Next Article in Journal
The Intestinal Mechanisms in the Excretion of Pepsinogen, Amylase and Lipase in Coprofiltrate in Women During Pregnancy and the Postpartum Period
Previous Article in Journal
Recent Advances in the Structural Studies of the Proteolytic ClpP/ClpX Molecular Machine
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiac Myosin Inhibitors in the Treatment of Hypertrophic Cardiomyopathy: Clinical Trials and Future Challenges

1
Department of Pharmacokinetics and Therapeutic Drug Monitoring, Pomeranian Medical University, 72 Powstańców Wielkopolskich Avenue, 70-111 Szczecin, Poland
2
Department of Pharmacology, Pomeranian Medical University, 72 Powstańców Wielkopolskich Avenue, 70-111 Szczecin, Poland
*
Author to whom correspondence should be addressed.
Biomolecules 2025, 15(8), 1098; https://doi.org/10.3390/biom15081098
Submission received: 30 May 2025 / Revised: 11 July 2025 / Accepted: 14 July 2025 / Published: 29 July 2025

Abstract

Hypertrophic cardiomyopathy (HCM) is a prevalent and often underdiagnosed genetic cardiac disorder characterized by left ventricular hypertrophy and, in many cases, dynamic left ventricular outflow tract obstruction (LVOTO). The development of cardiac myosin inhibitors (CMIs) represents an emerging therapeutic approach in the pharmacological management of obstructive HCM (oHCM). This review offers an integrated and up-to-date synthesis of the cardiac myosin inhibitor class, with a focus on mavacamten, aficamten, and the broader landscape of emerging agents. It also highlights recent clinical trial outcomes, pharmacokinetic and pharmacogenetic considerations, and potential future directions in therapy. Furthermore, we incorporate the most recent data up to May 2025, including late-breaking trial results and real-world safety findings, aiming to provide clinicians with a practical and comprehensive perspective on this evolving drug class. A narrative review was conducted by systematically searching PubMed, Scopus, Google Scholar, and ClinicalTrials.gov for English-language articles and trials published between January 2016 and May 2025. Keywords included “cardiac myosin inhibitor”, mavacamten”, “aficamten”, “MYK-224”, and “hypertrophic cardiomyopathy.” Inclusion criteria encompassed clinical trials and comprehensive reviews specifically addressing CMIs in cardiac applications. CMIs such as mavacamten and aficamten have demonstrated significant clinical benefits in reducing LVOT gradients, improving exercise capacity, and alleviating symptoms in patients with oHCM. Mavacamten is currently approved for clinical use, while aficamten is in advanced regulatory review. Comparative data suggest potential advantages of aficamten in the onset of action, pharmacokinetic profile, and tolerability. Emerging evidence supports the exploration of CMIs in pediatric populations, heart failure with preserved ejection fraction (HFpEF), and non-obstructive HCM (nHCM), although results are still preliminary. Cardiac myosin inhibitors offer a novel, pathophysiology-targeted approach to managing oHCM. While mavacamten has established efficacy, next-generation agents like aficamten may offer improved safety and versatility. Further long-term studies are needed to clarify their role across broader patient populations.

1. Introduction

According to the guidelines of the European Society of Cardiology (ESC), hypertrophic cardiomyopathy (HCM) is diagnosed when an increased thickness of the left ventricular (LV) wall is present, in the absence of secondary causes (such as hypertension or tachyarrhythmias). In order to diagnose HCM in adult patients, the LV wall thickness should be equal to or higher than 15mm in any myocardial segment. For children, LV wall thickness is required to be greater than two standard deviations (SD) from the predicted mean (z-score more than 2) [1].
From a statistical standpoint, it is estimated that 1 in 500 individuals may be affected by HCM. However, this figure may be underestimated, as only 10–20% of cases are currently identified [2]. Recent data suggest that the prevalence of HCM may be approximately 1 in 200 individuals, highlighting a growing clinical problem. More widespread use of echocardiographic diagnostics in the general population is the primary reason for the increased HCM diagnosis [3,4].
For example, in Poland, it is estimated that approximately 180,000 people are affected by HCM. Data on the prevalence of hypertrophic cardiomyopathy in Poland between 2016 and 2020 revealed a yearly increase in newly diagnosed HCM cases, except in 2020, which is attributed to the COVID-19 pandemic [5].
In addition to secondary causes of hypertrophic cardiomyopathy, two primary etiologies of HCM can be distinguished: genetic mutations and diseases that mimic HCM in imaging studies (e.g., echocardiography and cardiac magnetic resonance), referred to as phenocopies. These phenocopies exhibit imaging features similar to those seen in genetically determined HCM but have different underlying causes. They are most commonly either storage diseases, such as Fabry disease and Danon disease, or infiltrative conditions, such as amyloidosis.
Regarding genetic mutations, HCM is most frequently associated with mutations in genes encoding cardiac sarcomere proteins. The most commonly affected genes are MYH7, MYBPC3 and TNNT2. Mutations in MYH7 and MYBPC3 account for approximately 60–70% of HCM cases. In contrast, mutations in TNNT2 represent around 3–5% [1,6,7]
The treatment of hypertrophic cardiomyopathy is a complex clinical challenge. Current therapeutic algorithms distinguish two main treatment pathways, and the choice between them is closely linked to two key aspects: The severity of heart failure symptoms, classified using the New York Heart Association (NYHA) functional classification [8], and the presence or absence of left ventricular outflow tract obstruction (LVOTO).
The parameter used to assess LVOTO is the pressure gradient across the left ventricular outflow tract (LVOT), as measured by echocardiography. A normal LVOT pressure gradient should not exceed 30 mmHg, whereas a value equal to or greater than 50 mmHg is considered hemodynamically significant, leading to marked impairment of blood flow. This threshold (50 mmHg) serves as a critical decision point in both pharmacological and invasive treatment strategies according to current clinical guidelines. Yet, it is the value of 30 mmHg of LVOT gradient that is the value used to differentiate patients into two clinical subtypes: those with obstructive hypertrophic cardiomyopathy (oHCM), characterized by LVOT obstruction, and those with non-obstructive hypertrophic cardiomyopathy (nHCM), in whom obstruction is absent. European Society of Cardiology guidelines underline that most patients in the range of 30 mmHg to 49 mmHg LVOT gradient can be treated according to the non-obstructive hypertrophic cardiomyopathy pathway. Cardiac myosin inhibitors (CMIs) are currently used exclusively in the treatment of oHCM. The LVOT gradient is highly relevant in the context of heart failure symptoms. Its elevation can lead to worsening of heart failure manifestations, such as fatigue, increased dyspnea, and a significant reduction in exercise capacity, due to impeded delivery of oxygenated blood to peripheral tissues [1,9,10,11].
Treatment of LVOTO is guided by the presence of heart failure symptoms. In symptomatic patients with HCM, the first-line therapy is based on beta-blockers. If symptoms persist, non-dihydropyridine calcium channel blockers (such as verapamil or diltiazem) may be added. Subsequently, cardiac myosin inhibitors or disopyramide can be introduced. If symptoms remain refractory to pharmacotherapy, invasive procedures are recommended, either alcohol septal ablation or surgical myectomy (see Figure 1) [1].
The introduction of a new class of medications, i.e., cardiac myosin inhibitors, is a major breakthrough in the treatment of HCM [12]. This article provides a detailed discussion of CMIs. The emergence of this novel pharmacological option not only enables improved symptom control but also contributes to delaying the need for invasive intervention. The development and clinical benefits of CMIs have been acknowledged by the leading cardiology societies, i.e., the European Society of Cardiology (ESC) and the American College of Cardiology (ACC), both of which have issued positive recommendations for their use, in 2023 and 2024, respectively [1,13,14,15].
Currently, the most widely used CMIs is mavacamten, which was approved by the U.S. Food and Drug Administration (FDA) in 2022 [16] and by the European Medicines Agency (EMA) in 2023 [17]. Other agents in this class currently undergoing clinical investigation include the following: aficamten, which has completed Phase III clinical trials [18], and MYK-224, which is currently in Phase II clinical trials [19]. It is noteworthy that, in parallel with the growing population of individuals affected by HCM, there is a steady increase in the number of clinical trials focused on the pharmacological treatment of this condition [20]. Although several recent reviews have focused primarily on mavacamten or aficamten, the present article provides a broader, comparative overview of the entire class of cardiac myosin inhibitors, including mavacamten, aficamten, and investigational agents such as MYK-224 and MYK-581. This review also incorporates the most up-to-date clinical trial data available as of May 2025. In addition, we highlight important aspects such as CYP2C19 pharmacogenetic considerations, regulatory milestones, and the potential future use of CMIs in non-obstructive HCM, heart failure with preserved ejection fraction, and pediatric populations. By addressing these elements, this review aims to deliver a comprehensive and clinically relevant summary of current evidence and future directions in the pharmacological management of HCM with CMIs.

Physiology of Cardiac Muscle Contraction and Pathophysiology of HCM

Under physiological conditions, cardiac muscle contraction occurs as a result of interactions between myosin filaments (thick filaments) and actin filaments (thin filaments) within a structural unit known as the sarcomere. A key component of myosin is the myosin head, which contains two crucial binding sites: one for actin and one for ATP (adenosine triphosphate).
When ATP binds to the myosin head, it undergoes hydrolysis catalyzed by cardiac myosin ATPase, releasing energy. This energy drives a conformational change in the myosin head, enabling it to bind to actin, forming a cross-bridge. Following the release of inorganic phosphate and then the ADP (adenosine diphosphate), which are products of ATP hydrolysis, the thin filament is pulled toward the center of the sarcomere, resulting in muscle contraction. A new ATP molecule then binds to the myosin head, causing it to detach from actin, allowing the cycle to repeat [21,22]. The basic physiology of cardiac muscle contraction is shown in Figure 2.
It is also important to note that, physiologically, two resting states of myosin can be distinguished: the SRX (super-relaxed) state and the DRX (disordered-relaxed) state. These states differ significantly in their ATP consumption rates: SRX consumes far less ATP compared to DRX (0.003 s−1 vs. 0.03 s−1). Furthermore, the DRX state represents a condition in which the myosin head is primed for activity, i.e., capable of forming cross-bridges and initiating contraction. In contrast, in the SRX state, the myosin head cannot bind to actin, thereby inhibiting contraction [23]. Under pathophysiological conditions, the underlying cause of HCM is often genetic mutations, such as those affecting the β-myosin heavy chain gene (MYH7). This mutation may lead to an increased number of myosin heads in the DRX state, which in turn causes a higher number of cross-bridges between myosin and actin in cardiac sarcomeres [24]. Figure 3 shows the pathophysiological mechanism of HCM. It also includes cardiac myosin inhibitors, which are described more specifically later in this article.
The clinical consequences of the dysfunction include the following: increased myocardial contractility, impaired diastolic relaxation, narrowing of the left ventricular outflow tract (due to increased myocardial fiber mass), and myocardial fibrosis [12,25,26,27].

2. Materials and Methods

2.1. Data Sources

We searched PubMed, Scopus, and Google Scholar to gather relevant literature. We selected a data frame from January 2016 to May 2025. These platforms were chosen simply because they offer a wide range of peer-reviewed biomedical research and are well-established databases in biomedical sciences. We also took a thorough look at ClinicalTrials.gov to find any relevant clinical trials, mainly 3-phase clinical trials for CMIs.

2.2. Search Strategy

The search was performed using a narrative desk review approach. The following keywords and combinations were used: “cardiac myosin inhibitor”, “mavacamten,” “aficamten”, “myosin ATPase”, “hypertrophic cardiomyopathy”, “HCM”, and “hyperertrophic cardiomyopathy treatment”. Boolean operators (AND/OR) were applied to enhance search precision. Only articles published in English were considered. For additional depth, manual screening of reference lists from key articles was performed. The final search was completed on 15 May 2025.

2.3. Inclusion and Exclusion Criteria

Included materials encompassed original research articles, clinical trials, pharmacological studies, and comprehensive review articles specifically addressing cardiac myosin inhibitors. Studies were excluded if they provided only general discussions on sarcomeric proteins without specific reference to pharmacological inhibitors, were editorials, commentaries, or opinion pieces without original data or comprehensive synthesis, or were non-English publications or non-peer-reviewed sources (e.g., blogs or preprints without peer review).

2.4. Article Selection and Quality Assessment

Chosen articles were investigated manually for relevance and quality. Emphasis was placed on peer-reviewed studies from reputable journals with transparent methodology and well-described objectives. Clinical trials were evaluated based on the design phase, registration status, and reported outcomes. No formal scoring system was applied, but a qualitative assessment was conducted to prioritize clinically meaningful publications.

2.5. Bias Assessment

Bias was minimized through the use of multiple databases and a defined inclusion protocol. Critical source analysis was applied to assess the scientific validity, originality, and contextual relevance of each article. These measures aimed to accurately represent the current state of knowledge on cardiac myosin inhibition.

2.6. Transparency and Reproducibility

The methodology employed in this review follows a qualitative narrative approach, ensuring reproducibility through defined search parameters and source documentation. No new experiments, datasets, or protocols were created. All data cited are publicly available or accessible via institutional subscriptions. No ethical approval was required. Generative AI was not used for content generation, data analysis, or synthesis in this manuscript.

3. Cardiac Myosin Inhibitors

Cardiac myosin ATPase inhibitors are more commonly referred to in the scientific literature as cardiac myosin inhibitors. They are small-molecule drugs that act by inhibiting a key enzyme responsible for cardiac muscle contraction, namely cardiac myosin ATPase. This inhibition results in a reduced formation of actin–myosin cross-bridges. Additionally, drugs in this class help maintain myosin in its super-relaxed state (SRX). Collectively, these mechanisms counteract the pathophysiological basis of HCM by decreasing myocardial contractility, improving diastolic function, and reducing left ventricular outflow tract obstruction. These effects translate into clinical benefits for patients, including symptom relief, reduction in the LVOT pressure gradient, and improvement in exercise capacity. Importantly, unlike other drug classes used in HCM, CMIs act not only on symptoms but also target the underlying pathophysiology of the disease [28,29,30].
The primary agents in this class include the following: mavacamten (Camzyos ®), which currently approved by both the FDA [16] and the European Medicines Agency [17]; aficamten (CK-274), which is currently undergoing clinical investigation [18]; MYK-224, which is in Phase II clinical trials [19]; MYK-581, which is currently in preclinical studies [31]; Blebbastatin, which is used for research purposes [32]. All of these compounds are small-molecule drugs [33]. A summary of the compounds classified as cardiac myosin inhibitors is presented in Table 1.

3.1. Mavacamten

Mavacamten, also known as MYK-461, is an allosteric inhibitor of cardiac myosin ATPase and the first approved drug in this class [30], making it also the most extensively studied. The structure of mavacamten is shown in Figure 4, and its molecular weight is 273.33 g/mol [34].
Its mechanism of action, which involves reducing the formation of actin–myosin cross-bridges, has been described previously [30]. From a pharmacokinetic perspective, mavacamten demonstrates high oral bioavailability (>85%), allowing for once-daily oral administration. It has a long half-life, estimated at 6–9 days. Mavacamten is metabolized primarily by two cytochrome P450 isoenzymes: CYP2C19 (≈74%), for which genotyping is recommended by the EMA to identify poor metabolizers [17], and to a lesser extent, CYP3A4 (≈18%) and CYP2C9 (≈8%). Excretion is primarily via the biliary route [35,36]. In preclinical studies on mice, the early administration of mavacamten significantly reduced the progression of hypertrophic cardiomyopathy. Moreover, the same study showed that mavacamten could reverse cardiac hypertrophy in subjects where hypertrophy was already established [37]. In Phase II clinical trials, the PIONEER-HCM study and its long-term extension PIONEER-OLE (n = 13) demonstrated that mavacamten significantly reduced LVOT gradients, improved exercise capacity, and enhanced quality of life [38,39]. In Phase III, the EXPLORER-HCM trial (n = 251) confirmed its clinical efficacy by showing a mean reduction of 37 mmHg in the post-exercise LVOT gradient (compared to 14 mmHg in the placebo group), improvement in peak oxygen consumption (pVO2), an improvement of ≥1 NYHA class in 80% of patients, and a 9.1-point increase in quality-of-life scores versus 5.1 points in the placebo group. Mavacamten demonstrated a safety profile comparable to the placebo [40]. Another clinical trial, The HORIZON-HCM, was conducted on the Japanese population. The study had similar outcomes regarding efficacy and safety of mavacamten when compared to EXPLORER-HCM [41].
The MAVA-LTE trial (ongoing through 2029), a long-term extension of EXPLORER-HCM, aims to assess long-term efficacy and safety. Of the 231 patients completing EXPLORER-HCM, 211 were enrolled in MAVA-LTE. The initial dose was 5 mg, with titration based on ejection fraction and LVOT gradient. Preliminary data show sustained LVOT gradient reduction, left atrial volume reduction (~5.5 mL/m2), which is a marker of improved diastolic function, at week 180, 66.3% of patients were in NYHA class I, a low discontinuation rate (5.6%), with main adverse effects being atrial fibrillation (14.3%), and worsening heart failure (6.1%) [42]. The VALOR-HCM, an important clinical trial regarding mavacamten and its impact on the treatment of HCM, evaluated its potential to delay or prevent invasive procedures (alcohol septal ablation or surgical myectomy) in patients with NYHA class III/IV of HCM already scheduled for such interventions. At week 16, only 17.9% of patients in the mavacamten group still qualified for invasive treatment, offering a significant clinical benefit [14,43]. However, an important limitation of mavacamten is its potential to reduce left ventricular ejection fraction (LVEF). It is not recommended for patients with LVEF < 55%, and echocardiographic monitoring is essential, especially during the initial treatment phase [17,35]. Following the approval of mavacamten by the FDA and EMA, the number of real-world experience studies evaluating this drug has increased. For example, a study conducted by Ramonfaur et al. [44] in a racially diverse population over an 18-month period demonstrated efficacy and safety outcomes consistent with those observed in clinical trials. The study also found that patients with HCM and obesity were less likely to experience symptom improvement with mavacamten. Another real-world study by Lim et al. [45] demonstrated that mavacamten can be a safe and effective treatment in the Korean population diagnosed with oHCM. Acknowledging mavacamten metabolism, it should be avoided or used with special caution at lower doses in patients co-administered with moderate or strong CYP2C19 or CYP3A4 inhibitors/inducers due to its metabolic profile. Because of the polymorphic nature of the gene coding for CYP2C19, patients should be genotyped for the CYP2C19 gene to determine the appropriate dose of mavacamten. Patients with a poor metabolizer phenotype may experience up to a threefold increase in mavacamten exposure, which may lead to an increased risk of systolic dysfunction compared to normal metabolizers. If treatment is initiated before the CYP2C19 phenotype is determined, patients should follow the dosing recommendations for poor metabolizers until their CYP2C19 status is known [17,35,46]. This issue has been especially relevant in Asian populations, where the prevalence of poor CYP2C19 metabolism is higher, as observed in the HORIZON-HCM trial (Japanese population) [39], as well as in Chinese [47] and Korean cohorts [45]. In summary, regarding CYP2C19 polymorphism, patients who are poor metabolizers should initiate treatment at a lower dose than normal metabolizers (2.5 mg vs. 5 mg), with subsequent dose adjustment based on echocardiographic imaging [17,46]. The most common adverse events during mavacamten treatment were dizziness, syncope, and atrial fibrillation (AF) [35,36,40]. Due to the occurrence of AF during mavacamten therapy, there is a need for clinical guidelines regarding the safety of anticoagulants and antiarrhythmic (AAD) drugs. A review by Ricci et al. [48] addressed this issue, noting that commonly used anticoagulants, such as vitamin K antagonists and non-vitamin K oral anticoagulants (NOACs), are generally considered safe. However, given the lack of large-scale studies, patients should be closely monitored for potential complications when combining anticoagulation therapy with mavacamten. Regarding AADs, it is important to consider replacing flecainide or propafenone with alternative agents due to their proarrhythmic properties in the HCM population [48,49]. Additionally, mavacamten use has been associated with other arrhythmias, such as premature atrial contractions (PACs), premature ventricular contractions (PVCs), and supraventricular tachycardia (SVT). These arrhythmias were observed primarily at the start of treatment, but the correlation was transient and not present over the long term [50]. To conclude, mavacamten has shown beneficial effects not only on clinical symptoms but also on echocardiographic parameters, with a favorable safety profile. It also offers a non-invasive alternative in treatment pathways.

3.2. Aficamten

Aficamten, currently under FDA review [51], is a structural analog of mavacamten with a molecular weight equal to 337.4 g/mol (see Figure 5) [52].
Overall, aficamten’s mechanism of action is similar to mavacamten, but it exhibits greater selectivity for the cardiac myosin active site [53]. Thanks to its pharmacokinetic properties, the steady-state drug concentration is set within 2 weeks of treatment, which translates to a faster clinical effect, including NYHA class improvement and LVOT gradient reduction, and is typically achieved in this period of two weeks of treatment [54], compared to 6–9 weeks for mavacamten [40]. Pharmacokinetically, aficamten is metabolized by multiple cytochrome P450 enzymes, reducing the risk of drug–drug interactions and the need for pharmacogenetics testing. It has a half-life of ~4 days, reaches a steady state in ~2 weeks, shows less plasma protein binding (compared to mavacamten), and is renally excreted (~32%), unlike mavacamten, which undergoes minimal renal elimination [55,56,57]. The SEQUOIA-HCM Phase III trial (n = 282) demonstrated that aficamten improved pVO2 by +1.8 mL/kg/min, reduced resting LVOT gradient by 34.7mmHg, improved NYHA class in 59% of patients, and enhanced overall quality of life [54,58,59]. Earlier trials also showed improvements in LVOT gradient and NYHA class, though some did not reach statistical significance [60,61,62]. In terms of safety, aficamten appears better tolerated than mavacamten. The most common adverse effects of aficamten are dizziness and LVEF reduction below 50%, which occurred in only 3.5% of patients (n = 144) after 28 weeks vs. 8.7% in the EXPLORE-HCM study with mavacamten (n = 180) [39,52,54,63]. A safety meta-analysis by Davis et al. [64] found that atrial fibrillation occurred in 4.1/100 patients with aficamten vs. 11.5/100 for mavacamten. LVEF-related discontinuation occurred only once for aficamten (n = 188), and was transient; in contrast, mavacamten was discontinued completely in eight patients and temporarily in 28 out of 414 [64]. In the context of the cardiac conduction system, aficamten did not prolong the QTc interval and did not influence heart rate [65]. Moreover, regarding aficamten pharmacokinetics and safety, it is worth mentioning that patients with mild to moderate kidney and/or liver impairment may not require dose adjustment, as the pharmacokinetics of aficamten were not significantly different from those observed in individuals with normal liver and kidney function [66]. These findings suggest pharmacokinetic and pharmacodynamic differences between aficamten and mavacamten. Overall, it seems that aficamten acts faster, is cleared more rapidly, and may offer a safer profile, though larger studies are needed. FDA approval is expected by late 2025, and EMA approval by mid-2026 [51,64,67].

3.3. Experimental and Emerging CMIs

Blebbistatin is a prototype myosin II inhibitor widely used in experimental research, including studies of muscle physiology and cancer models, due to its fluorescent properties [32,68]. Despite its value as a research tool, its utility in vivo is limited by phototoxicity, poor solubility, and cytotoxicity [69]. To overcome these drawbacks, several derivatives, such as para-nitroblebbistatin and para-aminoblebbistatin, have been developed, offering improved solubility and reduced phototoxicity while retaining inhibitory activity. However, these analogs still exhibit residual cytotoxicity and genotoxicity, limiting their suitability for preclinical development [68,70]. While blebbistatin itself is not clinically viable, its molecular scaffold remains a valuable template for the development of future isoform-selective myosin inhibitors.
Building on these early insights, newer cardiac myosin inhibitors have progressed into clinical development. MYK-224, a structural analog of mavacamten, is currently in Phase II trials. It is designed with a shorter half-life to offer greater dosing flexibility, although detailed preclinical data remain limited [71]. The MERCUTIO trial evaluating MYK-224 in obstructive HCM was terminated for business-related reasons [72]; however, the AURORA-HFpEF trial remains ongoing, assessing its safety and tolerability in patients with heart failure with preserved ejection fraction (HFpEF) [31]. Notably, mavacamten has also shown promise in this population: in the EMBARK-HFpEF trial (n = 30), treatment led to a 13% reduction in troponin I and a 26% reduction in NT-proBNP levels, suggesting potential efficacy in HFpEF [73]. In parallel, MYK-581, another CMI analog, is currently in preclinical development and has demonstrated inhibition of HCM progression in animal models [74].

4. Discussion

The future of pharmacotherapy for hypertrophic cardiomyopathy using cardiac myosin inhibitors remains filled with open questions. Furthermore, ongoing studies continue to investigate new agents in this class, such as the aforementioned MYK-224 or MYK-581.
To determine the definitive role of CMIs in oHCM, comparative trials with beta-blockers are necessary. The MAPLE-HCM trial, comparing aficamten vs. metoprolol, is completed, but results are pending [75]. However, valuable insights have already emerged from post hoc analyses of existing trial data. In a detailed subgroup analysis of the EXPLORER-HCM and MAVA-LTE trials, Wheeler et al. [76] investigated the impact of background beta-blocker use on the efficacy of mavacamten. Their findings revealed that while the improvement in peak oxygen consumption (VO2) was attenuated in patients on beta-blockers due to chronotropic incompetence, the overall clinical benefit of mavacamten was preserved. Key endpoints such as reductions in LVOT gradients, improvements in NYHA functional class, and decreases in NT-proBNP levels were largely unaffected by beta-blocker use. Notably, the data raise the hypothesis that in certain patients, beta-blocker withdrawal or dose reduction could potentially unmask additional benefits of CMI therapy, particularly in terms of exercise capacity. Nevertheless, given the multifactorial indications for beta-blockers in HCM (e.g., arrhythmia management, coronary disease, and comorbid hypertension), this approach must be individualized. The upcoming results from head-to-head trials such as MAPLE-HCM are therefore essential to establish whether CMIs could supplant beta-blockers as first-line pharmacotherapy in symptomatic obstructive HCM.
The role of CMIs in nHCM is under investigation in the ACACIA-HCM trial (aficamten). This is the first aficamten study in the nHCM population [77]. Regarding aficamten, an interesting study was conducted by Masri et al. [78], in which they evaluated the combined efficacy of aficamten administered concurrently with disopyramide, a drug with a well-established role in the treatment of hypertrophic cardiomyopathy. In this study, researchers divided patients from clinical trials of aficamten (REDWOOD-HCM, SEQUOIA-HCM, and FOREST-HCM) into four groups: patients receiving continuous treatment with both aficamten and disopyramide; patients receiving both aficamten and disopyramide, with subsequent discontinuation of disopyramide; patients receiving disopyramide and the placebo; patients receiving disopyramide and aficamten, with subsequent discontinuation of aficamten. The results were very positive for aficamten. The study showed that the combination of disopyramide and aficamten did not demonstrate superiority compared to aficamten alone. Moreover, the discontinuation of aficamten (which left patients treated only with disopyramide) resulted in a higher left ventricular outflow tract gradient, recurrence of symptoms, and NT-proBNP levels returning to baseline levels (before starting aficamten treatment). Thus, the use of aficamten alone may be sufficient to alleviate symptoms of obstructive HCM, potentially allowing for the discontinuation of disopyramide when patients begin treatment with aficamten. In addition, these results may lead to a reduced number of drugs in the patient’s treatment, which offers benefits such as a lower risk of drug interactions and fewer treatment complications. [79,80]. Mavacamten has already been evaluated in nHCM through the MAVERICK-HCM trial [81], which showed improved biomarkers (NT-proBNP and troponin I), but no significant improvements in pVO2 or NYHA class, which was limited by the small sample size (n = 59), short duration (16 weeks), and reliance on surrogate endpoints. To address these limitations, the ODYSSEY-HCM trial (n = 580) was launched in 2022, with extended follow-up through 2029. However, interim results were negative, showing no improvement in pVO2 or KCCQ-23 CSS scores, leading to early termination. Investigators suggested that nHCM may represent a distinct disease requiring different pharmacologic strategies [82,83]. A study by Amr et al. [84] also addressed a similar issue, but from a different perspective. Among patients with obstructive HCM, only a portion of this population was included in clinical trials of cardiac myosin inhibitors (CMIs) during phases 2 and 3. According to the authors’ observations, the most common reason for exclusion was an insufficient LVOT gradient. This highlights an important issue, particularly for patients with a resting LVOT gradient in the 30–49 mmHg range [1,84]. For example, in the SEQUOIA-HCM trial (a clinical trial for aficamten), the inclusion criteria were a resting LVOT gradient ≥30 mmHg and <50 mmHg and a post-Valsalva (provoked) LVOT gradient ≥50 mmHg [13]. In contrast, the EXPLORER-HCM trial (a clinical trial for mavacamten) used stricter criteria, requiring a peak LVOT gradient of at least 50 mmHg (either at rest or after Valsalva maneuver/exercise) [40]. These differences raise an important question: from what LVOT gradient should CMIs be considered? Currently, the general consensus is that 50 mmHg serves as the primary threshold. Another key area of CMIs is their pediatric use (<18 years). The CEDAR-HCM trial, currently recruiting, is testing aficamten in this population [85]. Given promising animal model data with mavacamten [35] and aficamten’s potentially superior safety [64], this study may be highly impactful in reshaping HCM therapy. The clinical development of cardiac myosin inhibitors continues to evolve, with next-generation agents such as MYK-224 and MYK-581 under investigation and expanding interest in broader indications (for well-established CMIs such as mavacamten and aficamten), including HFpEF, non-obstructive HCM, and pediatric populations. Despite some setbacks and unanswered questions, ongoing trials reflect a hopeful trajectory for the future of CMI-based pharmacotherapy in hypertrophic cardiomyopathy.

5. Conclusions

Cardiac myosin inhibitors (CMIs) reduce myocardial contractility and are currently indicated for a narrow subset of patients with obstructive hypertrophic cardiomyopathy (oHCM). As of now, mavacamten is the only CMI approved for clinical use. Its newer analog, aficamten, may reach the market by late 2025, pending regulatory approval. Both agents have demonstrated favorable clinical and hemodynamic effects, along with acceptable safety profiles in clinical trials. CMIs may expand their therapeutic scope in the future to include pediatric populations and patients with HFpEF or nHCM. However, as a relatively new drug class, CMIs will require further large-scale clinical trials to establish their long-term efficacy, safety, and optimal place in therapy.

Author Contributions

Conceptualization, A.K.; methodology, A.K.; validation, A.K. and M.D.; formal analysis, A.K.; investigation, A.K.; resources, A.K.; data curation, A.K.; writing—original draft preparation, A.K.; writing—review and editing, A.K. and M.D.; visualization, A.K.; supervision, M.D.; project administration, A.K.; funding acquisition, M.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

All of the figures (except Figure 4 and Figure 5) in this article were created on the BioRender.com website. The authors declare that for each figure, a publication license was obtained, generated via the official site (BioRender.com). ChatGPT (GPT-4 omni) was only used to assess grammatical and spelling errors, it was not used for any other reason.

Conflicts of Interest

The authors declare no conflicts of interest.

Abbreviations

The following abbreviations are used in this manuscript:
HFpEF Heart Failure with Preserved Ejection Fraction
HCM Hypertrophic Cardiomyopathy
oHCM Obstructive Hypertrophic Cardiomyopathy
nHCM Non-Obstructive Hypertrophic Cardiomyopathy
CMI Cardiac Myosin Inhibitor

References

  1. Arbelo, E.; Protonotarios, A.; Gimeno, J.R.; Arbustini, E.; Barriales-Villa, R.; Basso, C.; Bezzina, C.R.; Biagini, E.; Blom, N.A.; de Boer, R.A.; et al. 2023 ESC Guidelines for the management of cardiomyopathies. Eur. Heart J. 2023, 44, 3503–3626. [Google Scholar] [CrossRef]
  2. Maron, B.J.; Desai, M.Y.; Nishimura, R.A.; Spirito, P.; Rakowski, H.; Towbin, J.A.; Rowin, E.J.; Maron, M.S.; Sherrid, M.V. Diagnosis and Evaluation of Hypertrophic Cardiomyopathy: JACC State-of-the-Art Review. J. Am. Coll. Cardiol. 2022, 79, 372–389. [Google Scholar] [CrossRef] [PubMed]
  3. Semsarian, C.; Ingles, J.; Maron, M.S.; Maron, B.J. New perspectives on the prevalence of hypertrophic cardiomyopathy. J. Am. Coll. Cardiol. 2015, 65, 1249–1254. [Google Scholar] [CrossRef] [PubMed]
  4. Mandeş, L.; Roşca, M.; Ciupercă, D.; Popescu, B.A. The role of echocardiography for diagnosis and prognostic stratification in hypertrophic cardiomyopathy. J. Echocardiogr. 2020, 18, 137–148. [Google Scholar] [CrossRef] [PubMed]
  5. Mizia-Stec, K.; Leszek, P.; Cegłowska, U.; Wiśniewska, A.; Hałgas, K.; Wybraniec, M.; Pachciński, O.; Stec, M.; Cieśla, D.; Gąsior, M.; et al. Incidence and prevalence of cardiomyopathies in Poland and outcomes for patients in the years 2016–2020. Kardiol. Pol. 2024, 82, 217–219. [Google Scholar] [CrossRef]
  6. Chumakova, O.S.; Baklanova, T.N.; Zateyshchikov, D.A. Clinical Features and Prospective Outcomes of Thin-Filament Hypertrophic Cardiomyopathy: Intrinsic Data and Comparative Insights from Other Cohorts. J. Clin. Med. 2025, 14, 866. [Google Scholar] [CrossRef]
  7. Hong, Y.; Xi, H.T.; Yang, X.Y.; Su, W.W.; Li, X.P. Pathogenic genes and clinical prognosis in hypertrophic cardiomyopathy. World J. Cardiol. 2025, 17, 99595. [Google Scholar] [CrossRef]
  8. The Criteria Committee of the New York Heart Association. Nomenclature and Criteria for Diagnosis of Diseases of the Heart and Great Vessels, 9th ed.; Little, Brown & Co.: Boston, MA, USA, 1994; pp. 253–256. [Google Scholar]
  9. Vilcant, V.; Hai, O. Left Ventricular Outflow Tract Obstruction. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022. [Google Scholar]
  10. Huffman, M.D.; Roth, G.A.; Sliwa, K.; Yancy, C.W.; Prabhakaran, D. Heart Failure. In Cardiovascular, Respiratory, and Related Disorders, 3rd ed.; Prabhakaran, D., Anand, S., Gaziano, T.A., Mbanya, J.C., Wu, Y., Nugent, R., Eds.; The International Bank for Reconstruction and Development/The World Bank: Washington, DC, USA, 2017. [Google Scholar]
  11. Elliott, P.M.; Gimeno, J.R.; Tomé, M.T.; Shah, J.; Ward, D.; Thaman, R.; Mogensen, J.; McKenna, W.J. Left ventricular outflow tract obstruction and sudden death risk in patients with hypertrophic cardiomyopathy. Eur. Heart J. 2006, 27, 1933–1941. [Google Scholar] [CrossRef]
  12. Kalinski, J.K.; Xu, B.; Boyd, R.; Tasseff, N.; Rutkowski, K.; Ospina, S.; Smedira, N.; Thamilarasan, M.; Popovic, Z.B.; Desai, M.Y. Novel Cardiac Myosin Inhibitor Therapy for Hypertrophic Cardiomyopathy in Adults: A Contemporary Review. Am. J. Cardiovasc. Drugs 2024, 24, 591–602. [Google Scholar] [CrossRef]
  13. Sherrod Iv, C.F.; Saberi, S.; Nassif, M.E.; Claggett, B.L.; Coats, C.J.; Garcia-Pavia, P.; Januzzi, J.L.; Lewis, G.D.; Ma, C.; Maron, M.S.; et al. Effect of Aficamten on Health Status Outcomes in Obstructive Hypertrophic Cardiomyopathy: Results From SEQUOIA-HCM. J. Am. Coll. Cardiol. 2024, 84, 1773–1785. [Google Scholar] [CrossRef]
  14. Desai, M.Y.; Owens, A.; Wolski, K.; Geske, J.B.; Saberi, S.; Wang, A.; Sherrid, M.; Cremer, P.C.; Lakdawala, N.K.; Tower-Rader, A.; 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, 968–977. [Google Scholar] [CrossRef]
  15. Ommen, S.R.; Ho, C.Y.; Asif, I.M.; Balaji, S.; Burke, M.A.; Day, S.M.; Dearani, J.A.; Epps, K.C.; Evanovich, L.; Ferrari, V.A.; 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–e1311. [Google Scholar] [CrossRef] [PubMed]
  16. U.S. Foods and Drugs Administration (FDA). Available online: https://www.fda.gov/drugs/news-events-human-drugs/fda-approves-new-drug-improve-heart-function-adults-rare-heart-condition (accessed on 15 May 2025).
  17. European Medicine Agency. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/camzyos (accessed on 15 May 2025).
  18. Clinical Trails. Available online: https://clinicaltrials.gov/study/NCT05186818 (accessed on 15 May 2025).
  19. Clinical Trails. Available online: https://clinicaltrials.eu/inn/bms-986435/ (accessed on 15 May 2025).
  20. Zhang, H.; Yu, C.; Cheng, Y.; Chen, Z.; Chen, M.; He, W.; Jin, Z.; Cai, S.; Yu, L. Clinical Trials in Hypertrophic Cardiomyopathy Therapy: A Comprehensive Analysis of Trials Registered in Global Clinical Databases. Drug Des. Devel. Ther. 2023, 17, 1863–1877. [Google Scholar] [CrossRef] [PubMed]
  21. Gordon, A.M.; Homsher, E.; Regnier, M. Regulation of contraction in striated muscle. Physiol. Rev. 2000, 80, 853–924. [Google Scholar] [CrossRef] [PubMed]
  22. Squire, J. Special Issue: The Actin-Myosin Interaction in Muscle: Background and Overview. Int. J. Mol. Sci. 2019, 20, 5715. [Google Scholar] [CrossRef] [PubMed]
  23. Jani, V.P.; Song, T.; Gao, C.; Gong, H.; Sadayappan, S.; Kass, D.A.; Irving, T.C.; Ma, W. The structural OFF and ON states of myosin can be decoupled from the biochemical super- and disordered-relaxed states. PNAS Nexus 2024, 3, 39. [Google Scholar] [CrossRef] [PubMed]
  24. Cheng, Z.; Fang, T.; Huang, J.; Guo, Y.; Alam, M.; Qian, H. Hypertrophic cardiomyopathy: From phenotype and pathogenesis to treatment. Front. Cardiovasc. Med. 2021, 8, 722340. [Google Scholar] [CrossRef]
  25. Trivedi, D.V.; Adhikari, A.S.; Sarkar, S.S.; Ruppel, K.M.; Spudich, J.A. Hypertrophic cardiomyopathy and the myosin mesa: Viewing an old disease in a new light. Biophys. Rev. 2018, 10, 27–48. [Google Scholar] [CrossRef]
  26. Kim, K.H.; Kim, H.K.; Hwang, I.C.; Lee, S.P.; Park, E.A.; Lee, W.; Kim, Y.J.; Park, J.H.; Sohn, D.W. Myocardial scarring on cardiovascular magnetic resonance in asymptomatic or minimally symptomatic patients with “pure” apical hypertrophic cardiomyopathy. J. Cardiovasc. Magn. Reson. 2012, 14, 52. [Google Scholar] [CrossRef]
  27. Ingles, J.; Burns, C.; Bagnall, R.D.; Lam, L.; Yeates, L.; Sarina, T.; Puranik, R.; Briffa, T.; Atherton, J.J.; Driscoll, T.; et al. Nonfamilial Hypertrophic Cardiomyopathy: Prevalence, Natural History, and Clinical Implications. Circ. Cardiovasc. Genet. 2017, 10, e001620. [Google Scholar] [CrossRef]
  28. Turer, A.T.; Wang, A. Cardiac Myosin Inhibitors: Unlocking Potential to Improve Treatment in Hypertrophic Cardiomyopathy. Circulation 2023, 147, 700–702. [Google Scholar] [CrossRef] [PubMed]
  29. Aman, A.; Akram, A.; Akram, B.; Maham, M.; Bokhari, M.Z.; Akram, A.; Akram, S.; Yaqub, F. Efficacy of cardiac myosin inhibitors mavacamten and aficamten in hypertrophic cardiomyopathy: A systematic review and meta-analysis of randomised controlled trials. Open Heart 2025, 12, e003215. [Google Scholar] [CrossRef] [PubMed]
  30. Braunwald, E.; Saberi, S.; Abraham, T.P.; Elliott, P.M.; Olivotto, I. Mavacamten: A first-in-class myosin inhibitor for obstructive hypertrophic cardiomyopathy. Eur. Heart J. 2023, 44, 4622–4633. [Google Scholar] [CrossRef] [PubMed]
  31. Del Rio, C.L.; Aprajita, Y.; Ferguson, B.S.; Christopher, Z.; Trisha, S.; Frank, R.; John, S.; Lee-Jae, G.; Allison, H.; Julie, G.; et al. Abstract 14585: Chronic Treatment with a Mavacamten-Like Myosin-Modulator (MYK-581) Blunts Disease Progression in a Mini-Pig Genetic Model of Non-Obstructed Hypertrophic Cardiomyopathy: In Vivo Evidence for Improved Relaxation and Functional Reserve. Circulation 2019, 140, A14585. [Google Scholar]
  32. Swift, L.M.; Asfour, H.; Posnack, N.G.; Arutunyan, A.; Kay, M.W.; Sarvazyan, N. Properties of blebbistatin for cardiac optical mapping and other imaging applications. Pflug. Arch. 2012, 464, 503–512. [Google Scholar] [CrossRef]
  33. Lehman, S.J.; Crocini, C.; Leinwand, L.A. Targeting the sarcomere in inherited cardiomyopathies. Nat. Rev. Cardiol. 2022, 19, 353–363. [Google Scholar] [CrossRef]
  34. National Center for Biotechnology Information. PubChem Compound Summary for CID 117761397, Mavacamten. PubChem Identifier. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Mavacamten#section=2D-Structure&fullscreen=true (accessed on 21 May 2025).
  35. Keam, S.J. Mavacamten: First Approval. Drugs 2022, 82, 1127–1135, Erratum in Drugs 2022, 82, 1235. [Google Scholar] [CrossRef]
  36. Available online: https://www.accessdata.fda.gov/drugsatfda_docs/label/2022/214998s000lbl.pdf (accessed on 15 May 2025).
  37. Green, E.M.; Wakimoto, H.; Anderson, R.L.; Evanchik, M.J.; Gorham, J.M.; Harrison, B.C.; Henze, M.; Kawas, R.; Oslob, J.D.; Rodriguez, H.M.; et al. A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice. Science 2016, 351, 617–621. [Google Scholar] [CrossRef]
  38. Heitner, S.B.; Jacoby, D.; Lester, S.J.; Owens, A.; Wang, A.; Zhang, D.; Lambing, J.; Lee, J.; Semigran, M.; Sehnert, A.J. Mavacamten Treatment for Obstructive Hypertrophic Cardiomyopathy: A Clinical Trial. Ann. Intern. Med. 2019, 170, 741–748. [Google Scholar] [CrossRef]
  39. Masri, A.; Lester, S.J.; Stendahl, J.C.; Hegde, S.M.; Sehnert, A.J.; Balaratnam, G.; Shah, A.; Fox, S.; Wang, A. Long-Term Safety and Efficacy of Mavacamten in Symptomatic Obstructive Hypertrophic Cardiomyopathy: Interim Results of the PIONEER-OLE Study. J. Am. Heart Assoc. 2024, 13, e030607. [Google Scholar] [CrossRef]
  40. Olivotto, I.; Oreziak, A.; Barriales-Villa, R.; Abraham, T.P.; Masri, A.; Garcia-Pavia, P.; Saberi, S.; Lakdawala, N.K.; Wheeler, M.T.; Owens, A.; et al. Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (EXPLORER-HCM): A randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2020, 396, 759–769, Erratum in Lancet 2020, 396, 758. [Google Scholar] [CrossRef]
  41. Kitaoka, H.; Ieda, M.; Ebato, M.; Kozuma, K.; Takayama, M.; Tanno, K.; Komiyama, N.; Sakata, Y.; Maekawa, Y.; Minami, Y.; et al. Phase 3 Open-Label Study Evaluating the Efficacy and Safety of Mavacamten in Japanese Adults with Obstructive Hypertrophic Cardiomyopathy—The HORIZON-HCM Study. Circ. J. 2024, 89, 130–138. [Google Scholar] [CrossRef]
  42. Rader, F.; Oręziak, A.; Choudhury, L.; Saberi, S.; Fermin, D.; Wheeler, M.T.; Abraham, T.P.; Garcia-Pavia, P.; Zwas, D.R.; Masri, A.; et al. Mavacamten Treatment for Symptomatic Obstructive Hypertrophic Cardiomyopathy: Interim Results From the MAVA-LTE Study, EXPLORER-LTE Cohort. JACC Heart Fail. 2024, 12, 164–177. [Google Scholar] [CrossRef]
  43. Inestroza, K.; Mijares-Rojas, I.; Matute-Martínez, C.; Ergui, L.; Albosta, M.; Vergara-Sanchez, C.; Dangl, M.; Hernandez, R.J.; Ebner, B.; Vincent, L.T.; et al. In-hospital outcomes of septal myectomy vs. alcohol septal ablation for hypertrophic cardiomyopathy with outflow tract obstruction: An update and insights from the national inpatient sample from 2011 to 2019. J. Investig. Med. 2024, 72, 262–269. [Google Scholar] [CrossRef]
  44. Ramonfaur, D.; Gasperetti, A.; Blake, V.E.; Rivers, B.; Kassamali, A.A.; Kasper, E.K.; Barouch, L.A.; Wu, K.C.; Madrazo, J.A.; Carrick, R.T. Eighteen-Month Real-World Experience Using Mavacamten for Treatment of Obstructive Hypertrophic Cardiomyopathy in a Racially Diverse Population. J. Am. Heart Assoc. 2024, 13, e034069. [Google Scholar] [CrossRef] [PubMed]
  45. Lim, J.; Cho, J.Y.; Kwak, S.; Park, C.S.; Park, J.; Choi, H.M.; Cho, G.Y.; Choi, G.H.; Kim, J.; Na, J.O. Real-World Experience of Mavacamten for Patients With Obstructive Hypertrophic Cardiomyopathy in South Korea: A Prospective Multi-Center Observational Study. Korean Circ. J. 2025, 55, 339–354. [Google Scholar] [CrossRef] [PubMed]
  46. Scholtz, S.; Coppée, C.; Mohemed, K.; Potratz, M.; Sequeira, V.; Rudolph, V.; Scholtz, W.; Reil, J.C. Mavacamten maintenance dose determination: Insights into individualised therapy for hypertrophic cardiomyopathy. Open Heart 2025, 12, e003192. [Google Scholar] [CrossRef] [PubMed]
  47. Wu, X.; Chen, N.; Hsu, P.; Sun, J.; Li, W.T.; Wang, Q.; Samira, M.; Wei, Q.; Yu, J.C.; Cao, G.Y.; et al. Pharmacokinetics and safety of mavacamten in healthy Chinese participants with different CYP2C19 phenotypes. Clin. Transl. Sci. 2024, 17, e13877. [Google Scholar] [CrossRef]
  48. Ricci, F.; Molinari, L.V.; Mansour, D.; Galanti, K.; Vagnarelli, F.; Renda, G.; Gallina, S.; Owens, A.; Luzum, J.A.; Olivotto, I.; et al. Managing drug-drug interactions with mavacamten: A focus on combined use of antiarrhythmic drugs and anticoagulants. Heart Rhythm 2025, 22, 510–525. [Google Scholar] [CrossRef]
  49. Ammirati, E.; Contri, R.; Coppini, R.; Cecchi, F.; Frigerio, M.; Olivotto, I. Pharmacological treatment of hypertrophic cardiomyopathy: Current practice and novel perspec-tives. Eur. J. Heart Fail. 2016, 18, 1106–1118. [Google Scholar] [CrossRef]
  50. Badr, A.; Roehl, K.; Suppah, M.; Abdullah, H.A.; Arsanjani, R.; Siontis, K.C.; Geske, J.B.; Ommen, S.R.; Giudicessi, J.R.; Alsidawi, R. Temporal Patterns of Holter-Detected Arrhythmias in Hypertrophic Cardiomyopathy Patients Treated with Mavacamten. Biomedicines 2025, 13, 1005. [Google Scholar] [CrossRef]
  51. Cytokinetics. Available online: https://pnr-files.pro1.gus.wdc.dianum.io/globenewswire/articles/2989688/en/cytokinetics-announces-fda-acceptance-of-new-drug.pdf (accessed on 15 May 2025).
  52. National Center for Biotechnology Information. PubChem Compound Summary for CID 139331495, Aficamten. Available online: https://pubchem.ncbi.nlm.nih.gov/compound/Aficamten (accessed on 21 May 2025).
  53. Hartman, J.J.; Hwee, D.T.; Robert-Paganin, J.; Chuang, C.; Chin, E.R.; Edell, S.; Lee, K.H.; Madhvani, R.; Paliwal, P.; Pernier, J.; et al. Aficamten is a small-molecule cardiac myosin inhibitor designed to treat hypertrophic cardiomyopathy. Nat. Cardiovasc. Res. 2024, 3, 1003–1016. [Google Scholar] [CrossRef]
  54. Maron, M.S.; Masri, A.; Nassif, M.E.; Barriales-Villa, R.; Arad, M.; Cardim, N.; Choudhury, L.; Claggett, B.; Coats, C.J.; Düngen, H.D.; et al. Aficamten for Symptomatic Obstructive Hypertrophic Cardiomyopathy. N. Engl. J. Med. 2024, 390, 1849–1861. [Google Scholar] [CrossRef] [PubMed]
  55. Chuang, C.; Collibee, S.; Ashcraft, L.; Wang, W.; Wal, M.V.; Wang, X.L.; Hwee, D.T.; Wu, Y.S.; Wang, J.Y.; Chin, E.R.; et al. Discovery of Aficamten (CK-274), a Next-Generation Cardiac Myosin Inhibitor for the Treatment of Hypertrophic Cardiomyopathy. J. Med. Chem. 2021, 64, 14142–14152. [Google Scholar] [CrossRef] [PubMed]
  56. Malik, F.I.; Robertson, L.A.; Armas, D.R.; Robbie, E.P.; Osmukhina, A.; Xu, D.; Li, H.; Solomon, S.D. A Phase 1 Dose-Escalation Study of the Cardiac Myosin Inhibitor Aficamten in Healthy Participants. JACC Basic Transl. Sci. 2022, 7, 763–775. [Google Scholar] [CrossRef] [PubMed]
  57. Xu, D.; Divanji, P.; Griffith, A.; Sukhun, R.; Cheplo, K.; Li, J.; German, P. Pharmacokinetics, disposition, and biotransformation of the cardiac myosin inhibitor aficamten in humans. Pharmacol. Res. Perspect. 2024, 12, e70006. [Google Scholar] [CrossRef]
  58. Maron, M.S.; Masri, A.; Nassif, M.E.; Barriales-Villa, R.; Abraham, T.P.; Arad, M.; Cardim, N.; Choudhury, L.; Claggett, B.; Coats, C.J.; et al. Impact of Aficamten on Disease and Symptom Burden in Obstructive Hypertrophic Cardiomyopathy: Results From SEQUOIA-HCM. J. Am. Coll. Cardiol. 2024, 84, 1821–1831. [Google Scholar] [CrossRef]
  59. Schulze, C.; Abraham, T.P.; Barriales-Villa, R.; Claggett, B.; Coats, C.; García-Pavía, P.; Hagège, A.; Januzzi, J.L.; Kulac, I.J.; Kwong, R.Y.; et al. Abstract 4140102: Changes in EQ-5D-5L with Aficamten in Obstructive Hypertrophic Cardiomyopathy (oHCM): The SEQUOIA-HCM Trial. Circulation 2024, 150, Suppl_1. [Google Scholar] [CrossRef]
  60. Saberi, S.; Abraham, T.P.; Choudhury, L.; Owens, A.T.; Tower-Rader, A.; Rader, F.; Pavia, P.G.; Olivotto, I.; Coats, C.; Fifer, M.A.; et al. Efficacy and safety of Aficamten in the first cohort of patients with symptomatic obstructive hypertrophic cardiomyopathy completing 1-year follow-up: Findings from the FOREST-HCM study. J. Am. Coll. Cardiol. 2024, 83, 356. [Google Scholar] [CrossRef]
  61. Owens, A.T.; Masri, A.; Abraham, T.P.; Choudhury, L.; Rader, F.; Symanski, J.D.; Turer, A.T.; Wong, T.C.; Tower-Rader, A.; Coats, C.J.; et al. Aficamten for Drug-Refractory Severe Obstructive Hypertrophic Cardiomyopathy in Patients Receiving Disopyramide: REDWOOD-HCM Cohort 3. J. Card. Fail. 2023, 29, 1576–1582. [Google Scholar] [CrossRef]
  62. Maron, M.S.; Masri, A.; Choudhury, L.; Olivotto, I.; Saberi, S.; Wang, A.; Garcia-Pavia, P.; Lakdawala, N.K.; Nagueh, S.F.; Rader, F.; et al. Phase 2 Study of Aficamten in Patients With Obstructive Hypertrophic Cardiomyopathy. J. Am. Coll. Cardiol. 2023, 81, 34–45. [Google Scholar] [CrossRef]
  63. Coats, C.J.; Masri, A.; Nassif, M.E.; Barriales-Villa, R.; Arad, M.; Cardim, N.; Choudhury, L.; Claggett, B.; Düngen, H.; Garcia-Pavia, P.; et al. Dosing and Safety Profile of Aficamten in Symptomatic Obstructive Hypertrophic Cardiomyopathy: Results From SEQUOIA-HCM. J. Am. Heart Assoc. 2024, 13, e035993. [Google Scholar] [CrossRef] [PubMed]
  64. Davis, B.J.; Volk, H.; Nguyen, O.; Kamna, D.; Chen, H.; Barriales-Villa, R.; Garcia-Pavia, P.; Olivotto, I.; Owens, A.T.; Coats, C.J.; et al. Safety and Efficacy of Mavacamten and Aficamten in Patients With Hypertrophic Cardiomyopathy. J. Am. Heart Assoc. 2025, 14, e038758. [Google Scholar] [CrossRef] [PubMed]
  65. Maharao, N.; Xu, D.; Simkins, T.J.; Bowles, O.; Liu, G.; Benattia, Y.; Griffith, A.; Heitner, S.B.; Kupfer, S.; German, P. Clinical Evaluation of the Effect of Aficamten on QT/QTc Interval in Healthy Participants. Clin. Transl. Sci. 2025, 18, e70218. [Google Scholar] [CrossRef] [PubMed]
  66. Xu, D.; Lutz, J.D.; Divanji, P.; Li, J.; Benattia, Y.; Griffith, A.; Heitner, S.B.; Kupfer, S.; German, P. Effect of Hepatic Impairment or Renal Impairment on the Pharmacokinetics of Aficamten. Clin. Pharmacokinet. 2025, 64, 397–406. [Google Scholar] [CrossRef]
  67. Cytokinetics. Available online: https://pnr-files.pro1.gus.wdc.dianum.io/globenewswire/articles/3001161/en/cytokinetics-announces-european-medicines-agency.pdf (accessed on 15 May 2025).
  68. Rauscher, A.Á.; Gyimesi, M.; Kovács, M.; Málnási-Csizmadia, A. Targeting Myosin by Blebbistatin Derivatives: Optimization and Pharmacological Potential. Trends. Biochem. Sci. 2018, 43, 700–713. [Google Scholar] [CrossRef]
  69. Roman, B.I.; Verhasselt, S.; Stevens, C.V. Medicinal Chemistry and Use of Myosin II Inhibitor (S)-Blebbistatin and Its Derivatives. J. Med. Chem. 2018, 61, 9410–9428. [Google Scholar] [CrossRef]
  70. Gyimesi, M.; Rauscher, A.Á.; Suthar, S.K.; Hamow, K.Á.; Oravecz, K.; Lőrincz, I.; Borhegyi, Z.; Déri, M.T.; Kiss, Á.F.; Monostory, K.; et al. Improved Inhibitory and Absorption, Distribution, Metabolism, Excretion, and Toxicology (ADMET) Properties of Blebbistatin Derivatives Indicate That Blebbistatin Scaffold Is Ideal for drug Development Targeting Myosin-2. J. Pharmacol. Exp. Ther. 2021, 376, 358–373. [Google Scholar] [CrossRef]
  71. Alsulami, K.; Marston, S. Small Molecules acting on Myofilaments as Treatments for Heart and Skeletal Muscle Diseases. Int. J. Mol. Sci. 2020, 21, 9599. [Google Scholar] [CrossRef]
  72. AdisInsight. Available online: https://adisinsight.springer.com/drugs/800056091 (accessed on 15 May 2025).
  73. Clinical Trails. Available online: https://clinicaltrials.gov/study/NCT06122779 (accessed on 15 May 2025).
  74. Shah, S.J.; Rigolli, M.; Javidialsaadi, A.; Patel, R.B.; Khadra, S.; Goyal, P.; Little, S.; Wever-Pinzon, O.; Owens, A.T.; Skali, H.; et al. Cardiac Myosin Inhibition in Heart Failure with Normal and Supranormal Ejection Fraction: Primary Results of the EMBARK-HFpEF Trial. JAMA Cardiol. 2025, 10, 170–175. [Google Scholar] [CrossRef]
  75. Garcia-Pavia, P.; Bilen, O.; Burroughs, M.; Costabel, J.P.; Correia, E.d.B.; Dybro, A.M.; Elliott, P.; Lakdawala, N.K.; Mann, A.; Nair, A.; et al. Aficamten vs Metoprolol for Obstructive Hypertrophic Cardiomyopathy: MAPLE-HCM Rationale, Study Design, and Baseline Characteristics. JACC Heart Fail. 2025, 13, 346–357. [Google Scholar] [CrossRef]
  76. Wheeler, M.T.; Jacoby, D.; Elliott, P.M.; Saberi, S.; Hegde, S.M.; Lakdawala, N.K.; Myers, J.; Sehnert, A.J.; Edelberg, J.M.; Li, W.; et al. Effect of beta-blocker therapy on the response to mavacamten in patients with symptomatic obstructive hypertrophic cardiomyopathy. Eur. J. Heart Fail. 2023, 25, 260–270. [Google Scholar] [CrossRef] [PubMed]
  77. Clinical Trails. Available online: https://clinicaltrials.gov/study/NCT06081894 (accessed on 15 May 2025).
  78. Masri, A.; Maron, M.S.; Abraham, T.P.; Nassif, M.E.; Barriales-Villa, R.; Bilen, O.; Coats, C.J.; Elliott, P.; Garcia-Pavia, P.; Massera, D.; et al. Concomitant Aficamten and Disopyramide in Symptomatic Obstructive Hypertrophic Cardiomyopathy. JACC Heart Fail. 2025. [Google Scholar] [CrossRef] [PubMed]
  79. Hughes, C. Appropriate and inappropriate polypharmacy-Choosing the right strategy. Br. J. Clin. Pharmacol. 2021, 87, 84–86. [Google Scholar] [CrossRef] [PubMed]
  80. Delara, M.; Murray, L.; Jafari, B.; Bahji, A.; Goodarzi, Z.; Kirkham, J.; Chowdhury, M.; Seitz, D.P. Prevalence and factors associated with polypharmacy: A systematic review and Meta-analysis. BMC Geriatr. 2022, 22, 601, Erratum in BMC Geriatr. 2022, 22, 742. [Google Scholar] [CrossRef]
  81. Ho, C.Y.; Mealiffe, M.E.; Bach, R.G.; Bhattacharya, M.; Choudhury, L.; Edelberg, J.M.; Hegde, S.M.; Jacoby, D.; Lakdawala, N.K.; Lester, S.J.; et al. Evaluation of Mavacamten in Symptomatic Patients With Nonobstructive Hypertrophic Cardiomyopathy. J. Am. Coll. Cardiol. 2020, 75, 2649–2660. [Google Scholar] [CrossRef]
  82. Desai, M.Y.; Nissen, S.E.; Abraham, T.; Olivotto, I.; Garcia-Pavia, P.; Lopes, R.D.; Verheyen, N.; Wever-Pinzon, O.; Wolski, K.; Jaber, W.; et al. Mavacamten in Symptomatic Nonobstructive Hypertrophic Cardiomyopathy: Design, Rationale, and Baseline Characteristics of ODYSSEY-HCM. JACC Heart Fail. 2025, 13, 358–370. [Google Scholar] [CrossRef]
  83. Bristol Myers Squibb. Available online: https://news.bms.com/news/details/2025/Bristol-Myers-Squibb-Provides-Update-on-Phase-3-ODYSSEY-HCM-Trial/ (accessed on 15 May 2025).
  84. Amr, A.; Kayvanpour, E.; Reich, C.; Koelemen, J.; Asokan, S.; Frey, N.; Meder, B.; Sedaghat-Hamedani, F. Assessing the Applicability of Cardiac Myosin Inhibitors for Hypertrophic Cardiomyopathy Management in a Large Single Center Cohort. Rev. Cardiovasc. Med. 2024, 25, 225. [Google Scholar] [CrossRef]
  85. Clinical Trails. Available online: https://clinicaltrials.gov/study/NCT06412666 (accessed on 15 May 2025).
Figure 1. Simplified diagram of the therapeutic pathway for hypertrophic cardiomyopathy based on the guidelines of the European Society of Cardiology [1]. * depending on ejection fraction; ** described in the main text. Created in BioRender: https://BioRender.com/wzb0fkf (accessed on 30 May 2025).
Figure 1. Simplified diagram of the therapeutic pathway for hypertrophic cardiomyopathy based on the guidelines of the European Society of Cardiology [1]. * depending on ejection fraction; ** described in the main text. Created in BioRender: https://BioRender.com/wzb0fkf (accessed on 30 May 2025).
Biomolecules 15 01098 g001
Figure 2. Basic physiology of cardiac muscle contraction: from myofibril structure to sarcomere and actin–myosin interaction. Troponin complex describes the combination of troponin C, I, and T. The colors used in this figure: blue describes myosin (thick filament), red describes actin (thin filament), purple describes tropomyosin, and green describes ion calcium (Ca2+). Created in BioRender: https://BioRender.com/y3xg904 (accessed on 30 May 2025).
Figure 2. Basic physiology of cardiac muscle contraction: from myofibril structure to sarcomere and actin–myosin interaction. Troponin complex describes the combination of troponin C, I, and T. The colors used in this figure: blue describes myosin (thick filament), red describes actin (thin filament), purple describes tropomyosin, and green describes ion calcium (Ca2+). Created in BioRender: https://BioRender.com/y3xg904 (accessed on 30 May 2025).
Biomolecules 15 01098 g002
Figure 3. Pathological actin–myosin interaction in hypertrophic cardiomyopathy and its modulation by cardiac myosin inhibitors. The left side of Figure 3 presents the pathological state of cardiac myosin-actin complex in HCM with a larger number of cross-bridges, which results in hypercontraction (as described in the text). The right side shows a state of myosin–actin complex in the presence of CMI: the cardiac myosin inhibitors block excessive myosin heads, and as a result, there is a lower number of cross-bridges. The colors used in this figure: blue describes myosin (thick filament), red describes actin (thin filament), and purple describes tropomyosin. CMI: cardiac myosin inhibitor. The chemical structure shown is symbolic and does not represent any actual compound. Created in BioRender (2025): https://BioRender.com/pie1hc8 (accessed on 30 May 2025).
Figure 3. Pathological actin–myosin interaction in hypertrophic cardiomyopathy and its modulation by cardiac myosin inhibitors. The left side of Figure 3 presents the pathological state of cardiac myosin-actin complex in HCM with a larger number of cross-bridges, which results in hypercontraction (as described in the text). The right side shows a state of myosin–actin complex in the presence of CMI: the cardiac myosin inhibitors block excessive myosin heads, and as a result, there is a lower number of cross-bridges. The colors used in this figure: blue describes myosin (thick filament), red describes actin (thin filament), and purple describes tropomyosin. CMI: cardiac myosin inhibitor. The chemical structure shown is symbolic and does not represent any actual compound. Created in BioRender (2025): https://BioRender.com/pie1hc8 (accessed on 30 May 2025).
Biomolecules 15 01098 g003
Figure 4. Structure of Mavacamten [34].
Figure 4. Structure of Mavacamten [34].
Biomolecules 15 01098 g004
Figure 5. Structure of aficamten [52].
Figure 5. Structure of aficamten [52].
Biomolecules 15 01098 g005
Table 1. Regulatory status and clinical development stage of selected cardiac myosin inhibitors (references in the text).
Table 1. Regulatory status and clinical development stage of selected cardiac myosin inhibitors (references in the text).
Drug NameFDA ApprovalEMA ApprovalClinical Development Stage
MavacamtenApproved (2022)Approved (2023)Available on the market
AficamtenPending approvalPending approvalPhase III completed
MYK-224Not approvedNot approvedPhase II
MYK-581Not approvedNot approvedPreclinical phase
BlebbistatinNot approvedNot approvedResearch use only
FDA—Food and Drug Administration; EMA—European Medicines Agency.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Kukowka, A.; Droździk, M. Cardiac Myosin Inhibitors in the Treatment of Hypertrophic Cardiomyopathy: Clinical Trials and Future Challenges. Biomolecules 2025, 15, 1098. https://doi.org/10.3390/biom15081098

AMA Style

Kukowka A, Droździk M. Cardiac Myosin Inhibitors in the Treatment of Hypertrophic Cardiomyopathy: Clinical Trials and Future Challenges. Biomolecules. 2025; 15(8):1098. https://doi.org/10.3390/biom15081098

Chicago/Turabian Style

Kukowka, Arnold, and Marek Droździk. 2025. "Cardiac Myosin Inhibitors in the Treatment of Hypertrophic Cardiomyopathy: Clinical Trials and Future Challenges" Biomolecules 15, no. 8: 1098. https://doi.org/10.3390/biom15081098

APA Style

Kukowka, A., & Droździk, M. (2025). Cardiac Myosin Inhibitors in the Treatment of Hypertrophic Cardiomyopathy: Clinical Trials and Future Challenges. Biomolecules, 15(8), 1098. https://doi.org/10.3390/biom15081098

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop