can provide one of the most imaging benefits for patients with DMD; however, it
accurate assessments of left ventricular size and can also be challenging in the pediatric population
2538 Kamdar and Garry JACC VOL. 67, NO. 21, 2016

DMD and BMD Cardiomyopathy MAY 31, 2016:2533–46

F I G U R E 3 Absence of Dystrophin in DMD Skeletal Muscle

Photomicrographs of cross sections of normal and DMD human skeletal muscle. (A) Normal muscle biopsy shows relatively uniform fiber
diameter and peripherally located nuclei, with no fiber degeneration, inflammation, or endomysial fibrosis. (B) DMD muscle biopsy shows varied
fiber diameter, inflammation, necrotic changes with fat, and fibrous replacement of normal muscle tissue. (C) Normal muscle biopsy stained for
dystrophin, which is located at the plasmalemma (brown). (D) DMD muscle biopsy stained for dystrophin, demonstrating the absence of
dystrophin expression. DMD ¼ Duchenne muscular dystrophy.

due to the need for sedation, cost, and lack of acces-


sibility. Thus, we propose that CMR be obtained at the
time of diagnosis and annually after 10 years of age,
F I G U R E 4 Interventions Prolong Survival in DMD unless contraindicated.

30 PHARMACOLOGICAL TREATMENT OF
CARDIOMYOPATHY

CORTICOSTEROIDS. There is evidence that cortico-


20 steroids can improve not only muscular function, but
Age At Death

also cardiac function in dystrophinopathies (61–63).


One of the earliest studies was an evaluation of 21
patients with DMD treated with deflazacort for more
10 than 3 years who had a significantly lower incidence of
cardiomyopathy at 3 years than DMD control subjects
(5% vs. 58%) (62). Similarly, in a study of 48 patients
with DMD who had received either deflazacort or
0
prednisone, the steroid-treated DMD group had
SS

s
0

90

80

70

60

higher fractional shortening than patients with un-


99
t+

19
19

19
19
1
n

ed
nt

ed

d
ed
Ve

treated DMD (32). Another small study of children


e
Ve

Di
Di

Di
Di

with DMD demonstrated that the 14 patients treated

A marked increase in survival of patients with DMD has been achieved with the addition
with steroids through the 4.5-year follow-up had less
of ventilation strategies (vent) and spinal stabilization surgeries (SS). Adapted with ventricular dilation and systolic dysfunction than
permission from Eagle et al. (21). DMD ¼ Duchenne muscular dystrophy. those who had not received steroids (53). In one of
the largest retrospective studies that included 462
JACC VOL. 67, NO. 21, 2016 Kamdar and Garry 2539
MAY 31, 2016:2533–46 DMD and BMD Cardiomyopathy

F I G U R E 5 DMD Cardiac Disease Progression

Schematic outlining DMD cardiomyopathy disease progression. Initially, patients with DMD have structurally normal hearts. Subsequently, patients with
DMD develop fibrosis of the inferobasal wall as the earliest sign of myocardial involvement. Over time, this leads to progressive fibrosis, left ventricular (LV)
dysfunction, and dilation leading to end-stage heart failure. DMD ¼ Duchenne muscular dystrophy.

patients with DMD, those treated with steroids had a who received steroids had significantly lower cardio-
delay in the onset of cardiomyopathy (64). Addition- vascular mortality and incidence of new cardiomy-
ally, another retrospective study that included 86 opathy than control subjects, which was largely
patients with DMD concluded that patients with DMD driven by a reduction in heart failure–related

F I G U R E 6 ECG Changes in DMD

Electrocardiogram (ECG) from a patient with DMD. The ECG demonstrates sinus tachycardia, Q waves in leads I, aVL, and V4 to V6, and large R waves in
V1 and V2. DMD ¼ Duchenne muscular dystrophy.
2540 Kamdar and Garry JACC VOL. 67, NO. 21, 2016

DMD and BMD Cardiomyopathy MAY 31, 2016:2533–46

ventricular function. They randomized 57 children


F I G U R E 7 CMR Demonstrating Dilated Cardiomyopathy and
Fibrosis in a Patient With DMD
with DMD (mean age 10.7 years) to perindopril (2 to 4
mg/day) or placebo. At 3 years of follow-up, there was
no significant difference in LV function between the
children treated with perindopril or placebo. How-
ever, 3 years following enrollment, all patients were
switched to perindopril treatment and followed for an
additional 2 years. After crossing over to perindopril
at 2 years, there was no difference in mean LV func-
tion between those patients treated with perindopril
initially versus those initially treated with placebo.
However, in the initial placebo group, 8 of 29 patients
had LVEF <45%, whereas only 1 of 27 patients had LV
dysfunction in the perindopril group (p ¼ 0.02),
supporting the notion that early treatment with per-
indopril was effective in preventing progression to
left ventricular dysfunction in DMD. After a 10-year
follow-up period, only 65% of patients in the initial
placebo group were alive versus 92.9% in the initial
perindopril group (p ¼ 0.013), which emphasized that
early initiation of an ACEI reduced mortality in pa-
CMR delayed enhancement image in the basal short-axis view, tients with DMD (67). Current guidelines for DMD
performed on a 1.5-T MRI. This image demonstrates near trans- recommend initiation of ACEI in patients with DMD
mural enhancement (gray) in the left ventricular (LV) basal- only after LV dysfunction has developed (27); how-
midanterolateral, inferolateral, and lateral wall (area denoted by
ever, on the basis of studies by Duboc et al. (66,67),
dashed white line) consistent with myocardial scar or fibrosis. This
is in an 18-year-old male patient with DMD and exon 24 deletion
we would recommend initiation of an ACEI (by 10
with associated cardiomyopathy (LV ejection fraction 33%). CMR ¼ years of age) before the development of left ventric-
cardiac magnetic resonance; DMD ¼ Duchenne muscular dystrophy; ular dysfunction in patients with DMD, as they are at
MRI ¼ magnetic resonance imaging; RV ¼ right ventricle. high risk for developing left ventricular dysfunction
(ACC heart failure stages A and B). Additionally, for
those patients who are intolerant to ACEIs, ARBs can
deaths (65). However, a confounding variable in this also be used, as they were demonstrated to be as
study is that the patients with DMD treated with ste- effective as ACEIs in DMD (68).
roids also began taking angiotensin-converting BETA-ADRENERGIC RECEPTOR BLOCKERS. Although
enzyme inhibitors (ACEIs) approximately 3 years the benefit of ACEIs in DMD cardiomyopathy has been
earlier than the control subjects, which may have also definitively established, the efficacy of beta-blockers
been cardioprotective and had an effect on the results. in DMD cardiomyopathy has been less clear. The use
Although several studies have demonstrated a of carvedilol has been assessed in pediatric patients
delay in onset of cardiomyopathy in patients with with DMD who have elevated atrial natriuretic peptide
DMD, these have all been retrospective, observational (ANP) or BNP and a low ejection fraction (EF <40%) by
studies with inherent limitations. Additionally, the echocardiography with no significant difference in
variability of steroid dosing and duration poses a carvedilol-treated patients with respect to symptoms
challenge to interpretation of the data and broad or left ventricular dysfunction (69). However, in a
application of the results. To rigorously assess the study by Rhodes et al. (70), carvedilol was shown to be
effect of corticosteroid use on ventricular function in efficacious in patients with DMD cardiomyopathy.
patients with DMD, a prospective randomized When superimposed on background therapy of ACEIs,
controlled trial will be necessary. the use of carvedilol in this patient population re-
ACEIs AND ANGIOTENSIN RECEPTOR BLOCKERS. mains unclear. In an analysis of 13 patients with DMD
ACEIs and angiotensin receptor blockers (ARBs) are a who were treated with ACEI versus ACEI and carve-
cornerstone of neurohormonal modulation in heart dilol, echocardiography revealed a beneficial effect of
failure and were shown to be effective in the reduc- beta-blocker therapy in increasing left ventricular
tion of cardiovascular mortality in patients with heart shortening and decreasing left ventricular
failure (51). Duboc et al. (66) assessed the effect of end-diastolic dimensions (71). In contrast, a recent
ACEIs in patients with DMD with preserved left study by Viollet et al. (72) tested ACEI alone versus
JACC VOL. 67, NO. 21, 2016 Kamdar and Garry 2541
MAY 31, 2016:2533–46 DMD and BMD Cardiomyopathy

ACEI and metoprolol; in this study, a low-dose


F I G U R E 8 Cardiac Fibrosis in DMD
beta-blocker was added only for heart rates above
100 beats/min or if arrhythmias occurred. The results
showed an improvement from pre-treatment LVEF
in both groups, but no difference between them.
Further research with larger groups of patients and
more robust trial designs are needed to definitively
address the use of beta-blockers in DMD; however, on
the basis of current guidelines, we would recommend
that beta-blockers be initiated in patients with DMD
who have left ventricular dysfunction (51).
MINERALOCORTICOID RECEPTOR ANTAGONISTS.
Aldosterone inhibitors, such as spironolactone or
eplerenone, are standard heart failure therapy for
patients with LVEF <35% and New York Heart Asso-
ciation functional class II to VI symptoms (51). In a
recently completed randomized double-blinded clin-
ical trial, eplerenone, an aldosterone antagonist, or
placebo was added to background therapy of ACEIs or
ARBs in patients with DMD who had normal LV Photomicrograph of the basal posterior wall of the left ventricle in a patient with DMD
function to assess the efficacy of eplerenone in pre- demonstrating subepicardial fibrotic replacement (blue) of the myocardium using Masson
venting cardiomyopathy in DMD. Twenty patients trichrome stain. DMD ¼ Duchenne muscular dystrophy.
were randomized to eplerenone and 20 to placebo,
and they were followed for 6 and 12 months with
CMR. The primary endpoint was change in left ven- immunosuppression, had no difference in post-
tricular circumferential strain, which is a marker operative intubation, and were able to be rehabili-
of contractility, at 12 months. The investigators tated (74). Ruiz-Cano et al. (75) described a Spanish
observed that the decline in LV circumferential strain single-center experience with heart transplantation in
was lower in the group treated with eplerenone than 3 patients with BMD who underwent cardiac trans-
in the placebo group, although there was no overall plantation with a mean follow-up duration of 57
change in left ventricular function in either group months; this study also demonstrated that patients
(73). This small study demonstrated attenuation of with BMD had an intraoperative and post-operative
progressive left ventricular dysfunction that occurs in course comparable to nonmuscular dystrophy
DMD; although this study is positive, further studies patients undergoing heart transplantation. Patanè
to assess the effect of eplerenone on DMD survival are et al. (76) also described a single case of successful
warranted. Table 2 summarizes cardiac pharmaco- transplantation in a patient with cardiomyopathy
logical treatments for DMD cardiomyopathy. secondary to BMD. We performed a more recent
multicenter registry analysis of cardiac trans-
CARDIAC TRANSPLANTATION AND plantation using the Cardiac Transplant Research
LEFT VENTRICULAR ASSIST DEVICES Database and identified 29 patients with muscular
dystrophies, of whom 15 had BMD and 3 had DMD, who
The only curative therapy for end-stage heart failure underwent cardiac transplantation between 1995 and
remains cardiac transplantation. Heart failure with 2005 and compared them with 275 nonmuscular dys-
multisystem organ involvement and inability to trophy, nonischemic patients matched for age, body
rehabilitate after cardiac transplantation has been a
relative contraindication for orthotopic heart trans-
T A B L E 2 DMD Cardiomyopathy Treatment
plantation, and thus has limited the broad use of this
therapy in the DMD/BMD population. Rees et al. (74) Level of Evidence

were the first to describe heart transplantation Corticosteroids þþ


in patients with muscular dystrophies in a single ACE inhibitors þþþ

German center. Of 582 transplants performed, 3 Beta-blockers þ


Mineralocorticoid receptor antagonists þ
patients with DMD and 1 patient with BMD underwent
cardiac transplantation with a mean duration of ACE ¼ angiotensin-converting enzyme; DMD ¼ Duchenne muscular dystrophy.
follow-up of 40 months. These patients tolerated
2542 Kamdar and Garry JACC VOL. 67, NO. 21, 2016

DMD and BMD Cardiomyopathy MAY 31, 2016:2533–46

mass index, sex, and race (77). We demonstrated that BMD AND CARDIOMYOPATHY
there was no significant difference in 1- or 5-year sur-
vival, transplant rejection, infection, or allograft vas- In 1955, German physicians Becker and Kiener (82)
culopathy between the patients with and without described an X-linked muscular dystrophy with a
muscular dystrophy. These studies described compa- milder clinical course than DMD, now known as
rable outcomes of cardiac transplantation in a small Becker muscular dystrophy (83). In 1984, it was
and select group of patients with DMD and BMD with demonstrated that the gene responsible for BMD is
end-stage cardiomyopathy; however, the functional located on the X chromosome (84) and, subsequently,
status of these patients prior to transplantation was that mutations in the DMD gene resulted in BMD (85).
not documented, and these studies may have a BMD has an incidence of 1:19,000 and is an X-linked
selection bias. We propose that orthotopic heart recessive disorder resulting from a mutation of the
transplantation should be considered for the patient dystrophin gene (86). However, in comparison with
with DMD or BMD who has end-stage heart failure, patients with DMD who have a complete absence of
provided that other comorbidities do not limit survival dystrophin, dystrophin mutations in BMD tend to be
(i.e., respiratory failure). Further research regarding in-frame and result in misfolded or abnormal and less
cardiac transplantation in patients with DMD and BMD functional protein (87). Patients with BMD have a
who have end-stage cardiomyopathy is warranted. typically later age of onset and less severe clinical
Given the scarcity of organs for heart trans- involvement compared with patients with DMD
plantation, the use of left ventricular assist devices (Table 1) (88). Although the muscular symptoms may
(LVADs), demonstrated to be to be effective in treat- be less severe, over 70% of patients with BMD also
ing patients with end-stage or advanced heart failure, develop cardiomyopathy (89), and it is the leading
is applicable to a larger population, including those cause of death in patients with BMD (27). Cardiomy-
with muscular dystrophies, as LVADs can be used as opathy may be more severe in patients with BMD than
destination therapy without the need for trans- in patients with DMD (90,91). The onset of cardio-
plantation (78,79). Two groups recently reported myopathy is variable in BMD and is not correlated to
cases of successful implantation of LVADs as desti- skeletal muscle involvement (89,92). CMR studies
nation therapy in patients with DMD (80,81). Amodeo have demonstrated an inferobasal fibrotic pattern in
et al. (80) were the first to describe LVAD implanta- patients with BMD similar to that seen in DMD (93).
tion in 2 pediatric patients with DMD. These in- Patients with BMD cardiomyopathy should receive
vestigators implanted the Jarvik 2000 LVAD (Jarvik standard medical heart failure therapy (51). As pa-
Heart Inc., New York, New York) in a 15-year-old boy tients with BMD have a relatively milder skeletal
with DMD who had inotrope-refractory heart failure muscle phenotype, selected patients with BMD have
and in a 14-year-old boy with DMD who was bridged received LVADs and heart transplantation for severe
from extracorporeal membrane oxygenation to a Jar- cardiomyopathy with good outcomes (74–77).
vik 2000 LVAD. These patients were discharged 3 and
6 months after LVAD implantation, respectively. CARRIER STATUS AND CARDIOMYOPATHY
Ryan et al. (81) subsequently described implantation
of the HeartMate II LVAD (Thoratec, Pleasanton, As DMD and BMD affect males, given that it is
California) in a 29-year-old male patient with DMD inherited in an X-linked recessive manner, females
and end-stage heart failure and of the HeartWare can be carriers of DMD and BMD. The majority of DMD
LVAD (HeartWare, Framingham, Massachusetts) in a and BMD is inherited, and thus there are a significant
23-year-old female symptomatic DMD carrier with number of female carriers of DMD and BMD. The
end-stage heart failure. majority of female carriers of DMD and BMD are
The LVAD as destination therapy is a potentially asymptomatic; however, female carriers can become
promising therapy to address end-stage heart failure symptomatic or become manifesting carriers. Mani-
in patients with dystrophin-deficient heart failure; festing carriers can have symptoms, such as mild
however, post-operative complications, including muscle weakness, elevated serum creatinine kinase,
respiratory failure, rehabilitation, bleeding, stroke, and cardiomyopathy (94,95). Hoogerwaard et al.
and arrhythmias, will need to be evaluated further in (96,97) evaluated 90 women who were carriers of
this population. Extensive pre- and post-operative dystrophin mutations and identified 22% with symp-
management in an experienced center would be toms, including muscle weakness, and 18% with evi-
necessary for LVAD implantation in the DMD popu- dence of dilated left ventricles. The age of onset of
lation, and larger studies will be needed to evaluate carriers is variable, and the proportion of symptomatic
the efficacy and outcomes in this population. carriers has ranged from 2.5% of 22% in prior studies
JACC VOL. 67, NO. 21, 2016 Kamdar and Garry 2543
MAY 31, 2016:2533–46 DMD and BMD Cardiomyopathy

(96,97). X-inactivation is the mechanism where 1 of the United Kingdom; Prosensa, Leiden, the Netherlands)
2 X chromosomes in female cells randomly becomes and eteplirsen (AVI 4658, Sarepta Therapeutics,
transcriptionally inactive. It is postulated that carriers Cambridge, Massachusetts), are currently being
can become symptomatic on the basis of the extent of examined in clinical trials. Initial phase I and II trials
random X-inactivation of the normal X chromosome for both eteplirsen and drisapersen were promising
versus the dystrophic X chromosome. Although DMD in restoring dystrophin expression, but did not
and BMD carriers have been identified as having car- demonstrate a significant change in the primary
diomyopathy, the prevalence and severity of disease endpoint, the 6-min walk test. Major side effects
are incompletely defined (96,98). We recommend that included renal toxicity and thrombocytopenia
all female dystrophinopathy carriers be screened for (105,106). These drugs are being further evaluated in
cardiomyopathy, including CMR, at the time of diag- phase III clinical trials.
nosis of their children (99,100).
DYSTROPHIN GENE AND CELL THERAPY. As dystro-
X-LINKED DILATED CARDIOMYOPATHY phin mutations lead to complete absence of the
functional dystrophin protein in DMD, 1 of the stra-
X-linked dilated cardiomyopathy (XLDCM) was first tegies to mitigate the disease sequelae is gene
described in 1987 and is an extremely rare cardiomy- replacement therapy using recombinant adenoviral
opathy resulting from mutations in the DMD gene that virus vectors (rAAV). In comparison to the exon
affects teenage males and their mothers (101,102). skipping strategy, which currently is available only
Patients with XLDCM have normal dystrophin levels in for exon 51 skipping, gene replacement strategies can
skeletal muscle, but a complete loss of dystrophin in be beneficial for all patients with DMD, regardless of
cardiac muscle, thus making it a distinct phenotype their specific DMD mutation. A limitation of rAAV
(i.e., cardiomyopathy without overt skeletal muscle technology is the inability to deliver the large,
disease) from DMD or BMD (103). The rapidly pro- full-length dystrophin gene due to packaging limita-
gressive cardiomyopathy associated with XLDCM tions (107). However, the observation of mildly
results in death between 10 and 20 years of age. affected patients with BMD who had very large dys-
trophin gene deletions led to the understanding that
EMERGING THERAPIES
truncated dystrophins could be functional (85,108).
This led to the development of micro- or mini-
A major challenge for treatment of DMD is therapies
dystrophins, which contain only the essential, func-
that address dystrophin deficiency and target both
tional regions of the gene and can be packaged
cardiac and skeletal muscle (Central Illustration). A
within the rAAV (109). Dystrophin gene replacement
number of promising therapies are emerging,
therapy has been effective in mouse models; howev-
including exon skipping, dystrophin gene replace-
er, in large animal models, efficacy has been limited
ment, and gene-editing strategies.
due to the host immune response (110,111). In a hu-
EXON SKIPPING. DMD mutations are heterogeneous,
man pre-clinical trial of mini-dystrophin gene trans-
but the majority of mutations arise from an
fer, 6 patients with DMD received injections of
out-of-frame deletion or duplication within the DMD
mini-dystrophin, but none demonstrated significant
gene’s 79 exons. These mutations affect the normal
increases in dystrophin levels and 1 patient had an
transcription of dystrophin messenger ribonucleic
immune response (112,113). In addition, cell therapy
acid open reading frame and subsequently prevent
initiatives for DMD cardiomyopathy using
the full-length dystrophin protein from being tran-
cardiosphere-derived cells and/or induced pluripo-
scribed. A novel therapy for the treatment of DMD
tent stem cell derivatives have shown improvement
involves exon skipping within the central rod
in both cardiac function and exercise capacity of the
domain, which can restore the normal messenger
mdx (dystrophin knockout) mouse model. Gene
ribonucleic acid reading frame and convert an
replacement and cell therapies, although limited by
out-of-frame mutation to a less severe in-frame
immune side effects, may be overcome by immuno-
mutation, comparable to those seen in BMD. This
suppressant regimens and may represent promising
can be achieved through the use of antisense oligo-
therapies for DMD cardiomyopathy.
nucleotides (AONs), which are short, synthetic
nucleic acid fragments that regulate ribonucleic acid DYSTROPHIN GENE EDITING. Gene editing is an
splicing (104). Antisense oligonucleotides for exon emerging technology for DMD. Gene editing can be
skipping that target exon 51 of the DMD gene, which mediated by clustered regularly interspaced short
accounts for w13% of DMD mutations, including dri- palindromic repeats (CRISPR)/CRISPR-associated
sapersen (GSK-2402698, GlaxoSmithKline, Brentford, systems (Cas) to alter or edit the genome (114,115).
2544 Kamdar and Garry JACC VOL. 67, NO. 21, 2016

DMD and BMD Cardiomyopathy MAY 31, 2016:2533–46

The CRISPR/Cas9 system binds to the target gene and with medical and device therapies are warranted.
generates a double-stranded deoxyribonucleic acid Given the significant cardiomyopathic phenotype in
break, which then can be replaced by the corrected the dystrophinopathy population, we have estab-
gene sequence. This system precisely removes the lished a neurocardiomyopathy clinic where these
mutated gene of interest and replaces it with a func- patients have a comprehensive evaluation with heart
tional copy of the gene. Gene editing using a failure cardiologists and neuromuscular neurologists.
CRISPR/Cas9 strategy was utilized in vivo to correct We believe that increasing attention by heart failure–
the DMD gene mutation in the germline of mdx mice trained subspecialists and the establishment of
(116). In mosaic mdx mice with only 17% correction of dedicated neurocardiomyopathy clinics will enhance
dystrophin by gene editing, a normal muscle pheno- our understanding of the mechanisms of DMD car-
type was observed. This nascent technology has diomyopathy and ultimately have an effect on the
possible therapeutic benefits in patients with DMD, morbidity and mortality of this disease.
and further animal studies evaluating cardiac func- ACKNOWLEDGMENT The authors are grateful for
tion are needed. Recently, adult mdx mice were also artwork assistance by Ms. Cynthia DeKay.
successfully treated using a gene-editing strategy
(117,118).
REPRINT REQUESTS AND CORRESPONDENCE: Dr.
CONCLUSIONS Daniel J. Garry, Cardiovascular Division, Lillehei
Heart Institute, University of Minnesota, 2231 6th
Cardiomyopathy is a leading cause of death in the Street Southeast, 4-146 CCRB, Minneapolis, Minne-
DMD population. Early detection and intervention sota 55455. E-mail: garry@umn.edu.

REFERENCES

1. Meryon E. On granular and fatty degeneration 12. Rybakova IN, Patel JR, Ervasti JM. The dys- improvements in life expectancy since 1967 and
of the voluntary muscles. Medico-Chirugical Trans trophin complex forms a mechanically strong link the impact of home nocturnal ventilation. Neuro-
1852;35:73–4. between the sarcolemma and costameric actin. muscul Disord 2002;12:926–9.
J Cell Biol 2000;150:1209–14.
2. Duchenne GBA. Recherches sur la paralysie 22. Nigro G, Comi LI, Politano L, et al. The inci-
musculaire pseudo-hypertrophique ou paralysie 13. Ervasti JM, Campbell KP. A role for the dence and evolution of cardiomyopathy in
myo-sclerosique. Arch Gen Med 1868;11:5–25. dystrophin-glycoprotein complex as a trans- Duchenne muscular dystrophy. Int J Cardiol 1990;
membrane linker between laminin and actin. J Cell 26:271–7.
3. Gowers WR. A manual of disease of the nervous
Biol 1993;122:809–23. 23. Angelini C. The role of corticosteroids in
system1. London, UK: Churchill, 1886:391–4.
14. Cohn RD, Campbell KP. Molecular basis of muscular dystrophy: a critical appraisal. Muscle
4. Monaco AP, Neve RL, Colletti-Feener C, et al. Nerve 2007;36:424–35.
muscular dystrophies. Muscle Nerve 2000;23:
Isolation of candidate cDNAs for portions of the
1456–71. 24. Bushby K, Finkel R, Birnkrant DJ, et al., for the
Duchenne muscular dystrophy gene. Nature 1986;
15. Klietsch R, Ervasti JM, Arnold W, et al. DMD Care Considerations Working Group. Diag-
323:646–50.
Dystrophin-glycoprotein complex and laminin nosis and management of Duchenne muscular
5. Hoffman EP, Brown RH Jr., Kunkel LM. Dys- dystrophy, part 1: diagnosis, and pharmacological
colocalize to the sarcolemma and transverse tubules
trophin: the protein product of the Duchenne and psychosocial management. Lancet Neurol
of cardiac muscle. Circ Res 1993;72:349–60.
muscular dystrophy locus. Cell 1987;51:919–28. 2010;9:77–93.
16. Johnson EK, Zhang L, Adams ME, et al.
6. Emery AE. The muscular dystrophies. Lancet 25. Manzur AY, Kuntzer T, Pike M, et al. Gluco-
Proteomic analysis reveals new cardiac-specific
2002;359:687–95. corticoid corticosteroids for Duchenne muscular
dystrophin-associated proteins. PloS One 2012;7:
e43515. dystrophy. Cochrane Database Syst Rev 2008:
7. Cox GF, Kunkel LM. Dystrophies and heart dis-
CD003725.
ease. Curr Opin Cardiol 1997;12:329–43. 17. Cox GA, Cole NM, Matsumura K, et al. Over-
26. Bonifati MD, Ruzza G, Bonometto P, et al.
8. Mendell JR, Shilling C, Leslie ND, et al. expression of dystrophin in transgenic mdx mice
A multicenter, double-blind, randomized trial of
Evidence-based path to newborn screening for eliminates dystrophic symptoms without toxicity.
deflazacort versus prednisone in Duchenne
Duchenne muscular dystrophy. Ann Neurol 2012; Nature 1993;364:725–9.
muscular dystrophy. Muscle Nerve 2000;23:
71:304–13. 18. Petrof BJ, Shrager JB, Stedman HH, et al. 1344–7.
9. Koenig M, Hoffman EP, Bertelson CJ, et al. Dystrophin protects the sarcolemma from stresses
27. Bushby K, Finkel R, Birnkrant DJ, et al., for the
Complete cloning of the Duchenne muscular dys- developed during muscle contraction. Proc Natl
DMD Care Considerations Working Group. Diag-
trophy (DMD) cDNA and preliminary genomic or- Acad Sci U S A 1993;90:3710–4.
nosis and management of Duchenne muscular
ganization of the DMD gene in normal and 19. Yasuda S, Townsend D, Michele DE, et al. dystrophy, part 2: implementation of multidisci-
affected individuals. Cell 1987;50:509–17. Dystrophic heart failure blocked by membrane plinary care. Lancet Neurol 2010;9:177–89.
10. Ahn AH, Kunkel LM. The structural and func- sealant poloxamer. Nature 2005;436:1025–9.
28. Gozal D. Pulmonary manifestations of neuro-
tional diversity of dystrophin. Nat Genet 1993;3: 20. Konagaya M, Takayanagi T. Regularity in the muscular disease with special reference to
283–91. change of serum creatine kinase level in Duchenne Duchenne muscular dystrophy and spinal muscular
muscular dystrophy. A study with long-term atrophy. Pediatr Pulmonol 2000;29:141–50.
11. Dent KM, Dunn DM, von Niederhausern AC,
follow-up cases. Jpn J Med 1986;25:2–8.
et al. Improved molecular diagnosis of dystrophi- 29. Eagle M, Bourke J, Bullock R, et al. Managing
nopathies in an unselected clinical cohort. Am J 21. Eagle M, Baudouin SV, Chandler C, et al. Duchenne muscular dystrophy—the additive effect
Med Genet A 2005;134:295–8. Survival in Duchenne muscular dystrophy: of spinal surgery and home nocturnal ventilation
JACC VOL. 67, NO. 21, 2016 Kamdar and Garry 2545
MAY 31, 2016:2533–46 DMD and BMD Cardiomyopathy

in improving survival. Neuromuscul Disord 2007; 45. Yotsukura M, Fujii K, Katayama A, et al. Nine- with Duchenne muscular dystrophy: effect of age
17:470–5. year follow-up study of heart rate variability in and left ventricular systolic function. J Cardiovasc
patients with Duchenne-type progressive muscular Magn Reson 2013;15:107.
30. Finder JD, Birnkrant D, Carl J, et al. Respira-
dystrophy. Am Heart J 1998;136:289–96.
tory care of the patient with Duchenne muscular 60. Tandon A, Villa CR, Hor KN, et al. Myocardial
dystrophy: ATS consensus statement. Am J Respir 46. Thomas TO, Morgan TM, Burnette WB, et al. fibrosis burden predicts left ventricular ejection
Crit Care Med 2004;170:456–65. Correlation of heart rate and cardiac dysfunction in fraction and is associated with age and steroid
Duchenne muscular dystrophy. Pediatr Cardiol treatment duration in Duchenne muscular dystro-
31. American Academy of Pediatrics Section on
2012;33:1175–9. phy. J Am Heart Assoc 2015;4:e001338.
Cardiology and Cardiac Surgery. Cardiovascular
health supervision for individuals affected by 47. Dalmaz Y, Peyrin L, Mamelle JC, et al. The 61. Dec GW. Steroid therapy effectively delays
Duchenne or Becker muscular dystrophy. Pediat- pattern of urinary catecholamines and their me- Duchenne’s cardiomyopathy. J Am Coll Cardiol
rics 2005;116:1569–73. tabolites in Duchenne myopathy, in relation to 2013;61:955–6.
disease evolution. J Neural Transm 1979;46:17–34.
32. Markham LW, Spicer RL, Khoury PR, et al. 62. Silversides CK, Webb GD, Harris VA, et al. Ef-
Steroid therapy and cardiac function in Duchenne 48. Villa CR, Czosek RJ, Ahmed H, et al. Ambula- fects of deflazacort on left ventricular function in
muscular dystrophy. Pediatr Cardiol 2005;26: tory monitoring and arrhythmic outcomes in patients with Duchenne muscular dystrophy. Am J
768–71. pediatric and adolescent patients with Duchenne Cardiol 2003;91:769–72.
muscular dystrophy. J Am Heart Assoc 2015;5:
33. Jefferies JL, Eidem BW, Belmont JW, et al. 63. Mavrogeni S, Papavasiliou A, Douskou M, et al.
e002620.
Genetic predictors and remodeling of dilated car- Effect of deflazacort on cardiac and sternocleido-
49. Corrado G, Lissoni A, Beretta S, et al. Prog- mastoid muscles in Duchenne muscular dystrophy:
diomyopathy in muscular dystrophy. Circulation
nostic value of electrocardiograms, ventricular a magnetic resonance imaging study. Eur J Pae-
2005;112:2799–804.
late potentials, ventricular arrhythmias, and left diatr Neurol 2009;13:34–40.
34. Romfh A, McNally EM. Cardiac assessment in ventricular systolic dysfunction in patients with
Duchenne and Becker muscular dystrophies. Curr 64. Barber BJ, Andrews JG, Lu Z, et al. Oral cor-
Duchenne muscular dystrophy. Am J Cardiol
Heart Fail Rep 2010;7:212–8. ticosteroids and onset of cardiomyopathy in
2002;89:838–41.
Duchenne muscular dystrophy. J Pediatr 2013;163:
35. Thrush PT, Allen HD, Viollet L, et al. Re- 50. Yotsukura M, Yamamoto A, Kajiwara T, et al. 1080–4.e1.
examination of the electrocardiogram in boys with QT dispersion in patients with Duchenne-type
65. Schram G, Fournier A, Leduc H, et al. All-cause
Duchenne muscular dystrophy and correlation progressive muscular dystrophy. Am Heart J
mortality and cardiovascular outcomes with pro-
with its dilated cardiomyopathy. Am J Cardiol 1999;137:672–7.
phylactic steroid therapy in Duchenne muscular
2009;103:262–5.
51. Yancy CW, Jessup M, Bozkurt B, et al. 2013 dystrophy. J Am Coll Cardiol 2013;61:948–54.
36. Perloff JK, Roberts WC, de Leon AC Jr., et al. The ACCF/AHA guideline for the management of heart
66. Duboc D, Meune C, Lerebours G, et al. Effect
distinctive electrocardiogram of Duchenne’s progres- failure: a report of the American College of Car-
diology Foundation/American Heart Association of perindopril on the onset and progression of left
sive muscular dystrophy. An electrocardiographic-
Task Force on Practice Guidelines. J Am Coll Car- ventricular dysfunction in Duchenne muscular
pathologic correlative study. Am J Med 1967;42:
diol 2013;62:e147–239. dystrophy. J Am Coll Cardiol 2005;45:855–7.
179–88.
52. Rapezzi C, Leone O, Biagini E, et al. Echocar- 67. Duboc D, Meune C, Pierre B, et al. Perindopril
37. Sanyal SK, Johnson WW, Thapar MK, et al. An
diographic clues to diagnosis of dystrophin related preventive treatment on mortality in Duchenne
ultrastructural basis for electrocardiographic
dilated cardiomyopathy. Heart 2007;93:10. muscular dystrophy: 10 years’ follow-up. Am
alterations associated with Duchenne’s progres-
Heart J 2007;154:596–602.
sive muscular dystrophy. Circulation 1978;57: 53. Markham LW, Kinnett K, Wong BL, et al.
1122–9. Corticosteroid treatment retards development of 68. Allen HD, Flanigan KM, Thrush PT, et al.
ventricular dysfunction in Duchenne muscular A randomized, double-blind trial of lisinopril and
38. Frankel KA, Rosser RJ. The pathology of the
dystrophy. Neuromuscul Disord 2008;18:365–70. losartan for the treatment of cardiomyopathy in
heart in progressive muscular dystrophy: epi-
Duchenne muscular dystrophy [published correction
myocardial fibrosis. Human Pathol 1976;7:375–86. 54. Silva MC, Meira ZM, Gurgel Giannetti J, et al.
appears in PLoS Curr 2015;7]. PLoS Curr 2013;5.
Myocardial delayed enhancement by magnetic
39. Markham LW, Michelfelder EC, Border WL, 69. Saito T, Matsumura T, Miyai I, Nozaki S,
resonance imaging in patients with muscular dys-
et al. Abnormalities of diastolic function precede
trophy. J Am Coll Cardiol 2007;49:1874–9. Shinno S. [Carvedilol effectiveness for left
dilated cardiomyopathy associated with Duchenne
ventricular-insufficient patients with Duchenne
muscular dystrophy. J Am Soc Echocardiogr 2006; 55. Hagenbuch SC, Gottliebson WM, Wansapura J,
muscular dystrophy]. Rinsho Shinkeigaku 2001;41:
19:865–71. et al. Detection of progressive cardiac dysfunction
691–4.
by serial evaluation of circumferential strain in
40. Boas EP, Lowenburg H. The heart rate in 70. Rhodes J, Margossian R, Darras BT, et al.
patients with Duchenne muscular dystrophy. Am J
progressive muscular dystrophy. Arch Intern Med Safety and efficacy of carvedilol therapy for pa-
Cardiol 2010;105:1451–5.
1931;47:376–83. tients with dilated cardiomyopathy secondary to
56. Hor KN, Wansapura J, Markham LW, et al.
41. Fitch CW, Ainger LE. The Frank vectorcardio- muscular dystrophy. Pediatr Cardiol 2008;29:
Circumferential strain analysis identifies strata of
gram and the electrocardiogram in Duchenne 343–51.
cardiomyopathy in Duchenne muscular dystrophy:
progressive muscular dystrophy. Circulation 1967; a cardiac magnetic resonance tagging study. J Am 71. Kajimoto H, Ishigaki K, Okumura K, et al. Beta-
35:1124–40. Coll Cardiol 2009;53:1204–10. blocker therapy for cardiac dysfunction in patients
42. Kovick RB, Fogelman AM, Abbasi AD, et al. with muscular dystrophy. Circ J 2006;70:991–4.
57. Puchalski MD, Williams RV, Askovich B, et al.
Echocardiographic evaluation of posterior left Late gadolinium enhancement: precursor to car- 72. Viollet L, Thrush PT, Flanigan KM, et al. Effects
ventricular wall motion in muscular dystrophy. diomyopathy in Duchenne muscular dystrophy? Int of angiotensin-converting enzyme inhibitors
Circulation 1975;52:447–54. J Cardiovasc Imaging 2009;25:57–63. and/or beta blockers on the cardiomyopathy in
Duchenne muscular dystrophy. Am J Cardiol 2012;
43. Nomura H, Hizawa K. Histopathological study 58. Verhaert D, Richards K, Rafael-Fortney JA,
110:98–102.
of the conduction system of the heart in Duchenne et al. Cardiac involvement in patients with
progressive muscular dystrophy. Acta Pathol Jpn muscular dystrophies: magnetic resonance imag- 73. Raman SV, Hor KN, Mazur W, et al. Eplerenone
1982;32:1027–33. ing phenotype and genotypic considerations. Circ for early cardiomyopathy in Duchenne muscular
Cardiovasc Imaging 2011;4:67–76. dystrophy: a randomised, double-blind, placebo-
44. Yotsukura M, Sasaki K, Kachi E, et al. Circadian
controlled trial. Lancet Neurol 2015;14:153–61.
rhythm and variability of heart rate in Duchenne- 59. Hor KN, Taylor MD, Al-Khalidi HR, et al.
type progressive muscular dystrophy. Am J Car- Prevalence and distribution of late gadolinium 74. Rees W, Schüler S, Hummel M, et al.
diol 1995;76:947–51. enhancement in a large population of patients Heart transplantation in patients with muscular
2546 Kamdar and Garry JACC VOL. 67, NO. 21, 2016

DMD and BMD Cardiomyopathy MAY 31, 2016:2533–46

dystrophy associated with end-stage cardiomy- patients affected with subclinical Becker’s 105. Goemans NM, Tulinius M, van den Akker JT,
opathy. J Heart Lung Transplant 1993;12:804–7. muscular dystrophy. Circulation 1996;94:3168–75. et al. Systemic administration of PRO051 in
Duchenne’s muscular dystrophy. N Engl J Med
75. Ruiz-Cano MJ, Delgado JF, Jiménez C, et al. 90. Finsterer J, Stöllberger C. Cardiac involvement
2011;364:1513–22.
Successful heart transplantation in patients with in Becker muscular dystrophy. Can J Cardiol 2008;
inherited myopathies associated with end-stage 24:786–92. 106. Cirak S, Arechavala-Gomeza V, Guglieri M,
cardiomyopathy. Transplant Proc 2003;35:1513–5. et al. Exon skipping and dystrophin restoration in
91. Hoogerwaard EM, de Voogt WG, Wilde AA,
patients with Duchenne muscular dystrophy after
76. Patanè F, Zingarelli E, Attisani M, et al. Suc- et al. Evolution of cardiac abnormalities in Becker
systemic phosphorodiamidate morpholino olig-
cessful heart transplantation in Becker’s muscular muscular dystrophy over a 13-year period.
omer treatment: an open-label, phase 2, dose-
dystrophy. Eur J Cardiothorac Surg 2006;29:250. J Neurol 1997;244:657–63.
escalation study. Lancet 2011;378:595–605.
77. Wu RS, Gupta S, Brown RN, et al. Clinical 92. Nigro G, Politano L, Nigro V, et al. Mutation of
107. Pichavant C, Aartsma-Rus A, Clemens PR,
outcomes after cardiac transplantation in muscular dystrophin gene and cardiomyopathy. Neuromusc
et al. Current status of pharmaceutical and genetic
dystrophy patients. J Heart Lung Transplant 2010; Disord 1994;4:371–9.
therapeutic approaches to treat DMD. Mol Ther
29:432–8.
93. Yilmaz A, Gdynia HJ, Baccouche H, et al. Car- 2011;19:830–40.
78. Rose EA, Gelijns AC, Moskowitz AJ, et al., for diac involvement in patients with Becker muscular
108. England SB, Nicholson LV, Johnson MA, et al.
the Randomized Evaluation of Mechanical Assis- dystrophy: new diagnostic and pathophysiological
Very mild muscular dystrophy associated with the
tance for the Treatment of Congestive Heart insights by a CMR approach. J Cardiovasc Magn
deletion of 46% of dystrophin. Nature 1990;343:
Failure (REMATCH) Study Group. Long-term use Reson 2008;10:50.
180–2.
of a left ventricular assist device for end-stage
heart failure. N Engl J Med 2001;345:1435–43. 94. Norman A, Harper P. A survey of manifesting 109. Harper SQ, Hauser MA, DelloRusso C, et al.
carriers of Duchenne and Becker muscular dys- Modular flexibility of dystrophin: implications for
79. Slaughter MS, Rogers JG, Milano CA, et al., for
trophy in Wales. Clin Genet 1989;36:31–7. gene therapy of Duchenne muscular dystrophy.
the HeartMate II Investigators. Advanced heart
95. Comi LI, Nigro G, Politano L, et al. The car- Nat Med 2002;8:253–61.
failure treated with continuous-flow left ventric-
ular assist device. N Engl J Med 2009;361: diomyopathy of Duchenne/Becker consultands. Int 110. Gregorevic P, Allen JM, Minami E, et al.
2241–51. J Cardiol 1992;34:297–305. rAAV6-microdystrophin preserves muscle function
96. Hoogerwaard EM, van der Wouw PA, and extends lifespan in severely dystrophic mice.
80. Amodeo A, Adorisio R. Left ventricular assist
Wilde AA, et al. Cardiac involvement in carriers of Nat Med 2006;12:787–9.
device in Duchenne cardiomyopathy: can we
change the natural history of cardiac disease? Int J Duchenne and Becker muscular dystrophy. Neu- 111. Banks GB, Chamberlain JS, Froehner SC.
Cardiol 2012;161:e43. romusc Disord 1999;9:347–51. Truncated dystrophins can influence neuromus-
97. Hoogerwaard EM, Bakker E, Ippel PF, et al. cular synapse structure. Mol Cell Neurosci 2009;
81. Ryan TD, Jefferies JL, Sawnani H, et al. Im-
Signs and symptoms of Duchenne muscular dys- 40:433–41.
plantation of the HeartMate II and HeartWare left
ventricular assist devices in patients with trophy and Becker muscular dystrophy among 112. Mendell JR, Campbell K, Rodino-Klapac L,
Duchenne muscular dystrophy: lessons learned carriers in The Netherlands: a cohort study. Lancet et al. Dystrophin immunity in Duchenne’s muscular
from the first applications. ASAIO J 2014;60: 1999;353:2116–9. dystrophy. N Engl J Med 2010;363:1429–37.
246–8.
98. Schade van Westrum SM, Hoogerwaard EM, 113. Bowles DE, McPhee SW, Li C, et al. Phase 1
82. Becker PE, Kiener F. [A new x-chromosomal Dekker L, et al. Cardiac abnormalities in a follow- gene therapy for Duchenne muscular dystrophy
muscular dystrophy]. Archiv Psychiatr Nervenkr Z up study on carriers of Duchenne and Becker using a translational optimized AAV vector. Mol
Gesamte Neurol Psychiatr 1955;193:427–48. muscular dystrophy. Neurology 2011;77:62–6. Ther 2012;20:443–55.

83. Becker PE. Two families of benign sex-linked 99. Schelhorn J, Schoenecker A, Neudorf U, et al. 114. Jinek M, Chylinski K, Fonfara I, et al.
recessive muscular dystrophy. Rev Can Biol 1962; Cardiac pathologies in female carriers of Duchenne A programmable dual-RNA-guided DNA endonu-
21:551–66. muscular dystrophy assessed by cardiovascular clease in adaptive bacterial immunity. Science
magnetic resonance imaging. Eur Radiol 2015;25: 2012;337:816–21.
84. Kingston HM, Sarfarazi M, Thomas NS, et al.
3066–72. 115. Cong L, Ran FA, Cox D, et al. Multiplex
Localisation of the Becker muscular dystrophy
gene on the short arm of the X chromosome by 100. Lang SM, Shugh S, Mazur W, et al. Myocardial genome engineering using CRISPR/Cas systems.
linkage to cloned DNA sequences. Hum Genet fibrosis and left ventricular dysfunction in Science 2013;339:819–23.
1984;67:6–17. Duchenne muscular dystrophy carriers using car- 116. Long C, McAnally JR, Shelton JM, et al. Pre-
85. Koenig M, Beggs AH, Moyer M, et al. The diac magnetic resonance imaging. Pediatr Cardiol vention of muscular dystrophy in mice by CRISPR/
molecular basis for Duchenne versus Becker 2015;36:1495–501. Cas9-mediated editing of germline DNA. Science
muscular dystrophy: correlation of severity with 101. Berko BA, Swift M. X-linked dilated cardio- 2014;345:1184–8.
type of deletion. Am J Hum Genet 1989;45: myopathy. N Engl J Med 1987;316:1186–91. 117. Long C, Amoasii L, Mireault AA, et al. Post-
498–506.
102. Muntoni F, Cau M, Ganau A, et al. Brief natal genome editing partially restores dystrophin
86. Worton R. Muscular dystrophies: diseases of report: deletion of the dystrophin muscle- expression in a mouse model of muscular dystro-
the dystrophin-glycoprotein complex. Science promoter region associated with X-linked dilated phy. Science 2016;351:400–3.
1995;270:755–6. cardiomyopathy. N Engl J Med 1993;329:921–5. 118. Nelson CE, Hakim CH, Ousterout DG, et al.
87. Monaco AP, Bertelson CJ, Liechti-Gallati S, 103. Towbin JA, Hejtmancik JF, Brink P, et al. In vivo genome editing improves muscle function
et al. An explanation for the phenotypic differ- X-linked dilated cardiomyopathy. Molecular in a mouse model of Duchenne muscular dystro-
ences between patients bearing partial deletions genetic evidence of linkage to the Duchenne phy. Science 2016;351:403–7.
of the DMD locus. Genomics 1988;2:90–5. muscular dystrophy (dystrophin) gene at the Xp21
88. Bradley WG, Jones MZ, Mussini JM, locus. Circulation 1993;87:1854–65.
Fawcett PR. Becker-type muscular dystrophy.
104. Aartsma-Rus A, Fokkema I, Verschuuren J, KEY WORDS Becker muscular dystrophy
Muscle Nerve 1978;1:111–32.
et al. Theoretic applicability of antisense-mediated cardiomyopathy, Duchenne muscular
89. Melacini P, Fanin M, Danieli GA, et al. exon skipping for Duchenne muscular dystrophy dystrophy cardiomyopathy, muscular
Myocardial involvement is very frequent among mutations. Hum Mutat 2009;30:293–9. dystrophy cardiomyopathy

You might also like