Inherited cardiomyopathies – novel therapies Dror B. Leviner, Edith Hochhauser, Michael Arad PII: DOI: Reference:
S0163-7258(15)00161-8 doi: 10.1016/j.pharmthera.2015.08.003 JPT 6807
To appear in:
Pharmacology and Therapeutics
Please cite this article as: Leviner, D.B., Hochhauser, E. & Arad, M., Inherited cardiomyopathies – novel therapies, Pharmacology and Therapeutics (2015), doi: 10.1016/j.pharmthera.2015.08.003
This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
ACCEPTED MANUSCRIPT 0
P&T 22456
T
Inherited cardiomyopathies – novel therapies
SC R
IP
Dror B. Leviner1,2 MD, Edith Hochhauser PhD2, Michael Arad3 MD
Department of Cardiothoracic Surgery, Rabin Medical Center, Petah Tikva, Israel
2
Cardiac Research Laboratory, Felsenstein Medical Research Center, Petah Tikva and
NU
1
Sackler School of Medicine, Tel Aviv University
Leviev Heart Center, Sheba Medical Center, Tel Hashomer and Sackler School of
D
Medicine, Tel Aviv University
MA
3
CE P
Michael Arad MD,
TE
Address and details of the corresponding author:
Leviev Heart Center, Sheba Medical Center, Tel Hashomer, Ramat-Gan 52621, Israel
AC
+972-3-5304560, fax +972-3-5304540 E-mail:
[email protected]
ACCEPTED MANUSCRIPT 1
Abstract Cardiomyopathies arising due to a single gene defect represent various pathways that
T
evoke adverse remodeling and cardiac dysfunction. While the gene therapy approach
IP
is slowly evolving and has not yet reached clinical "prime time" and gene correction
SC R
approaches are applicable at the bench but not at the bedside, major advances are being made with molecular and drug therapies. This review summarizes the contemporary drugs introduced or being tested to help manage these unique disorders
MA
NU
bearing a major impact on the quality of life and survival of the affected individuals.
The restoration of the RNA reading frame facilitates the expression of partly
D
functional protein to salvage or alleviate the disease phenotype. Chaperones are used
TE
to prevent the degradation of abnormal but still functional proteins, while other molecules are given for pathogen silencing, to prevent aggregation or to enhance
CE P
clearance of protein deposits. Absence of protein may be managed by viral gene delivery or protein therapy. Enzyme replacement therapy is already a clinical reality
AC
for a series of metabolic diseases. The progress in molecular biology, based on the knowledge of the gene defect, helps generate small molecules and pharmaceuticals targeting the key events occurring in the malfunctioning element of the sick organ. Cumulatively, these tools augment the existing armamentarium of phenotype oriented symptomatic and evidence-based therapies for patients with inherited cardiomyopathies.
Key words: 6 Hypertrophic cardiomyopathy, Dilated cardiomyopathy, Arrhythmogenic cardiomyopathy, Amyloidosis, Duchenne Muscular Dystrophy, Gene therapy
ACCEPTED MANUSCRIPT 2
Introduction
4
2.
Hypertrophic cardiomyopathy (HCM)
4
3.
Novel therapeutic strategies in HCM
4.
Dilated cardiomyopathy (DCM)
5.
Novel therapeutic strategies in familial DCM
14
6.
Dystrophinopathy
18
7.
Arrhythmogenic cardiomyopathy (ACM)
21
8.
Novel therapeutic strategies in familial ACM
23
9.
Restrictive cardiomyopathy (RCM)
24
10.
Novel therapeutic strategies in familial RCM
25
11.
Transthyretin amyloidosis
26
12.
Metabolic and related cardiomyopathies
28
Pompe disease
28
7 10
Anderson Fabry disease
30
Danon disease and disorders of autophagy
32
Summary
35
14.
AC
CE P
D
MA
NU
SC R
IP
T
1.
TE
Table of Contents
Conflict of Interest Statement
37
15.
References
37
Figure legend
45
13.
ACCEPTED MANUSCRIPT 3
Abbreviations ACM - arrhythmogenic cardiomyopathy
T
AON - antisense oligo-nucleotide
IP
α-Gal A - α-galactosidase-A
SC R
BMD - Becker Muscular Dystrophy cMyBPC - myosin- binding protein C
DMD - Duchenne muscular dystrophy ERT – enzyme replace therapy
MA
GAA - lysosomal acid α-glucosidase
NU
DCM – dilated cardiomyopathy
Gb3 - Globotriaosylceramide
HF - heart failure
CE P
LV - left ventricle
TE
D
HCM - hypertrophic cardiomyopathy
LVOT - LV outflow tract
AC
LVEF – left ventricular ejection fraction PLN - phospholamban RCM - restrictive cardiomyopathy SAP - serum amyloid P component SCD - sudden cardiac death SR - sarcoplasmic reticulum TTR-FAP - familial amyloidotic polyneuropathy WPW – Wolff Parkinson White
ACCEPTED MANUSCRIPT 4
1.
Introduction Cardiomyopathies arising due to a single gene defect represent various
T
pathways to evoke adverse remodeling and cardiac dysfunction. While the gene
IP
therapy approach is slowly evolving and has not yet reached clinical "prime time" and
SC R
gene correction approaches are being developed at the bench but not at the bedside, progress is gradually being made with medical therapies. This review summarizes the contemporary drugs being tested and introduced into the clinical arena to help manage
NU
these diseases. We selected drugs soon to be used in the clinic, those being tested in
MA
clinical trials or having abundant experimental evidence in their favor and are about to start clinical experimentation in various cardiomyopathy groups (Table 1).
D
We hereby grouped the cardiomyopathies according to the principal subtypes as
TE
defined by the most recent cardiomyopathy classification (Elliott, et al., 2008), while paying specific attention to unique subtypes characterized by Red Flags useful for
CE P
disease diagnosis (Rapezzi, et al., 2013). There are numerous diverse diseases caused by metabolic defects, substrate accumulation and protein aggregates. Being unable to
AC
address every particular gene-variant, we describe the representative examples to demonstrate the status of current therapy and future directions (see Table 1).
2.
Hypertrophic cardiomyopathy (HCM)
Clinical overview HCM is the most common genetic cardiovascular disease with an estimated prevalence of 1 in 500. It is characterized by a thickened, but not dilated, left ventricle (LV) without a secondary cause for the hypertrophy (such as aortic stenosis). HCM is the most common cause of sudden cardiac death (SCD) in the young and in athletes. Adults may suffer from effort intolerance due to angina or heart failure while in the
ACCEPTED MANUSCRIPT 5
elderly atrial fibrillation and strokes are common complications. Yet, many individuals with the disease have a normal life expectancy and have little, if any,
T
medical limitations (Maron, et al., 2009; Maron, et al., 2013).
IP
Genetics: HCM is the result of a mutation in one of 11 genes encoding components of
SC R
the sarcomere or (sometimes) the Z-disc. Of the genetically diagnosed patients, 70% have a mutation in the β-myosin heavy chain and myosin-binding protein C
NU
(cMyBPC) (Richard, et al., 2003). Abnormalities in myofilament contraction and in the intracellular calcium management are considered to trigger the hypertrophic
MA
response leading to an abnormal ventricular remodeling. Although transmitted in an autosomal dominant fashion, there is significant phenotypic heterogeneity, implying
D
that there is a role for modifier genetic and environmental factors. Known mutations
TE
can only be identified in ~50% of affected probands, a fact that complicates genetic counseling. Moreover, there is limited correlation between mutations and clinical
CE P
outcome (Landstrom, et al., 2010). Thus, the major contribution for genetic testing today is in ruling out the carrier state in family members of affected individuals and
AC
identifying patients with an alternate condition mimicking HCM (such as Fabry's disease, familial amyloidosis) (Weidemann, et al., 2009). Diagnosis and clinical course: Diagnosis might be a consequence of a cardiac event, family screening, or an abnormal physical examination or ECG (Adabag, et al., 2006). Currently, diagnosis is made with echocardiography or cardiovascular magnetic resonance imaging which show an unexplained increase of LV wall thickness ≥ 15 mm (21-22mm on average) (Rickers, et al., 2005). Hypertrophy develops in different patterns, even in patients with the same mutation, and might be accompanied with obstruction of the LV outflow tract (LVOT). As previously noted, there is a variable clinical course ranging from asymptomatic patients to SCD at a young age,
ACCEPTED MANUSCRIPT 6
development of heart failure (HF) or atrial fibrillation with stroke. HF is usually the result of diastolic dysfunction, LVOT obstruction and atrial fibrillation (Melacini, et
T
al., 2010). Only a small percentage develops systolic dysfunction, often commencing
IP
in severe heart failure where the only effective treatment is heart transplantation
SC R
(Maron, et al., 2010). SCD is more common in patients under 35 years of age and can be the first presentation of HCM. While cases occurring during competitive sports get most publicity, sudden death most commonly occurs while the patient is sedentary or
NU
engaged in mild activity.
MA
Management: Symptomatic left ventricular outflow obstruction is usually managed by a combination of beta adrenergic blocker with disopyramide. Treating obstructive
D
cardiomyopathy is often a challenge, in particular in the elderly with coexisting
TE
hypertension of conduction system disease. For patients with persistent symptoms despite optimal medical therapy, there is an indication for surgical myectomy (or
CE P
alcohol septal reduction as an alternative) with the goal of relieving LVOT obstruction (Kwon, et al., 2008; Ommen, et al., 2005). Treatment of HF with diastolic
AC
dysfunction consists of beta blockers or verapamil with a cautious use of diuretics. Once the patient develops systolic failure, the treatment is similar to other forms of HF with reduced left ventricular ejection fraction, although none of the "evidence based" therapies has been validated in this population. Patients with hypokinetic HCM often develop severe symptoms and require heart transplantation rather early because of a coexistent diastolic dysfunction and the limited capacity of the fibrotichypertrophic ventricle to increase its volume by chamber dilatation. Identification of at-risk family members of a diagnosed patient via clinical or genetic screening is a key component of management. Defibrillators are implanted for primary or secondary prevention of sudden death. The indications for primary
ACCEPTED MANUSCRIPT 7
prevention are based on clinical criteria such as SCD in the family, massive LV hypertrophy, unexplained syncopal events and recurrent non sustained ventricular
T
tachycardia on Holter monitoring (Gersh et al., 2011). An algorithm numerically
IP
assessing the 5-year risk of life-threatening arrhythmia has recently been introduced.
SC R
In addition to the above clinical criteria it takes into account age, left atrial side and left ventricular outflow obstruction (Elliott, et al., 2014).
Novel therapeutic strategies in HCM
NU
3.
MA
Following are several examples of drugs/procedures aimed at alleviating HCM symptoms.
D
- One mechanism that underlies arrhythmia leading to SCD in HCM is thought to be
TE
the increased sensitivity of the myofilaments to Ca2+. Blebbistatin is an inhibitor of actin-myosin interaction functioning independently of Ca2+ influx (Dou, et al., 2007).
CE P
Use of blebbistatin in a murine model of troponin T mutation reduced arrhythmia susceptibility, establishing Ca2+ sensitization as a potential therapeutic target in HCM
AC
(Baudenbacher, et al., 2008). - Parvalbumin is a Ca2+ buffering molecule, which is not normally expressed in cardiac muscle, and also binds Mg2+. When Ca2+ concentration rises, Parvalbumin releases magnesium and bind calcium. Forced expression of parvalbumin in cardiac muscle was able to correct the slow relaxation (and thus improve diastolic dysfunction) in the rat and mouse models of HCM due to a mutation in α-tropomyosin (Coutu, et al., 2004). MYK-461, a novel allosteric modulator of the myosin molecule, attenuates abnormal excessive contraction and enhances relaxation. MYK-461 has recently entered into Phase I of a clinical program (MyoKardia, NCT02329184).
ACCEPTED MANUSCRIPT 8
- Enhanced Ca2+ uptake by the sarcoplasmic reticulum (SR) can also alter the progression of HCM phenotype. This was achieved by two means in a HCM mouse
T
model caused by a tropomyosin mutation. The first was by an adenoviral delivery of
IP
SERCA2a (an SR protein) to 1-day-old transgenic mice. This single injection gene
SC R
delivery resulted in a significantly increased expression of total cardiac SERCA2a protein up to a few weeks of age, and was sufficient to improve morphology and response to β-adrenergic stimulation up to 3–4 months of age (Pena, et al., 2010). The
NU
second method used was a knockout of the phospholamban (PLN) gene, a SERCA2a
MA
inhibitor. Thus, a knockout of PLN resulted in increased Ca2+ uptake by the SR (Gaffin, et al., 2011).
D
- A recent work has shown the feasibility and effectiveness of gene-transfer of deleted
TE
genes by recombinant viral vectors of cMyBPC (Merkulov, et al., 2012), one of the most common causes of HCM. The increased levels of cMyBPC resulted in improved
CE P
contractile function both in vitro and in vivo. - Another mechanism which may affect the altered Ca2+ homeostasis is the inhibition
AC
of the L-type Ca2+ channel by diltiazem (Semsarian, et al., 2002). The mutation in the α-cardiac myosin heavy chain in mice resulted in altered Ca2+ regulation on multiple levels. These changes appeared in advance of changes in the cardiac histology or morphology. Administration of diltiazem at an early stage resulted in the upregulation of SR proteins and attenuated the development of the HCM phenotype. A recently published study in humans suggests that diltiazem may alleviate left ventricular remodeling in asymptomatic sarcomere mutation carriers (Ho, et al., 2015). - Diversion of myocardial substrate utilization from free fatty acid oxidation to carbohydrates by perhexilin, which inhibits mitochondrial uptake of long-chain fatty
ACCEPTED MANUSCRIPT 9
acids, resulted in a more efficient ATP production and subsequent improvement in cardiac function (Abozguia, et al., 2010), (NCT00500552). Similarly, trimetazidine,
T
can switch the heart metabolism from free fatty acid to carbohydrates utilization in
IP
parallel with its Ca2+ antagonism properties (clinical trial NCT01696370).
SC R
- Ranolazine is another metabolic modulator having a similar effect to that of perhexilin and trimetazidine. In addition, ranolazine has recently been shown to shorten the action potential duration and reduce arrhythmias in human HCM
NU
cardiomyocytes by inhibiting the late sodium current INa (Coppini, et al., 2013).
MA
Ranolazine is currently approved for refractory angina in patients with coronary disease and has been advocated for HCM patients suffering from myocardial ischemia
D
(in the absence of epicardial coronary artery disease). A multicentre, double-blind,
TE
placebo-controlled pilot study is currently underway to test the efficacy of ranolazine on exercise tolerance and diastolic function in symptomatic HCM patients (the
CE P
RESTYLE-HCM, EUDRA-CT 2011-004507-20). Another selective inhibitor of INa, GS-6615, is being investigated as a means to improve exercise capacity in
AC
symptomatic HCM (Liberty trial, NCT02291237). - Aldosterone is elevated in humans with HCM. Spironolactone, an aldosterone inhibitor, has been found to reduce myocardial fibrosis, attenuate the extent of myocyte disarray and improve diastolic function in experimental models (Tsybouleva, et al., 2004). Eplerenone, another aldosterone antagonist, is similar in structure to spironolactone, but has far fewer adverse effects. Both compounds have an established role in post-myocardial infarction and in HF with reduced ejection fraction. Both are currently being tested in HCM. [Clinical trials, (NCT00879060) and (NCT01521546), respectively]
ACCEPTED MANUSCRIPT 10
- Losartan, an angiotensin II receptor blocker, has been found to ameliorate morphological changes in LV mass (as measured by MR imaging) in a small group of
T
patients with HCM (Yamazaki, et al., 2007). Another angiotensin receptor blocker,
IP
candesartan, also had a favorable effect after one year of treatment, with a reduction
SC R
in LV hypertrophy and an improvement of LV function (Penicka, et al., 2009). The greatest response was seen in patients with β-myosin heavy chain mutations. In disagreement with these, the recently published INHERIT trial (Axelsson, et al.,
NU
2015) examined 100 mg/day losartan in a large number of patients (n=133). The drug
MA
was well tolerated but did not affect the left ventricular mass after 12 months of therapy.
D
- N-acetylcysteine, a precursor of glutathione, the largest intracellular thiol pool
TE
against oxidative stress, reversed cardiac hypertrophy and fibrosis in transgenic rabbits with a HCM phenotype (Lombardi, et al., 2009). A clinical trial to test its
CE P
effect on HCM patients is now being conducted and is at the recruiting stage
4.
AC
(NCT01537926).
Dilated cardiomyopathy (DCM)
Clinical overview DCM is a myocardial disease characterized by LV (or biventricular) enlargement and decreased systolic function. The majority of cases in the Western hemisphere are caused by coronary artery disease. The prevalence of non-ischemic DCM is 1:2000-3000 and it constitutes a major cause of HF and heart transplantation. DCM may arise via multiple, sometimes interacting etiologies including hypertension, inflammation, endocrine-metabolic, toxic and even infiltrative lesions. In about 25-
ACCEPTED MANUSCRIPT 11
35% of cases it is a familial disease having more than 40 causative genes identified to date (Burkett, et al., 2005).
T
Genetics: in contrast to HCM, the genes identified in DCM encode for a number of
IP
cellular components, such as the nuclear envelope, gene transcription, calcium
SC R
handling, the sarcomere, cytoskeleton and sarcolemmal proteins (Dellefave, et al., 2010). These gene mutations evoke diverse mechanisms of cell damage triggering molecular responses which become maladaptive leading to decreased function and
NU
adverse remodeling. Eventually, various causes of DCM converge to a final common
MA
pathway which comprises decreased calcium stores and myofilament calcium sensitivity, reduced ejection fraction and ventricular dilatation. Yet, there are
D
important differences in the propensity for ventricular arrhythmia, the presence of
TE
conduction system disease, skeletal myopathy, unique morphologic features such as left ventricular non-compaction and, rarely, extracardiac manifestations (deafness,
CE P
etc.). Inheritance is mostly autosomal dominant, although x-linked, autosomal recessive and mitochondrial forms have been reported.
AC
Diagnosis and clinical course: Medical evaluation starts with a detailed clinical history that is aimed at ruling out risk factors and other causes of DCM (such as coronary disease, chemotherapy exposure or excess alcohol consumption). Physical examination will reveal signs of HF (left-sided or biventricular) but may be rather unremarkable at an early stage of the disease. The ECG may show LBBB, left axis deviation or a nonspecific cardiomyopathy pattern (low limb lead but high precordial lead voltage and poor R wave progression in the chest leads). Echocardiography will show a globally hypokinetic LV with variable degrees of ventricular enlargement and atrio-ventricular valve regurgitation. The right ventricle works against a lower afterload and often develops the disease on a later stage but may deteriorate
ACCEPTED MANUSCRIPT 12
concomitantly with the LV. Coronary angiography or radionuclide perfusion imaging is important to rule out ischemic heart disease as a cause for DCM in clinically
T
relevant scenarios. Cardiac MRI may help distinguish between ischemic and non-
IP
ischemic DCM and provide clues to a specific diagnosis such as arrhythmogenic
SC R
cardiomyopathy, myocarditis, sarcoidosis, left ventricular non-compaction, amyloid and hemochromatosis. Endomyocardial biopsy is sometimes necessary when myocardial inflammatory or infiltrative process are suspected or have to be ruled out.
NU
Meticulous family history and screening of first degree family members by ECG and
MA
echo-Doppler are important to identify a familial disease. Familial DCM is typically defined when there is at least one first degree family member affected by the same
D
disease or died suddenly before age 35 years (Mestroni, et al., 1999).
TE
Management: Genetically determined DCM is treated according to contemporary HF guidelines (Yancy, et al., 2013). No etiology-specific treatment exists today except
CE P
several rare metabolic conditions (such as carnitine deficiency, fatty acid acyl-Co A dehydrogenase deficiency) which respond to a specific therapy. Patients are treated
AC
with angiotensin converting enzyme (ACE) inhibitors, -blockers and diuretics similar to that used with other causes of systolic HF. Mineralocorticoid antagonism is indicated in patients with symptomatic HF. LCZ696, comprising an angiotensin receptor blocker with a neutral endopeptidase inhibitor (McMurray, et al., 2014), is about to be approved for clinical use in HF with reduced systolic function instead of ACE inhibitors/ARBs. Cardiac resynchronization therapy is warranted in patients with a prolonged QRS and left ventricular dyssynchrony (Chung, et al., 2008). Family management starts with screening of first order relatives of affected individuals in order to find members at risk. Screening should include history, physical examination, ECG and echocardiography since asymptomatic DCM is
ACCEPTED MANUSCRIPT 13
common. Age-dependent and incomplete penetrance is a common feature in familial DCM, so most patients will present in the forth-to-seventh decade of life. Some
T
require an additional stress such as hypertension or pregnancy to express the disease.
IP
Genetic testing may be accomplished by gene sequencing of panels of multiple DCM-
SC R
related candidate genes (40 and up to 130) (Sturm, et al., 2013). Founder mutations are uncommon but shall be considered as a cause of DCM in certain populations (Dhandapany, et al., 2009; van der Zwaag, et al., 2012).
NU
The optimal time of initiating "evidence based" cardioprotective therapy in
MA
DCM gene carriers is not well established. Carvedilol may have some efficacy in preventing left ventricular dilatation in asymptomatic individuals with 'early' familial
D
DCM (Yeoh, et al., 2011). In case of a severe disease represented by Duchenne
TE
Muscular Dystrophy (DMD), perindopril delayed the onset and progression of LV dysfunction (Duboc, et al., 2005) and reduced mortality as shown by a 10-year
CE P
follow-up study.
The yield of gene testing in DCM is quite low: only ~ 30% have their disease-
AC
causing mutation identified. Several genes and certain phenotypes warrant particular attention:
Mutations in a nuclear envelope protein lamin A/C (LMNA) are associated with conduction system disease and early life-threatening ventricular arrhythmia occurring when the ventricular function is relatively preserved. LMNA accounts to 5-7% of all DCM and to 30% of DCM with conduction system disease (van Rijsingen, et al., 2012).
Dystrophin (DYS) gene on X chromosome is responsible for DMD and Becker Muscular Dystrophy (BMD), but is also the cause of 5-8% of DCM evaluated in cardiology clinic. While these patients are characterized by severe
ACCEPTED MANUSCRIPT 14
ventricular dilatation and mild myopathy and/or CK elevation, ventricular arrhythmias are uncommon (Diegoli, et al., 2011). Sarcomere protein gene mutations cumulatively account for 5-10% of dilated
T
IP
cardiomyopathy and are typically characterized by early disease onset. The
SC R
giant protein titin constitutes a molecular ruler responsible for structural integrity and diastolic tension. While known to be a DCM gene, the TTN gene was considered to be a rare cause of disease. The introduction of
NU
contemporary sequencing techniques led to identification of truncating TTN
MA
mutations in 25% of familial and 18% of sporadic DCM cases (Herman, et al., 2012). This genocopy is typically characterized by a late onset presentation
D
and variable expression which is affected by hemodynamic load as well as
5.
CE P
TE
other environmental and genetic modifiers.
Novel therapeutic strategies in familial DCM
AC
The role of humoral immunity in DCM is well established not only in inflammatory cardiomyopathy but also in genetically determined disease (Caforio, et al., 2007). It has been shown that anti--adrenergic receptor autoantibodies have a pathogenic role in the development of DCM in mice (Jahns, et al., 2004) and humans (Iwata, et al., 2001). This led to trials of immunomodulation as a treatment strategy. Immunoadsorption which removes circulating auto-antibodies was used to treat DCM in 45 patients (Staudt, et al., 2004). A significant result was only achieved in those patients in whom antibodies proved to be cardio-depressing in mice. This treatment option is currently being tested in a randomized trial of 200 patients (NCT00558584).
ACCEPTED MANUSCRIPT 15
As previously noted in DCM patients, the presence of anti-1-receptor autoantibodies has been shown to predict increased depressed left ventricular function, a
T
raised prevalence of serious ventricular arrhythmias, SCD and cardiovascular
IP
mortality (Iwata, et al., 2001). In the rat model of autoimmune cardiomyopathy, COR-
SC R
1 cyclopeptide has been shown to bind to, and therefore decrease, the anti--receptor autoantibody effect (Jahns, et al., 2010). Recently, a phase I trial of COR-1 was proved safe and effective in humans with no unwanted side effects on the immune
NU
system (Munch, et al., 2012).
MA
N-3 polyunsaturated fatty acids (nPUFA) are fatty acids derived from fish oil which are important for the normal human metabolism. PUFA are under investigation
D
in diverse clinical settings, from gastric cancer to arrhythmias. PUFA administration
TE
prevented cardiac remodeling in a rat aortic banding model (Duda, et al., 2009) and
CE P
resulted in a small decrease in mortality in patients with HF from any cause (Tavazzi, et al., 2008). PUFA were recently tested in a randomized trial of DCM patients where they improved LV systolic and diastolic function, as well as functional capacity
AC
(Nodari, et al., 2011).
Gene therapy can target different stages in the course of the development of
DCM. One target that has been tested in humans is the defective SR calcium handling as a result of decreased SERCA2a levels (discussed in the chapter on HCM). In a study using an adeno-associated virus (AAV) serotype 1 vector to carry the SERCA2a gene to cardiac muscle, a 3-year follow-up of 39 patients showed a tendency towards improved survival and reduced cardiovascular events, especially in the high dose group. There was no increase in arrhythmias in the treatment group and a long term vector persistence was demonstrated on cardiac biopsies (Jaski, et al., 2009; Zsebo, et al., 2014). Of note, only patients without preformed, naturally occurring antibodies to
ACCEPTED MANUSCRIPT 16
AAV1 are eligible for this treatment. A larger study is now being conducted (NCT0164330), as well as a study in patients with LVAD (NCT00534703).
T
Intracoronary stem cells transplantation was first used in the setting of
IP
ischemic heart disease (IHD) and acute MI. Evidence of microvascular dysfunction as
SC R
well as dysfunction of bone marrow cells (BMCs) and endothelial progenitor cells in DCM was the trigger for the use of stem cells in DCM. After harvesting CD34+ cells (previously by aspiration but currently by peripheral harvesting after administration of
NU
G-CSF), the cells were introduced via intracoronary infusion to predefined
MA
myocardial areas with contractile dysfunction. This resulted in a 1-year improvement of both clinical and echocardiographic parameters (6-minute walk, LVEF, survival
D
(Vrtovec, et al., 2011). The same group recently published a 5-year follow up
TE
showing consistent results with most of the benefits of therapy in the first year. This study also demonstrated a positive correlation between myocardial homing of the
CE P
stem cells and the response to therapy (Vrtovec, et al., 2013). The mechanisms involved are still unknown but might include paracrine effects, such as attenuation of
AC
apoptosis of endogenous cardiomyocytes and endothelial cells, promotion of angiogenesis, activation of resident cardiac stem cells, or induction of an antiinflammatory effect (Ebelt, et al., 2007). As mentioned in the HCM treatment section, ranolazine, an inhibitor of the late sodium current INa, L is being tested in multiple clinical scenarios such as HCM, as an anti-arrhythmic and angina relief. Another recently initiated trial, aims to determine if ranolazine improves myocardial perfusion in patients with DCM (NCT02133911). Ixmyelocel-T is an autologous multicellular therapy expanded from bone marrow mononuclear cells. As a result of the manufacturing process, it comprises
ACCEPTED MANUSCRIPT 17
both lymphoid and myeloid cells with a large number of M2-like macrophages and mesenchymal stromal cells (Bartel, et al., 2012). Due to this multicellular
T
composition, Ixmyelocel-T has demonstrated multiple activities relevant to tissue
IP
repair and regeneration (Ledford, et al., 2013). Ixmyelocel-T was recently tested in
SC R
Phase IIA clinical trials in patients with ischemic and non-ischemic DCM. After harvesting bone marrow cells from the iliac crest patients underwent intramyocardial injection of Ixmyelocel-T, either surgically or with a percutaneous technique. In both
NU
studies there was no effect in the non-ischemic DCM group but a clinically significant
MA
effect in the ischemic group. The latter clinical improvement is in contrast to the lack of improvement in LVEF, LVEDD and other laboratory measures (NCT00765518,
D
NCT01020968). This might be a result of the lack of a true placebo group (i.e., no
TE
bone marrow aspiration was done in the control group). Ixmyelocel-T is being further tested in ischemic DCM (NCT01670981).
CE P
Pharmacological myofilament Ca2+ sensitization by agents such as levosimendan has been shown to be of clinical benefit in advanced HF with
AC
hemodynamic compromise (NCT01290146). A recent study has suggested that intermittent administration may improve functional status and prevent hospitalization. Newer drugs from the same category (Pimobendan, EMD 53998 and MCI-154) are being developed. Another class of drugs are the cardiac myosin activators, which act by accelerating the transition from weekly bound myosin to strongly bound myosin without altering the Ca2+ transient and without being affected by -blockers (Malik, et al., 2011). Omecamtiv mecarbil, the representative drug in this class, was compared to dobutamine in a canine model of HF. Its use resulted in an increase in stroke volume and cardiac output without affecting oxygen consumption (Shen, et al., 2010). In a phase II clinical trial in 45 patients with DCM (both ischemic and non-ischemic)
ACCEPTED MANUSCRIPT 18
treatment with IV Omecamtiv mecarbil resulted in a dose dependent increase in the duration of left ventricular systole. The drug was generally well tolerated unless high
T
plasma concentrations were reached, when signs and symptoms of ischemia were
IP
noted (Cleland, et al., 2011). Omecamtiv mecarbil is currently being tested in a trial of
SC R
acute HF (NCT01300013).
Qili qiangxin, a Chinese traditional medicine, enhances heart function in chronic HF, in part by regulating the balance of TNF- and IL-10 (Xiao, et al., 2009).
NU
Qili qiangxin was approved for use in CHF patients by the Chinese food and drug
MA
administration in 2004. In a randomized, double blind, placebo controlled trial of nearly 500 patients with DCM (with over 50% of them with non-ischemic DCM),
D
addition of Qili qiangxin to standard medical therapy resulted in a significant decrease
TE
in NT-proBNP levels concomitant with improvement in the 6-minute walk test and EF (X. Li, et al., 2013). This drug is now being examined as a supplement to standard
CE P
therapy in a large cohort of DCM patients (NCT01293903). Disease-specific treatment strategies are just beginning to evolve. Abnormal
AC
activation of mitogen-activated protein (MAP) kinase signaling pathway through p38α was detected in LMNA mutant mice prior to the onset of significant cardiac impairment. Pharmacological inhibition of p38α prevented LV dilation and deterioration of fractional shortening in mice with DCM caused by LMNA (Muchir, et al., 2012). Approaches to correct the gene defect are described in detail in the chapter on dystrophinopathies.
6. Dystrophinopathy
ACCEPTED MANUSCRIPT 19
DMD, an X-linked disease of skeletal and cardiac muscle caused by dystrophin gene defects is the prototype of dystrophynopathies. This lethal disease has
T
a high incidence among newborn males (approximately 1:3500). The disease follows
IP
a course of progressive muscle weakness, which further involves the cardiac and
SC R
respiratory muscles, resulting in early-age mortality for the vast majority of affected boys, and to a lesser extent, motion disability at the age of 12. To date, no curative treatment for DMD is present, though the molecular basis of the disease is highly
NU
elucidated. Over the years, the life expectancy of DMD has immensely increased,
MA
probably as a result of the effect of combined corticosteroids, antibiotics therapy, noninvasive ventilation and improved airway hygiene. DCM is a characteristic feature of
D
DMD and currently constitutes a major cause of morbidity and mortality (Fayssoil, et
TE
al., 2010). Dystrophin gene mutations have been established as the sole cause of DMD, resulting in the complete or partial abrogation of the protein. The dystrophin
CE P
gene located on the short arm of the X chromosome constitutes the largest gene in the human genome, covering 2.2 mega-bases and composed of 79 exons and 7 promoter
AC
regions. Dystrophin protein is localized in the sarcolemma of muscle fibers and postulated as an essential molecular shock absorber of the contractile apparatus, thus protecting the sarcolemma from mechanically-induced damage. Various types of mutations leading to DMD have been reported, such as deletions (most common), duplication of exons and point mutations (usually stop codons). Classical DMD patients are characterized by the complete lack of the dystrophin protein, because the mutation leads to a loss of a reading frame. By contrast, mutations resulting in less defective, albeit still partially functional dystrophin protein, lead to a milder phenotype, and are typically diagnosed as BMD, where cardiac manifestations often precede skeletal muscle weakness.
ACCEPTED MANUSCRIPT 20
Much of the ongoing research is focused on gene therapy. The development of highly efficient and specific viral vectors for heart gene transfer, mainly AAV
T
serotypes 5, 6, 8 and 9, enabled the insertion of synthetic gene fragments into the
IP
heart in order to either replace or repair the defective dystrophin gene. Genetically
SC R
engineered minimized (mini- or micro-) dystrophin or utrophin (the dystrophin homolog) gene transduction has recently provided encouraging findings showing improved sarcolemmal integrity in cardiomyocytes in vivo and displaying reduced
NU
fibrosis and a normalized heart rate in dystrophin-null mdx mice. Significant
MA
improvement of calcium homeostasis was observed in mice to which an overexpression vector of SERCA2a was delivered, supporting the potential of gene-
D
therapy as a game changer. Another encouraging strategy aims to exclude or skip an
TE
exon by taking advantage of antisense oligo-nucleotide (AON), which bind to specific splicing sites on pre-RNA (Kole, et al., 2012; Koo, et al., 2013). AON leads to the
CE P
restoration of the reading frame (which was distorted by the mutation, usually causing exons-deletion) and to the production of smaller, though a sufficiently functional
AC
dystrophin protein. Deletions between exons 44-55, corresponding to the rod domain of the protein, account for nearly 75% of DMD patients and therefore most exon skipping therapies were directed to this region (45 PRO045). Importantly, since cardiomyocytes exhibit a poor AON uptake rate compared with skeletal muscle cells, the additional advantage of chemical modifications, including conjugating AON to nano-particles or to membrane penetrating peptides, provide great physiological benefits in mdx mice, thus paving the way for clinical trials. Stop codons causing protein truncation are another type of lethal mutation. There have been several attempts to 'read through' a stop codon by either gentamycin or drugs interacting with ribosomal subunits. Ataluren is the first drug in this class of
ACCEPTED MANUSCRIPT 21
oral medications, and has been recently approved as a potential therapy for ~ 13% of DMD patients who carry a nonsense mutation. Ataluren’s effect was demonstrated in
T
a wide spectrum of diseases caused by nonsense mutations (Welch, et al., 2007) as
IP
well as in Phase IIa trials in DMD and cystic fibrosis patients (Finkel, et al., 2013). In
SC R
a phase IIb study, 174 patients were randomized to different doses of ataluren or placebo. After a 48-week follow-up there was a reduction in the rate of decline in the 6-minute walk test in the treated patients (medium dose but not high dose). It is
NU
important to acknowledge that cardiac function was not targeted in any of these trials.
MA
A Phase III study is currently under way (NCT01826487) Since the defective protein is degraded through the misfolded protein response
D
in the endoplasmic reticulum and through the proteasome, a research effort is made to
TE
prevent complete degradation by interfering with these pathways (Guerriero, et al., 2012). Proteasome inhibitors increased the expression of dystrophin and of associated
CE P
membrane proteins in a murine model and in muscle biopsies of patients with DMD
7.
AC
and BMD (Gazzerro, et al., 2010).
Arrhythmogenic cardiomyopathy (ACM) Ventricular arrhythmias and fatty replacement of myocardium are the hallmarks
of ACM. This disease was previously called arrhythmogenic right ventricular cardiomyopathy but that term is inapt since up to 50% of cases develop biventricular involvement (Rizzo, et al., 2012). Rarely, the predominant feature of the disease is the involvement of the LV with ventricular arrhythmia (which is disproportionate to the degree of left ventricular dysfunction). The prevalence of ACM is about 1:5000 but due to variable disease expression many cases remain underdiagnosed and this estimate may be inaccurate (Peters, et al., 2004). The disease is an important cause of
ACCEPTED MANUSCRIPT 22
life threatening ventricular arrhythmia in young and middle-age adults and constitutes an important cause of sudden death in athletes. At a later stage, once the arrhythmia is
T
controlled, HF becomes the main contributor to morbidity and mortality in this
IP
disease.
SC R
Genetics: The principal genetic cause of ACM are mutations in the genes that encode for the desmosomal proteins which are responsible for adhesion on adjacent
NU
cardiomyocytes (Swope, et al., 2013). When sequencing DNA from a patient with the disease, definite mutations will be found in only ~50%, but many patients will have a
MA
single nucleotide change of unknown significance in one of the candidate genes. These variants may sometimes be found in healthy controls and have a modifying
D
effect on the disease phenotype (Kapplinger, et al., 2011). Moreover, at least the
TE
major known disease genes (PKP2, JUP, DSG2, DSC2, DSP) must be sequenced
CE P
because patients with more than one variant have a higher likelihood of disease (Quarta, et al., 2011).
Diagnosis and clinical course: The diagnosis of ACM is based on the combination of
AC
clinical, morphological, electrocardiographic and genetic findings, which are divided into major and minor diagnostic criteria as proposed in 2010 (Marcus, et al., 2010). Ventricular arrhythmias, usually of right ventricular origin, are the hallmark of the disease and are usually the presenting symptom. Ventricular dysfunction is common at a later stage but only a minority of patients develop overt HF (Rizzo, et al., 2012). Of note, SCD may occur even before major structural abnormalities occur. This fact emphasizes the importance of identifying family members of a proband (Tabib, et al., 2003). The ECG may show disease-specific epsilon waves or non-specific findings such as RBBB or T wave inversion in the right precordial leads. Visualization of right
ACCEPTED MANUSCRIPT 23
ventricular dilatation with regional dysfunction, often associated with wall thinning and aneurysm formation, is usually required for the diagnosis. Cardiac magnetic
T
resonance is particularly useful to identify the right ventricular pathology and the
IP
extensive patchy delayed gadolinium enhancement which characterizes the left
SC R
ventricular involvement. Cardiac biopsy and electrophysiological studies may be necessary.
Management: Currently, management is primarily focused on the early identification
NU
and control of ventricular arrhythmias. Competitive athletic activity and strenuous
MA
physical activity are forbidden since not only may they induce a life-threatening ventricular arrhythmia but may also enhance disease progression (Cruz, et al., 2015;
D
James, et al., 2013; Saberniak, et al., 2014). -blockers or antiarrhythmic drugs such
TE
as sotalol or amiodarone are often used for arrhythmia control and prevention. Patients with aborted SCD and syncope have an indication for implantable
CE P
cardioverter-defibrillator implantation. Another option for recurrent arrhythmia would be a catheter ablation of VT focus (Bai, et al., 2011; Tabib, et al., 2003). There is no
AC
established therapy for isolated right ventricular dysfunction. Once the LV ventricle starts to fail, patients are treated according to HF guidelines.
8.
Novel therapeutic strategies in familial ACM Based on the theory that reduction in wall stress can prevent progression of the
disease in genetically predisposed patients, a blind trial of preload reduction was held in plakoglobin-deficient mice (Fabritz, et al., 2011). Preload reduction with nitrates and diuretics prevented right ventricular enlargement, right ventricular conduction slowing and the induction of right ventricular arrhythmias. A clinical trial called PreVENT-ARVC, that is based on this approach is about to commence. The human
ACCEPTED MANUSCRIPT 24
trial will use a long-acting nitrate that does not induce nitrate tolerance (PETN) rather than isosorbid nitrates and a combination of a thiazide diuretic and a potassium-saving
T
diuretic will be used instead of furosemide (Fabritz, et al., 2012).
IP
PLN is a regulator of the cardiac sarcoplasmic reticulum Ca2+ pump
SC R
(SERCA2a), and is thus important for maintaining Ca2+ homeostasis. A mutation in the gene for PLN, R14del, was identified in 12% of Dutch patients with ACM and
NU
15% with DCM (van der Zwaag, et al., 2012), implying an overlap between these two seemingly separate clinical entities. This cohort of patients was more prone to
MA
arrhythmias with a generally poor prognosis (van Rijsingen, et al., 2014). On the microscopic level, a diminished plakoglobin signal at intercalated disks was
D
associated with the ACM phenotype. An ongoing trial (NCT01857856) attempts to
TE
slow disease progression in presymptomatic patients with R14del using eplerenone, a mineralocorticoid with antifibrotic properties.
CE P
The Wnt / β–catenin signaling pathway is modified by desmosomal proteins. Increased nuclear translocation of plakoglobin suppresses Wnt signaling of cardiac
AC
progenitor cells. Thus, plakoglobin inhibits myogenesis, while (instead) triggering adipogenesis. Pharmacological or genetic alterations which positively regulate the pathway or prevent plakoglobin nuclear translocation could be a good candidate for the treatment of ARVC.
Finally, PPAR¥s, an adipogenic transcription factor,
demonstrated an important role in the fibro-fatty change during ARVC progression, constituting a therapeutic target.
9.
Restrictive cardiomyopathy (RCM) This is apparently the least common of the inherited cardiomyopathies. It
usually presents as heart failure with severe diastolic dysfunction and normal or
ACCEPTED MANUSCRIPT 25
'preserved' systolic function (indicating LVEF on the lower side of the normal range such as 50%). Enlarged atria and atrial fibrillation are common features (Ammash, et
T
al., 2000). The disease may evolve because of a pathological process causing
IP
cardiomyocyte stiffening, injuries (such as irradiation) which trigger extensive
SC R
interstitial fibrosis or an infiltrative process within the interstitial layer. It is very important to differentiate between restriction and extra-myocardial causes such as constriction and compression as well as to distinguish between the acquired and
NU
inherited causes of restrictive cardiomyopathy.
MA
Genetics: The genetics of RCM have some overlap with HCM and indeed, some authors refer to RCM as HCM with a restrictive pattern (Kubo, et al., 2007).
D
Regardless of the classification, there are a few sarcomere gene mutations that have
TE
been linked to RCM or to a mixed RCM/HCM phenotype. Mutations in Troponin I were the first to be identified in RCM families and are thought to cause the disease by
CE P
impaired calcium dissociation from the myofilaments (Menon, et al., 2008). We have recently described a family with a lethal RCM by a de novo mutation in the giant
AC
filament titin (TTN), which is responsible for the diastolic tension of the sarcomere (Peled, et al., 2014). Other genetic variants are mediated by the intracellular accumulation of a defective protein (desmin), iron, various metabolites in several storage diseases (glycolipid in Fabry’s or mucopolysaccaride in MPS) or an extracellular protein polymer in hereditary amyloidosis.
Diagnosis and clinical course: The common clinical features are effort intolerance, atrial arrhythmias (due to enlarged atria), thromboembolic phenomena and frequent progression to HF with pulmonary and prominent systemic venous congestion. Systemic manifestations may accompany certain subtypes and lead to a specific
ACCEPTED MANUSCRIPT 26
diagnosis. Conduction disease is quite common. When occurring early in the course of the disease, certain conditions should be suspected, such as desminopathy or
T
sarcoidosis. The prognosis for RCM is worse than that of HCM patients, death being
IP
the result of HF, SCD, or cerebrovascular accident (Ammash, et al., 2000; Kubo, et
SC R
al., 2007).
Management: There is no specific treatment for RCM. Treatment focuses on treatment for arrhythmias, symptomatic treatment of HF and, in end stage cases, heart
NU
transplantation. Hemochromatosis may respond to iron chelation or phlebotomies,
MA
AL amyloidosis partly responds to hematological therapy and certain metabolic diseases have an enzyme therapy which may stop or slow the disease progression. As
TE
D
a rule, once the restrictive phenotype has evolved the prognosis is invariably poor.
10. Novel therapeutic strategies in familial RCM
CE P
Fibrosis constitutes a central mechanism in the evolution of restrictive cardiomyopathy although it has prominent functional and arrhythmic impacts in other
AC
cardiomyopathy forms. While most evidence based therapies attenuate fibrosis secondary to their principle mechanism of action, so far no specific drug was developed to directly target fibrosis. Several attempts are being made in other forms with restrictive cardiomyopathy, such as diabetic and uremic myocardium and they might eventually be applicable for genetically determined RCM (Gonzalez-Quesada, et al., 2013; C. J. Li, et al., 2012; Lin, et al., 2015).
11. Familial amyloidosis Familial amyloidosis is caused by extracellular deposition of misfolded protein aggregates in various organs such as heart, kidney, gut and nervous tissue. The
ACCEPTED MANUSCRIPT 27
deposition of natural protein involved in the transport of retinol and thyroxine, transthyretine (TTR), causes a slowly progressive cardiomyopathy in the elderly
T
called senile amyloidosis. Mutant TTR has a lower stability and gives rise to a
IP
prevalent form of familial amyloidosis, a maturity-onset disease which may manifest
SC R
as familial amyloidotic polyneuropathy (TTR-FAP) or as cardiomyopathy. Abnormal protein production may be targeted through RNA interference (RNAi) or gene silencing molecule.
This is a novel therapeutic modality that utilizes cellular
NU
mechanisms of protein production inhibition. The ability to give the small interference
MA
RNA (siRNA) as a drug has been limited. Lately, however, there is the ability to target these molecules to the relevant organs. Delivery of a siRNA in a lipid
D
nanoparticle to patients, as a treatment of TTR-FAP, has recently been tested in a
TE
phase I trial (Coelho, et al., 2013). This study assessed two different formulations of liver-targeted siRNA in a group of patients with early TTR-FAP and healthy
CE P
volunteers. The result was a significant decline in hepatic TTR production after administration of a single dose with few adverse events. Tafamidis meglumine binds
AC
the two thyroxine binding sites of TTR thus preventing its dissociation into monomers. Two trials of tafamidis for the treatment of TTR-FAP showed modest, but promising results (Coelho, et al., 2012; Merlini, et al., 2013). The trials differed in the patients' mutations and the follow-up period. Both studies focused on neurological outcomes and quality of life measurements, but the more recently published one also measured cardiac function using several parameters. No deterioration in cardiac function was noted in a follow-up after 12 months, even in patients at high risk for cardiac complications (as assessed by NT-pro-BNP levels). A study of 400 patients to assess the cardiovascular benefit of Tafamidis is expected to end in 2018 (NCT01994889).
ACCEPTED MANUSCRIPT 28
Another strategy for stabilizing the TTR tetramer is by using diflunisal, a nonsteroidal anti-inflammatory (NSAID) drug. Naturally, treatment of patients with CHF
T
with an NSAID causes concern, especially due to the risks of hypertension and
IP
deterioration of renal function. To assess safety and to gather primary efficacy data,
SC R
an open label, single arm and short term trial in 13 patients was conducted (Castano, et al., 2012). A modest decline in renal function was the only adverse effect and in comparison to the TRACS cohort (Ruberg, et al., 2012) there was a trend toward a
NU
better outcome. A much larger, randomized, double blind study has recently been
MA
published (Berk, et al., 2013). The outcomes in this trial were focused on neurological parameters. The improvement in these parameters over the course of two years,
D
accompanied by only a few serious adverse events holds promise for cardiac
TE
improvement as well. This study is currently being continued as an observational study with cardiac function being a secondary outcome (NCT01432587).
CE P
A combination of an antibiotic doxycycline with a bile acid TUDCA was shown to remove TTR amyloid deposits in mice (Cardoso, et al., 2010; Obici, et al.,
AC
2012) and is currently under a phase I trial in humans with Transthyretin Amyloid Cardiomyopathy (NCT01855360). A different target for treatment of amyloidosis is the serum amyloid P component (SAP). This plasma glycoprotein binds to amyloid fibrils and enhances fibrillogenesis. In knock-out mouse for SAP there was a reduction in amyloid deposits (Botto, et al., 1997), and a therapeutic compound CPHPC was thus designed to target SAP. CPHPC forms a complex with SAP that is quickly cleared by the liver. In the first attempt to treat amyloidosis patients of various etiologies there was a dramatic reduction of serum SAP and a sub-total reduction of tissue SAP (Gillmore, et al., 2010). Assessing clinical benefit was more difficult, especially as the patients treated were mostly in advanced stages of the
ACCEPTED MANUSCRIPT 29
disease. In a mouse model of amyloidosis, therapy with CPHAP was augmented by the use of anti-SAP antibodies which resulted in macrophage dependent clearance of
T
tissue amyloid (Bodin, et al., 2010). The synergistic effect of CPHPC and an anti-SAP
IP
antibody is now being tested in a clinical trial (NCT01777243). This might emerge as
12. Metabolic and related cardiomyopathies
SC R
the first effective therapy to reverse the tissue damage inflicted by AL amyloidosis.
NU
These rare diseases are designated Orphan Diseases and the drugs developed
MA
are defined as Orphan Drugs. While being unable to account for all possible metabolic defects eventually leading to cardiomyopathy, we chose several classical
D
examples, in particular those which may be encountered in an adult cardiomyopathy
CE P
Pompe disease
TE
clinic.
Glycogen storage disease type II, commonly referred as Pompe disease, is a
AC
rare autosomal recessive inherited disorder, caused by a deficiency of the lysosomal acid α-glucosidase (acid maltase, GAA) enzyme, resulting in a massive lysosomal glycogen accumulation in cardiac and skeletal muscles. In general, two forms of the disease are considered: the infantile form, characterized by massive cardiac hypertrophy and muscle weakness with a prominent mortality rate during the first year of life due to cardiac and/or respiratory failure; and the late-onset disease form, which is typified by a progressive course of muscle weakness in children. The involvement of respiratory muscles, ultimately leads to premature death. The difference between these two forms of the disease has been shown to originate from the wide variety of mutations on the GAA gene and the level of residual enzyme
ACCEPTED MANUSCRIPT 30
activity. Palliative treatments for Pompe disease include substrate reduction therapy and supportive care (van der Ploeg, et al., 2008).
T
Enzyme replacement therapy (ERT) predominantly relies on the ability of
IP
cells to internalize synthetically recombinant lysosomal enzymes via the mannose-6-
SC R
phosphate receptor pathway followed by their delivery to the lysosomes, where the replacement of the defective enzyme with the functional enzyme occurs. Clinical studies using a recombinant analog of -glucosidase, alglucosidase alfa have shown a
NU
significant increase in life-span in parallel with prominent decrease of cardiac
MA
hypertrophy, in both infantile and late-onset forms (Kishnani, et al., 2007). Furthermore, when administered before overt clinical symptoms become apparent, the
D
beneficial effect is largely increased. Nevertheless, there are some drawbacks of this
TE
strategy: the high cost due to the need for life-long repeated infusions of the recombinant enzyme, poor targeting to muscles and enhanced immune response. All
CE P
these led scientists to develop alternative therapeutic strategies for Pompe disease. The combination of ERT with recently introduced innovative strategies holds great
AC
promise in changing the natural history of the disease. While gene therapy using various viral vectors significantly elevated persistency (over 1 year) and transduction level after only a single systemic delivery, other methods combining ERT with immunomodulatory agents such as methotrexate, account for the immense reduction of immune response against the recombinant enzyme (Joly, et al., 2014). Strikingly, a work by van Til et al., has provided the first evidence for combining both beneficial features by harnessing the immune-tolerance and whole-body distribution capabilities of autologous hematopoietic stem cells (van Til, et al., 2010). Introducing the recombinant enzyme under the strong promotors of viral vectors into these cells, prior to transplantation enabled the major clearance of glycogen in the heart, diaphragm
ACCEPTED MANUSCRIPT 31
and liver and improved muscular strength and cardiac function and remodeling. Another strategy was developed based on the assumption that better delivery of the
T
mutant enzyme to the lysosome (where it functions to breakdown glycogen) and
IP
increased stability of circulating protein would eventually lead to better glycogen
SC R
clearance and improved phenotypic outcome. To address these issues, various pharmacological chaperones were used in conjunction with ERT in Pompe disease fibroblasts and GAA-KO mice, and the findings are extremely encouraging.
NU
Consequently, a Phase II clinical trial has been initiated for Pompe patients (Khanna,
MA
et al., 2012). Finally, pioneering experiments in GAA-KO mice have shown that the inhibition of glycogen synthase by either RNA interference techniques or by
D
negatively regulating it via its signaling pathway both enhanced the beneficial effect
TE
of ERT, leading to an immense reduction in lysosomal glycogen storage. Hence, these techniques might be considered as a novel therapeutic option for Pompe patients
CE P
(Clayton, et al., 2014).
AC
Anderson Fabry disease
Fabry's disease is another X-linked lysosomal storage disease where early
diagnosis is important because of the availability of an effective enzyme therapy. The disease is caused by a deficiency of the lysosomal enzyme β-galactosidase A, leading to the accumulation of globotriaosylceramide (Gb3) in various tissues. Principal manifestations include peripheral and autonomic neuropathy, nephropathy, premature stroke and white matter lesions and cardiomyopathy. Cardiomyopathy usually develops at a relatively advanced stage but constitutes a major cause of mortality. It typically manifests in middle aged males and elderly females as cardiac hypertrophy with diastolic dysfunction and involvement of the conduction tissue which eventually
ACCEPTED MANUSCRIPT 32
leads to conduction block and arrhythmia. While substrate manipulation and supporting care comprising HF medications, pacemaker/defibrillators, dialysis and
T
pain control are part of the therapeutic arsenal, ERT revolutionized the natural history
IP
of this disease, in particular when initiated before the occurrence of irreversible organ
SC R
damage. Novel approaches are developed to potentiate the effect of ERT. A recent study in a mouse model of Fabry disease suggests that abnormal androgen receptor activity is one of the mitigating factors in the development of the disease
NU
manifestations. In this study (Shen, et al., 2015), increased levels of IGF-1 and
MA
decreased levels of TGF-β1 were measured in the cardiac tissue of Fabry mice. Castration of asymptomatic mice prevented the development of cardiac and renal
D
hypertrophy in the animals. Furthermore, castration of 12-month-old mice (who
TE
already had cardiac hypertrophy) resulted in a decrease of cardiac weight compared to WT mice. The availability of oral antiandrogens approved for treatment of other
CE P
disorders renders this treatment strategy a viable option. PRX-102 is a novel enzyme produced by BY2 tobacco cell culture. It is a
AC
homo-dimer with a function equivalent to the current enzyme replacement available for clinical use, but showing a superior stability profile and pharmacokinetics (Kizhner, et al., 2015). PRX-102 is currently being tested in humans (NCT01981720). AT-1001 (Migalastat HCl) is a pharmacological chaperone which improves the folding, stability and lysosomal trafficking of mutant forms of α-galactosidase-A (α-Gal A). It was previously shown to reduce the levels of globotriaosylsphingosine when given as monotherapy in mice and humans (Young-Gqamana, et al., 2013). AT-1001 was recently given in co-formulation with α-Gal A, which resulted in higher tissue levels of the enzyme and lower levels of globotriaosylsphingosine when compared to enzyme replacement therapy alone (Xu, et al., 2015). This drug is
ACCEPTED MANUSCRIPT 33
currently under an extended period trial as monotherapy for certain genetic variants (NCT02194985).
T
Efficacy of enzyme and substrate reduction therapy for Fabry disease with a
IP
novel antagonist of glucosylceramide synthase has been reported (Ashe, et al., 2015).
SC R
A recent study discovered two new drugs that might be potential enhancers of pharmacological chaperones. The expectorant ambroxol, especially in combination with migalastat, raised α-Gal A levels in a cell free thermal denaturation test. . The
NU
PPAR-γ agonist rosiglitazone, was also tested in a similar modal, and showed
MA
promising results, especially in combination with migalastat (Lukas, et al., 2015).
D
Danon disease
TE
Danon disease is caused by LAMP2 defects leading to the inability to complete the autophagic process. The lysosomes fail to fuse with autophagosomes
CE P
and the cell is left with undegraded contents such as protein aggregates, malfunctional organelles and old glycogen. For this reason the disease was originally named Pompe
AC
with normal acid maltase. Danon disease is a natural example of a primary failure of autophagy. It is now appreciated that secondary impairment of autophagy also contributes to the pathophysiology of other lysosomal storage diseases by the accumulation of toxic protein aggregates and defective mitochondria (Choi, et al., 2013). Danon's is an X linked disease and therefore males are more severely affected. Typically, a teenage boy has rapidly progressive hypertrophy which may reach extreme dimensions and be associated with Wolff–Parkinson–White (WPW) syndrome and arrhythmia (Arad, et al., 2005). Over several years, the heart progresses to the hypokinetic stage and develops severe HF leading to death unless heart transplantation takes place. Mental issues ranging from mild learning deficits to overt
ACCEPTED MANUSCRIPT 34
retardation and skeletal involvement ranging from asymptomatic CK elevation to overt myopathy are present
concomitantly or precede the diagnosis of
T
cardiomyopathy. Male patients also have transaminase elevation without clinically
IP
significant liver disease (Arad, et al., 2005). Women present in adulthood as
SC R
hypertrophic or dilated cardiomyopathy, sometimes in association with WPW syndrome or AV block. Teenage and young-adult presentation is also possible due to skewed X inactivation. Electrophysiological abnormalities follow, but may precede
NU
the development of cardiomyopathy. Enzyme abnormalities are uncommon in
MA
females. Because the stress of pregnancy and labor may lead to HF, some cases are wrongly diagnosed as peripartum cardiomyopathy and correct diagnosis is reached
D
only after the development of a typical disease in a male offspring (D'Souza R, et al.,
TE
2014). Much needs to be learned about the fascinating process of autophagy wherein the cell recycles its defective components, removes its waste and adjusts to ongoing
CE P
stresses. Modification of the processes triggering autophagy may help modify the course of this lethal disease while a targeted protein therapy would facilitate the
AC
restoration of the lysosomal function in Danon's disease. Impaired autophagy has a secondary role in other cardiomyopathies such as classical lysosomal storage diseases, protein aggregate diseases (such as desminopathy which causes dilated or restrictive cardiomyopathy with AV block), hypertrophic cardiomyopathy by MyBPC3 mutations and dilated cardiomyopathy by titin splice-variants (Choi, et al., 2013; Lieberman, et al., 2012; Schlossarek, et al., 2014). Interestingly, activating autophagy (Bhuiyan, et al., 2013) helps prevent the progression of desmin-like cardiomyopathy while inhibiting autophagy actually potentiates the effect of enzyme therapy in experimental Pompe disease. These findings are very important because autophagy
ACCEPTED MANUSCRIPT 35
appears to be a modifiable process amenable to drug therapy (Kroemer, 2015;
IP
T
Vakifahmetoglu-Norberg, et al., 2015).
SC R
Mitochondrial DNA mutations and defects of oxidative phosphorylation manifest as a multisystem disease. As opposed to defects of oxidative phosphorylation caused by nuclear DNA mutations, which quite uniformly present in infancy, the presentation of
NU
mitochondrial DNA defects is more variable and age-dependent, related to tissue distribution of defective mitochondria, mutation load and mitochondrial aging
MA
(Holmgren, et al., 2003; Scaglia, et al., 2004). Organ dysfunction is caused by energy deficiency, oxygen radical damage and proapoptotic signaling by the damaged
D
mitochondria. Oxygen radical scavengers have a variable efficacy. Novel
TE
mitochondrial protective agents such as bendavia (Brown, et al., 2014; Eirin, et al.,
CE P
2014) may be useful in future to modify the disease course until a definitive therapy to correct the underlying defect is available.
AC
Friedreich's ataxia is an autosomal recessive neurodegenerative disease caused by frataxine mutations. Frataxin is located within the mitochondria and its defects are considered to cause abnormal iron ion accumulation and oxygen radical damage. Cardiac presentation is variable but usually manifests as hypertrophic cardiomyopathy eventually progressing to HF with conduction abnormalities. Oxygen radical scavengers (idebenon) have a variable effect on neurological function while the iron chelator diferipron seems to attenuate cardiac hypertrophy. Approaches to promote frataxine expression by small molecules or histone deacetylase inhibitors are also being promoted but have not yet reached the clinical experimentation stage (Gottesfeld, 2007).
ACCEPTED MANUSCRIPT 36
13. Summary
T
The spectrum of potential approaches to treat inherited disorders, in particular
IP
genetically-determined cardiomyopathies, is rapidly evolving due to an access to
SC R
molecular targets which were not available in the past (Table 1, Figure 1). While gene editing strategies to eliminate the mutation are effective at the bench but have not yet been accomplished at the bedside, techniques to restore the RNA reading frame or to
NU
silence pathogen expression have already arrived at the clinical arena. Chaperones are
MA
used to prevent degradation of abnormal but still functional proteins while other molecules are being introduced to prevent aggregation or enhance the clearance of
D
protein deposits. Absence of protein may be managed by viral gene delivery or
TE
protein therapy. Enzyme replacement therapy is already a reality for a series of metabolic diseases. The progress in molecular biology, triggered by knowledge of the
CE P
gene defect and analysis of the secondary responses ameliorates the understanding of the molecular pathways involved in disease pathophysiology. This helps generate
AC
small molecules and pharmaceuticals targeting the key events occurring in the malfunctioning element of the sick organ. Finally, improved symptomatic medical care and phenotype-based evidence-based medicine developed to treat common disorders, is being applied to inherited cardiomyopathies, offering a greater choice of available options for every one of these rare conditions (Figure 1). Introducing a novel therapy for a rare disease is typically hampered by the difficulty of enrolling an adequate number of patients for a clinical trial to demonstrate an unequivocal benefit of the new medicine. Ethical issues such as administration of placebo to critically ill subjects, as well as delaying drug registration until obtaining an unequivocal proof of efficacy, have to be dealt with sense but with
ACCEPTED MANUSCRIPT 37
determination. Drugs used for rare genetic diseases are often designated Orphan Drugs necessitating huge resource allocation for a small subgroup of patients and for
T
protracted/lifelong periods of care. It is the role of the regulatory authorities to define
IP
the population which is expected to benefit most from any given intervention and the
SC R
reasonable threshold for cost-efficacy. Some patients are always discouraged by such restrictions but their hope has to be mobilized to motivate participation in clinical
MA
14. Conflict of Interest Statement
NU
trials facilitating the optimization of care and new drug development.
References
TE
15.
D
The authors declare that there are no conflicts of interest.
AC
CE P
Abozguia, K., Elliott, P., McKenna, W., Phan, T. T., Nallur-Shivu, G., Ahmed, I., et al. (2010). Metabolic modulator perhexiline corrects energy deficiency and improves exercise capacity in symptomatic hypertrophic cardiomyopathy. Circulation, 122, 1562-1569. Adabag, A. S., Kuskowski, M. A., & Maron, B. J. (2006). Determinants for clinical diagnosis of hypertrophic cardiomyopathy. Am J Cardiol, 98, 1507-1511. Ammash, N. M., Seward, J. B., Bailey, K. R., Edwards, W. D., & Tajik, A. J. (2000). Clinical profile and outcome of idiopathic restrictive cardiomyopathy. Circulation, 101, 24902496. Arad, M., Maron, B. J., Gorham, J. M., Johnson, W. H., Jr., Saul, J. P., Perez-Atayde, A. R., et al. (2005). Glycogen storage diseases presenting as hypertrophic cardiomyopathy. N Engl J Med, 352, 362-372. Ashe, K. M., Budman, E., Bangari, D. S., Siegel, C. S., Nietupski, J. B., Wang, B., et al. (2015). Efficacy of enzyme and substrate reduction therapy with a novel antagonist of glucosylceramide synthase for Fabry disease. Mol Med. Axelsson, A., Iversen, K., Vejlstrup, N., Ho, C., Norsk, J., Langhoff, L., et al. (2015). Efficacy and safety of the angiotensin II receptor blocker losartan for hypertrophic cardiomyopathy: the INHERIT randomised, double-blind, placebo-controlled trial. Lancet Diabetes Endocrinol, 3, 123-131. Bai, R., Di Biase, L., Shivkumar, K., Mohanty, P., Tung, R., Santangeli, P., et al. (2011). Ablation of ventricular arrhythmias in arrhythmogenic right ventricular dysplasia/cardiomyopathy: arrhythmia-free survival after endo-epicardial substrate based mapping and ablation. Circ Arrhythm Electrophysiol, 4, 478-485. Bartel, R. L., Cramer, C., Ledford, K., Longcore, A., Parrish, C., Stern, T., et al. (2012). The Aastrom experience. Stem Cell Res Ther, 3, 26. Baudenbacher, F., Schober, T., Pinto, J. R., Sidorov, V. Y., Hilliard, F., Solaro, R. J., et al. (2008). Myofilament Ca2+ sensitization causes susceptibility to cardiac arrhythmia in mice. J Clin Invest, 118, 3893-3903.
ACCEPTED MANUSCRIPT 38
AC
CE P
TE
D
MA
NU
SC R
IP
T
Berk, J. L., Suhr, O. B., Obici, L., Sekijima, Y., Zeldenrust, S. R., Yamashita, T., et al. (2013). Repurposing diflunisal for familial amyloid polyneuropathy: a randomized clinical trial. JAMA, 310, 2658-2667. Bhuiyan, M. S., Pattison, J. S., Osinska, H., James, J., Gulick, J., McLendon, P. M., et al. (2013). Enhanced autophagy ameliorates cardiac proteinopathy. J Clin Invest, 123, 52845297. Bodin, K., Ellmerich, S., Kahan, M. C., Tennent, G. A., Loesch, A., Gilbertson, J. A., et al. (2010). Antibodies to human serum amyloid P component eliminate visceral amyloid deposits. Nature, 468, 93-97. Botto, M., Hawkins, P. N., Bickerstaff, M. C., Herbert, J., Bygrave, A. E., McBride, A., et al. (1997). Amyloid deposition is delayed in mice with targeted deletion of the serum amyloid P component gene. Nat Med, 3, 855-859. Brown, D. A., Hale, S. L., Baines, C. P., del Rio, C. L., Hamlin, R. L., Yueyama, Y., et al. (2014). Reduction of early reperfusion injury with the mitochondria-targeting peptide bendavia. J Cardiovasc Pharmacol Ther, 19, 121-132. Burkett, E. L., & Hershberger, R. E. (2005). Clinical and genetic issues in familial dilated cardiomyopathy. J Am Coll Cardiol, 45, 969-981. Caforio, A. L., Mahon, N. G., Baig, M. K., Tona, F., Murphy, R. T., Elliott, P. M., et al. (2007). Prospective familial assessment in dilated cardiomyopathy: cardiac autoantibodies predict disease development in asymptomatic relatives. Circulation, 115, 76-83. Cardoso, I., Martins, D., Ribeiro, T., Merlini, G., & Saraiva, M. J. (2010). Synergy of combined doxycycline/TUDCA treatment in lowering Transthyretin deposition and associated biomarkers: studies in FAP mouse models. J Transl Med, 8, 74. Castano, A., Helmke, S., Alvarez, J., Delisle, S., & Maurer, M. S. (2012). Diflunisal for ATTR cardiac amyloidosis. Congest Heart Fail, 18, 315-319. Choi, A. M., Ryter, S. W., & Levine, B. (2013). Autophagy in human health and disease. N Engl J Med, 368, 651-662. Chung, E. S., Leon, A. R., Tavazzi, L., Sun, J. P., Nihoyannopoulos, P., Merlino, J., et al. (2008). Results of the Predictors of Response to CRT (PROSPECT) trial. Circulation, 117, 2608-2616. Clayton, N. P., Nelson, C. A., Weeden, T., Taylor, K. M., Moreland, R. J., Scheule, R. K., et al. (2014). Antisense Oligonucleotide-mediated Suppression of Muscle Glycogen Synthase 1 Synthesis as an Approach for Substrate Reduction Therapy of Pompe Disease. Mol Ther Nucleic Acids, 3, e206. Cleland, J. G., Teerlink, J. R., Senior, R., Nifontov, E. M., Mc Murray, J. J., Lang, C. C., et al. (2011). The effects of the cardiac myosin activator, omecamtiv mecarbil, on cardiac function in systolic heart failure: a double-blind, placebo-controlled, crossover, doseranging phase 2 trial. Lancet, 378, 676-683. Coelho, T., Adams, D., Silva, A., Lozeron, P., Hawkins, P. N., Mant, T., et al. (2013). Safety and efficacy of RNAi therapy for transthyretin amyloidosis. N Engl J Med, 369, 819-829. Coelho, T., Maia, L. F., Martins da Silva, A., Waddington Cruz, M., Plante-Bordeneuve, V., Lozeron, P., et al. (2012). Tafamidis for transthyretin familial amyloid polyneuropathy: a randomized, controlled trial. Neurology, 79, 785-792. Coppini, R., Ferrantini, C., Yao, L., Fan, P., Del Lungo, M., Stillitano, F., et al. (2013). Late sodium current inhibition reverses electromechanical dysfunction in human hypertrophic cardiomyopathy. Circulation, 127, 575-584. Coutu, P., Bennett, C. N., Favre, E. G., Day, S. M., & Metzger, J. M. (2004). Parvalbumin corrects slowed relaxation in adult cardiac myocytes expressing hypertrophic cardiomyopathy-linked alpha-tropomyosin mutations. Circ Res, 94, 1235-1241.
ACCEPTED MANUSCRIPT 39
AC
CE P
TE
D
MA
NU
SC R
IP
T
Cruz, F. M., Sanz-Rosa, D., Roche-Molina, M., Garcia-Prieto, J., Garcia-Ruiz, J. M., Pizarro, G., et al. (2015). Exercise Triggers ARVC Phenotype in Mice Expressing a Disease-Causing Mutated Version of Human Plakophilin-2. J Am Coll Cardiol, 65, 1438-1450. D'Souza R, S., Levandowski, C., Slavov, D., Graw, S. L., Allen, L. A., Adler, E., et al. (2014). Danon disease: clinical features, evaluation, and management. Circ Heart Fail, 7, 843-849. Dellefave, L., & McNally, E. M. (2010). The genetics of dilated cardiomyopathy. Curr Opin Cardiol, 25, 198-204. Dhandapany, P. S., Sadayappan, S., Xue, Y., Powell, G. T., Rani, D. S., Nallari, P., et al. (2009). A common MYBPC3 (cardiac myosin binding protein C) variant associated with cardiomyopathies in South Asia. Nat Genet, 41, 187-191. Diegoli, M., Grasso, M., Favalli, V., Serio, A., Gambarin, F. I., Klersy, C., et al. (2011). Diagnostic work-up and risk stratification in X-linked dilated cardiomyopathies caused by dystrophin defects. J Am Coll Cardiol, 58, 925-934. Dou, Y., Arlock, P., & Arner, A. (2007). Blebbistatin specifically inhibits actin-myosin interaction in mouse cardiac muscle. Am J Physiol Cell Physiol, 293, C1148-1153. Duboc, D., Meune, C., Lerebours, G., Devaux, J. Y., Vaksmann, G., & Becane, H. M. (2005). Effect of perindopril on the onset and progression of left ventricular dysfunction in Duchenne muscular dystrophy. J Am Coll Cardiol, 45, 855-857. Duda, M. K., O'Shea, K. M., Tintinu, A., Xu, W., Khairallah, R. J., Barrows, B. R., et al. (2009). Fish oil, but not flaxseed oil, decreases inflammation and prevents pressure overload-induced cardiac dysfunction. Cardiovasc Res, 81, 319-327. Ebelt, H., Jungblut, M., Zhang, Y., Kubin, T., Kostin, S., Technau, A., et al. (2007). Cellular cardiomyoplasty: improvement of left ventricular function correlates with the release of cardioactive cytokines. Stem Cells, 25, 236-244. Eirin, A., Williams, B. J., Ebrahimi, B., Zhang, X., Crane, J. A., Lerman, A., et al. (2014). Mitochondrial targeted peptides attenuate residual myocardial damage after reversal of experimental renovascular hypertension. J Hypertens, 32, 154-165. Elliott, P., Andersson, B., Arbustini, E., Bilinska, Z., Cecchi, F., Charron, P., et al. (2008). Classification of the cardiomyopathies: a position statement from the European Society Of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J, 29, 270-276. Elliott, P. M., Anastasakis, A., Borger, M. A., Borggrefe, M., Cecchi, F., Charron, P., et al. (2014). 2014 ESC Guidelines on diagnosis and management of hypertrophic cardiomyopathy: the Task Force for the Diagnosis and Management of Hypertrophic Cardiomyopathy of the European Society of Cardiology (ESC). Eur Heart J, 35, 27332779. Fabritz, L., Fortmuller, L., Yu, T. Y., Paul, M., & Kirchhof, P. (2012). Can preload-reducing therapy prevent disease progression in arrhythmogenic right ventricular cardiomyopathy? Experimental evidence and concept for a clinical trial. Prog Biophys Mol Biol, 110, 340-346. Fabritz, L., Hoogendijk, M. G., Scicluna, B. P., van Amersfoorth, S. C., Fortmueller, L., Wolf, S., et al. (2011). Load-reducing therapy prevents development of arrhythmogenic right ventricular cardiomyopathy in plakoglobin-deficient mice. J Am Coll Cardiol, 57, 740750. Fayssoil, A., Nardi, O., Orlikowski, D., & Annane, D. (2010). Cardiomyopathy in Duchenne muscular dystrophy: pathogenesis and therapeutics. Heart Fail Rev, 15, 103-107. Finkel, R. S., Flanigan, K. M., Wong, B., Bonnemann, C., Sampson, J., Sweeney, H. L., et al. (2013). Phase 2a study of ataluren-mediated dystrophin production in patients with nonsense mutation Duchenne muscular dystrophy. PLoS One, 8, e81302.
ACCEPTED MANUSCRIPT 40
AC
CE P
TE
D
MA
NU
SC R
IP
T
Gaffin, R. D., Pena, J. R., Alves, M. S., Dias, F. A., Chowdhury, S. A., Heinrich, L. S., et al. (2011). Long-term rescue of a familial hypertrophic cardiomyopathy caused by a mutation in the thin filament protein, tropomyosin, via modulation of a calcium cycling protein. J Mol Cell Cardiol, 51, 812-820. Gazzerro, E., Assereto, S., Bonetto, A., Sotgia, F., Scarfi, S., Pistorio, A., et al. (2010). Therapeutic potential of proteasome inhibition in Duchenne and Becker muscular dystrophies. Am J Pathol, 176, 1863-1877. Gillmore, J. D., Tennent, G. A., Hutchinson, W. L., Gallimore, J. R., Lachmann, H. J., Goodman, H. J., et al. (2010). Sustained pharmacological depletion of serum amyloid P component in patients with systemic amyloidosis. Br J Haematol, 148, 760-767. Gonzalez-Quesada, C., Cavalera, M., Biernacka, A., Kong, P., Lee, D. W., Saxena, A., et al. (2013). Thrombospondin-1 induction in the diabetic myocardium stabilizes the cardiac matrix in addition to promoting vascular rarefaction through angiopoietin-2 upregulation. Circ Res, 113, 1331-1344. Gottesfeld, J. M. (2007). Small molecules affecting transcription in Friedreich ataxia. Pharmacol Ther, 116, 236-248. Guerriero, C. J., & Brodsky, J. L. (2012). The delicate balance between secreted protein folding and endoplasmic reticulum-associated degradation in human physiology. Physiol Rev, 92, 537-576. Herman, D. S., Lam, L., Taylor, M. R., Wang, L., Teekakirikul, P., Christodoulou, D., et al. (2012). Truncations of titin causing dilated cardiomyopathy. N Engl J Med, 366, 619628. Ho, C. Y., Lakdawala, N. K., Cirino, A. L., Lipshultz, S. E., Sparks, E., Abbasi, S. A., et al. (2015). Diltiazem treatment for pre-clinical hypertrophic cardiomyopathy sarcomere mutation carriers: a pilot randomized trial to modify disease expression. JACC Heart Fail, 3, 180-188. Holmgren, D., Wahlander, H., Eriksson, B. O., Oldfors, A., Holme, E., & Tulinius, M. (2003). Cardiomyopathy in children with mitochondrial disease; clinical course and cardiological findings. Eur Heart J, 24, 280-288. Iwata, M., Yoshikawa, T., Baba, A., Anzai, T., Mitamura, H., & Ogawa, S. (2001). Autoantibodies against the second extracellular loop of beta1-adrenergic receptors predict ventricular tachycardia and sudden death in patients with idiopathic dilated cardiomyopathy. J Am Coll Cardiol, 37, 418-424. Jahns, R., Boivin, V., Hein, L., Triebel, S., Angermann, C. E., Ertl, G., et al. (2004). Direct evidence for a beta 1-adrenergic receptor-directed autoimmune attack as a cause of idiopathic dilated cardiomyopathy. J Clin Invest, 113, 1419-1429. Jahns, R., Schlipp, A., Boivin, V., & Lohse, M. J. (2010). Targeting receptor antibodies in immune cardiomyopathy. Semin Thromb Hemost, 36, 212-218. James, C. A., Bhonsale, A., Tichnell, C., Murray, B., Russell, S. D., Tandri, H., et al. (2013). Exercise increases age-related penetrance and arrhythmic risk in arrhythmogenic right ventricular dysplasia/cardiomyopathy-associated desmosomal mutation carriers. J Am Coll Cardiol, 62, 1290-1297. Jaski, B. E., Jessup, M. L., Mancini, D. M., Cappola, T. P., Pauly, D. F., Greenberg, B., et al. (2009). Calcium upregulation by percutaneous administration of gene therapy in cardiac disease (CUPID Trial), a first-in-human phase 1/2 clinical trial. J Card Fail, 15, 171-181. Joly, M. S., Martin, R. P., Mitra-Kaushik, S., Phillips, L., D'Angona, A., Richards, S. M., et al. (2014). Transient low-dose methotrexate generates B regulatory cells that mediate antigen-specific tolerance to alglucosidase alfa. J Immunol, 193, 3947-3958. Kapplinger, J. D., Landstrom, A. P., Salisbury, B. A., Callis, T. E., Pollevick, G. D., Tester, D. J., et al. (2011). Distinguishing arrhythmogenic right ventricular
ACCEPTED MANUSCRIPT 41
AC
CE P
TE
D
MA
NU
SC R
IP
T
cardiomyopathy/dysplasia-associated mutations from background genetic noise. J Am Coll Cardiol, 57, 2317-2327. Khanna, R., Flanagan, J. J., Feng, J., Soska, R., Frascella, M., Pellegrino, L. J., et al. (2012). The pharmacological chaperone AT2220 increases recombinant human acid alphaglucosidase uptake and glycogen reduction in a mouse model of Pompe disease. PLoS One, 7, e40776. Kishnani, P. S., Corzo, D., Nicolino, M., Byrne, B., Mandel, H., Hwu, W. L., et al. (2007). Recombinant human acid [alpha]-glucosidase: major clinical benefits in infantileonset Pompe disease. Neurology, 68, 99-109. Kizhner, T., Azulay, Y., Hainrichson, M., Tekoah, Y., Arvatz, G., Shulman, A., et al. (2015). Characterization of a chemically modified plant cell culture expressed human alphaGalactosidase-A enzyme for treatment of Fabry disease. Mol Genet Metab, 114, 259267. Kole, R., Krainer, A. R., & Altman, S. (2012). RNA therapeutics: beyond RNA interference and antisense oligonucleotides. Nat Rev Drug Discov, 11, 125-140. Koo, T., & Wood, M. J. (2013). Clinical trials using antisense oligonucleotides in duchenne muscular dystrophy. Hum Gene Ther, 24, 479-488. Kroemer, G. (2015). Autophagy: a druggable process that is deregulated in aging and human disease. J Clin Invest, 125, 1-4. Kubo, T., Gimeno, J. R., Bahl, A., Steffensen, U., Steffensen, M., Osman, E., et al. (2007). Prevalence, clinical significance, and genetic basis of hypertrophic cardiomyopathy with restrictive phenotype. J Am Coll Cardiol, 49, 2419-2426. Kwon, D. H., Kapadia, S. R., Tuzcu, E. M., Halley, C. M., Gorodeski, E. Z., Curtin, R. J., et al. (2008). Long-term outcomes in high-risk symptomatic patients with hypertrophic cardiomyopathy undergoing alcohol septal ablation. JACC Cardiovasc Interv, 1, 432438. Landstrom, A. P., & Ackerman, M. J. (2010). Mutation type is not clinically useful in predicting prognosis in hypertrophic cardiomyopathy. Circulation, 122, 2441-2449; discussion 2450. Ledford, K. J., Zeigler, F., & Bartel, R. L. (2013). Ixmyelocel-T, an expanded multicellular therapy, contains a unique population of M2-like macrophages. Stem Cell Res Ther, 4, 134. Li, C. J., Lv, L., Li, H., & Yu, D. M. (2012). Cardiac fibrosis and dysfunction in experimental diabetic cardiomyopathy are ameliorated by alpha-lipoic acid. Cardiovasc Diabetol, 11, 73. Li, X., Zhang, J., Huang, J., Ma, A., Yang, J., Li, W., et al. (2013). A multicenter, randomized, double-blind, parallel-group, placebo-controlled study of the effects of qili qiangxin capsules in patients with chronic heart failure. J Am Coll Cardiol, 62, 1065-1072. Lieberman, A. P., Puertollano, R., Raben, N., Slaugenhaupt, S., Walkley, S. U., & Ballabio, A. (2012). Autophagy in lysosomal storage disorders. Autophagy, 8, 719-730. Lin, C. Y., Hsu, Y. J., Hsu, S. C., Chen, Y., Lee, H. S., Lin, S. H., et al. (2015). CB1 cannabinoid receptor antagonist attenuates left ventricular hypertrophy and Akt-mediated cardiac fibrosis in experimental uremia. J Mol Cell Cardiol. Lombardi, R., Rodriguez, G., Chen, S. N., Ripplinger, C. M., Li, W., Chen, J., et al. (2009). Resolution of established cardiac hypertrophy and fibrosis and prevention of systolic dysfunction in a transgenic rabbit model of human cardiomyopathy through thiolsensitive mechanisms. Circulation, 119, 1398-1407. Lukas, J., Pockrandt, A. M., Seemann, S., Sharif, M., Runge, F., Pohlers, S., et al. (2015). Enzyme enhancers for the treatment of fabry and pompe disease. Mol Ther, 23, 456464.
ACCEPTED MANUSCRIPT 42
AC
CE P
TE
D
MA
NU
SC R
IP
T
Malik, F. I., Hartman, J. J., Elias, K. A., Morgan, B. P., Rodriguez, H., Brejc, K., et al. (2011). Cardiac myosin activation: a potential therapeutic approach for systolic heart failure. Science, 331, 1439-1443. Marcus, F. I., McKenna, W. J., Sherrill, D., Basso, C., Bauce, B., Bluemke, D. A., et al. (2010). Diagnosis of arrhythmogenic right ventricular cardiomyopathy/dysplasia: proposed modification of the Task Force Criteria. Eur Heart J, 31, 806-814. Maron, B. J., & Maron, M. S. (2013). Hypertrophic cardiomyopathy. Lancet, 381, 242-255. Maron, B. J., Maron, M. S., Wigle, E. D., & Braunwald, E. (2009). The 50-year history, controversy, and clinical implications of left ventricular outflow tract obstruction in hypertrophic cardiomyopathy from idiopathic hypertrophic subaortic stenosis to hypertrophic cardiomyopathy: from idiopathic hypertrophic subaortic stenosis to hypertrophic cardiomyopathy. J Am Coll Cardiol, 54, 191-200. Maron, M. S., Kalsmith, B. M., Udelson, J. E., Li, W., & DeNofrio, D. (2010). Survival after cardiac transplantation in patients with hypertrophic cardiomyopathy. Circ Heart Fail, 3, 574-579. McMurray, J. J., Packer, M., Desai, A. S., Gong, J., Lefkowitz, M. P., Rizkala, A. R., et al. (2014). Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med, 371, 993-1004. Melacini, P., Basso, C., Angelini, A., Calore, C., Bobbo, F., Tokajuk, B., et al. (2010). Clinicopathological profiles of progressive heart failure in hypertrophic cardiomyopathy. Eur Heart J, 31, 2111-2123. Menon, S. C., Michels, V. V., Pellikka, P. A., Ballew, J. D., Karst, M. L., Herron, K. J., et al. (2008). Cardiac troponin T mutation in familial cardiomyopathy with variable remodeling and restrictive physiology. Clin Genet, 74, 445-454. Merkulov, S., Chen, X., Chandler, M. P., & Stelzer, J. E. (2012). In vivo cardiac myosin binding protein C gene transfer rescues myofilament contractile dysfunction in cardiac myosin binding protein C null mice. Circ Heart Fail, 5, 635-644. Merlini, G., Plante-Bordeneuve, V., Judge, D. P., Schmidt, H., Obici, L., Perlini, S., et al. (2013). Effects of tafamidis on transthyretin stabilization and clinical outcomes in patients with non-Val30Met transthyretin amyloidosis. J Cardiovasc Transl Res, 6, 1011-1020. Mestroni, L., Maisch, B., McKenna, W. J., Schwartz, K., Charron, P., Rocco, C., et al. (1999). Guidelines for the study of familial dilated cardiomyopathies. Collaborative Research Group of the European Human and Capital Mobility Project on Familial Dilated Cardiomyopathy. Eur Heart J, 20, 93-102. Muchir, A., Wu, W., Choi, J. C., Iwata, S., Morrow, J., Homma, S., et al. (2012). Abnormal p38alpha mitogen-activated protein kinase signaling in dilated cardiomyopathy caused by lamin A/C gene mutation. Hum Mol Genet, 21, 4325-4333. Munch, G., Boivin-Jahns, V., Holthoff, H. P., Adler, K., Lappo, M., Truol, S., et al. (2012). Administration of the cyclic peptide COR-1 in humans (phase I study): ex vivo measurements of anti-beta1-adrenergic receptor antibody neutralization and of immune parameters. Eur J Heart Fail, 14, 1230-1239. Nodari, S., Triggiani, M., Campia, U., Manerba, A., Milesi, G., Cesana, B. M., et al. (2011). Effects of n-3 polyunsaturated fatty acids on left ventricular function and functional capacity in patients with dilated cardiomyopathy. J Am Coll Cardiol, 57, 870-879. Obici, L., Cortese, A., Lozza, A., Lucchetti, J., Gobbi, M., Palladini, G., et al. (2012). Doxycycline plus tauroursodeoxycholic acid for transthyretin amyloidosis: a phase II study. Amyloid, 19 Suppl 1, 34-36. Ommen, S. R., Maron, B. J., Olivotto, I., Maron, M. S., Cecchi, F., Betocchi, S., et al. (2005). Long-term effects of surgical septal myectomy on survival in patients with obstructive hypertrophic cardiomyopathy. J Am Coll Cardiol, 46, 470-476.
ACCEPTED MANUSCRIPT 43
AC
CE P
TE
D
MA
NU
SC R
IP
T
Peled, Y., Gramlich, M., Yoskovitz, G., Feinberg, M. S., Afek, A., Polak-Charcon, S., et al. (2014). Titin mutation in familial restrictive cardiomyopathy. Int J Cardiol, 171, 2430. Pena, J. R., Szkudlarek, A. C., Warren, C. M., Heinrich, L. S., Gaffin, R. D., Jagatheesan, G., et al. (2010). Neonatal gene transfer of Serca2a delays onset of hypertrophic remodeling and improves function in familial hypertrophic cardiomyopathy. J Mol Cell Cardiol, 49, 993-1002. Penicka, M., Gregor, P., Kerekes, R., Marek, D., Curila, K., & Krupicka, J. (2009). The effects of candesartan on left ventricular hypertrophy and function in nonobstructive hypertrophic cardiomyopathy: a pilot, randomized study. J Mol Diagn, 11, 35-41. Peters, S., Trummel, M., & Meyners, W. (2004). Prevalence of right ventricular dysplasiacardiomyopathy in a non-referral hospital. Int J Cardiol, 97, 499-501. Quarta, G., Muir, A., Pantazis, A., Syrris, P., Gehmlich, K., Garcia-Pavia, P., et al. (2011). Familial evaluation in arrhythmogenic right ventricular cardiomyopathy: impact of genetics and revised task force criteria. Circulation, 123, 2701-2709. Rapezzi, C., Arbustini, E., Caforio, A. L., Charron, P., Gimeno-Blanes, J., Helio, T., et al. (2013). Diagnostic work-up in cardiomyopathies: bridging the gap between clinical phenotypes and final diagnosis. A position statement from the ESC Working Group on Myocardial and Pericardial Diseases. Eur Heart J, 34, 1448-1458. Richard, P., Charron, P., Carrier, L., Ledeuil, C., Cheav, T., Pichereau, C., et al. (2003). Hypertrophic cardiomyopathy: distribution of disease genes, spectrum of mutations, and implications for a molecular diagnosis strategy. Circulation, 107, 2227-2232. Rickers, C., Wilke, N. M., Jerosch-Herold, M., Casey, S. A., Panse, P., Panse, N., et al. (2005). Utility of cardiac magnetic resonance imaging in the diagnosis of hypertrophic cardiomyopathy. Circulation, 112, 855-861. Rizzo, S., Pilichou, K., Thiene, G., & Basso, C. (2012). The changing spectrum of arrhythmogenic (right ventricular) cardiomyopathy. Cell Tissue Res, 348, 319-323. Ruberg, F. L., Maurer, M. S., Judge, D. P., Zeldenrust, S., Skinner, M., Kim, A. Y., et al. (2012). Prospective evaluation of the morbidity and mortality of wild-type and V122I mutant transthyretin amyloid cardiomyopathy: the Transthyretin Amyloidosis Cardiac Study (TRACS). Am Heart J, 164, 222-228 e221. Saberniak, J., Hasselberg, N. E., Borgquist, R., Platonov, P. G., Sarvari, S. I., Smith, H. J., et al. (2014). Vigorous physical activity impairs myocardial function in patients with arrhythmogenic right ventricular cardiomyopathy and in mutation positive family members. Eur J Heart Fail, 16, 1337-1344. Scaglia, F., Towbin, J. A., Craigen, W. J., Belmont, J. W., Smith, E. O., Neish, S. R., et al. (2004). Clinical spectrum, morbidity, and mortality in 113 pediatric patients with mitochondrial disease. Pediatrics, 114, 925-931. Schlossarek, S., Frey, N., & Carrier, L. (2014). Ubiquitin-proteasome system and hereditary cardiomyopathies. J Mol Cell Cardiol, 71, 25-31. Semsarian, C., Ahmad, I., Giewat, M., Georgakopoulos, D., Schmitt, J. P., McConnell, B. K., et al. (2002). The L-type calcium channel inhibitor diltiazem prevents cardiomyopathy in a mouse model. J Clin Invest, 109, 1013-1020. Shen, J. S., Meng, X. L., Wight-Carter, M., Day, T. S., Goetsch, S. C., Forni, S., et al. (2015). Blocking hyperactive androgen receptor signaling ameliorates cardiac and renal hypertrophy in Fabry mice. Hum Mol Genet, 24, 3181-3191. Shen, Y. T., Malik, F. I., Zhao, X., Depre, C., Dhar, S. K., Abarzua, P., et al. (2010). Improvement of cardiac function by a cardiac Myosin activator in conscious dogs with systolic heart failure. Circ Heart Fail, 3, 522-527.
ACCEPTED MANUSCRIPT 44
AC
CE P
TE
D
MA
NU
SC R
IP
T
Staudt, A., Staudt, Y., Dorr, M., Bohm, M., Knebel, F., Hummel, A., et al. (2004). Potential role of humoral immunity in cardiac dysfunction of patients suffering from dilated cardiomyopathy. J Am Coll Cardiol, 44, 829-836. Sturm, A. C., & Hershberger, R. E. (2013). Genetic testing in cardiovascular medicine: current landscape and future horizons. Curr Opin Cardiol, 28, 317-325. Swope, D., Li, J., & Radice, G. L. (2013). Beyond cell adhesion: the role of armadillo proteins in the heart. Cell Signal, 25, 93-100. Tabib, A., Loire, R., Chalabreysse, L., Meyronnet, D., Miras, A., Malicier, D., et al. (2003). Circumstances of death and gross and microscopic observations in a series of 200 cases of sudden death associated with arrhythmogenic right ventricular cardiomyopathy and/or dysplasia. Circulation, 108, 3000-3005. Tavazzi, L., Maggioni, A. P., Marchioli, R., Barlera, S., Franzosi, M. G., Latini, R., et al. (2008). Effect of n-3 polyunsaturated fatty acids in patients with chronic heart failure (the GISSI-HF trial): a randomised, double-blind, placebo-controlled trial. Lancet, 372, 1223-1230. Tsybouleva, N., Zhang, L., Chen, S., Patel, R., Lutucuta, S., Nemoto, S., et al. (2004). Aldosterone, through novel signaling proteins, is a fundamental molecular bridge between the genetic defect and the cardiac phenotype of hypertrophic cardiomyopathy. Circulation, 109, 1284-1291. Vakifahmetoglu-Norberg, H., Xia, H. G., & Yuan, J. (2015). Pharmacologic agents targeting autophagy. J Clin Invest, 125, 5-13. van der Ploeg, A. T., & Reuser, A. J. (2008). Pompe's disease. Lancet, 372, 1342-1353. van der Zwaag, P. A., van Rijsingen, I. A., Asimaki, A., Jongbloed, J. D., van Veldhuisen, D. J., Wiesfeld, A. C., et al. (2012). Phospholamban R14del mutation in patients diagnosed with dilated cardiomyopathy or arrhythmogenic right ventricular cardiomyopathy: evidence supporting the concept of arrhythmogenic cardiomyopathy. Eur J Heart Fail, 14, 1199-1207. van Rijsingen, I. A., Arbustini, E., Elliott, P. M., Mogensen, J., Hermans-van Ast, J. F., van der Kooi, A. J., et al. (2012). Risk factors for malignant ventricular arrhythmias in lamin a/c mutation carriers a European cohort study. J Am Coll Cardiol, 59, 493-500. van Rijsingen, I. A., van der Zwaag, P. A., Groeneweg, J. A., Nannenberg, E. A., Jongbloed, J. D., Zwinderman, A. H., et al. (2014). Outcome in phospholamban R14del carriers: results of a large multicentre cohort study. Circ Cardiovasc Genet, 7, 455-465. van Til, N. P., Stok, M., Aerts Kaya, F. S., de Waard, M. C., Farahbakhshian, E., Visser, T. P., et al. (2010). Lentiviral gene therapy of murine hematopoietic stem cells ameliorates the Pompe disease phenotype. Blood, 115, 5329-5337. Vrtovec, B., Poglajen, G., Lezaic, L., Sever, M., Domanovic, D., Cernelc, P., et al. (2013). Effects of intracoronary CD34+ stem cell transplantation in nonischemic dilated cardiomyopathy patients: 5-year follow-up. Circ Res, 112, 165-173. Vrtovec, B., Poglajen, G., Sever, M., Lezaic, L., Domanovic, D., Cernelc, P., et al. (2011). Effects of intracoronary stem cell transplantation in patients with dilated cardiomyopathy. J Card Fail, 17, 272-281. Weidemann, F., Niemann, M., Breunig, F., Herrmann, S., Beer, M., Stork, S., et al. (2009). Long-term effects of enzyme replacement therapy on fabry cardiomyopathy: evidence for a better outcome with early treatment. Circulation, 119, 524-529. Welch, E. M., Barton, E. R., Zhuo, J., Tomizawa, Y., Friesen, W. J., Trifillis, P., et al. (2007). PTC124 targets genetic disorders caused by nonsense mutations. Nature, 447, 87-91. Xiao, H., Song, Y., Li, Y., Liao, Y. H., & Chen, J. (2009). Qiliqiangxin regulates the balance between tumor necrosis factor-alpha and interleukin-10 and improves cardiac function in rats with myocardial infarction. Cell Immunol, 260, 51-55.
ACCEPTED MANUSCRIPT 45
AC
CE P
TE
D
MA
NU
SC R
IP
T
Xu, S., Lun, Y., Brignol, N., Hamler, R., Schilling, A., Frascella, M., et al. (2015). Coformulation of a Novel Human alpha-Galactosidase A With the Pharmacological Chaperone AT1001 Leads to Improved Substrate Reduction in Fabry Mice. Mol Ther. Yamazaki, T., Suzuki, J., Shimamoto, R., Tsuji, T., Ohmoto-Sekine, Y., Ohtomo, K., et al. (2007). A new therapeutic strategy for hypertrophic nonobstructive cardiomyopathy in humans. A randomized and prospective study with an Angiotensin II receptor blocker. Int Heart J, 48, 715-724. Yancy, C. W., Jessup, M., Bozkurt, B., Butler, J., Casey, D. E., Jr., Drazner, M. H., et al. (2013). 2013 ACCF/AHA guideline for 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, 62, e147-239. Yeoh, T., Hayward, C., Benson, V., Sheu, A., Richmond, Z., Feneley, M. P., et al. (2011). A randomised, placebo-controlled trial of carvedilol in early familial dilated cardiomyopathy. Heart Lung Circ, 20, 566-573. Young-Gqamana, B., Brignol, N., Chang, H. H., Khanna, R., Soska, R., Fuller, M., et al. (2013). Migalastat HCl reduces globotriaosylsphingosine (lyso-Gb3) in Fabry transgenic mice and in the plasma of Fabry patients. PLoS One, 8, e57631. Zsebo, K., Yaroshinsky, A., Rudy, J. J., Wagner, K., Greenberg, B., Jessup, M., et al. (2014). Long-term effects of AAV1/SERCA2a gene transfer in patients with severe heart failure: analysis of recurrent cardiovascular events and mortality. Circ Res, 114, 101108.
ACCEPTED MANUSCRIPT 46
Figure legend Figure 1: Potential approaches to target inherited cardiomyopathies.
T
Mutation-based approaches try to fix the mutation or ameliorate the defect on the
IP
level of protein translation. Alternatively an attempt may be made to prevent a
SC R
complete loss of a defective, but still functioning protein. Proteins where structural integrity is essential for function (such as enzymes) may need to be replaced. A phenotype-based approach tries to identify specific key molecular mechanisms which
NU
may be targeted by small molecules or overexpression of gene products. The entire
MA
cardiac tissue may be modified by means of cell therapy and extracellular matrix modification. Finally, gradual progress is continuously made in defining evidence-
AC
CE P
TE
D
based therapies for different types of cardiac diseases.
ACCEPTED MANUSCRIPT 47
Table 1: Mechanisms and treatment strategies of main subtypes of inherited cardiomyopathies Type Genetics Principal Current Novel interventions and future mechanisms treatment targets
T
Symptomatic, ICD implantation, stroke prevention, myectomy/septal reduction for persistent symptoms.
IP
Increased Ca2+ sensitivity, energy deficit
SC R
Mostly autosomal dominant: Mutations in genes encoding components of the sarcomere or Zdisc. Significant phenotypic heterogeneity.
Mostly autosomal dominant: results from defects in multiple cellular components with a final common pathway of deranged cardiomyocytes contraction
Variable: decreased Ca2+ sensitivity: disruption of sarcolemmal integrity and ion function; impaired force transmission due to cytoskeletal damage; altered gene expression; apoptosis
Early diagnosis treatment as in other forms of systolic dysfunction.
Symptomatic. Corticosteroids, ACE inhibitors/beta blockers to prevent systolic dysfunction, non-invasive ventilation and removing secretions Antiarrythmics, banning of intense physical activity, ICD implantation
AC
CE P
TE
D
Dilated CM
MA
NU
Hypertrophic CM
Duchenne/Becker Muscular Dystrophy (DMD)
X-linked inheritance: Dystrophin gene mutations
disruption of sarcolemmal and cytoskeletal structure and function
Arrhythmogenic CM
Mutations in the genes that encode for the desmosomal proteins which are responsible for cell to cell adhesion
Impaired adhesion and electrical conductance between adjacent cardiomyocytes
- Ca2+ homeostasis: Blebbistatin, parvalbumin adenoviral delivery of SERCA2a, dialtiazem - Gene transfer: recombinant viral vectors of cMyBPC - Diversion of myocardial substrate utilization: perhexilin, trimetazidine, ranolazine - Preventing fibrosis: Aldosterone/ARB blocakade: Spironolactone, eplerenone, losartan, candesartan - Immune system modulation: immunoabsorption, COR-1 cyclopeptide - N-3 polyunsaturated fatty acids (nPUFA) - Gene-therapy: SERCA2 gene - Intracoronary stem cells transplantation (CD34+ cells) - Tissue repair and regeneration: Ixmyelocel-T - Ca2+ sensitization: levosimendan, pimobendan, EMD 53998, MCI-154 - Cardiac myosin activators: omecamtiv mecarbil - Gene-therapy: Dystrophin or utrophin, SERCA2a - Reading frame restoration: Antisense Oligo-Nucleotide (AON) - ‘Read through’ therapy: ataluren - Proteasome inhibitors
- Wall stress reduction: combining afterload reduction and diuretics - Mineralocorticoid therapy - Wnt / β–catenin signaling pathway - PPAR-ɣ
ACCEPTED MANUSCRIPT 48
TE CE P AC
T
Cardiomyocyte dysfunction and death due to storage, arrested autophagy, energetic deficit and ROS damage
IP
Autosomal recessive, x-linked or maternal (in mitochondrial diseases). Caused by enzyme deficiency or defects in oxidative phosphorylation
Symptomatic treatment for heart failure, arrhythmias and hypotension
- Fibrosis attenuation - Small interference RNA - Prevention of TTR fibril formation: tafamidis meglumine, diflunisal - Removal of TTR amyloid deposits: doxycycline plus TUDCA Clearance of serum amyloid protein (SAP): CPHAP
NU
SC R
Wall stiffening due to intracellular or extracellular substrate accumulation, or myocardial fibrosis
D
Metabolic CM
Mutations in sarcomere genes causing severe fibrosis or gene defects that cause intracellular or extracellular accumulation of various substances (e.g., desmin, glycolipid, iron, amyloid protein)
MA
Restrictive CM
Substrate modification, substrate supplementation, enzyme replacement therapy, antioxidants
- Novel enzyme replacement therapy: PRX-102 - Pharmacological chaperone: Migalastat HCl - Enhancers of pharmacological chaperones: Ambroxol, Rosiglitazone Mitochondrial protectors
ACCEPTED MANUSCRIPT 49
AC
CE P
TE
D
MA
NU
SC R
IP
T
Figure 1