Cardiac myosin inhibitors in hypertrophic cardiomyopathy: gaps in knowledge and future opportunities
Introduction
Hypertrophic cardiomyopathy (HCM) is the most common monogenic cardiovascular disease with an estimated prevalence of at least 1 in 500 (0.2%) (1). HCM patients have increased left ventricular (LV) wall thickness of ≥15 mm (≥13 mm in patients with a family history or pathogenic sarcomere variant), and can have small ventricular size, and normal or hyperdynamic LV ejection fraction (LVEF). Other manifestations may include systolic anterior motion (SAM) of the mitral valve resulting in mitral regurgitation and LV outflow tract (LVOT) obstruction (LVOTO). Obstructive HCM is defined by an LVOT gradient of 30 mmHg or more at rest and/or 50 mmHg or more with provocation (2,3).
LVOTO leads to reduced LV compliance and LV diastolic dysfunction (LVDD) (4). Eventually, LVDD, along with other factors such as increased pulmonary artery pressure, contributes to heart failure (HF) symptoms (5). HCM is an important cause of HF, atrial fibrillation, embolic stroke, and arrhythmic sudden death (6). Sudden cardiac death (SCD) may be the first manifestation of the disease; young patients are at the highest risk of SCD (7,8). Cardiac myosin inhibitors selectively address the pathophysiological mechanisms of HCM. The primary objective of this study was to comprehensively examine the latest research pertaining to therapeutic alternatives for HCM, with a specific focus on cardiac myosin inhibitors.
HCM phenotypes and genomics
There are four phenotypic types of HCM, each characterized by distinct patterns of hypertrophy location: (I) sigmoid type: features a prominent basal septal protuberance, a concave septum, and an ovoid LV cavity, which presents with LVOTO; (II) reverse curvature type: exhibits a convex septum, an increasing LV cavity, typically corresponds to mid-ventricular hypertrophy, and may involve mid-ventricular obstruction and/or apical aneurysm; (III) apical type: characterized by hypertrophy of the apical portion, sometimes extending to the middle segments, resulting in an “ace of spades” shaped cavity, and is more frequently observed in Asia; (IV) neutral type: involves uniform hypertrophy of the septum (9).
Importantly, these phenotypic types may coexist within the same patient or exhibit atypical or mixed localization components. While a greater extent of LV hypertrophy has been associated with younger age and a mitral valve with a higher degree of SAM and outflow obstruction, there is no clear evidence linking hypertrophy extent to symptom severity or gender (10). Thus, planning a successful treatment strategy requires precise diagnosis and phenotyping.
Approximately 60% of HCM patients exhibit a clear familial pattern, typically following an autosomal dominant inheritance with a single gene mutation responsible for the condition (11). The most prevalent causal genes, MYH7 (encodes β-Myosin heavy chain ATPase activity and forces generation) and MYBPC3 (encodes myosin binding protein-C and participates in cardiac contraction) account for around half of familial HCM cases, while TNNT2, TNNI3, and TPM1 mutations are rarer, contributing to less than 10% of cases. Additional genes like ACTC1, MYL2, MYL3, and CSRP3 are also implicated, albeit infrequently (12,13). Most prevalent genes associated with HCM’s physiopathology is demonstrated in Table 1.
Table 1
Gene | Protein encoded | Chromosomal location | Protein function | % HCM caused by pathogenic variants | Pathogenic mechanism | Therapeutic target | References |
---|---|---|---|---|---|---|---|
MYBPC3 | Cardiac myosin-binding protein C | 11p11.2 | Key regulator of cardiac contraction | 50% | Haploinsufficiency leading to sarcomere dysfunction | Yes | (14) |
MYH7 | Cardiac β-myosin heavy chain | 14q11.2 | Contractile velocity of cardiac muscle | 30–35% | Altered contractile function, increased cardiomyocyte size, and reduced the maximum twitch forces | Yes | (15) |
TNNI3 | Cardiac troponin I | 19q13.42 | Inhibits actin-myosin interactions | ~5% | Altered muscle contraction and relaxation dynamics | Potential | (16) |
TNNT2 | Cardiac troponin T | 1q32.1 | Tropomyosin-binding subunit of the troponin complex | ~5% | Weakening of sarcomeric tension | Potential | (17) |
TPM1 | Tropomyosin 1 | 15q22.2 | Stabilizes actin filaments | <3% | Disruption of actin-tropomyosin interaction | Potential | (18) |
MYL2 (MLC-2) | Myosin light chain 2 | 12q24.11 | Regulates cardiac myosin cycling kinetics | <3% | Altered phosphorylation state | Potential | (19) |
MYL3 | Myosin light chain 3 | 3p21.31 | Supports the neck domain and the lever arm with myosin regulatory light chain | <3% | Interfere with binding of calcium or magnesium | Potential | (20) |
ACTC1 | Actin alpha cardiac muscle 1 | 15q14 | Major component of the contractile apparatus | ~1% | Disrupted actin-myosin interaction | Potential | (21) |
CSRP3 | Cysteine and glycine rich protein 3 (muscle LIM protein) | 11p15.1 | Integral role in sarcomeric structure | <1% | Impaired calcium handling | No | (22) |
TNNC1 | Cardiac TnC | 3p21.1 | Binding of calcium to TnC regulate contraction | <1% | Calcium binding abnormalities | Potential | (23) |
ACTN2 | Actinin alpha 2 | 1q42.13 | Help anchor the myofibrillar actin filaments | <1% | Impaired sarcomere stability | No | (24) |
JPH2 | Junctophilin-2 | 20q13.12 | Approximates the plasmalemmal L-type calcium channel and the sarcoplasmic reticulum | Rare | Protein mis-localization, impaired calcium-handling | No | (25) |
This table has been reproduced with permission from BMJ Publishing Group Ltd. This table originally appeared in the article “Cardiovascular genetics: the role of genetic testing in diagnosis and management of patients with hypertrophic cardiomyopathy” by Ahluwalia et al. (26). The use of this table is authorized under the BMJ RightsLink license number 5835380853667. TnC, troponin C; HCM, hypertrophic cardiomyopathy.
Moreover, phenocopies of HCM, such as Noonan syndrome, Anderson-Fabry disease, and Danon disease, can present with various clinical manifestations, including those similar to the phenotypic types of HCM (27). The prevalence of phenocopy conditions is significant (28). These phenocopies are important to consider as they can be distinguished from HCM by their associated genetic and clinical features. For instance, mutations in lysosome-associated membrane protein 2 (LAMP-2) can cause Danon disease, a lethal disease associated with Wolff-Parkinson-White syndrome (29).
Stage-specific therapy
In 2012, Olivotto et al. classified HCM into 4 clinical stages to achieve a systematic clinical staging of the disease: non-hypertrophic HCM, classic phenotype, adverse remodeling, and overt dysfunction (30). The initial stage of addressing HCM involves the prevention of the disease in individuals who carry HCM-causing mutations but have yet to develop the hypertrophic phenotype. This can be accomplished through measures such as genetic testing, familial screening, and the use of pharmaceuticals that target pathways implicated in HCM development.
The second stage is characterized by a fully expressed hypertrophic phenotype, in which hypercontractility predominates without extensive fibrotic replacement and LV is hyperdynamic (EF >65%). The main cause of symptoms in this stage is LVOTO (31). Drugs with negative inotropic effects such as β-blockers, non-dihydropyridine calcium channel blocker (CCB), and disopyramide are commonly used to treat symptoms caused by hypercontractility. However, CCB should be avoided in patients with vasodilator effect symptoms, such as hypotension, dyspnea at rest, and very high resting gradients (e.g., >100 mmHg) (32). For refractory symptoms despite optimal medical therapy, septal reduction therapy (SRT) [surgical myectomy and alcohol septal ablation (ASA)] is considered. Allosteric cardiac myosin inhibitors such as mavacamten are considered treatment options in this stage.
In stage III (adverse remodeling), patients may develop structural abnormalities such as LV fibrosis and worsening diastolic and systolic function in a low-normal range of EF 50–65%, which can predict progression to overt LV systolic dysfunction (33). Replacement fibrosis is a therapeutic target but attempts with anti-fibrotic drugs have been elusive with spironolactone and losartan (34). Perhexiline and ranolazine, anti-anginal medications, have shown promise in reducing the 24-hour ventricular arrhythmic burden and improving angina symptoms, but their ability to mitigate the long-term effects of microvascular ischemia, including fibrosis development, is unknown (35). Mavacamten has shown some promise in reducing N-terminal pro-B-type natriuretic peptide (NT-proBNP) and troponin I in symptomatic non-obstructive HCM patients, but more definite answers are expected from the ongoing Phase 3 ODYSSEY-HCM trial (36).
Stage IV is characterized by overt LV systolic dysfunction (LVEF <50%) seen in 5–8% of HCM patients (37). Optimal guideline-directed therapy for HF should be considered, including sodium-glucose transporter 2 inhibitors, ACE inhibitors and angiotensin II receptor blockers (ARBs), HF-specific β-blockers, spironolactone, loop diuretics, cardiac resynchronization therapy (CRT) (mostly biventricular pacing), and oral anticoagulants [in case of concurrent atrial fibrillation (AF) or an apical aneurysm]. In patients with refractory HF symptoms, options are largely confined to heart transplants (38). Careful follow-up with regular cardiac function assessment and symptoms monitoring is necessary to avoid misrecognition of the disease severity.
Current management
Primary management of HCM includes symptom relief and prevention of adverse events. This may include β-blockers to control symptoms such as dyspnea and angina, as well as CCBs (verapamil or diltiazem) to slow heart rate and prolong LV filling time (Figure 1) (39). Regular monitoring of the heart function to detect any potential complications, such as the development of incident AF or HF is also needed. Up to 15% of HCM patients may develop signs and symptoms of HF despite preserved systolic function. Standard HF therapy should be introduced if LVEF is <50%, including ACE inhibitors, β-blockers, and loop diuretics. CRT and cardiac transplant may be considered in end-stage HCM patients (40,41).
For obstructive HCM, nonvasodilating β-blockers are the most effective agents for the treatment of provokable LVOTO demonstrated in small randomized studies (42). β-blockers should be titrated to maximally tolerated dose in patients with LVOTO until resting heart rate is suppressed (43). Side effects such as fatigue should be carefully investigated to assess an optimal individual dose (39,42). Non-dihydropyridine CCBs such as verapamil and diltiazem can be used in patients who are intolerant to β-blockers (39). Disopyramide, a class IA antiarrhythmic agent, is the most effective drug for resting obstruction through its negative inotropic effect and in association with β-blockers improve symptoms and reduces intraventricular gradients (42,44). Long-term use of disopyramide has been associated with reduced subaortic gradient by 50% and reduced cardiovascular deaths, including arrhythmic sudden death, indicating an anti-arrhythmic effect (45,46).
SRT
SRT encompasses surgical septal myectomy (SM) and ASA, aimed at treating HCM with obstructive symptoms. ASA is an alternative to SM, particularly in older patients or those with significant comorbidities. ASA involves the infusion of alcohol into the septal artery to induce an iatrogenic infarction, thereby reducing septal thickness and LVOTO (47,48).
SRT, surgical SM, or ASA is indicated in patients with an LVOT gradient of more than 50 mmHg, associated with a New York Heart Association (NYHA) class ≥ III or refractory symptoms despite maximal medical therapy (Figure 1) (49). SRT is also considered for patients with less severe LVOT gradients of ≥30 to <50 mmHg who have no explanation of symptoms except HCM (50,51). Obstructive HCM with concomitant mitral valve, chordal and/or papillary muscle abnormalities may require both a myectomy and a mitral valve leaflet reduction or repair particularly if the anterior leaflet is long and causing LVOTO (Figure 1) (52). According to 2024 American Heart Association (AHA)/American College of Cardiology (ACC) guideline for the diagnosis and treatment of patients with HCM, SRT can be performed in obstructive HCM patients with persistent symptoms despite receiving β-blockers or nondihydropyridine CCBs (Class I recommendation) (43).
Apical myectomy may have a role in some patients with apical HCM with no subaortic or intraventricular gradients but develop refractory HF symptoms (53). However, apical myectomy is considered for apical variant. A trans-apical approach has been considered for mid-ventricular variant where transaortic exposure is limited, apical variants, and in patients who have limited LV cavity (54,55). SM complications include pacemaker implantation, particularly in patients with prior right bundle branch block. Low-volume centers have significantly greater odds of all-cause mortality and bleeding complications after myectomy (56).
A study of 5,679 obstructive HCM patients aged >65 years showed SM was associated with lower mortality after 2 years of follow-up and lower need for redo SRT after adjustment of confounding variables (57). Notably, patients who underwent ASA were significantly older and had a higher prevalence of comorbidities. SM was associated with higher 3-month mortality and morbidity. Additionally, both groups had reduced HF readmissions and improved quality of life in the follow-up period compared to the year before the SRT. The study also found that higher-volume centers had lower long-term mortality, but most SRT procedures (70%) were performed in low-volume centers (57). Also, it is important to note that there is reduced access to myectomy and implantable cardioverter-defibrillator (ICD) implantation in minority populations (58).
The data on long-term mortality with a follow-up of 5 years or more in seven studies exhibited higher mortality with ASA compared with myectomy (59). However, surgical myectomy can lead to complications, including atrioventricular block requiring permanent pacemaker implantation, tamponade, stroke, and death, among others, prompting a growing interest in finding an alternative therapy to reduce the need for SRT (60).
Myosin inhibitors
Myosin inhibitors are drugs that dose-dependently inhibit myosin ATPase. This class of drugs reduces the myosin duty ratio (strongly actin-bound state time to total chemo mechanical cycle time of myosin) (61). Thus, these drugs reversibly decrease the number of actin-myosin cross-bridge (Figure 2) reducing hypercontractility seen in HCM, improving myocardial energetics and ventricular compliance (62). Mavacamten, produced by Bristol Myers SquibbTM, New York City, New York, U.S. under the tradename Camzyos, and Aficamten, produced by Cytokinetics®, South San Francisco, California, U.S. are two drugs in this class of medication (63).
Mutations in key sarcomeric genes are responsible for causing HCM, with a notable presence in MYBP3, MYH7, TNNT2, and TNNI3 genes (26). The efficacy of mavacamten in ameliorating cellular manifestations of HCM has been demonstrated for gene variants in MYBPC3 and MYH7, achieved by enhancing the stabilization of myosin super relaxation (64,65). Mavacamten effectively counteracts both excessive contractility and compromises relaxation in the context of MYH7 (cardiac β-myosin heavy chain) mutations. By directly addressing myosin’s disorderly relaxation, it progressively diminishes the amplitude of active tension in simulated MYH7 scenarios. This leads to the deactivation of the propagated impact on thin filament activation within MYH7, thereby rectifying the decelerated relaxation. Importantly, mavacamten has exhibited the ability to reinstate contractile function, regardless of the specific mechanism driving the heightened contractility (66).
Mavacamten, with the chemical name MYK-461, was first introduced in the early 2010s. Mavacamten was granted breakthrough therapy designation by the U.S. Food and Drug Administration (FDA) in 2018 and received FDA approval for the treatment of adults with symptomatic NYHA class II–III obstructive HCM on 28 April 2022, as the first-in-class myosin inhibitor approved for this indication in the U.S. (67). It is well tolerated with minimal side effects including dizziness, syncope, fatigue, exertional dyspnea, chest pain, and flushing (68,69). Studies on cardiac myosin inhibitors are summarized in Table 2.
Table 2
Study | Active drug | Trial design/location | Study population & treatment | Concomitant treatment | Treatment duration | Outcomes |
---|---|---|---|---|---|---|
PIONEER-HCM NCT02842242 (69) | Mavacamten | Open-label, non-randomized, phase II/U.S. | 21 patients, age 18–70 years, symptomatic obstructive HCM diagnosis, resting LVOT gradient ≥30 mgHg and post-exercise peak LVOT gradient ≥50 mmHg | Cohort A (proof of concept): no drug allowed, Cohort B (dose response): β-blocker allowed | 16 weeks | • Change in postexercise peak LVOT gradient from baseline to week 12 |
• Change in dyspnea symptom score from baseline to week 12 | ||||||
• Change in LVEF 2D and 3D, global longitudinal strain, and LV fractional shortening from baseline to week 12 | ||||||
• Plasma pharmacokinetic profile of mavacamten (16 weeks) | ||||||
• Change in postexercise peak LVOT gradient from week 12 to week 16 | ||||||
• Change in pVO2 and VE/VCO2 from baseline to week 12 | ||||||
• Proportion of subjects achieving an LVOT gradient response of postexercise peak gradient | ||||||
MAVERICK-HCM NCT02842242 (70) | Mavacamten | Randomized, double-blind, placebo-controlled, phase II/U.S. | 59 patients, age 18+ years with body weight ≥45 kg with symptomatic non-obstructive HCM and preserved LVEF | Allowed to continue BB or CCB if the dose remained stable for at least 2 weeks before screening and was anticipated to remain unchanged throughout the treatment period | 16 weeks | • Safety and tolerability at week 16 (treatment-emergent adverse event) |
• Week 16: pVO2 change from baseline | ||||||
• Week 16: composite functional endpoint (1.5 mL/kg per min or greater increase in pVO2 and at least one NYHA class reduction or a 3.0 mL/kg per min or greater pVO2 increase without NYHA class worsening) | ||||||
EXPLORER-HCM NCT02842242 (68) | Mavacamten | Randomized, double-blind, placebo-controlled, phase III/global | 251 patients, age >18 years with symptomatic obstructive HCM | Allowed to continue standard HCM medical therapy except disopyramide (for safety reasons), if dosing remained stable for at least 2 weeks before screening and no changes were anticipated during the study | 30 weeks | • 1.5 mL/kg per min or greater increase in pVO2 and at least one NYHA class reduction or a 3.0 mL/kg per min or greater pVO2 increase without NYHA class worsening |
• Change in postexercise LVOT gradient | ||||||
• Change in pVO2 | ||||||
• Change in HCMSQSoB | ||||||
• Change in KCCQ-CSS | ||||||
• Change in NYHA class | ||||||
VALOR-HCM NCT02842242 (71) | Mavacamten | Randomized, double-blind, placebo-controlled, phase III/U.S. | 112 patients, age >18 years and body weight >45 kg diagnosed with symptomatic obstructive HCM. Referred or under active consideration for and willing to have SRT procedure | Allowed to continue HCM medical therapy, if dosing remained stable for at least 2 weeks before screening and no changes were anticipated during the study | 32 weeks | • Change in number of subjects who proceed or remain guideline-eligible for SRT at 16 and 32 weeks |
• Change in NYHA class | ||||||
• Change in KCCQ-CSS | ||||||
• Change in NT-proBNP and troponin | ||||||
• Change in LVOT gradient | ||||||
• Change in diastolic function grade | ||||||
• Change in average E/e’ ratio and indexed left atrial volumes (mL/m2) | ||||||
REDWOOD-HCM NCT02842242 (63) | Aficamten | Randomized, double-blind, placebo-controlled, phase II/North America and Europe | 41 patients with obstructive HCM diagnosis | Required to be on stable doses for >4 weeks before enrollment (beta-blockers, calcium-channel blockers, or ranolazine) | 10 weeks, followed by a 2-week washout period | • Safety and tolerability |
• Change in NT-proBNP | ||||||
• Dose-response on LVOT gradients | ||||||
• Reduction in Valsalva LVOT gradient | ||||||
• Improvement by at least one NYHA class in most patients | ||||||
SEQUOIA-HCM NCT05186818 (72) | Aficamten | Randomized, phase III, double-blind, placebo-controlled trial, 14 countries, including locations in North America, Asia, and Europe | 282 patients with obstructive HCM diagnosis | Patients were followed during the washout period at the end of the trial | 24 weeks, followed during a 4-week | • Change in KCCQ-CSS at 24 weeks |
• Improvement of ≥1 NYHA functional class at 24 weeks | ||||||
• Reduction in Valsalva LVOT gradient at 24 weeks | ||||||
• Change in total duration of septal reduction therapy eligibility during treatment period (days) | ||||||
• Change in total workload during cardiopulmonary exercise testing at 24 weeks (watts) |
HCM, hypertrophic cardiomyopathy; LVOT, left ventricular outflow tract; LVEF, left ventricular ejection fraction; LV, left ventricular; pVO2, peak oxygen consumption; VE/VCO2, ventilatory efficiency/carbon dioxide output ratio; BB, beta-blocker; CCB, calcium channel blocker; NYHA, New York Heart Association; HCMSQSoB, Hypertrophic Cardiomyopathy Symptom Questionnaire Shortness of Breath; KCCQ-CSS, Kansas City Cardiomyopathy Questionnaire Clinical Summary Score; SRT, septal reduction therapy; NT-proBNP, N-terminal pro-B-type natriuretic peptide; E/e’, ratio of transmitral E velocity to the myocardial e’ velocity.
Aficamten has a shorter half-life (75–85 hours vs. 7–9 days in mavacamten) and, as a result, more rapid dose titrations (individualized adjustments), drug washout, and hence has a wider therapeutic window (73,74). Unlike mavacamten, it does not have significant drug-drug interactions and the β-blocker use is not associated with reduced therapeutic effect in patients receiving aficamten (68,72).
Myosin inhibitors in patients with obstructive HCM
The PIONEER-HCM phase 2 proof-of-concept trial found that mavacamten reduced post-exercise LVOT gradients and symptoms while improving exercise capacity in patients with obstructive HCM (69). Based on these promising results, the phase 3 EXPLORER-HCM trial, a multicenter, randomized, double-blind, placebo-controlled trial, was conducted. In this trial, a significantly higher proportion of patients in the mavacamten group met the primary endpoint of improved functional capacity and symptom burden after 30 weeks of treatment when compared to the placebo group. Additionally, a greater number of patients in the mavacamten group experienced symptom improvement by at least one NYHA class. Mavacamten was well-tolerated with a significant reduction in post-exercise LVOT gradient (68). Approximately 6–10% of patients in clinical trials experienced a reversible reduction in LVEF to below 50%, necessitating temporary discontinuation of the therapy (68,70).
VALOR-HCM trial phase 3 randomized trial study showed that mavacamten significantly reduced the need for SRT in the short term. After 16 weeks of treatment, 76.8% of the placebo group still met guideline criteria for SRT or elected to undergo the procedure, while 46 of 56 (82.1%) mavacamten-treated patients improved and became ineligible for SRT based on guidelines (P<0.001). Mavacamten significantly reduced both LVOT gradient at rest and after the Valsalva maneuver. The quality-of-life questionnaire [Kansas City Cardiomyopathy Questionnaire (KCCQ)] showed improvement in the mavacamten group.
The study extended the drug therapy for a maximum of 32 weeks in the mavacamten group, while the placebo group transitioned to receiving dose-blinded mavacamten from weeks 16 to 32. Eighty-eight percent of all patients (both groups) did not meet SRT criteria after week 32. The study was conducted at high-volume referral centers specializing in HCM treatment, known for achieving favorable results in SRT procedures (75). At week 56, after either 56 weeks (original mavacamten group) or 40 weeks of treatment (cross-over group), authors reported a sustained reduction in resting and Valsalva LVOT gradients. Additionally, there was an improvement in the NYHA class of 1 or higher (76). As of 2024, guidelines for management of HCM recommend myosin inhibitors for patients with obstructive HCM who have persistent symptoms due to LVOTO despite taking beta blockers or nondihydropyridine CCBs (Class I recommendation) (43).
The SEQUOIA-HCM Study on aficamten for symptomatic obstructive HCM, which was a phase III, double-blind, randomized, placebo-controlled trial, reported a significant increase in peak oxygen consumption (pVO2) (mean change 1.8 mL/kg/min) compared to placebo (0.0 mL/kg/min) (P<0.001) in aficamten group. Additionally, there were significant improvements in Kansas City Cardiomyopathy Questionnaire Clinical Summary Score (KCCQ-CSS), of at least one NYHA functional class, a reduction in the LVOT gradient after the Valsalva maneuver, and a decrease in the total duration of eligibility for SRT at week 24 of follow-up. There were no significant difference in adverse events between two groups; 3.5% of patients in the aficamten group experienced a transient reduction in LVEF to below 50% and 4.9% of patients in the aficamten group required a per-protocol dose reduction. Of note, patients in the aficamten group with an LVEF below 50% did not experience treatment interruption or a worsening of HF symptoms (72,77). Myosin inhibitors in patients with non-obstructive HCM.
MAVERICK-HCM was a phase II multicenter, exploratory, dose-ranging, double-blind, randomized, placebo-controlled study evaluating mavacamten in non-obstructive HCM. At week 16, participants would have achieved the exploratory composite endpoint if they had an improvement of at least 1.5 mL/kg/min in pVO2 and a reduction of ≥1 NYHA functional class or an improvement of ≥3.0 mL/kg/min in pVO2 with no worsening in NYHA functional class compared to baseline. Mavacamten was associated with reduced NT-proBNP and cTnI levels. The mavacemten group did not show changes in the KCCQ-CSS score, NYHA functional class, pVO2, or the composite endpoint compared with the placebo group. However, this study was likely underpowered to detect clinical benefits.
Ongoing studies
Results of treatment with mavacamten from the EXPLORER-LTE cohort of the MAVA-LTE study showed that 4.3% (10/231) patients had permanent treatment discontinuations due to treatment-emergent adverse event (TEAE). LVEF reduction, cardiac failure and arrest, acute myocardial infarction, muscular weakness, systemic lupus erythematosus, fatigue, bacterial endocarditis, and prolonged corrected QT interval (QTc) were reported TEAEs that led to permanent discontinuation (78). The EXPLORER-HCM patients who participated in the MAVA-LTE study underwent cardiac magnetic resonance imaging, which revealed mild reductions in LV mass index and LVEF between weeks 24 to 96. The myocardial contraction fraction remained unchanged, and LV end-systolic volume index increased (79).
PIONEER-OLE study reported results from 156 weeks of mavacamten treatment, indicating improvements in LVOT gradients, ratio of transmitral E velocity to the myocardial e’ velocity (E/e’), NT-proBNP, NYHA class, and Kansas City Cardiomyopathy Questionnaire Overall Summary Score (KCCQ-OSS). Reductions in left atrial volume index (LAVI) and maximum wall thickness were observed, while no notable changes occurred in posterior wall thickness. They also noted mild reductions in LVEF, which remained within the normal range, and only one patient experienced a reduction of LVEF to less than 50% (80).
REDWOOD-HCM Cohort assessed the safety and tolerability of aficamten in patients with symptomatic non-obstructive HCM (81). The LVEF reversibly dropped at an average rate of 5.5%, and no participant discontinued aficamten. The mean improvement in KCCQ-CSS was 10.6 points and 58% of the patients experienced clinical improvement. After 10 weeks of treatment, there was a significant reduction in high-sensitive cardiac Troponin and NT-proBNP levels by an average of 21% and 55%, respectively.
EXPLORER-CN, a multicenter, phase III, randomized, double-blind, placebo-controlled registration trial has been designed and implemented to evaluate the efficacy and safety of mavacamten in approximately 81 Chinese adults with symptomatic obstructive HCM during a 48-week follow-up period. This study is the first trial that is being conducted outside of Europe and the U.S. (82).
MAPLE-HCM, an ongoing trial sponsored by cytokinetics, intends to compare the efficacy and safety of aficamten with metoprolol succinate in adults with symptomatic HCM and LVOTO in a 24-week follow-up. The primary objective of this study is to assess and compare the changes in pVO2 by cardiopulmonary exercise testing (CPET) in two groups (83).
Gaps in the knowledge
Drop in LVEF and the possibility of obstruction recurrence
As a myosin inhibitor, mavacamten disrupts the formation of myosin-actin bridges and leading to a drop in LVEF (62). However, it is unclear if this drop in EF causes patients to develop HF with reduced LVEF. As a result, the U.S. FDA issued a warning stating that the patient’s clinical status and LVEF should be assessed before and regularly during treatment, and the drug’s dosage should be adjusted accordingly (84). This program falls under the Risk Evaluation and Mitigation Strategy (REMS), which is a critical safety measure implemented by the FDA to ensure that the benefits of certain medications surpass their potential risks. Currently, mavacamten is administered only under the REMS program (85,86).
According to the 2024 guideline for the management of HCM, cardiac myosin inhibitors should be discontinued in patients who develop persistent systolic dysfunction (LVEF <50%) or interrupted and resumed at a lower dose if LVEF improves (43). Nevertheless, according to the EXPLORER-HCM and the MAVERICK-HCM trials, changes in the LVEF are temporary, and patients’ LVEF returned to baseline after mavacamten treatment was interrupted (68,70). In the VALOR-HCM trial, LVEF decreased in two patients, but returned to normal after medication discontinuation (71). In aficamten phase 2 trial, LVEF dropped in two patients, but adjustments were made to the dosage and discontinuation was not necessary (63). This indicates that LVEF drop is often asymptomatic and reversible with discontinuation or dose adjustments. Also, it is unknown whether discontinuing the drug would cause a recurrence of the obstruction, especially in patients who have undergone dose adjustment due to the LVEF drop.
Limited data in underrepresented racial and ethnic populations
Variations in disease presentation and outcomes across different racial and ethnic groups have been increasingly recognized (Figure 3). Black patients with HCM exhibit certain characteristics more frequently than White patients, such as sub-basal and diffuse hypertrophy, greater LV maximal thickness, and mid-ventricular obstruction. However, LVOTO at rest was less common in Blacks. Black patients show pathogenic and likely pathogenic HCM variants in the MYH7 and MBPC3 genes (87). Black patients with HCM are younger at the time of diagnosis and have a higher prevalence of NYHA class III or IV HF at presentation (88). Hispanic patients undergoing SRT experience higher in-hospital mortality compared with White patients receiving this treatment (89).
Assessment of 2,467 patients in the U.S.-based sites of the Sarcomeric Human Cardiomyopathy Registry (SHaRe) from 1989 through 2018 showed black patients with HCM were less likely to undergo SRT compared with White patients. Black patients had lower odds of atrial fibrillation and Black race was independently associated with the development of NYHA class III or IV HF (88).
Length of stay, pacemaker and ICD implantation, acute kidney injury, blood transfusion, hospitalization costs, and discharge disposition were significantly different between different races (89). In contrast, another study assessing the trends in mortality of patients with HCM through 39,200 HCM-related deaths, reported that males, Black patients, and patients ≥75 years of age had higher mortality rates (90). Black patients with HCM have a higher rate of appropriate ICD anti-tachycardia pacing therapy or discharges, despite having similar rates of ICD insertion as White patients for primary prevention. Black patients are more likely to have massive LV hypertrophy and two or more SCD risk factors compared to White patients (87).
Although the U.S. Black population has grown to approximately 47.2 million as of 2021, racially and ethnically diverse patients are frequently underrepresented in large, multi-center studies (91,92). In the Hypertrophic Cardiomyopathy Registry (HCMR), a prospective registry across Europe and North America, only 204 (7.4%) were Black. The study population was composed of 84.4% White individuals (93). In SHaRe, of 4,591 patients with HCM studies between 1960 and December 2016, only 141 (3%) patients were Black. Hospitals from Italy, the Netherlands, the U.S., and Brazil participated in this study (94). Aside from HCMR and SHaRe, other registries on HCM patients include the European Society of Cardiology Registry of Hypertrophic Cardiomyopathy (EURObservational Research Program) and the Japanese Society of Hypertrophic Cardiomyopathy (JHCM) Registry, with the former situated in various European centers while the latter is specifically for Japan (95,96). The number of studies conducted in countries where the majority of the population is non-White (e.g., African countries) is severely limited, which could be due to a lack of financial and human resources or cultural and social constraints (97).
Only 2 (5%) patients in the MAVERIK-HCM trial, 3 (5.4%) patients in the VALOR-HCM trial, and 1 (1%) patient in the EXPLORER-HCM trial were Black patients who received mavacamten (68,71,98). In the REDWOOD-HCM trial, all patients in the aficamten-receiving group were White (63).
Better phenotyping of HCM in black patients, including echocardiography, advanced imaging data, genomics, and social determinants of health, may lead to better stratification of these patients (87,90). As mentioned earlier, machine learning (ML) can play an important role in improving the phenotyping of patients with HCM (99).
Conducting further studies on the use of mavacamten in a diverse population is needed to comprehend the interaction between biological factors (including cytochrome P450 genotypes) and behavioral, cultural, and social determinants of health. This will ensure that the administration of the drug is both safe and effective for all patients (92). Moreover, U.S. FDA will soon require researchers seeking approval for late-stage clinical trials to submit a plan that ensures diversity among trial participants (92,100).
Cost-effectiveness
As a novel and expensive drug, mavacamten’s cost may limit its use, particularly in countries with limited healthcare resources. It is worth noting that mavacamten is a relatively new and expensive drug, with a price reported to be $245.20 per capsule. For each additional quality-adjusted life year (QALY), the estimated cost would be $1.2 million (101). According to the VALOR-HCM study authors, the average cost of SRT is approximately $100,000 in the U.S. (71). For every reduction in death from HCM, mavacamten would cost $5.6 million more than myectomy and $7.0 million more than ASA (102).
While mavacamten shows clinical effectiveness, its cost is significantly higher compared to standard treatments like disopyramide (103). Based on the Institute for Clinical and Economic Review (ICER) study findings, the use of mavacamten, along with standard first-line treatment, was projected to lead to a higher number of QALYs compared to standard treatment alone. Assuming a placeholder cost of $75,000, the incremental cost-effectiveness ratio was much higher than commonly accepted thresholds ($1.2 million/QALY). When compared to disopyramide, the incremental cost per QALY was even higher ($1.5 million/QALY), and mavacamten was found to be inferior to both myectomy and ASA. The study estimated the health benefit price benchmark (HBPB) for mavacamten to be between $12,000 and $15,000 per year, and it is expected that the actual cost-effectiveness of the treatment will depend on its actual price (102). Long-term data for HF hospitalizations and mortality is needed to confidently establish the cost-effectiveness of myosin inhibitors.
Future directions
In future HCM research, prioritizing diversity, sex-specific analyses, and a global perspective is crucial for inclusivity and generalizability. HCM is a complex genetic disease with variations among different ethnic and racial groups, emphasizing the need for expanded research and understanding. Global collaboration, comprehensive registries, and data-sharing initiatives are key to advancing HCM research and improving patient outcomes. Leveraging ML and artificial intelligence (AI) can enhance HCM research by integrating various data sources and identifying biomarkers and personalized treatments. Identifying responders to myosin inhibitors therapy, analyzing real-world data, and exploring selective treatment approaches can provide valuable insights. ML tools can aid in precise phenotyping and patient selection, transforming our understanding of HCM and improving outcomes. To better understand the comparative benefits and risks of myosin inhibitors, such as mavacamten, over myectomy further investigation is warranted. It is crucial to identify patients who would benefit most from myosin inhibitors or myectomy.
Conclusions
The existing literature suggests that myosin inhibitors may provide short-term relief for HCM patients by reducing hypercontractility and improving diastolic dysfunction and myocardial energetics, thus ameliorating symptoms such as dyspnea. Additionally, myosin inhibitors may prevent the progression of HCM by slowing the rate of hypertrophy, leading to a decrease in the need for SRT. The optimal treatment modality for obstructive HCM patients, whether SRT or mavacamten, will depend on various factors, such as the functional status and co-morbidities, and concomitant mitral valve and/or papillary muscle abnormalities, which warrants further investigation.
Mavacamten might be a viable option, particularly for patients who are not eligible for SRT due to anatomical constraints or inaccessibility to specialized procedures. However, comprehensive, large-scale studies from more well-represented populations are imperative to elucidate the long-term efficacy and safety of mavacamten in managing HCM patients in diverse populations.
Acknowledgments
Figure 1, Figure 2, and Figure 3 were created with BioRender (https://www.biorender.com/).
Funding: None.
Footnote
Peer Review File: Available at https://shc.amegroups.com/article/view/10.21037/shc-24-12/prf
Conflicts of Interest: All authors have completed the ICMJE uniform disclosure form (available at https://shc.amegroups.com/article/view/10.21037/shc-24-12/coif). The authors have no conflicts of interest to declare.
Ethical Statement: The authors are accountable for all aspects of the work in ensuring that questions related to the accuracy or integrity of any part of the work are appropriately investigated and resolved.
Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0/.
References
- Marsiglia JD, Pereira AC. Hypertrophic cardiomyopathy: how do mutations lead to disease? Arq Bras Cardiol 2014;102:295-304. [Crossref] [PubMed]
- Tower-Rader A, Kramer CM, Neubauer S, et al. Multimodality Imaging in Hypertrophic Cardiomyopathy for Risk Stratification. Circ Cardiovasc Imaging 2020;13:e009026. [Crossref] [PubMed]
- Elliott PM, Gimeno JR, Tomé MT, et al. Left ventricular outflow tract obstruction and sudden death risk in patients with hypertrophic cardiomyopathy. Eur Heart J 2006;27:1933-41. [Crossref] [PubMed]
- Maron BJ, Desai MY, Nishimura RA, et al. Diagnosis and Evaluation of Hypertrophic Cardiomyopathy: JACC State-of-the-Art Review. J Am Coll Cardiol 2022;79:372-89. [Crossref] [PubMed]
- Maron BJ, Rowin EJ, Udelson JE, et al. Clinical Spectrum and Management of Heart Failure in Hypertrophic Cardiomyopathy. JACC Heart Fail 2018;6:353-63. [Crossref] [PubMed]
- Maron BJ, Maron MS. Hypertrophic cardiomyopathy. Lancet 2013;381:242-55. [Crossref] [PubMed]
- Schmied C, Borjesson M. Sudden cardiac death in athletes. J Intern Med 2014;275:93-103. [Crossref] [PubMed]
- Maron BJ, Rowin EJ, Casey SA, et al. How Hypertrophic Cardiomyopathy Became a Contemporary Treatable Genetic Disease With Low Mortality: Shaped by 50 Years of Clinical Research and Practice. JAMA Cardiol 2016;1:98-105. [Crossref] [PubMed]
- Maron MS, Maron BJ, Harrigan C, et al. Hypertrophic cardiomyopathy phenotype revisited after 50 years with cardiovascular magnetic resonance. J Am Coll Cardiol 2009;54:220-8. [Crossref] [PubMed]
- Llamas-Esperón GA, Llamas-Delgado G. Hypertrophic cardiomyopathy. Proposal for a new classification. Arch Cardiol Mex 2022;92:377-89. [PubMed]
- Greaves SC, Roche AH, Neutze JM, et al. Inheritance of hypertrophic cardiomyopathy: a cross sectional and M mode echocardiographic study of 50 families. Br Heart J 1987;58:259-66. [Crossref] [PubMed]
- Marian AJ, Braunwald E. Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res 2017;121:749-70. [Crossref] [PubMed]
- Morita H, Rehm HL, Menesses A, et al. Shared genetic causes of cardiac hypertrophy in children and adults. N Engl J Med 2008;358:1899-908. [Crossref] [PubMed]
- Suay-Corredera C, Pricolo MR, Herrero-Galán E, et al. Protein haploinsufficiency drivers identify MYBPC3 variants that cause hypertrophic cardiomyopathy. J Biol Chem 2021;297:100854. [Crossref] [PubMed]
- Loiben AM, Chien WM, Friedman CE, et al. Cardiomyocyte Apoptosis Is Associated with Contractile Dysfunction in Stem Cell Model of MYH7 E848G Hypertrophic Cardiomyopathy. Int J Mol Sci 2023;24:4909. [Crossref] [PubMed]
- England J, Loughna S. Heavy and light roles: myosin in the morphogenesis of the heart. Cell Mol Life Sci 2013;70:1221-39. [Crossref] [PubMed]
- Madan A, Viswanathan MC, Woulfe KC, et al. TNNT2 mutations in the tropomyosin binding region of TNT1 disrupt its role in contractile inhibition and stimulate cardiac dysfunction. Proc Natl Acad Sci U S A 2020;117:18822-31. [Crossref] [PubMed]
- Kopylova GV, Berg VY, Kochurova AM, et al. The effects of the tropomyosin cardiomyopathy mutations on the calcium regulation of actin-myosin interaction in the atrium and ventricle differ. Biochem Biophys Res Commun 2022;588:29-33. [Crossref] [PubMed]
- Sheikh F, Lyon RC, Chen J. Functions of myosin light chain-2 (MYL2) in cardiac muscle and disease. Gene 2015;569:14-20. [Crossref] [PubMed]
- Moore JR, Leinwand L, Warshaw DM. Understanding cardiomyopathy phenotypes based on the functional impact of mutations in the myosin motor. Circ Res 2012;111:375-85. [Crossref] [PubMed]
- Vang S, Corydon TJ, Børglum AD, et al. Actin mutations in hypertrophic and dilated cardiomyopathy cause inefficient protein folding and perturbed filament formation. FEBS J 2005;272:2037-49. [Crossref] [PubMed]
- Li X, Lu WJ, Li Y, et al. MLP-deficient human pluripotent stem cell derived cardiomyocytes develop hypertrophic cardiomyopathy and heart failure phenotypes due to abnormal calcium handling. Cell Death Dis 2019;10:610. [Crossref] [PubMed]
- Parvatiyar MS, Landstrom AP, Figueiredo-Freitas C, et al. A mutation in TNNC1-encoded cardiac troponin C, TNNC1-A31S, predisposes to hypertrophic cardiomyopathy and ventricular fibrillation. J Biol Chem 2012;287:31845-55. [Crossref] [PubMed]
- Ranta-Aho J, Olive M, Vandroux M, et al. Mutation update for the ACTN2 gene. Hum Mutat 2022;43:1745-56. [Crossref] [PubMed]
- Landstrom AP, Weisleder N, Batalden KB, et al. Mutations in JPH2-encoded junctophilin-2 associated with hypertrophic cardiomyopathy in humans. J Mol Cell Cardiol 2007;42:1026-35. [Crossref] [PubMed]
- Ahluwalia M, Ho CY. Cardiovascular genetics: the role of genetic testing in diagnosis and management of patients with hypertrophic cardiomyopathy. Heart 2021;107:183-9. [Crossref] [PubMed]
- Keren A, Syrris P, McKenna WJ. Hypertrophic cardiomyopathy: the genetic determinants of clinical disease expression. Nat Clin Pract Cardiovasc Med 2008;5:158-68. [Crossref] [PubMed]
- Ruiz-Guerrero L, Barriales-Villa R. Storage diseases with hypertrophic cardiomyopathy phenotype. Glob Cardiol Sci Pract 2018;2018:28. [Crossref] [PubMed]
- Maron BJ, Roberts WC, Arad M, et al. Clinical outcome and phenotypic expression in LAMP2 cardiomyopathy. JAMA 2009;301:1253-9. [Crossref] [PubMed]
- Olivotto I, Cecchi F, Poggesi C, et al. Patterns of disease progression in hypertrophic cardiomyopathy: an individualized approach to clinical staging. Circ Heart Fail 2012;5:535-46. [Crossref] [PubMed]
- Olivotto I, Maron BJ, Appelbaum E, et al. Spectrum and clinical significance of systolic function and myocardial fibrosis assessed by cardiovascular magnetic resonance in hypertrophic cardiomyopathy. Am J Cardiol 2010;106:261-7. [Crossref] [PubMed]
- Argirò A, Zampieri M, Marchi A, et al. Stage-specific therapy for hypertrophic cardiomyopathy. Eur Heart J Suppl 2023;25:C155-61. [Crossref] [PubMed]
- Marstrand P, Han L, Day SM, et al. Hypertrophic Cardiomyopathy With Left Ventricular Systolic Dysfunction: Insights From the SHaRe Registry. Circulation 2020;141:1371-83. [Crossref] [PubMed]
- Maron MS, Chan RH, Kapur NK, et al. Effect of Spironolactone on Myocardial Fibrosis and Other Clinical Variables in Patients with Hypertrophic Cardiomyopathy. Am J Med 2018;131:837-41. [Crossref] [PubMed]
- Argirò A, Zampieri M, Dei LL, et al. Safety and efficacy of ranolazine in hypertrophic cardiomyopathy: Real-world experience in a National Referral Center. Int J Cardiol 2023;370:271-8. [Crossref] [PubMed]
- U.S. National Library of Medicine. A Study of Mavacamten in Non-Obstructive Hypertrophic Cardiomyopathy (ODYSSEY_HCM). 2022. Available online: https://clinicaltrials.gov/ct2/show/NCT05582395
- Harris KM, Spirito P, Maron MS, et al. Prevalence, clinical profile, and significance of left ventricular remodeling in the end-stage phase of hypertrophic cardiomyopathy. Circulation 2006;114:216-25. [Crossref] [PubMed]
- Rowin EJ, Maron BJ, Carrick RT, et al. Outcomes in Patients With Hypertrophic Cardiomyopathy and Left Ventricular Systolic Dysfunction. J Am Coll Cardiol 2020;75:3033-43. [Crossref] [PubMed]
- Authors/Task Force members. 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 2014;35:2733-79. [Crossref] [PubMed]
- Pasqualucci D, Fornaro A, Castelli G, et al. Clinical Spectrum, Therapeutic Options, and Outcome of Advanced Heart Failure in Hypertrophic Cardiomyopathy. Circ Heart Fail 2015;8:1014-21. [Crossref] [PubMed]
- McMurray JJ, Adamopoulos S, Anker SD, et al. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur Heart J 2012;33:1787-847. [Crossref] [PubMed]
- Nistri S, Olivotto I, Maron MS, et al. β Blockers for prevention of exercise-induced left ventricular outflow tract obstruction in patients with hypertrophic cardiomyopathy. Am J Cardiol 2012;110:715-9. [Crossref] [PubMed]
- Ommen SR, Ho CY, Asif IM, et al. 2024 AHA/ACC/AMSSM/HRS/PACES/SCMR Guideline for the Management of Hypertrophic Cardiomyopathy: A Report of the American Heart Association/American College of Cardiology Joint Committee on Clinical Practice Guidelines. Circulation 2024;149:e1239-311. [Crossref] [PubMed]
- Verlinden NJ, Coons JC. Disopyramide for Hypertrophic Cardiomyopathy: A Pragmatic Reappraisal of an Old Drug. Pharmacotherapy 2015;35:1164-72. [Crossref] [PubMed]
- Coppini R, Ferrantini C, Pioner JM, et al. Electrophysiological and Contractile Effects of Disopyramide in Patients With Obstructive Hypertrophic Cardiomyopathy: A Translational Study. JACC Basic Transl Sci 2019;4:795-813. [Crossref] [PubMed]
- Sherrid MV, Barac I, McKenna WJ, et al. Multicenter study of the efficacy and safety of disopyramide in obstructive hypertrophic cardiomyopathy. J Am Coll Cardiol 2005;45:1251-8. [Crossref] [PubMed]
- Gragnano F, Pelliccia F, Guarnaccia N, et al. Alcohol Septal Ablation in Patients with Hypertrophic Obstructive Cardiomyopathy: A Contemporary Perspective. J Clin Med 2023;12:2810. [Crossref] [PubMed]
- Jahnlová D, Tomašov P, Adlová R, et al. Outcome of patients ≥ 60 years of age after alcohol septal ablation for hypertrophic obstructive cardiomyopathy. Arch Med Sci 2019;15:650-5. [Crossref] [PubMed]
- Maron BJ, Desai MY, Nishimura RA, et al. Management of Hypertrophic Cardiomyopathy: JACC State-of-the-Art Review. J Am Coll Cardiol 2022;79:390-414. [Crossref] [PubMed]
- Ommen SR, Mital S, Burke MA, et al. 2020 AHA/ACC Guideline for the Diagnosis and Treatment of Patients With Hypertrophic Cardiomyopathy: A Report of the American College of Cardiology/American Heart Association Joint Committee on Clinical Practice Guidelines. J Am Coll Cardiol 2020;76:e159-240. [Crossref] [PubMed]
- Authors/Task Force members. 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 2014;35:2733-79. [Crossref] [PubMed]
- Sherrid MV, Balaram S, Kim B, et al. The Mitral Valve in Obstructive Hypertrophic Cardiomyopathy: A Test in Context. J Am Coll Cardiol 2016;67:1846-58. [Crossref] [PubMed]
- Nguyen A, Schaff HV. Surgical Myectomy: Subaortic, Midventricular, and Apical. Cardiol Clin 2019;37:95-104. [Crossref] [PubMed]
- Schaff HV, Brown ML, Dearani JA, et al. Apical myectomy: a new surgical technique for management of severely symptomatic patients with apical hypertrophic cardiomyopathy. J Thorac Cardiovasc Surg 2010;139:634-40. [Crossref] [PubMed]
- Said MS, Quintana E, Salhab KF. Hypertrophic Cardiomyopathy: Surgical Perspectives. In: Said MS. editor. New Insights on Cardiomyopathy. Rijeka: IntechOpen; 2023.
- Kim LK, Swaminathan RV, Looser P, et al. Hospital Volume Outcomes After Septal Myectomy and Alcohol Septal Ablation for Treatment of Obstructive Hypertrophic Cardiomyopathy: US Nationwide Inpatient Database, 2003-2011. JAMA Cardiol 2016;1:324-32. [Crossref] [PubMed]
- Mentias A, Smedira NG, Krishnaswamy A, et al. Survival After Septal Reduction in Patients >65 Years Old With Obstructive Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2023;81:105-15. [Crossref] [PubMed]
- Maron BJ, Dearani JA, Smedira NG, et al. Ventricular Septal Myectomy for Obstructive Hypertrophic Cardiomyopathy (Analysis Spanning 60 Years Of Practice): AJC Expert Panel. Am J Cardiol 2022;180:124-39. [Crossref] [PubMed]
- Bytyçi I, Nistri S, Mörner S, et al. Alcohol Septal Ablation versus Septal Myectomy Treatment of Obstructive Hypertrophic Cardiomyopathy: A Systematic Review and Meta-Analysis. J Clin Med 2020;9:3062. [Crossref] [PubMed]
- Liebregts M, Vriesendorp PA, Mahmoodi BK, et al. A Systematic Review and Meta-Analysis of Long-Term Outcomes After Septal Reduction Therapy in Patients With Hypertrophic Cardiomyopathy. JACC Heart Fail 2015;3:896-905. [Crossref] [PubMed]
- Green EM, Wakimoto H, Anderson RL, et al. A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice. Science 2016;351:617-21. [Crossref] [PubMed]
- Anderson RL, Trivedi DV, Sarkar SS, et al. Deciphering the super relaxed state of human β-cardiac myosin and the mode of action of mavacamten from myosin molecules to muscle fibers. Proc Natl Acad Sci U S A 2018;115:E8143-52. [Crossref] [PubMed]
- Maron MS, Masri A, Choudhury L, et al. Phase 2 Study of Aficamten in Patients With Obstructive Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2023;81:34-45. [Crossref] [PubMed]
- Toepfer CN, Garfinkel AC, Venturini G, et al. Myosin Sequestration Regulates Sarcomere Function, Cardiomyocyte Energetics, and Metabolism, Informing the Pathogenesis of Hypertrophic Cardiomyopathy. Circulation 2020;141:828-42. [Crossref] [PubMed]
- Toepfer CN, Wakimoto H, Garfinkel AC, et al. Hypertrophic cardiomyopathy mutations in MYBPC3 dysregulate myosin. Sci Transl Med 2019;11:eaat1199. [Crossref] [PubMed]
- Margara F, Psaras Y, Wang ZJ, et al. Mechanism based therapies enable personalised treatment of hypertrophic cardiomyopathy. Sci Rep 2022;12:22501. [Crossref] [PubMed]
- Keam SJ. Mavacamten: First Approval. Drugs 2022;82:1127-35. [Crossref] [PubMed]
- Olivotto I, Oreziak A, Barriales-Villa R, et al. Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (EXPLORER-HCM): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020;396:759-69. [Crossref] [PubMed]
- Heitner SB, Jacoby D, Lester SJ, et al. Mavacamten Treatment for Obstructive Hypertrophic Cardiomyopathy: A Clinical Trial. Ann Intern Med 2019;170:741-8. [Crossref] [PubMed]
- Ho CY, Mealiffe ME, Bach RG, et al. Evaluation of Mavacamten in Symptomatic Patients With Nonobstructive Hypertrophic Cardiomyopathy. J Am Coll Cardiol 2020;75:2649-60. [Crossref] [PubMed]
- Desai MY, Owens A, Geske JB, et al. Myosin Inhibition in Patients With Obstructive Hypertrophic Cardiomyopathy Referred for Septal Reduction Therapy. J Am Coll Cardiol 2022;80:95-108. [Crossref] [PubMed]
- Maron MS, Masri A, Nassif ME, et al. Aficamten for Symptomatic Obstructive Hypertrophic Cardiomyopathy. N Engl J Med 2024;390:1849-61. [Crossref] [PubMed]
- Malik FI, Robertson LA, Armas DR, et al. A Phase 1 Dose-Escalation Study of the Cardiac Myosin Inhibitor Aficamten in Healthy Participants. JACC Basic Transl Sci 2022;7:763-75. [Crossref] [PubMed]
- Chuang C, Collibee S, Ashcraft L, et al. Discovery of Aficamten (CK-274), a Next-Generation Cardiac Myosin Inhibitor for the Treatment of Hypertrophic Cardiomyopathy. J Med Chem 2021;64:14142-52. [Crossref] [PubMed]
- Desai MY, Owens A, Geske JB, et al. Dose-Blinded Myosin Inhibition in Patients With Obstructive Hypertrophic Cardiomyopathy Referred for Septal Reduction Therapy: Outcomes Through 32 Weeks. Circulation 2023;147:850-63. [Crossref] [PubMed]
- Desai MY, Owens A, Wolski K, et al. Mavacamten in Patients With Hypertrophic Cardiomyopathy Referred for Septal Reduction: Week 56 Results From the VALOR-HCM Randomized Clinical Trial. JAMA Cardiol 2023;8:968-77. [Crossref] [PubMed]
- Coats CJ, Maron MS, Abraham TP, et al. Exercise Capacity in Patients With Obstructive Hypertrophic Cardiomyopathy: SEQUOIA-HCM Baseline Characteristics and Study Design. JACC Heart Fail 2024;12:199-215. [Crossref] [PubMed]
- American College of Cardiology. Updated cumulative results of treatment with mavacamten from the EXPLORER-LTE cohort of the MAVA-LTE study in patients with obstructive hypertrophic cardiomyopathy. 2022. Available online: https://www.tctmd.com/slide/updated-cumulative-results-treatment-mavacamten-explorer-lte-cohort-mava-lte-study-patients
- Saberi S, Kramer CM, Oreziak A, et al. 96-week cardiac magnetic resonance (CMR) results of treatment with mavacamten from the explorer cohort of the mava-long-term extension (LTE) study in patients (PTS) with obstructive hypertrophic cardiomyopathy (HCM). JACC 2023;81:326. [Crossref]
- Masri A, Lester SJ, Stendahl J, et al. Long-term safety and efficacy of mavacamten in patients (PTS) with symptomatic obstructive hypertrophic cardiomyopathy (HCM): updated results from the PIONEER-OLE study. JACC 2023;81:346. [Crossref]
- Masri A, Sherrid MV, Choudhury L, et al. Aficamten in patients with symptomatic non-obstructive hypertrophic cardiomyopathy (REDWOOD-HCM Cohort 4). JACC 2023;81:609. [Crossref]
- Tian Z, Wang F, Jin W, et al. Study design and rationale of EXPLORER-CN: a phase III, randomised, double-blind, placebo-controlled clinical study to evaluate the efficacy and safety of mavacamten in Chinese adults with symptomatic obstructive hypertrophic cardiomyopathy. BMJ Open 2023;13:e071473. [Crossref] [PubMed]
- U.S. National Library of Medicine. The Purpose of This Study is to Compare the Efficacy and Safety of Aficamten (CK-3773274) Compared With Metoprolol Succinate in Adults With Symptomatic Hypertrophic Cardiomyopathy and Left Ventricular Outflow Tract Obstruction (MAPLE-HCM). 2023. Available online: https://classic.clinicaltrials.gov/ct2/show/NCT05767346
. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/214998s000lbl.pdfU.S. Food and Drug Administration - U.S. Food and Drug Administration. Risk Evaluation and Mitigation Strategies | REMS. 2023. Available online: https://www.fda.gov/drugs/drug-safety-and-availability/risk-evaluation-and-mitigation-strategies-rems
- Bristol Myers Squibb. Understanding the CAMZYOS Risk Evaluation and Mitigation Strategy (REMS) program. Available online: https://www.camzyos.com/taking-camzyos/camzyos-rems#:~:text=In%20some%20patients%2C%20CAMZYOS%20can,safe%20use%20of%20the%20medication
- Arabadjian ME, Yu G, Sherrid MV, et al. Disease Expression and Outcomes in Black and White Adults With Hypertrophic Cardiomyopathy. J Am Heart Assoc 2021;10:e019978. [Crossref] [PubMed]
- Eberly LA, Day SM, Ashley EA, et al. Association of Race With Disease Expression and Clinical Outcomes Among Patients With Hypertrophic Cardiomyopathy. JAMA Cardiol 2020;5:83-91. [Crossref] [PubMed]
- Patlolla SH, Schaff HV, Nishimura RA, et al. Impact of Race and Ethnicity on Use and Outcomes of Septal Reduction Therapies for Obstructive Hypertrophic Cardiomyopathy. J Am Heart Assoc 2023;12:e026661. [Crossref] [PubMed]
- Minhas AMK, Wyand RA, Ariss RW, et al. Demographic and Regional Trends of Hypertrophic Cardiomyopathy-Related Mortality in the United States, 1999 to 2019. Circ Heart Fail 2022;15:e009292. [Crossref] [PubMed]
- Moslimani M, Tamir C, Budiman A, et al. Facts About the U.S. Black Population. 2023 [updated March 2, 2023]. Available online: https://www.pewresearch.org/social-trends/fact-sheet/facts-about-the-us-black-population/
- Ahluwalia M, Onwuanyi A, Agu E, et al. Advocating for a Path to Increase Diversity in Enrollment in Cardiovascular Clinical Trials. JACC Adv 2022;1:100152. [Crossref] [PubMed]
- Neubauer S, Kolm P, Ho CY, et al. Distinct Subgroups in Hypertrophic Cardiomyopathy in the NHLBI HCM Registry. J Am Coll Cardiol 2019;74:2333-45. [Crossref] [PubMed]
- Ho CY, Day SM, Ashley EA, et al. Genotype and Lifetime Burden of Disease in Hypertrophic Cardiomyopathy: Insights from the Sarcomeric Human Cardiomyopathy Registry (SHaRe). Circulation 2018;138:1387-98. [Crossref] [PubMed]
- Charron P, Elliott PM, Gimeno JR, et al. The Cardiomyopathy Registry of the EURObservational Research Programme of the European Society of Cardiology: baseline data and contemporary management of adult patients with cardiomyopathies. Eur Heart J 2018;39:1784-93. [Crossref] [PubMed]
- Kubo T, Takano H, Takayama M, et al. Baseline clinical features in a large-scale registration survey of patient with hypertrophic cardiomyopathy throughout Japan: J-HCM registry study. European Heart Journal 2020;41:ehaa946.2085.
- Zheng X, He Z, Li M, et al. Global research trends of hypertrophic cardiomyopathy from 2000 to 2022: Insights from bibliometric analysis. Front Cardiovasc Med 2023;10:1039098. [Crossref] [PubMed]
- Cremer PC, Geske JB, Owens A, et al. Myosin Inhibition and Left Ventricular Diastolic Function in Patients With Obstructive Hypertrophic Cardiomyopathy Referred for Septal Reduction Therapy: Insights From the VALOR-HCM Study. Circ Cardiovasc Imaging 2022;15:e014986. [Crossref] [PubMed]
- Seastedt KP, Moukheiber D, Mahindre SA, et al. A scoping review of artificial intelligence applications in thoracic surgery. Eur J Cardiothorac Surg 2022;61:239-48. [Crossref] [PubMed]
- U.S. Department of Health and Human Services, Food and Drug Administration, Oncology Center of Excellence (OCE), et al. Diversity Plans to Improve Enrollment of Participants from Underrepresented Racial and Ethnic Populations in Clinical Trials Guidance for Industry, Draft Guidance 2022. Available online: https://www.fda.gov/media/157635/download
- Dalo JD, Weisman ND, White CM. Mavacamten, a First-in-Class Cardiac Myosin Inhibitor for Obstructive Hypertrophic Cardiomyopathy. Ann Pharmacother 2023;57:489-502. [Crossref] [PubMed]
- Wasfy JH, Walton SM, Beinfeld M, et al. Mavacamten for Hypertrophic Cardiomyopathy: Effectiveness and Value; Final Evidence Report and Meeting Summary: Institute for Clinical and Economic Review; 2021 November 16, 2021. Available online: https://icer.org/wp-content/uploads/2021/04/ICER_HCM_Final_Evidence_Report_111621.pdf
- Beinfeld M, Wasfy JH, Walton S, et al. Mavacamten for hypertrophic cardiomyopathy: effectiveness and value. J Manag Care Spec Pharm 2022;28:369-75. [Crossref] [PubMed]
Cite this article as: Rahimi M, Ahluwalia M, Sulague RM, Kpodonu J. Cardiac myosin inhibitors in hypertrophic cardiomyopathy: gaps in knowledge and future opportunities. Shanghai Chest 2024;8:20.