Hypertrophic cardiomyopathy (HCM) is common and frequently encountered in various clinical settings.1 Traditionally, HCM has been thought of as a monogenic disease but, due to frequent genotype-negative evaluations, a polygenic component has been proposed and continues to be evaluated.2 HCM is characterised by enhanced cardiac actin–myosin cross-bridge cycling, leading to hypercontractility, myocardial hypertrophy, myocardial fibrosis and diastolic dysfunction.3 HCM with obstructive physiology (oHCM) is the more common phenotype, occurring in up to two-thirds of patients, and results from the combination of hypercontractility, hypertrophy, a small left ventricular (LV) cavity and abnormal mitral valve leaflet length and subvalvular apparatus leading, most commonly, to LV outflow tract (LVOT) obstruction.1,3 HCM has a wide range of phenotypic presentations, including apical HCM, mid-ventricular HCM and septal variant with LVOT obstruction (here termed oHCM).
This narrative review explores traditional and emerging pharmacological and invasive interventions for oHCM, with an emphasis on the latter.
Pharmacological Therapies
Pharmacological therapies are the preferred initial therapy in symptomatic oHCM patients. For decades, β-blockers, calcium channel blockers (CCBs) and disopyramide were mainstay therapies, based largely on results from small and observational studies (Figure 1 and Table 1).1 The discovery of the super-relaxed state of cardiac myosin and the eventual introduction of cardiac myosin inhibitors (CMIs) as the first sarcomere-selective therapeutic approach in HCM represent pivotal moments in the care of patients with oHCM.4,5 The regulatory approval of the first CMI, mavacamten, for the treatment of symptomatic oHCM has dramatically changed clinical practice.6,7
Traditional Medical Therapies
Non-vasodilating β-blockers and non-dihydropyridine CCBs are recommended as first- and second-line therapies respectively for symptomatic patients with oHCM.1 In cardiomyocytes, β-blockers bind to β-adrenoceptors and CCBs inhibit L-type calcium channels, decreasing calcium entry during action potentials and subsequently reducing calcium release by the sarcoplasmic reticulum.8,9 A decrease in the amount of calcium available for binding troponin C subsequently leads to negative inotropy (Figure 1).9 In addition, β-blockers and CCBs act on receptors in cardiac nodal tissue to prolong diastolic filling (Figure 1 ).8,9 Collectively, the increase in LV filling and stroke volume has the potential to reduce the LVOT gradient and symptoms in oHCM patients. However, studies evaluating the physiological and symptomatic effects of these agents in oHCM are lacking. In fact, the recommendation for these agents as first-line therapy is based primarily on long-standing clinical use and expert consensus.1 Available studies examining their outcomes report variable degrees of LVOT reduction, with a substantial proportion of patients classified as non-responders.10–19 To date, there have been no adequately powered randomised and placebo-controlled multicentre clinical trials of these agents.
Disopyramide is a sodium channel blocker with negative inotropic effects that is recommended for oHCM patients with refractory symptoms despite the use of β-blockers and/or CCBs (Figure 1).1 Disopyramide exerts negative inotropic effects by reducing the calcium transient amplitude, thereby lowering calcium-mediated myofilament activation and decreasing force generation.20–22 This occurs through disopyramide’s combined inhibition of peak and late sodium currents, L-type calcium currents, delayed rectifier potassium channels and selective cardiac ryanodine receptors (RyRs).20 Disopyramide also shortens the action potential and stabilises RyRs, thereby reducing early and delayed afterdepolarisations, but there is limited evidence that it can mitigate arrhythmias in HCM.20–22 Although there are some data supporting its symptomatic benefit, the use of disopyramide in clinical practice is limited by inconsistent efficacy and side effects such as QTc prolongation, drug–drug interactions, systemic anticholinergic symptoms and tachyphylaxis.21,22
Although these conventional therapies can potentially improve symptoms for some patients, they do not directly target the pathophysiological mechanisms underlying HCM. Consequently, these treatments neither modify the natural progression of the disease nor consistently achieve symptom relief.
Cardiac Myosin Inhibitors
The 2024 multisociety guidelines recommend CMIs as an alternative to disopyramide and invasive septal reduction therapy for oHCM patients with symptoms despite treatment with β-blockers and/or CCBs.1 CMIs are the first drug class designed to directly target the underlying pathophysiological mechanisms of HCM and the only HCM therapy with data from multiple placebo-controlled phase III clinical trials.23–27
The development of CMIs reflects the field’s growing understanding of myosin–actin interactions that contribute to the pathophysiology of oHCM. Myosin heavy chain heads shift between two states depending on the presence of ATP: a super-relaxed (SRX) state and a disordered-relaxed (DRX) state. The SRX state is characterised by myosin heads that are ‘off’ and unable to engage in actin–myosin cross-bridging. Meanwhile, in the DRX state, one of the myosin heads becomes available for binding. In HCM, a key mechanism driving aberrant sarcomere contraction is destabilisation of the SRX state.4 This increases myosin–actin cross-bridge formation and increases sarcomere force generation to produce hypercontractility.5 By selectively inhibiting cardiac myosin ATPase activity to stabilise the SRX state, CMIs decrease excessive myosin–actin cross-bridge formation to reduce hypercontractility and decrease LVOT obstruction to address symptoms (Figure 1 and Table 1).4,28 Our perspective on the clinical impact of CMIs is largely shaped by recent trials on mavacamten and aficamten.
Mavacamten
Mavacamten is the first and only commercially available CMI that is approved in North America, Asia, Europe, Australia and South America for the treatment of HCM with New York Heart Association (NYHA) functional class II–III symptoms.29 Mavacamten monitoring programs are dictated by the respective health and regulatory authorities. In the US, mavacamten is approved under a risk mitigation program. Mavacamten is started at a dose of 5 mg or lower with monitoring via an echocardiogram every 4 weeks, and the dose cannot be uptitrated the first 12 weeks.30 Subsequently, if the dose is stable, then the echocardiogram can be spaced out to every 12 weeks.30 However, whenever the dose is changed, an ad hoc 4-week echocardiogram is required.30 Any new decrease in LV ejection fraction (LVEF) below 50% requires at least a temporary interruption in therapy, with reinitiation of mavacamten at the next lower dose when LVEF recovers on a subsequent echocardiogram.30
Several randomised controlled trials support the efficacy of mavacamten.23,26,31 The phase III EXPLORER-HCM trial (NCT03470545) demonstrated that mavacamten can improve exercise capacity, reduce LVOT gradient and alleviate symptoms.23 A second phase III trial, VALOR-HCM (NCT04349072), showed that mavacamten therapy reduces the need for invasive septal reduction therapy in most patients.26 Substudies of EXPLORER-HCM showed favourable LV remodelling using imaging, with mavacamten contributing to significant positive myocardial remodelling effects, including an improvement in LV mass index, maximum LV wall thickness and left atrial volume index, which have the potential to reduce long-term cardiovascular events.24,32–34 Longer-term studies are needed to determine whether these findings translate to improved clinical outcomes.
Importantly, adverse events associated with mavacamten are mitigated through structured monitoring programs implemented in both clinical trials and commercial use. Permanent discontinuation due to mavacamten-related effects, such as LVEF <50%, heart failure, or prolonged QTc, has occurred in fewer than 5% of patients.23,25,26,35–39 However, mavacamten’s half-life and potential safety are dependent on metaboliser status.40 In addition, the relationship between mavacamten dose (pharmacokinetics) and its effect on LVEF (pharmacodynamics) is inconsistent, which leads to an unpredictable degree of LVEF reduction.36,41 This appears to be related to cumulative exposure to mavacamten. For example, the rate of developing LVEF <50% in EXPLORER-HCM was 5.6%, whereas the rate in its open-label extension was 8.7% even though that population included placebo-to-mavacamten crossover.39 Reassuringly, most patients with interrupted mavacamten therapy were able to restart mavacamten at the next lower dose, and the incidence of clinical heart failure in conjunction with LVEF <50% was low.23,26,31,36,38,42 In VALOR-HCM, which used echocardiogram-guided titration of mavacamten without monitoring of drug levels, up to 13.8% of patients had LVEF <50%, including two patients with LVEF <30% and one death that occurred in close proximity to such events.26 Furthermore, the rate of new-onset and recurrent AF is emerging as a potential side effect of mavacamten, and has thus become another important metric to monitor. Although AF is common in oHCM, and is expected to occur at a rate of 2–3% per year, the rates of new-onset AF seen with longer-term follow-up of mavacamten in the MAVA-LTE (NCT03723655) and VALOR-HCM trials (7.8% and 10.2%, respectively) are higher than expected and require further investigation.25,26,43
Recent data from the commercial use of mavacamten show somewhat similar conclusions from the clinical trials and their long-term extension studies.6,7,42 However, these analyses are fraught with bias towards patients who continue and restart mavacamten and frequent under-reporting of adverse events due to the lack of a requirement to submit data upon mavacamten discontinuation.42,44 Finally, the drug–drug interaction that can occur with mavacamten use requires significant monitoring and resources, and partly explains why the US Food and Drug Administration’s (FDA) risk mitigation program requires frequent pharmacy involvement.30
Aficamten
Aficamten is the second-in-class CMI. Aficamten binds to a distinct site compared to mavacamten and is currently under regulatory review for the indication of symptomatic oHCM.45 Compared with mavacamten, aficamten has a shorter half-life of 3.4 days and no significant drug–drug interactions.45
The pivotal phase III trial SEQUOIA-HCM (NCT05186818) demonstrated the efficacy and safety of aficamten in oHCM.27 In SEQUOIA-HCM, aficamten improved peak oxygen consumption to 1.74 ml/kg/min and over 95% of oHCM patients treated with aficamten experienced a clinically meaningful improvement in one or more of the following study measures: reduction in LVOT gradient, relief in symptom burden, improvement in exercise capacity and improvement in cardiac biomarkers.27,46 In addition, favourable LV remodelling was shown using both echocardiography and cardiac MRI.35
The collective evidence to date shows that aficamten’s properties result in a favourable safety profile. Aficamten studies in oHCM report a low rate (5.3%) of LVEF reduction to <50%, and downtitration rather than discontinuation of aficamten is safe in patients with LVEF 40–49%.27,46,47 In addition, the rate of AF is lower (2.4%) than that seen in VALOR-HCM and MAVA-LTE, but longer-term exposure data are required to enable more definitive conclusions.27
Challenges and Future Directions for Cardiac Myosin Inhibitors
As the use of CMIs continues to expand, several important considerations must be addressed. First, accurate diagnosis and phenotyping of HCM play a crucial role. Current data support the use of CMIs in sarcomeric oHCM. However, their efficacy in non-sarcomeric phenocopies, non-obstructive HCM, or less common variants of oHCM, including patients with apical or mid-ventricular obstruction, is not well studied.47,48 The potential role of CMIs in non-obstructive HCM is currently being investigated in ODYSSEY-HCM (NCT05582395) and the ACACIA-HCM trial (NCT06081894).
Second, there is currently no method to predict LVEF reduction and/or heart failure events. Initial exploratory data suggest that a history of AF and increased baseline index LV end-diastolic volume (>56 ml/m2) are associated with an increased risk of developing LVEF <50%.33 However, more expansive investigations into predictors of adverse outcomes associated with CMIs, especially mavacamten, are needed. A risk-mitigation strategy implemented by the European Medicines Agency involves assessing metaboliser status before initiating mavacamten.49 This approach aims to predict and reduce the likelihood of adverse events caused by excessive drug response or elevated drug levels. In addition, the emerging signal of increased burden of AF on mavacamten is concerning and requires further investigation.39
Finally, several logistical factors may limit the more widespread adoption of CMIs. The current list price of USD$89,500 for a year of treatment of mavacamten in the US is a challenge.50 In addition, the frequency of monitoring required for patients on mavacamten adds another layer of complexity and cost. Moreover, because of the complexities that relate to the use of mavacamten, HCM centres are typically tasked with prescribing it.
Pharmacotherapies in Early Development
CMIs mark a significant advance in the treatment of HCM, but they are still associated with side effects and logistical factors that limit their widespread use. Several novel pharmacotherapies in development, including EDG-7500, gene therapy and sotagliflozin, attempt to address the shortcomings of CMIs and have the potential to transform the field of HCM therapeutics.
EDG-7500 is a first-in-class selective cardiac sarcomere modulator designed to slow the rate of myocardial force generation in early systole and accelerate actin–myosin cross-bridge detachment in early diastole to improve ventricular filling. Unlike other pharmacotherapies, EDG-7500 was designed to preserve cardiac myosin function, and in vitro studies have shown that it does not affect myosin–actin proximity or cross-bridge reattachment rates.51
Data from studies in animal models show that EDG-7500 successfully slows early contraction and increases LV compliance to decrease LVOT obstruction.51–54 A reduction in LV systolic function below normal was not observed across multiple preclinical models, even at the highest exposure.51–54 Subsequently, a phase I study demonstrated that EDG-7500 was well tolerated in healthy subjects for 14 days in both the single ascending dose and multiple ascending dose cohorts.55 EDG-7500 had a half-life of approximately 30 hours, achieving steady-state concentrations within 4 days with once-daily dosing.55 Decreases in LVEF below 50% were not observed, even with increasing doses of EGD-7500 above the predicted target therapeutic exposure.55 CIRRUS-HCM, a multicentre open-label phase II trial, is currently underway to evaluate the safety, tolerability, pharmacokinetics and pharmacodynamics of EDG-7500 in adults with oHCM and non-obstructive HCM.56,57 Unpublished preliminary data from CIRRUS-HCM show promising results, with a reduction in resting and Valsalva LVOT gradient by 67% and 55%, respectively, for three of five patients in the combined 100/200 mg cohorts.56
Sodium–glucose cotransporter (SGLT) inhibitors, particularly the dual SGLT1 and SGLT2 inhibitor sotagliflozin, are another class of medications under investigation for their therapeutic potential in HCM. Sotagliflozin has been shown to reduce adverse cardiovascular events in diabetic patients with heart failure, prompting interest in its role in HCM.58 In HCM, impaired fatty acid uptake and a metabolic shift from fatty acid oxidation (FAO) to glycolysis have been associated with the progression of LV hypertrophy.59,60 Preclinical studies have yielded mixed results regarding the effects of SGLT2 inhibition on cardiac hypertrophy.61,62 Early studies in a murine model of HCM indicate that SGLT2 inhibition increases the glucagon/insulin ratio in cardiomyocytes, thereby increasing FAO and glycolysis–glucose oxidation coupling, which, in turn, attenuates LV hypertrophy and fibrosis.63 In addition, there is some evidence that chronic pressure overload increases SGLT1 expression to generate cardiac hypertrophy and that SGLT1 inhibition can reverse pathological hypertrophy and LV failure.64 Conversely, a recent study suggests SGLT1 downregulation does not affect hyperglycaemia-related hypertrophy in diabetic hearts.63 These discrepancies may stem from variations in study designs and animal models, and further investigations are needed to understand the impact of sotagliflozin and other SGLT inhibitors on HCM. The phase III SONATA-HCM (NCT06481891) trial is currently investigating the effects of sotagliflozin on symptoms and function in HCM.65
Finally, gene therapy targeting common mutations in HCM is a rapidly evolving field that holds promise for transforming HCM care. Two strategies being explored are gene replacement and gene editing. Gene replacement involves using a viral vector, commonly adenovirus 9 (AAV9), to introduce functional copies of MYBPC3 DNA. In preclinical studies in mice lacking the MYBPC3 gene, TN-201 treatment reduced heart mass, increased cardiac ejection fraction and improved survival.66 At present, multiple biotechnology companies are studying this therapeutic option. MyPEAK1 (NCT05836259) is a phase Ib/II trial led by Tenaya Therapeutics that aims to study the safety of MYBPC3 gene therapy with TN-201. Preliminary data from three patients showed that TN-201 treatment resulted in increased concentration of TN-201 mRNA and MYBPC protein.67 However, the degree of RNA expression was lower than predicted from preclinical models.68 Although there are no reports of cardiotoxicity, all three patients experienced elevations in liver enzymes without evidence of liver injury, with one patient requiring corticosteroid injection.69 Meanwhile, preclinical studies exploring gene-editing therapies have demonstrated some success using the AAV9 system with cardiac-specific promoters to deliver clustered regularly interspaced short palindromic repeat (CRISPR)–Cas9 adenine base editing to correct the MYH7 c1208G>A (p.Arg403Gln) mutation in mouse models.69–71 These studies showed a wide range of transcript correction (from 26% to 68%) owing to study design and the specific cardiac cell types targeted.69–71
Although initial preclinical data underscore the potential of gene therapies, several challenges limit their use in humans. The delivery method and unintended off-target effects pose a challenge because the unintentional consequences of gene therapies can be hazardous.71 In addition, the body’s immune system may be triggered by the introduction of foreign genetic material.72 In a recent gene therapy trial for Duchenne muscular dystrophy, a patient experienced a severe immune response to the AAV9 vector that ultimately led to their death.72 The adverse event was attributed to the immune response elicited by the viral vector, not the gene-editing technology itself.72
Invasive Therapies
Septal reduction therapies are invasive strategies to relieve LVOT obstruction in patients with oHCM whose symptoms are refractory to medical therapy (Table 2). Current standard-of-care approaches include surgical myectomy and alcohol septal ablation (ASA).1 Robust evidence supports surgical myectomy and ASA as an effective means to reduce septal thickness and LVOT obstruction, thereby improving symptoms in patients with oHCM (Table 2).73,74–77 However, these approaches have notable limitations. Recent advances in minimally invasive techniques, such as radiofrequency ablation and septal scoring along the midline endocardium (SESAME), have expanded the therapeutic landscape.
Patient Selection for Invasive Therapies
The multisociety 2024 HCM guidelines recommend septal reduction therapies (SRT) for patients with resting or provoked LVOT gradient ≥50 mmHg with NYHA class III/IV symptoms despite optimal medical therapies.1 Age and comorbidities are also factored into assessment of surgical candidacy. Myectomy is often favoured in patients with concomitant cardiac conditions requiring invasive intervention, such as papillary muscle disease or valvular pathologies.1,78–80 Skilled surgeons can frequently address multiple pathologies in a single operation with minimally added perioperative risk and potentially lower rates of postoperative complications.81–83 In patients who are deemed not surgical candidates, ASA has traditionally been used to alleviate LVOT obstruction. Advances in targeting alternative coronary branches, including septal perforators not originating from the left anterior descending artery, have expanded its applicability.84 However, ASA is ineffective in patients who have unsuitable coronary artery anatomy, massive LV hypertrophy, severely elevated LVOT gradients and concomitant abnormal mitral valve or papillary muscle anatomy.85–87 Due to the frequency of complete heart block, ASA also has a significant postoperative complication of permanent pacing.76,88 In addition, although these approaches are established and extensively studied in oHCM, they are less well-established in other phenotypes of HCM, such as apical and mid-ventricular HCM.89 Limitations of ASA and the invasive nature of septal myectomy have promoted evaluation of newer invasive techniques that are more robust than ASA but avoid the burden of open-heart surgery.
Radiofrequency Ablation
Radiofrequency ablation (RFA) is a newer innovation in minimally invasive therapies. RFA creates a focal lesion that can achieve a similar reduction in LVOT gradient as ablating a larger amount of myocardium (Table 2).90–92 RFA allows for greater precision compared with alcohol ablation, with decreased risks of conduction system injury and inadequate debulking, as well as better targeting of anatomy that is not confined by the coronary artery anatomy.90–92 Studies on RFA have varied in their approach and use of imaging modalities to ensure precision of the ablation target.93–99
Endocardial RFA of septal hypertrophy (ERASH) is one of the earlier RFA therapies, and involves applying high-frequency currents directly to endocardial tissue of the septum to cause localised tissue necrosis.97,100 Although ERASH did not achieve as large a reduction in septal thickness and LVOT gradient as surgical myectomy, it had a similar effect on symptom improvement.100,101 Percutaneous intramyocardial septal RFA (PIMSRA) is a more recent RFA approach that uses an echocardiography-guided electrode needle inserted transapically into the septal myocardial tissue to deliver high-frequency currents for targeted tissue ablation.94,97 Similar to ERASH, initial studies for PIMSRA have demonstrated success in its ability to reduce septal hypertrophy, decrease LVOT gradient and improve clinical symptoms in the majority of patients.93,96,97,101
The rate of intraoperative and 30-day mortality of RFA across several studies is approximately 1–2%, comparable to overall mortality rates of ASA and surgical myectomy.70–73,75–77,85 Method-specific complications include a paradoxical increase in LVOT gradient due to localised tissue oedema, occurring in up to 9% of patients based on one study of ERASH.100 Pericardial effusion is the most common complication of PIMSRA, likely due to injury of a coronary vein from the transapical approach.93 For both RFA methods, the development of postoperative arrhythmias and the need for pacemaker implantation are less frequent than after ASA, because RFA allows for greater precision when targeting areas of therapeutic effect.92,99 However, based on early studies, compared with myectomy, RFA carries an increased risk of high-grade atrioventricular block requiring pacemaker implantation.91,100 There is insufficient data to compare ventricular and/or atrial tachyarrhythmia rates between RFA and other septal reduction therapies.
Despite the theoretical advantages of RFA due to its targeted approach, its efficacy and safety are not as well studied as surgical myectomy or ASA, and RFA studies generally have smaller cohorts and a shorter follow-up duration.93,95–97,99,100 Data on specific haemodynamic or anatomical traits that predict the success of RFA compared with ASA or surgical myectomy remain limited, complicating the decision-making process among available septal reduction therapies. This knowledge gap underscores the need for further research to compare the efficacy and safety profiles of RFA to standard therapies and to identify patient-specific factors that optimise outcomes.
Septal Scoring Along the Midline Endocardium
SESAME is a recent development in transcatheter therapies.102 SESAME involves using a catheter guided by echocardiography and fluoroscopy to electrosurgically lacerate the myocardium to achieve septal myotomy (Table 2).102 SESAME addresses several shortcomings of ASA and RFA, with decreased risks of geographic miss, inadequate debulking and injury to the conduction system.103,104
The few available studies on SESAME have demonstrated successful reduction of septal thickness, enlargement of the LVOT area and symptom improvement in the majority of patients.102,103 The decline in residual LVOT gradients occurred progressively as septal myotomy continued to widen for at least 1 month postoperatively.103 Although initial results are promising, further research is needed to examine the degree of progressive septal splay over a longer follow-up period, as well as the impact of ablation depth on long-term outcomes.
Complication rates of SESAME are high relative to other septal reduction therapies, which is expected considering the early and evolving experience with this technique. In the largest study on SESAME to date, 30% of 76 patients experienced a major complication, such as significant bleeding, iatrogenic myocardial perforation or the need for a permanent pacemaker.103 The intraoperative mortality rate was 2.6%, which is higher than mortality rates of ASA and myectomy at high-volume centres, but comparable to those at lower-volume centres.78,103–105 SESAME’s high complication rates likely reflect its early stages of development in addition to the technical challenges of the technique. Adequate training of proceduralists will likely be a barrier to its widespread adoption.
Ongoing Developments in Septal Reduction Therapies and Future Directions
One technique in early development is high-intensity focused ultrasound, which is a growing area of interest for oHCM therapeutics that uses ultrasound energy for localised tissue ablation with the potential to generate deeper and larger areas of ablation compared with other minimally invasive septal reduction therapies.106–108 One study on live canines provides proof of concept, but no in-human trials have been published yet.106 Another technique being studied is localised ionising radiation for septal ablation.109 The first-in-human study showed promising outcomes without any serious adverse events reported.109 However, larger trials, such as the NIRA-HOCM (NCT0415316), are needed to establish the efficacy and safety of this technique.
Although the landscape of septal reduction therapies has evolved dramatically in the past few decades, surgical myectomy still offers superior outcomes, especially in patients with greater baseline LVOT gradients, and lower complication rates compared with its less invasive alternatives.75–77,110 However, as minimally invasive techniques continue to be refined to improve target precision and ensure adequate debulking, they could potentially expand access to septal reduction therapies to patients who are not surgical candidates, in addition to offering patients less invasive options.
In the era of CMIs, it is necessary to consider their potential impact on the field of septal reduction therapies. Septal reduction therapies are typically reserved for severely symptomatic patients and do not directly address the underlying pathophysiology of the disease. Moreover, CMIs may offer increased access for patients who are not surgical candidates or do not want invasive therapies, and CMI outcomes are not operator dependent. Given the potential impact of CMIs on this field, further exploration into non-invasive strategies is essential to advance the boundaries of septal reduction therapies.
Conclusion
The past decade has seen substantial advances in HCM therapies with the rise of CMIs and minimally invasive therapies. Although traditional pharmacological treatments remain the first-line therapy for symptomatic patients, this is likely to change as more robust evidence on CMIs accumulates. Emerging options, including sarcomere modulators and non-invasive reduction therapies, aim to address the limitations of existing treatments. Future efforts should focus on areas of unmet need, including treating special populations, expanding access, developing clinical tools to facilitate decision-making and studying the impact of available therapies on the risk of arrhythmia and sudden cardiac death.
