You are on page 1of 13

Seminar

Left ventricular non-compaction cardiomyopathy


Jeffrey A Towbin, Angela Lorts, John Lynn Jefferies

Left ventricular non-compaction, the most recently classified form of cardiomyopathy, is characterised by abnormal Published Online
trabeculations in the left ventricle, most frequently at the apex. It can be associated with left ventricular dilation or April 10, 2015
http://dx.doi.org/10.1016/
hypertrophy, systolic or diastolic dysfunction, or both, or various forms of congenital heart disease. Affected S0140-6736(14)61282-4
individuals are at risk of left or right ventricular failure, or both. Heart failure symptoms can be induced by exercise The Heart Institute, Cincinnati
or be persistent at rest, but many patients are asymptomatic. Patients on chronic treatment for compensated heart Children’s Hospital Medical
failure sometimes present acutely with decompensated heart failure. Other life-threatening risks of left ventricular Center, Cincinnati, OH, USA
non-compaction are ventricular arrhythmias or complete atrioventricular block, presenting clinically as syncope, and (J A Towbin MD, A Lorts MD,
J L Jefferies MD)
sudden death. Genetic inheritance arises in at least 30–50% of patients, and several genes that cause left ventricular
Correspondence to:
non-compaction have been identified. These genes seem generally to encode sarcomeric (contractile apparatus) or
Dr Jeffrey A Towbin, Le Bonheur
cytoskeletal proteins, although, in the case of left ventricular non-compaction with congenital heart disease, Children’s Hosptial, University of
disturbance of the NOTCH signalling pathway seems part of a final common pathway for this form of the disease. Tennessee Health Science Centre,
Disrupted mitochondrial function and metabolic abnormalities have a causal role too. Treatments focus on Memphis, TN 38103, USA
jtowbin1@uthsc.edu
improvement of cardiac efficiency and reduction of mechanical stress in patients with systolic dysfunction. Further,
treatment of arrhythmia and implantation of an automatic implantable cardioverter-defibrillator for prevention of
sudden death are mainstays of therapy when deemed necessary and appropriate. Patients with left ventricular non-
compaction and congenital heart disease often need surgical or catheter-based interventions. Despite progress in
diagnosis and treatment in the past 10 years, understanding of the disorder and outcomes need to be improved.

Introduction Pathological changes


Left ventricular non-compaction cardiomyopathy (LVNC), In the early embryo, the heart is a loose, interwoven
which was first described by Grant1 in 1926, is a mesh of muscle fibres.5,6,17,19–21 The developing myocardium
heterogeneous myocardial disorder characterised by gradually condenses, and the large spaces within the
prominent trabeculae, intratrabecular recesses, and a trabecular meshwork disappear, resulting in condensing
left ventricular myocardium with two distinct layers: and compaction of the ventricular myocardium and
compacted and non-compacted.2–4 Continuity exists solidification of the endocardial surfaces. Trabecular
between the left ventricular cavity and deep intra- compaction is usually more complete in the left than in
trabecular recesses, both of which are filled with blood, the right ventricular myocardium, and therefore right
and no evidence of communication with the epicardial ventricular trabeculations are usually noted in the mature
coronary artery system is noted.5,6 Although LVNC mainly heart. Failure in the compaction pathway is thought
affects the left ventricle, isolated right ventricular and to occur because of arrest of endomyocardial morpho-
biventricular non-compaction also occur.7–9 Imaging and genesis, causing postnatal LVNC.5,6,17,19–21 The gross
assessment of pathological changes show that the pathological appearance of LVNC is characterised by
disorder is characterised by a spongy left ventricular excessively prominent trabeculations and deep intra-
myocardium with abnormal trabeculations usually most trabecular recesses resembling the right ventricular
evident in the left ventricular apex.3,10 The American endomyocardial morphology. Histologically, the recesses
Heart Association formally classified LVNC as a distinct and their troughs are lined with endothelium. Zones
cardiomyopathy in 2006.11 of fibrous and elastic tissue might be scattered on the
LVNC has been known by several names, including endocardial surfaces, with extension into the recesses.
spongy myocardium, fetal myocardium, non-compaction Coronary arterial circulation is usually normal;
of the left ventricular myocardium, hypertrabeculation extramural myocardial blood supply is not thought
syndrome, and left ventricular non-compaction, among
others.2,3,6,10–16 The genesis of LVNC has been speculated
to represent arrest of the final stage of myocardial Search strategy and selection criteria
morphogenesis (myocardial compaction).5,6,17–22 However, We searched PubMed, Medline, Current Contents, and OMIM
this hypothesis does not explain why several types of with the search terms “left ventricular noncompaction”,
LVNC occur, such as primary myocardial forms, a form “noncompaction cardiomyopathy”, “spongioform
associated with arrhythmias, and a form associated with cardiomyopathy”, and “left ventricular hypertrabeculation”
congenital heart disease, including septal defects, right for articles in English published between Jan 1, 1950, and
heart obstructive abnormalities (eg, pulmonic stenosis July 1, 2014. We identified trials in progress from
and Ebstein’s anomaly), hypoplastic left heart syndrome, ClinicalTrials.gov and clinicaltrialsregister.eu. We excluded
and others.10,12,23–29 In all types of LVNC, metabolic outdated textbook chapters, but did use our own published
derangements can be noted, particularly in neonates and and unpublished data.
infants.10,30,31

www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4 1


Seminar

to have a role in these abnormalities.32 Intramural some reports, but a low frequency of death or
perfusion, however, could be adversely affected by the transplantation is noted in others. For instance, Ichida
prominent trabeculations and intratrabecular recesses, and colleagues14 reported good survival and few
particularly in the subendocardium, causing sub- symptoms in paediatric patients whereas Chin and
endocardial ischaemia. Although no clear evidence investigators2 reported three deaths in the eight children
shows that subendocardial ischaemia causes LVNC, an that they studied. In our research group,3 outcomes
ischaemic insult to critical signalling pathways between were poor in neonates but excellent in children aged
the myocardium and the endocardium could affect between 18 months and 3 years, with 75% alive and
ventricular trabeculation.33,34 During cardiogenesis, the transplant-free 5 years after diagnosis. The neonates who
cardiac jelly has an important role in the interaction died all had systemic disease (mitochondrial or other
between these two layers. Whether ischaemia could metabolic disorders).
result in a disruption or modification of signalling Clinical studies in adults have consistently shown a
pathways is unclear.35 Additionally, no definitive high risk of ventricular tachyarrhythmias and sudden
evidence shows that subendocardial ischaemia plays an cardiac death in LVNC. As much as 47% of adults (and
important part in the clinical course of patients with 75% of symptomatic patients) die within 6 years of
LVNC, although cardiac MRI with late gadolinium presentation.4,24,37–39 But newer studies show a more
enhancement shows fibrosis in some patients. The benign natural history and lower risk for ventricular
endomyocardial morphology of LVNC also lends itself arrhythmias, including malignant types, than previously
to development of mural thrombi within the recesses, reported.47 Bhatia and colleagues48 reviewed published
which can embolise and cause stroke or coronary studies of 241 adults with isolated LVNC diagnosed by
obstruction.3,10,14,36,37 Arrhythmias can also occur.24,25,38,39 echocardiographic criteria, who were followed up for a
mean duration of 39 months. They reported a yearly
Prevalence event rate of 4% for cardiovascular deaths, 6·2%
Although LVNC has been deemed rare by some for cardiovascular death and associated surrogates
investigators, and its incidence and prevalence are (transplantation, appropriate implantable cardioverter-
uncertain, it seems to be the third most commonly defibrillator shocks), and 8·6% for all cardiovascular
diagnosed cardiomyopathy. Ritter and colleagues40 events (death, stroke, implantable cardioverter-defib-
reported a prevalence of isolated LVNC of 0·05% among rillator shocks, transplantation). Familial LVNC was
all adult echocardiographic examinations in a large identified in 30% of first-degree relatives of index cases
institution. Aras and coworkers41 reported a prevalence of screened by echocardiography.48 The precise substrate
less than 0·14% in adults referred for echocardiograms. for malignant ventricular arrhythmias in patients with
By contrast, Sandhu and colleagues42 recorded a 3·7% LVNC patients is unknown, however.49
prevalence of definite or probable LVNC in adults with Although diagnosis has focused mainly on
left ventricular ejection fractions of 45% or less and identification and description of trabeculations, other
0·26% prevalence in all patients referred for features are crucial to define specific subtypes of LVNC.
echocardiography. Most reports focus on so-called isolated LVNC, or LVNC
In patients with heart failure, the prevalence of LVNC with congenital heart disease. However, both of these
has been reported as 3–4%.43,44 These wide variations in types have a wide range of features.10,50 This lumping-
the reported prevalence probably depend on clinical together approach (rather than splitting) has several
recognition. Diagnosis of LVNC is becoming more disadvantages, particularly in terms of outcomes and
frequent, probably because of increased awareness, treatment. At least eight different phenotypes of LVNC
improved imaging technology, and recommendations seem to exist, all of which have different outcomes
that at-risk family members who also have the LVNC (figures 1A–F, 2A, 2B).
phenotype be screened (the disease is hereditary).
Subtypes
Clinical features and diagnosis Benign LVNC
Clinical presentation of LVNC is highly variable. It can So-called benign LVNC is characterised by normal left
occur at any age, range from asymptomatic to end-stage ventricular size and wall thickness with preserved
heart failure, or be associated with lethal arrhythmias, systolic and diastolic function. This subtype accounts
sudden cardiac death, or thromboembolic events (or for roughly 35% of patients and is a predictor of good
combinations thereof).2–4,7–10,12–14,45,46 Many patients are outcomes in the absence of clinically significant
asymptomatic, and identified serendipitously by echo- arrhythmias.51 On the basis of this subtype, some adult
cardiography after referral because of a murmur or for cardiologists have stated that LVNC does not represent a
familial screening. Some patients with LVNC present cardiomyopathy and is a benign and normal variant.52 A
with clinically significant arrhythmias or conduction possible explanation for this conclusion is that the
system disease. Data for outcomes in children and severe forms of LVNC tend to occur in childhood, and
adults have been inconsistent; outcomes are poor in those affected are either successfully treated,

2 www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4


Seminar

A B

C D

E F

Figure 1: Benign LVNC in a 12-year-old (A), dilated LVNC in a 6-year-old (B), hypertrophic LVNC in a 14-month-old (C), hypertrophic and dilated LVNC in a
20-month-old (D), restrictive LVNC in a 5-year-old (E), and biventricular LVNC in a 16-year-old (F)
The arrow in (A) shows trabeculations and intratrabecular recesses at the apex of the left ventricle. The arrow in (C) shows asymmetrical septal hypertrophy. In (C),
although the left ventricle is dilated, focal septal hypertrophy is apparent. The arrow in (E) shows atrial enlargement, which is pathognomonic of restrictive disease.
Note the dilated left atrium in the absence of mitral regurgitation. The arrows in (F) show trabeculations in both ventricles. LVNC=left ventricular non-compaction
cardiomyopathy.

transplanted, or die, and thus do not present to adult underlying arrhythmias are present and usually identified
cardiologists with symptomatic disease. Patients with at diagnosis. The presence of ventricular arrhythmias is
benign LVNC seem to have the same outcome as the an independent risk factor for mortality, and many are not
healthy population. detected by surveillance techniques.10,24,25,37,38,51,53–57 Patients
with LVNC with arrhythmias seem to have worse
LVNC with arrhythmias outcomes than do the general population or those with
In LVNC with arrhythmias, systolic function is preserved similar forms of rhythm disturbance but who do not have
and left ventricular size and wall thickness are normal, but LVNC.

www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4 3


Seminar

A B

Figure 2: LVNC in association with congenital heart disease in a 3-year-old (A), and LVNC in association with Ebstein’s anomaly in a neonate (B)
The arrow in (A) shows a ventricular septal defect with colour doppler. The arrow in (B) shows tricuspid valve displacement with ventricular atrialisation. LVNC=left
ventricular non-compaction cardiomyopathy.

Dilated LVNC mimics the clinical behaviour of restrictive cardiomyopathy


The dilated subtype of LVNC is characterised by and affected patients have similarly poor outcomes,
concomitant left ventricular dilation and systolic typically because of arrhythmia-related sudden cardiac
dysfunction.3,10,14,42,43,47,50 During its clinical course, a so-called events or, less frequently, heart failure with preserved
undulating phenotype can occur, in which the left ventricle ejection fraction.51 Restrictive LVNC has a similar prognosis
becomes smaller with some wall hypertrophy and to that of similar forms of restrictive cardiomyopathy.
improved function before reversion to dilation.3 Adults
with this subtype have similar outcomes to those without Right ventricular or biventricular LVNC
LVNC who have a similar degree of dilated cardiomyopathy; Right ventricular, or biventricular, LVNC is characterised
outcomes in neonates and infants are worse than in those by hypertrabeculation of both the right and left ventricles.
with other types of dilated cardiomyopathy. No recognised diagnostic criteria are available for right
ventricular non-compaction. In a previous report,58 use
Hypertrophic LVNC of the diagnostic criteria for LVNC was suggested.58 We
The hypertrophic subtype is characterised by left diagnose this subtype on the basis of very heavy
ventricular thickening, usually with asymmetrical septal trabeculation and severe spongioform appearance of the
hypertrophy, in addition to diastolic dysfunction and right ventricle. In these cases, trabeculations are noted
hypercontractile systolic function.51 In some cases, left in the lateral wall of the right ventricle, with hyper-
ventricular dilation with systolic dysfunction can occur trabeculation up to the tricuspid valve, particularly
late in the course of the disease. Patients with this in severe cases.3 The implications of biventricular
subtype seem to have similar outcomes to the general involvement remain unknown.7–9,55,56,59,60
population or those with a similar degree of hypertrophic
cardiomyopathy who do not have LVNC. LVNC with congenital heart disease
LVNC has been reported in association with almost all
Hypertrophic dilated LVNC congenital heart lesions and might contribute to myocardial
Some patients have a mixed phenotype characterised by dysfunction or arrhythmias, or both. Right-sided lesions—
left ventricular thickening, dilation, and depressed especially Ebstein’s anomaly, pulmonic stenosis,
systolic function at presentation. This phenotype is pulmonary atresia, tricuspid atresia, and double outlet right
associated with an increased risk of mortality and, in ventricle—are more frequently associated with LVNC than
paediatric patients, with metabolic or mitochondrial are other congenital heart lesions, although septal defects
disease.51 Hypertrophic and dilated LVNC is the most and left-sided lesions are also not uncommon.10,23,26–29,61
common of the undulating types, and typically results in Prognosis depends on the specific type of congenital heart
a dilated left ventricle with poor function and heart disease. However, LVNC increases postoperative risk in
failure. Prognosis for this subtype is worse than that patients with congenital heart disease, and, if ventricular
associated with other mixed phenotype presentations, dysfunction occurs at any time, their outcomes worsen.
such as burned-out forms of hypertrophic cardiomyopathy.
Imaging
Restrictive LVNC Diagnosis of LVNC relies on non-invasive imaging
Restrictive LVNC is rare. It is characterised by left atrial or studies—usually transthoracic echocardiography and
biatrial dilation and diastolic dysfunction. This phenotype cardiac MRI. However, the diagnostic criteria used for

4 www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4


Seminar

both these methods are highly controversial. Transthoracic


echocardiography remains the most common diagnostic
strategy, largely because of its widespread availability,
ease of interpretability, and low cost. The most common
diagnostic method is based on a ratio of the thickness of
the non-compacted layer to that of the compacted layer,
with a ratio of greater than 2:1 at the end of diastole
deemed diagnostic.62 Alternative diagnostic criteria have
been proposed, including ratios ranging between 2:1 and
3:1, but all necessitate expert assessment to discern
between normal variants and LVNC.2,13,63
The use of any ratio of non-compact to compact layer
thickness or trabecular length is perplexing. Why would
the size of trabeculations or ratio of layer thicknesses
matter? However, perhaps the position and density of
Figure 3: Cardiac MRI of a 15-year-old with benign left-ventricular
trabeculations could be crucial for diagnosis. Some data non-compaction cardiomyopathy
exist that help to elucidate these diagnostic quandaries. A coarsely trabeculated left ventricle is visible in this short axis view.
Punn and Silverman used the 16-segment model described
by the American Heart Association and the American Notably, cardiac MRI has shown that, in LVNC, the
Society of Echocardiography to do a retrospective analysis64 compact layer is frequently abnormally thin, especially at
of children with LVNC. Left ventricular ejection fraction the apex, and can actually be confused with being an
was inversely related to the number of segments apical aneurysm.
implicated, and in younger patients, particularly those More recently (eg, since 2001, with increasing popularity
aged 0–3 years, poor outcomes (as defined by death or occurring after 2007), cardiac CT has been used, which can
transplantation) were related to the number of segments show the abnormal architecture of the left ventricular wall
affected. in non-compaction. Cardiac CT enables quantitative and
Advanced echocardiographic techniques, such as qualitative assessment of global and regional ventricular
strain, strain rate, and torsion, are now being used to function, and is excellent for the exclusion of coronary
assist diagnosis of LVNC.65–67 Researchers who use the artery disease or anomalies, which is usually not feasible
ratios of layers as the basis for diagnosis support their with cardiac MRI or echocardiography.73 Whereas
viewpoints with statistical evidence, but no gold standard echocardiography and MRI do not expose patients to
exists against which data can be compared. However, radiation, CT delivers very high radiation doses, which is
because of the absence of definitive diagnostic criteria, an important and limiting concern because of oncological
concerns have been raised that LVNC has gone from potential, particularly in children or in patients who need
being under-diagnosed to over-diagnosed. long-term, repeated surveillance.74
Cardiac MRI is increasingly used in diagnosis and Our group uses a matrix of data to diagnose LVNC. We
surveillance of LVNC in both children and adults, and is use transthoracic echocardiography to assess location and
associated with the same controversies as density of trabeculations and visualise blood flow into the
echocardiography (figure 3). The diagnostic criteria for intertrabecular recesses by colour doppler interrogation
LVNC are also based on the ratio of the thickness of the on apical four-chamber view. Additionally, we measure the
non-compacted layer to that of the compacted layer, with a layer thickness ratio for completeness and assess the
ratio of greater than 2·3:1 at the end of diastole typically thickness of the compact layer compared with that in
used.68 However, as with echocardiography, consensus is healthy people. We also use the parasternal short axis view
lacking for this definition. Thuny and colleagues69 to trigger suspicion of LVNC, resulting in a higher
assessed 16 patients with LVNC who underwent both likelihood that attention will be paid to this possible
echocardiography and cardiac MRI within the same week diagnosis. We use a similar approach for cardiac MRI, but
for comparison. They used a standard 17-segment also use gadolinium to measure scar burden. Furthermore,
anatomical model and reported that the extent of LVNC we carefully assess left ventricular size, thickness, and
was better defined by cardiac MRI, which also provided systolic and diastolic function, and look for any associated
additional morphological characterisation of the congenital heart defects. Finally, we carefully analyse the
myocardium. Jacquier and investigators70 proposed right ventricle and its trabeculation burden.
diagnostic MRI criteria based on the amount of left The electrocardiogram of patients with LVNC is usually
ventricular mass and burden of trabeculations— abnormal. In as much as 87% of patients, it will show
trabeculations comprising more than 20% of the total hypertrophy by voltage criteria (either left ventricular
mass is deemed diagnostic. Cardiac MRI can be used to hypertrophy or biventricular hypertrophy), T-wave
assess myocardial fibrosis, and delayed gadolinium inversion, ST segment abnormalities or strain, left atrial
enhancement could offer prognostic information.71,72 enlargement, left axis deviation, QTc prolongation, or

www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4 5


Seminar

I aVR V1 V4 V3r

II aVL V2 V5 V4r

III aVF V3 V6 V7

II

25 mm/s 10 mm/mV 150 Hz 7.1.1 ISL 241 HD

Figure 4: Electrocardiogram from a patient aged 17 months with left ventricular non-compaction cardiomyopathy
Prominent precordial voltage is notable.

pre-excitation (figure 4). In neonates and young children, presented with appropriate defibrillator shocks as a result
extreme QRS voltages might be noted.3,14,51,54,75 of sustained ventricular tachycardia after a median of 6·1
months (IQR 1–16), suggesting that people with LVNC
Arrhythmias could be at high risk for sudden cardiac death. All the
Supraventricular and ventricular arrhythmias, and appropriate defibrillator interventions in this cohort were
bradyarrhythmias, many of which are life threatening, associated with fast ventricular tachycardias, although
occur frequently in LVNC. The LVNC subtype associated whether the initial rhythm in patients who had sudden
with early-onset rhythm abnormalities generally has a cardiac death due to ventricular fibrillation was also
substantial risk of sudden death. Implantable cardioverter initiated by a ventricular tachycardia trigger is unknown.77
defibrillators are highly effective for the prevention In patients with LVNC presenting with sustained
of sudden arrhythmic death in patients with LVNC, ventricular arrhythmias, the risk of recurrent sustained
including those with severe left ventricular dysfunction, ventricular tachycardia followed by appropriate
a previous history of sustained ventricular tachycardia or defibrillator shocks was 33% after median follow-up of
fibrillation, recurrent syncope of unknown cause, or 26 months. Similarly, Kobza and colleagues78 reported
a family history of sudden cardiac death. Ventricular appropriate shocks in 37% of patients with LVNC who
tachyarrhythmias, including those in patients in whom had implantable cardioverter defibrillators at mean
ventricular fibrillation causes cardiac arrest, are reported follow-up of 40 months. In small children, antiarrhythmic
in 38–47% of adult patients with LVNC and in 13–18% of drugs might be indicated before implantation of a
those who die suddenly.18,28,29,41,43,59,61 cardioverter defibrillator because of the high rate of lead
Caliskan and coworkers76 investigated the indications fractures and inappropriate shocks in this population.
for, and outcomes of, implantable cardioverter defib-
rillator therapy in 77 adults with LVNC, 44 of whom had Clinical genetics
such a device implanted on the basis of standard implant Inheritance of LVNC is most often X-linked recessive or
guidelines for non-ischaemic cardiomyopathy. During a autosomal dominant, although autosomal recessive and
mean follow-up of 33 months (SD 24), eight patients mitochondrial (maternal) inheritance also occur.10,14,30,79

6 www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4


Seminar

X-linked LVNC, which is usually associated with the identified DTNA causative mutations. Mutations in
multisystem disorder Barth syndrome (caused by a NKX2-5 were identified in children with LVNC and atrial
mutation in the TAZ gene), affects men almost septal defects and MYH7 mutations have been reported
exclusively, although a woman with cardiomyopathy in patients with LVNC and Ebstein’s anomaly.82,83 In
has been described.80 When LVNC is associated with LVNC without congenital heart disease, we identified
congenital heart disease, the congenital cardiac defect mutations in the Z-line protein-encoding LDB3, and
might be heterogeneous; this type of LVNC is others have shown that the sarcomere-encoding genes
transmitted as an autosomal dominant trait along with MYH7, ACTC1, TNNT2, MYBPC3, TPM1, and TNNI3
the congenital heart abnormality.10 In some families seem to account for 20% or more of LVNC.23,84–86
with autosomal dominant LVNC associated with Hoedemaekers and investigators84 showed that LVNC
congenital heart disease, some affected family members was associated with genetic variants in two calcium
have no identifiable congenital heart disease when they handling genes, TAZ and LMNA. Probst and colleagues85
are initially assessed because the cardiac defects include further showed the importance of sarcomere gene
minor forms of disease (eg, small ventricular and atrial mutations, reporting a prevalence of 29%—MYH7 and
septal defects, or patent ductus arteriosus) that have MYBPC3 were most frequently mutated (13% and 8%,
spontaneously closed; other family members have respectively). Dellefave and coworkers86 also identified
severe forms of congenital heart disease (eg, hypoplastic sarcomere mutations in LVNC, including those that
left heart syndrome, Ebstein’s anomaly).28,29,31,61 Penetrance present as heart failure in infancy.
might be reduced in some families. Ichida and co- In addition to sarcomere-encoding and cytoskeleton-
workers14 reported that, of the 44% of patients who encoding genes, we showed that mutations in the
inherited LVNC, 70% had autosomal dominant sodium channel gene SCN5A are associated with LVNC
inheritance and 30% X-linked inheritance. Additionally, and rhythm disturbance.87 Another cytoskeletal protein
patients with various chromosomal abnormalities have associated with LVNC is dystrophin; mutations in the
been diagnosed with LVNC. gene encoding this protein cause Duchenne and Becker
muscular dystrophy in boys.88 Additionally, a
Molecular genetics homozygous 2 bp deletion (5208_5209delAG) in an
The genetic causes of LVNC are heterogeneous, but alternatively spliced region of DSP, a desmosomal-
share a final common pathway, similar to other forms protein-encoding gene that has been reported to cause
of cardiomyopathy with heterogeneous causes. The arrhythmogenic cardiomyopathy and dilated
specific final common pathway, however, depends on cardiomyopathy when mutated, has been identified in
the clinical phenotype and resembles the genetic patients with LVNC.
causes of the clinical subtype in cardiomyopathies Mitochondrial genome mutations have been
devoid of LVNC. In LVNC, not only a final common associated with LVNC,89 and, as noted, chromosomal
pathway seems to be disturbed: in most cases, a abnormalities and syndromic patients with LVNC have
primary pathway (such as the sarcomere) and a also been identified.3,90 Mutations include 1p36 deletion,
developmental pathway (such as the NOTCH pathway) 7p14·3p14·1 deletion, 18p subtelomeric deletion,
are affected, often via a disturbance of protein–protein 22q11·2 deletion, distal 22q11·2,90 trisomies 18 and
binding caused by the primary genetic mutation. 13,91,92 8p23·1 deletion,93 tetrasomy 5q35·2–5q35,
The first genetic abnormality that causes LVNC without RPS6KA3 mutation (Coffin-Lowry syndrome), NSD1
evidence of congenital heart disease was initially described mutation (Sotos syndrome), and PMP22 duplication
by Bleyl and coworkers,81 who identified mutations in the (Charcot-Marie-Tooth disease type 1A).94–98 Diagnostic
X-linked TAZ in affected men and female carriers. TAZ testing in patients with LVNC seems to detect clinically
encodes tafazzin, a phospholipid transacylase that is significant variants in 35–40% of individuals—
important for membrane function. Mutation typically sarcomere-encoding genes are most often shown to
results in Barth syndrome, which is characterised by be mutated.95–99
cardiomyopathy (frequently LVNC), skeletal myopathy,
cyclic neutropenia, 3-methylglutaconic aciduria (a marker Murine models
of mitochondrial dysfunction), and deficiency of a Several murine models are being used to discern the
key membrane phospholipid of cardiomyocytes and underlying mechanisms of LVNC. The most common
mitochondria called cardiolipin. This defect disturbs conclusion reached so far is that hypertrabeculation results
mitochondrial function, leading to abnormalities of from altered regulation of cell proliferation, differentiation,
energy production and utilisation and, because the and maturation during the formation of the ventricular
sarcomere needs ATP, sarcomeric dysfunction. wall, particularly if the NOTCH signalling pathway is
Several mutations causing autosomal dominant LVNC affected, but other hypotheses have been shared.
have been identified, including mutations in genes FKBP1A is a member of the immunophilin protein
causing congenital heart disease with LVNC. In patients family that interacts with several intracellular protein
with hypoplastic left heart syndrome and LVNC, we complexes, such as calcium release channels (inositol

www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4 7


Seminar

triphosphate receptor and ryanodine receptor), BMP/ (haemopoietic stem cells, muscle satellite cells, cancer
activin/TGFβ type-1 receptors, voltage-gated sodium stem cells, and haemangioblasts). They maintain the fate
channels, FK506, and rapamycin, and inhibits calcineurin of neural stem cells and regulate their differentiation.
and mTOR activity.100–103 FKBP1A-deficient mice develop NUMB is a component of the adherens junction that
ventricular hypertrabeculation and non-compaction. regulates cell adhesion and migration, complexes with
Mutant mice deficient in FKBP1A develop multiple β-catenin to regulate WNT signalling, and interacts with
abnormalities in cardiac structure, including lack of integrin-β subunits to promote their endocytosis for
compaction, thin ventricular walls, deep intertrabecular directional cell migration. Deletion of NUMB and
recesses, increased trabeculae, and ventricular septal NUMBL from mouse hearts results in LVNC with
defects.104 Chen and coworkers105 reported that FKBP1A is congenital heart disease.22,108 The congenital heart defects
a novel negative modulator of activated NOTCH1: include atrioventricular septal defects, truncus arteriosus,
overexpression of FKBP1A significantly reduced and double outlet right ventricle. This model shows that
NOTCH1 stability, and direct inhibition of NOTCH NUMB family proteins regulate trabecular thickness
signalling significantly reduced hypertrabeculation in by inhibiting NOTCH1 signalling, control cardiac
FKBP1A-deficient mice. These findings suggested that morphogenesis in a NOTCH1-independent manner, and
FKBP1A-mediated regulation of NOTCH1 has an regulate cardiac progenitor cell differentiation in an
important role in intercellular communication between endocytosis-dependent manner.
endocardium and myocardium, which is crucial in Ventricular-muscle-cell-restricted knockout of NKX2-5
control of the formation of ventricular walls.101 in mice leads to progressive atrioventricular block with
Further support for the role of disturbances in the conduction system cell dropout and fibrosis. LVNC is a
NOTCH1 pathway in LVNC development was shown in prominent feature in neonatal mice, with progressive
families with autosomal dominant LVNC and germline biventricular dilation and heart failure developing
mutations (Val943Phe and Arg530X) in MIB1,106 which early.109 NKX2-5, a cardiac homeobox gene, is a
encodes for an E3 ubiquitin ligase implicated in regulation transcription factor that regulates heart development,
of endocytosis of NOTCH ligands DELTA and JAGGED. working along with MEF2, HAND1, and HAND2
Patients with MIB1 mutations had reduced NOTCH1 transcription factors to direct heart looping during early
activity, biventricular non-compaction including LVNC heart development. It directly activates MEF2 to control
with a dilated phenotype, and heart failure. MIB1 cardiomyocyte differentiation and operates in a positive
inactivation in mice resulted in LVNC associated with feedback loop with GATA transcription factors to
arrest of trabecular maturation and ventricular compaction. regulate cardiomyocyte formation.
BMP10 is a growth factor and a member of the TGF-β TGF-β superfamily members exert their biological
superfamily that is upregulated in FKBP1A-deficient functions by binding to serine/threonine kinase receptors
mice.107 BMP10 is expressed only briefly in the at the cell surface, followed by signal transduction by
ventricular myocardium during a crucial time when intracellular transducers called SMADs.111–113 SMAD
development is shifting from patterning to chamber proteins can be classified into three functional subclasses:
maturation (E9·0 to E13·5); its expression is restricted the receptor-regulated SMADs (R-SMAD), which include
to the trabecular myocardium. BMP10-deficient mice SMAD1, SMAD2, SMAD3, SMAD5, and SMAD8/9; the
typically die in utero at around E10·5 with evidence of common-mediator SMAD (co-SMAD), comprising only
very hypoplastic ventricular walls without trabeculae.18 SMAD 4, which interacts with R-SMADs to participate in
BMP10 overexpression leads to LVNC and ventricular signalling; and the antagonistic or inhibitory SMADs
septal defect. Upregulation of BMP10 also results in a (I-SMAD), comprising SMAD6 and SMAD7, which block
hypertrabeculation phenotype in NUMB/NUMBL- the activation of R-SMADs and co-SMADs. SMAD7 is an
deficient and NKX2-5-knockout mice.22,108,109 These inhibitory SMAD, deficiencies in which led to in-utero
findings suggest that BMP10 is important in cardiac murine death because of defects in cardiovascular
trabeculation and compaction. development, outflow tract malformations, and heart
TBX20 is a member of the TBX1 subfamily of the T-box failure.114 SMAD7 mutant mice that reach adulthood have
family transcription factors. In murine embryos it can notable LVNC with impaired cardiac function and severe
be detected in the cardiac precursor cells at E7·5 and the arrhythmias.
developing myocardium and endocardium at E8·0; later Expression of early response genes in lymphocytes is
its expression is more abundant in the atria than the regulated by NFAT transcription factors. NFATC1 mutant
ventricles. TBX20 is a key mediator of BMP10 signalling mouse embryos have cardiac abnormalities including
in ventricular wall development and maturation. myocardial developmental abnormalities, narrowing or
Overexpression of TBX20 led to a severe dilated cardio- occlusion of the ventricular outflow tract, defective
myopathy with hypertrabeculation consistent with the septum morphogenesis, and underdevelopment of the
dilated cardiomyopathy type of LVNC.110 semilunar valves; 50% die at day E14·5 from circulatory
NUMB family proteins, including NUMB and NUMBL, failure.115,116 40% have ventricular hypertrophy and small
are cell fate determinants for several progenitor cell types chamber size and LVNC with hypertrabeculation,

8 www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4


Seminar

suggesting that NFAT signalling pathways are important congestive medications that help to favourably remodel
for development of valves, the septum, hypertrabeculation the left ventricle, including angiotensin-converting
and non-compaction.116 enzyme inhibitors and β blockers, and an aldosterone
Jumonji family proteins are histone demethylases.117 antagonist. Loop diuretics should be considered for
JARID2 critically regulates developmental processes, patients with evidence of congestion or volume overload,
including cardiovascular development, and knockout and aspirin is used to reduce the risk of thrombotic
mice have left ventricular non-compaction associated complications. Inpatient treatments include intravenous
with a thin compact layer, ventricular septal defects, and diuretics or vasodilatory drugs, or both, in the setting of
double outlet right ventricles.118–120 Mysliwiec and acute decompensated heart failure. Inotropes can be used
coworkers120 showed that the NOTCH1 pathway is in patients with evidence of low cardiac output and poor
directly controlled by JARID2, and failure to regulate end-organ perfusion.
NOTCH1 expression leads to proliferation and Implantable-cardioverter defibrillators are an option if
differentiation defects in the developing heart, which patients meet criteria for implantation as recommended
manifest as LVNC associated with a thin ventricular in published guidelines. Advanced pacing strategies,
wall. Deletion of NOTCH1 results in impaired such as cardiac resynchronisation, are also used, and
trabeculation and myocardial proliferation associated result in improvement in some patients.78 Ventricular
with embryonic death due to cardiac insufficiency.121 assist devices and cardiac transplantation are possibilities
Barth syndrome is caused by mutations in the X-linked for patients with end-stage disease. Patients with
TAZ,10,23,122 knockout of which leads to LVNC associated associated hypertrophic cardiomyopathy might benefit
with abnormal cardiolipin profiles and mitochondrial from symptomatic treatment with β blockers or calcium
structural abnormalities, suggesting that mitochondrial channel blockers when left ventricular outflow tract
function is important for proper myocardial development.123 obstruction is present. Internal cardioverter-defibrillator
placement should be considered for patients with
Treatment and outcome increased risk of sudden cardiac death.125
Treatment is predicated on making the correct phenotypic The thromboembolic risks associated with LVNC are
diagnosis because different phenotypes necessitate well known.14,126 They are mainly reported in adults,126 and
different surveillance and are associated with variable thus antiplatelets or systemic anticoagulation should be
outcomes.51 Because LVNC is heritable, at-risk first- considered in adults, especially when the left ventricle or
degree relatives are recommended to undergo screening, atria are dilated. The incidence of stroke or other embolic
resulting in diagnosis of people who otherwise would events in children remains poorly characterised, and
have never undergone non-invasive imaging. Clinically antiplatelet drugs might be an option in those with
available genetic testing has also affected management: depressed left ventricular systolic dysfunction, evidence
panels that assess for known sarcomeric gene mutations of spontaneous echocardiographic contrast, severe left
implicated in LVNC are used more often and are thought ventricular dilation, or dilated atria. The presence of
to be the standard of care in many institutions. In atrial fibrillation might also prompt use of systemic
patients with identified pathological mutations, targeted anticoagulation.
sequencing can then be done in first-degree relatives, In patients with primary diastolic dysfunction, drug
which has important implications. These sarcomeric treatment might be instituted but none has proven
genes present as varying cardiac phenotypes within benefit. In many cases, a combination of systolic and
families, meaning that a first-degree relative of a known diastolic dysfunction occurs, causing decompensated
patient with LVNC who harbours the identical mutation heart failure necessitating the therapeutic approaches
might have an LVNC phenotype or instead could have described. Some patients develop restrictive physiology,
isolated dilated, hypertrophic, or restrictive cardio- and they generally need a heart transplant. Patients with
myopathy without LVNC. These family members could mitochondrial disease or metabolic derangements can be
also have the same mutation as the affected individual managed with additional treatments, such as coenzyme
and have no phenotype at all. The presence of a Q10, L-carnitine, riboflavin, and thiamine. However, the
pathological mutation changes screening recommen- benefit is unclear.
dations and genetic counselling should be provided LVNC can affect treatment and confound outcomes in
about risk to future offspring. patients with associated congenital heart disease. Treatment
Therapy for LVNC is largely dictated by concomitant of congenital heart disease will be dictated by the severity of
clinical findings associated with myocardial dysfunction the lesion and might necessitate percutaneous catheter or
or significant arrhythmias, or both, or congenital heart surgical intervention. Overarching genetic causes should
disease. Patients with evidence of systolic or diastolic be considered, which might affect management, as should
dysfunction should be managed on the basis of existing screening of at-risk family members. LVNC can increase
recommendations.124 For patients with LVNC and the risk of myocardial dysfunction, arrhythmias, and
associated systolic dysfunction or dilated cardiomyopathy thromboembolic events, especially in the peri-operative
phenotypes, oral treatments typically include anti- period. The possibility of syndromic or metabolic diseases

www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4 9


Seminar

should be remembered, and might affect management of 14 Ichida F, Hamamichi Y, Miyawaki T, et al. Clinical features of
patients undergoing catheter-based interventions or isolated noncompaction of the ventricular myocardium: long-term
clinical course, hemodynamic properties, and genetic background.
surgical palliations or corrective surgery. Management J Am Coll Cardiol 1999; 34: 233–40.
will be directed at associated myocardial dysfunction 15 Cartoni D, Salvini P, De Rosa R, Cortese A, Nazzaro MS, Tanzi P.
with or without evidence of heart failure or clinically Images in cardiovascular medicine. Multiple coronary artery-left
ventricle microfistulae and spongy myocardium: the eagerly awaited
significant dysrhythmias. link? Circulation 2007; 116: e81–84.
Outcomes of patients with LVNC are largely associated 16 Reynen K, Bachmann K, Singer H. Spongy myocardium. Cardiology
with the presence of myocardial dysfunction or clinically 1997; 88: 601–02.
17 Icardo JM, Fernandez-Terán A. Morphologic study of ventricular
significant arrhythmias, or both. In 2013, Brescia and trabeculation in the embryonic chick heart. Acta Anat (Basel) 1987;
colleagues51 reported that of 242 children with LVNC at a 130: 264–74.
single centre, 150 (62%) had myocardial dysfunction 18 Chen H, Zhang W, Li D, Cordes TM, Mark Payne R, Shou W. Analysis
of ventricular hypertrabeculation and noncompaction using genetically
and 80 (33%) had a clinically significant arrhythmia, engineered mouse models. Pediatr Cardiol 2009; 30: 626–34.
both of which were strongly associated with mortality 19 Harvey RP. Patterning the vertebrate heart. Nat Rev Genet 2002;
(p<0·001 and p=0·002, respectively). Similar reports 3: 544–56.
cite myocardial dysfunction or ventricular arrhythmias 20 Risebro CA, Riley PR. Formation of the ventricles.
ScientificWorldJournal 2006; 6: 1862–80.
as predictors of mortality in adults.
21 Sedmera D, McQuinn T. Embryogenesis of the heart muscle.
Contributors Heart Fail Clin 2008; 4: 235–45.
All authors contributed equally to the search of published works and 22 Yang J, Bücker S, Jungblut B, et al. Inhibition of Notch2 by Numb/
writing of this Seminar. Numblike controls myocardial compaction in the heart.
Cardiovasc Res 2012; 96: 276–85.
Declaration of interests
23 Ichida F, Tsubata S, Bowles KR, et al. Novel gene mutations in
We declare that we have no competing interests. patients with left ventricular noncompaction or Barth syndrome.
References Circulation 2001; 103: 1256–63.
1 Grant RT. An unusual anomaly of the coronary vessels in the 24 Caliskan K, Ujvari B, Bauernfeind T, et al. The prevalence of early
malformed heart of a child. Heart 1926; 13: 273–83. repolarization in patients with noncompaction cardiomyopathy
2 Chin TK, Perloff JK, Williams RG, Jue K, Mohrmann R. Isolated presenting with malignant ventricular arrhythmias.
noncompaction of left ventricular myocardium. A study of J Cardiovasc Electrophysiol 2012; 23: 938–44.
eight cases. Circulation 1990; 82: 507–13. 25 Steffel J, Duru F. Rhythm disorders in isolated left ventricular
3 Pignatelli RH, McMahon CJ, Dreyer WJ, et al. Clinical characterization noncompaction. Ann Med 2012; 44: 101–08.
of left ventricular noncompaction in children: a relatively common 26 Attenhofer Jost CH, Connolly HM, Warnes CA, et al. Noncompacted
form of cardiomyopathy. Circulation 2003; 108: 2672–78. myocardium in Ebstein’s anomaly: initial description in
4 Engberding R, Yelbuz TM, Breithardt G. Isolated noncompaction of three patients. J Am Soc Echocardiogr 2004; 17: 677–80.
the left ventricular myocardium—a review of the literature two 27 Zuckerman WA, Richmond ME, Singh RK, Carroll SJ, Starc TJ,
decades after the initial case description. Clin Res Cardiol 2007; Addonizio LJ. Left-ventricular noncompaction in a pediatric
96: 481–88. population: predictors of survival. Pediatr Cardiol 2011; 32: 406–12.
5 Sedmera D, Pexieder T, Vuillemin M, Thompson RP, Anderson RH. 28 Stähli BE, Gebhard C, Biaggi P, et al. Left ventricular
Developmental patterning of the myocardium. Anat Rec 2000; non-compaction: prevalence in congenital heart disease.
258: 319–37. Int J Cardiol 2013; 167: 2477–81.
6 Dusek J, Ostádal B, Duskova M. Postnatal persistence of spongy 29 Madan S, Mandal S, Bost JE, et al. Noncompaction cardiomyopathy
myocardium with embryonic blood supply. Arch Pathol 1975; in children with congenital heart disease: evaluation using
99: 312–17. cardiovascular magnetic resonance imaging. Pediatr Cardiol 2012;
7 Fazio G, Lunetta M, Grassedonio E, et al. Noncompaction of the 33: 215–21.
right ventricle. Pediatr Cardiol 2010; 31: 576–78. 30 Scaglia F, Towbin JA, Craigen WJ, et al. Clinical spectrum,
8 Ranganathan A, Ganesan G, Sangareddi V, Pillai AP, Ramasamy A. morbidity, and mortality in 113 pediatric patients with
Isolated noncompaction of right ventricle—a case report. mitochondrial disease. Pediatrics 2004; 114: 925–31.
Echocardiography 2012; 29: E169–72. 31 Yaplito-Lee J, Weintraub R, Jamsen K, Chow CW, Thorburn DR,
9 Tigen K, Karaahmet T, Gurel E, Cevik C, Basaran Y. Biventricular Boneh A. Cardiac manifestations in oxidative phosphorylation
noncompaction: a case report. Echocardiography 2008; 25: 993–96. disorders of childhood. J Pediatr 2007; 150: 407–11.
10 Towbin JA. Left ventricular noncompaction: a new form of heart 32 Pepper MS. Transforming growth factor-beta: vasculogenesis,
failure. Heart Fail Clin 2010; 6: 453–69, viii. angiogenesis, and vessel wall integrity. Cytokine Growth Factor Rev
11 Maron BJ, Towbin JA, Thiene G, et al, and the American Heart 1997; 8: 21–43.
Association, and the Council on Clinical Cardiology, Heart Failure 33 Suri C, Jones PF, Patan S, et al. Requisite role of angiopoietin-1, a
and Transplantation Committee, and the Quality of Care and ligand for the TIE2 receptor, during embryonic angiogenesis.
Outcomes Research and Functional Genomics and Translational Cell 1996; 87: 1171–80.
Biology Interdisciplinary Working Groups, and the Council on 34 Ferrara N, Carver-Moore K, Chen H, et al. Heterozygous embryonic
Epidemiology and Prevention. Contemporary definitions and lethality induced by targeted inactivation of the VEGF gene.
classification of the cardiomyopathies: an American Heart Nature 1996; 380: 439–42.
Association Scientific Statement from the Council on Clinical 35 Zhang W, Chen H, Qu X, Chang CP, Shou W. Molecular
Cardiology, Heart Failure and Transplantation Committee; Quality mechanism of ventricular trabeculation/compaction and the
of Care and Outcomes Research and Functional Genomics and pathogenesis of the left ventricular noncompaction
Translational Biology Interdisciplinary Working Groups; and cardiomyopathy (LVNC). Am J Med Genet C Semin Med Genet 2013;
Council on Epidemiology and Prevention. Circulation 2006; 163C: 144–56.
113: 1807–16. 36 Stöllberger C, Blazek G, Dobias C, Hanafin A, Wegner C,
12 Stöllberger C, Finsterer J, Blazek G. Left ventricular Finsterer J. Frequency of stroke and embolism in left ventricular
hypertrabeculation/noncompaction and association with additional hypertrabeculation/noncompaction. Am J Cardiol 2011; 108: 1021–23.
cardiac abnormalities and neuromuscular disorders. Am J Cardiol 37 Greutmann M, Mah ML, Silversides CK, et al. Predictors of adverse
2002; 90: 899–902. outcome in adolescents and adults with isolated left ventricular
13 Stöllberger C, Finsterer J. Left ventricular hypertrabeculation/ noncompaction. Am J Cardiol 2012; 109: 276–81.
noncompaction. J Am Soc Echocardiogr 2004; 17: 91–100.

10 www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4


Seminar

38 Steffel J, Kobza R, Oechslin E, Jenni R, Duru F. Electrocardiographic 60 Wlodarska EK, Wozniak O, Konka M, Piotrowska-Kownacka D,
characteristics at initial diagnosis in patients with isolated left Walczak E, Hoffman P. Isolated ventricular noncompaction
ventricular noncompaction. Am J Cardiol 2009; 104: 984–89. mimicking arrhythmogenic right ventricular cardiomyopath—a
39 Celiker A, Ozkutlu S, Dilber E, Karagöz T. Rhythm abnormalities in study of nine patients. Int J Cardiol 2010; 145: 107–11.
children with isolated ventricular noncompaction. 61 Hughes ML, Carstensen B, Wilkinson JL, Weintraub RG.
Pacing Clin Electrophysiol 2005; 28: 1198–202. Angiographic diagnosis, prevalence and outcomes for left
40 Ritter M, Oechslin E, Sütsch G, Attenhofer C, Schneider J, Jenni R. ventricular noncompaction in children with congenital cardiac
Isolated noncompaction of the myocardium in adults. Mayo Clin Proc disease. Cardiol Young 2007; 17: 56–63.
1997; 72: 26–31. 62 Jenni R, Oechslin E, Schneider J, Attenhofer Jost C, Kaufmann PA.
41 Aras D, Tufekcioglu O, Ergun K, et al. Clinical features of isolated Echocardiographic and pathoanatomical characteristics of isolated
ventricular noncompaction in adults long-term clinical course, left ventricular non-compaction: a step towards classification as a
echocardiographic properties, and predictors of left ventricular distinct cardiomyopathy. Heart 2001; 86: 666–71.
failure. J Card Fail 2006; 12: 726–33. 63 Oechslin E, Jenni R. Left ventricular non-compaction revisited: a
42 Sandhu R, Finkelhor RS, Gunawardena DR, Bahler RC. Prevalence distinct phenotype with genetic heterogeneity? Eur Heart J 2011;
and characteristics of left ventricular noncompaction in a 32: 1446–56.
community hospital cohort of patients with systolic dysfunction. 64 Punn R, Silverman NH. Cardiac segmental analysis in left
Echocardiography 2008; 25: 8–12. ventricular noncompaction: experience in a pediatric population.
43 Kovacevic-Preradovic T, Jenni R, Oechslin EN, Noll G, Seifert B, J Am Soc Echocardiogr 2010; 23: 46–53.
Attenhofer Jost CH. Isolated left ventricular noncompaction as a 65 McMahon CJ, Pignatelli RH, Nagueh SF, et al. Left ventricular
cause for heart failure and heart transplantation: a single center non-compaction cardiomyopathy in children: characterisation of
experience. Cardiology 2009; 112: 158–64. clinical status using tissue Doppler-derived indices of left
44 Patrianakos AP, Parthenakis FI, Nyktari EG, Vardas PE. ventricular diastolic relaxation. Heart 2007; 93: 676–81.
Noncompaction myocardium imaging with multiple 66 Eidem BW. Noninvasive evaluation of left ventricular
echocardiographic modalities. Echocardiography 2008; 25: 898–900. noncompaction: what’s new in 2009? Pediatr Cardiol 2009;
45 Sarma RJ, Chana A, Elkayam U. Left ventricular noncompaction. 30: 682–89.
Prog Cardiovasc Dis 2010; 52: 264–73. 67 van Dalen BM, Caliskan K, Soliman OI, et al. Left ventricular solid
46 Niemann M, Störk S, Weidemann F. Left ventricular noncompaction body rotation in non-compaction cardiomyopathy: a potential new
cardiomyopathy: an overdiagnosed disease. Circulation 2012; objective and quantitative functional diagnostic criterion?
126: e240–43. Eur J Heart Fail 2008; 10: 1088–93.
47 Murphy RT, Thaman R, Blanes JG, et al. Natural history and 68 Petersen SE, Selvanayagam JB, Wiesmann F, et al. Left ventricular
familial characteristics of isolated left ventricular non-compaction. non-compaction: insights from cardiovascular magnetic resonance
Eur Heart J 2005; 26: 187–92. imaging. J Am Coll Cardiol 2005; 46: 101–05.
48 Bhatia NL, Tajik AJ, Wilansky S, Steidley DE, Mookadam F. Isolated 69 Thuny F, Jacquier A, Jop B, et al. Assessment of left ventricular
noncompaction of the left ventricular myocardium in adults: a non-compaction in adults: side-by-side comparison of cardiac
systematic overview. J Card Fail 2011; 17: 771–78. magnetic resonance imaging with echocardiography.
49 Caliskan K, Kardos A, Szili-Torok T. Empty handed: a call for an Arch Cardiovasc Dis 2010; 103: 150–59.
international registry of risk stratification to reduce the ‘sudden-ness’ 70 Jacquier A, Thuny F, Jop B, et al. Measurement of trabeculated left
of death in patients with non-compaction cardiomyopathy. Europace ventricular mass using cardiac magnetic resonance imaging in the
2009; 11: 1138–39. diagnosis of left ventricular non-compaction. Eur Heart J 2010;
50 Biagini E, Ragni L, Ferlito M, et al. Different types of cardiomyopathy 31: 1098–104.
associated with isolated ventricular noncompaction. Am J Cardiol 71 Uribe S, Cadavid L, Hussain T, et al. Cardiovascular magnetic
2006; 98: 821–24. resonance findings in a pediatric population with isolated left
51 Brescia ST, Rossano JW, Pignatelli R, et al. Mortality and sudden ventricular non-compaction. J Cardiovasc Magn Reson 2012; 14: 9.
death in pediatric left ventricular noncompaction in a tertiary 72 Paterick TE, Tajik AJ. Left ventricular noncompaction: a diagnostically
referral center. Circulation 2013; 127: 2202–08. challenging cardiomyopathy. Circ J 2012; 76: 1556–62.
52 Sen Chowdhry S, McKenna WJ. Left ventricular noncompaction 73 Melendez-Ramirez G, Castillo-Castellon F, Espinola-Zavaleta N,
and cardiomyopathy: cause, contributor, or epiphenomenon? Meave A, Kimura-Hayama ET. Left ventricular noncompaction:
Curr Opin Cardiol 2008; 23: 171–75. a proposal of new diagnostic criteria by multidetector computed
53 Ronvelia D, Greenwood J, Platt J, Hakim S, Zaragoza MV. tomography. J Cardiovasc Comput Tomogr 2012; 6: 346–54.
Intrafamilial variability for novel TAZ gene mutation: Barth 74 Hollingsworth CL, Yoshizumi TT, Frush DP, et al. Pediatric cardiac-
syndrome with dilated cardiomyopathy and heart failure in an gated CT angiography: assessment of radiation dose.
infant and left ventricular noncompaction in his great-uncle. AJR Am J Roentgenol 2007; 189: 12–18.
Mol Genet Metab 2012; 107: 428–32. 75 Nihei K, Shinomiya N, Kabayama H, et al. Wolff-Parkinson-White
54 Ergul Y, Nisli K, Varkal MA, et al. Electrocardiographic findings at (WPW) syndrome in isolated noncompaction of the ventricular
initial diagnosis in children with isolated left ventricular myocardium (INVM). Circ J 2004; 68: 82–84.
noncompaction. Ann Noninvasive Electrocardiol 2011; 16: 184–91. 76 Caliskan K, Szili-Torok T, Theuns DA, et al. Indications and
55 Onay OS, Yildirim I, Beken B, et al. Successful implantation of an outcome of implantable cardioverter-defibrillators for primary and
intracardiac defibrillator in an infant with long QT syndrome and secondary prophylaxis in patients with noncompaction
isolated noncompaction of the ventricular myocardium. cardiomyopathy. J Cardiovasc Electrophysiol 2011; 22: 898–904.
Pediatr Cardiol 2013; 34: 189–93. 77 Zipes DP, Camm AJ, Borggrefe M, et al, and the American College
56 Nakashima K, Kusakawa I, Yamamoto T, et al. A left ventricular of Cardiology/American Heart Association Task Force, and the
noncompaction in a patient with long QT syndrome caused by a European Society of Cardiology Committee for Practice Guidelines,
KCNQ1 mutation: a case report. Heart Vessels 2013; 28: 126–29. and the European Heart Rhythm Association, and the Heart
57 Nascimento BR, Vidigal DF, De Carvalho Bicalho Carneiro R, et al. Rhythm Society. ACC/AHA/ESC 2006 Guidelines for Management
Complete atrioventricular block as the first manifestation of of Patients With Ventricular Arrhythmias and the Prevention of
noncompaction of the ventricular myocardium. Sudden Cardiac Death: a report of the American College of
Pacing Clin Electrophysiol 2013; 36: e107–10. Cardiology/American Heart Association Task Force and the
European Society of Cardiology Committee for Practice Guidelines
58 Ulusoy RE, Kucukarslan N, Kirilmaz A, Demiralp E. Noncompaction
(writing committee to develop Guidelines for Management of
of ventricular myocardium involving both ventricles.
Patients With Ventricular Arrhythmias and the Prevention of
Eur J Echocardiogr 2006; 7: 457–60.
Sudden Cardiac Death): developed in collaboration with the
59 Williams T, Machann W, Kühler L, et al. Novel desmoplakin European Heart Rhythm Association and the Heart Rhythm
mutation: juvenile biventricular cardiomyopathy with left Society. Circulation 2006; 114: e385–484.
ventricular non-compaction and acantholytic palmoplantar
keratoderma. Clin Res Cardiol 2011; 100: 1087–93.

www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4 11


Seminar

78 Kobza R, Steffel J, Erne P, et al. Implantable cardioverter- 100 Schreiber SL, Crabtree GR. Immunophilins, ligands, and the
defibrillator and cardiac resynchronization therapy in patients with control of signal transduction. Harvey Lect 1995-1996; 91: 99–114.
left ventricular noncompaction. Heart Rhythm 2010; 7: 1545–49. 101 Wang T, Donahoe PK. The immunophilin FKBP12: a molecular
79 Sasse-Klaassen S, Gerull B, Oechslin E, Jenni R, Thierfelder L. guardian of the TGF-beta family type I receptors. Front Biosci
Isolated noncompaction of the left ventricular myocardium in the 2004; 9: 619–31.
adult is an autosomal dominant disorder in the majority of patients. 102 Cameron AM, Steiner JP, Roskams AJ, Ali SM, Ronnett GV,
Am J Med Genet A 2003; 119A: 162–67. Snyder SH. Calcineurin associated with the inositol
80 Cosson L, Toutain A, Simard G, et al. Barth syndrome in a female 1,4,5-trisphosphate receptor-FKBP12 complex modulates Ca2+ flux.
patient. Mol Genet Metab 2012; 106: 115–20. Cell 1995; 83: 463–72.
81 Bleyl SB, Mumford BR, Brown-Harrison MC, et al. Xq28-linked 103 Maruyama M, Li BY, Chen H, et al. FKBP12 is a critical regulator
noncompaction of the left ventricular myocardium: prenatal of the heart rhythm and the cardiac voltage-gated sodium current
diagnosis and pathologic analysis of affected individuals. in mice. Circ Res 2011; 108: 1042–52.
Am J Med Genet 1997; 72: 257–65. 104 Shou W, Aghdasi B, Armstrong DL, et al. Cardiac defects and
82 Ouyang P, Saarel E, Bai Y, et al. A de novo mutation in NKX2.5 altered ryanodine receptor function in mice lacking FKBP12.
associated with atrial septal defects, ventricular noncompaction, Nature 1998; 391: 489–92.
syncope and sudden death. Clin Chim Acta 2011; 412: 170–75. 105 Chen H, Zhang W, Sun X, et al. Fkbp1a controls ventricular
83 Postma AV, van Engelen K, van de Meerakker J, et al. Mutations in myocardium trabeculation and compaction by regulating
the sarcomere gene MYH7 in Ebstein anomaly. Circ Cardiovasc Genet endocardial Notch1 activity. Development 2013; 140: 1946–57.
2011; 4: 43–50. 106 Luxán G, Casanova JC, Martínez-Poveda B, et al. Mutations in the
84 Hoedemaekers YM, Caliskan K, Michels M, et al. The importance NOTCH pathway regulator MIB1 cause left ventricular
of genetic counseling, DNA diagnostics, and cardiologic family noncompaction cardiomyopathy. Nat Med 2013; 19: 193–201.
screening in left ventricular noncompaction cardiomyopathy. 107 Chen H, Shi S, Acosta L, et al. BMP10 is essential for maintaining
Circ Cardiovasc Genet 2010; 3: 232–39. cardiac growth during murine cardiogenesis. Development 2004;
85 Probst S, Oechslin E, Schuler P, et al. Sarcomere gene mutations in 131: 2219–31.
isolated left ventricular noncompaction cardiomyopathy do not 108 Zhao C, Guo H, Li J, et al. Numb family proteins are essential for
predict clinical phenotype. Circ Cardiovasc Genet 2011; 4: 367–74. cardiac morphogenesis and progenitor differentiation.
86 Dellefave LM, Pytel P, Mewborn S, et al. Sarcomere mutations in Development 2014; 141: 281–95.
cardiomyopathy with left ventricular hypertrabeculation. 109 Pashmforoush M, Lu JT, Chen H, et al. Nkx2-5 pathways and
Circ Cardiovasc Genet 2009; 2: 442–49. congenital heart disease; loss of ventricular myocyte lineage
87 Shan L, Makita N, Xing Y, et al. SCN5A variants in Japanese patients specification leads to progressive cardiomyopathy and complete
with left ventricular noncompaction and arrhythmia. Mol Genet Metab heart block. Cell 2004; 117: 373–86.
2008; 93: 468–74. 110 Zhang W, Chen H, Wang Y, et al. Tbx20 transcription factor is a
88 Finsterer J, Stöllberger C. Primary myopathies and the heart. downstream mediator for bone morphogenetic protein-10 in
Scand Cardiovasc J 2008; 42: 9–24. regulating cardiac ventricular wall development and function.
89 Tang S, Batra A, Zhang Y, Ebenroth ES, Huang T. Left ventricular J Biol Chem 2011; 286: 36820–29.
noncompaction is associated with mutations in the mitochondrial 111 Massagué J. TGF-beta signal transduction. Annu Rev Biochem
genome. Mitochondrion 2010; 10: 350–57. 1998; 67: 753–91.
90 Digilio MC, Bernardini L, Gagliardi MG, et al. Syndromic 112 Heldin CH, Miyazono K, ten Dijke P. TGF-beta signalling from
non-compaction of the left ventricle: associated chromosomal cell membrane to nucleus through SMAD proteins. Nature 1997;
anomalies. Clin Genet 2013; 84: 362–67. 390: 465–71.
91 Beken S, Cevik A, Turan O, et al. A neonatal case of left ventricular 113 Shi Y, Massagué J. Mechanisms of TGF-beta signaling from cell
noncompaction associated with trisomy 18. Genet Couns 2011; membrane to the nucleus. Cell 2003; 113: 685–700.
22: 161–64. 114 Chen Q, Chen H, Zheng D, et al. Smad7 is required for the
92 Yukifumi M, Hirohiko S, Fukiko I, Mariko M. Trisomy 13 in a development and function of the heart. J Biol Chem 2009;
9-year-old girl with left ventricular noncompaction. Pediatr Cardiol 284: 292–300.
2011; 32: 206–07. 115 Liu J, Farmer JD Jr, Lane WS, Friedman J, Weissman I,
93 Blinder JJ, Martinez HR, Craigen WJ, Belmont J, Pignatelli RH, Schreiber SL. Calcineurin is a common target of cyclophilin-
Jefferies JL. Noncompaction of the left ventricular myocardium in a cyclosporin A and FKBP-FK506 complexes. Cell 1991; 66: 807–15.
boy with a novel chromosome 8p23.1 deletion. Am J Med Genet A 116 de la Pompa JL, Timmerman LA, Takimoto H, et al. Role of the
2011; 155A: 2215–20. NF-ATc transcription factor in morphogenesis of cardiac valves
94 Martinez HR, Belmont JW, Craigen WJ, Taylor MD, Jefferies JL. and septum. Nature 1998; 392: 182–86.
Left ventricular noncompaction in Sotos syndrome. 117 Whetstine JR, Nottke A, Lan F, et al. Reversal of histone lysine
Am J Med Genet A 2011; 155A: 1115–18. trimethylation by the JMJD2 family of histone demethylases.
95 Martinez HR, Niu MC, Sutton VR, Pignatelli R, Vatta M, Jefferies JL. Cell 2006; 125: 467–81.
Coffin-Lowry syndrome and left ventricular noncompaction 118 Shen X, Kim W, Fujiwara Y, et al. Jumonji modulates polycomb
cardiomyopathy with a restrictive pattern. Am J Med Genet A 2011; activity and self-renewal versus differentiation of stem cells.
155A: 3030–34. Cell 2009; 139: 1303–14.
96 Zechner U, Kohlschmidt N, Kempf O, et al. Familial Sotos 119 Landeira D, Sauer S, Poot R, et al. Jarid2 is a PRC2 component in
syndrome caused by a novel missense mutation, C2175S, in NSD1 embryonic stem cells required for multi-lineage differentiation
and associated with normal intelligence, insulin dependent and recruitment of PRC1 and RNA Polymerase II to
diabetes, bronchial asthma, and lipedema. Eur J Med Genet 2009; developmental regulators. Nat Cell Biol 2010; 12: 618–24.
52: 306–10. 120 Mysliwiec MR, Bresnick EH, Lee Y. Endothelial Jarid2/Jumonji is
97 Sellars EA, Zimmerman SL, Smolarek T, Hopkin RJ. Ventricular required for normal cardiac development and proper Notch1
noncompaction and absent thumbs in a newborn with tetrasomy expression. J Biol Chem 2011; 286: 17193–204.
5q35.2-5q35.3: an association with Hunter-McAlpine syndrome? 121 Grego-Bessa J, Luna-Zurita L, del Monte G, et al. Notch signaling
Am J Med Genet A 2011; 155A: 1409–13. is essential for ventricular chamber development. Dev Cell 2007;
98 Corrado G, Checcarelli N, Santarone M, Stollberger C, Finsterer J. 12: 415–29.
Left ventricular hypertrabeculation/noncompaction with PMP22 122 Jefferies JL. Barth syndrome. Am J Med Genet C Semin Med Genet
duplication-based Charcot-Marie-Tooth disease type 1A. Cardiology 2013; 163C: 198–205.
2006; 105: 142–45.
123 Phoon CK, Acehan D, Schlame M, et al. Tafazzin knockdown in
99 Teekakirikul P, Kelly MA, Rehm HL, Lakdawala NK, Funke BH. mice leads to a developmental cardiomyopathy with early diastolic
Inherited cardiomyopathies: molecular genetics and clinical dysfunction preceding myocardial noncompaction.
genetic testing in the postgenomic era. J Mol Diagn 2013; J Am Heart Assoc 2012; 1: jah3-e000455.
15: 158–70.

12 www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4


Seminar

124 Yancy CW, Jessup M, Bozkurt B, et al, and the American College 126 Pitta S, Thatai D, Afonso L. Thromboembolic complications of left
of Cardiology Foundation, and the American Heart Association ventricular noncompaction: case report and brief review of the
Task Force on Practice Guidelines. 2013 ACCF/AHA guideline for literature. J Clin Ultrasound 2007; 35: 465–68.
the management of heart failure: a report of the American College
of Cardiology Foundation/American Heart Association Task Force
on Practice Guidelines. J Am Coll Cardiol 2013; 62: e147–239.
125 Gersh BJ, Maron BJ, Bonow RO, et al, and the American College of
Cardiology Foundation/American Heart Association Task Force on
Practice Guidelines. 2011 ACCF/AHA Guideline for the Diagnosis
and Treatment of Hypertrophic Cardiomyopathy: a report of the
American College of Cardiology Foundation/American Heart
Association Task Force on Practice Guidelines. Developed in
collaboration with the American Association for Thoracic Surgery,
American Society of Echocardiography, American Society of
Nuclear Cardiology, Heart Failure Society of America, Heart
Rhythm Society, Society for Cardiovascular Angiography and
Interventions, and Society of Thoracic Surgeons. J Am Coll Cardiol
2011; 58: e212–60.

www.thelancet.com Published online April 10, 2015 http://dx.doi.org/10.1016/S0140-6736(14)61282-4 13

You might also like