Abstract
Variants in >12 genes encoding sarcomeric proteins can cause various cardiomyopathies. The two most common are hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM). Current therapeutics do not target the root causes of these diseases, but attempt to prevent disease progression and/or to manage symptoms. Accordingly, novel approaches are being developed to treat the cardiac muscle dysfunction directly. Challenges to developing therapeutics for these diseases include the diverse mechanisms of pathogenesis, some of which are still being debated and defined. Four small molecules that modulate the myosin motor protein in the cardiac sarcomere have shown great promise in the settings of HCM and DCM, regardless of the underlying genetic pathogenesis, and similar approaches are being developed to target other components of the sarcomere. In the setting of HCM, mavacamten and aficamten bind to the myosin motor and decrease the ATPase activity of myosin. In the setting of DCM, omecamtiv mecarbil and danicamtiv increase myosin activity in cardiac muscle (but omecamtiv mecarbil decreases myosin activity in vitro). In this Review, we discuss the therapeutic strategies to alter sarcomere contractile activity and summarize the data indicating that targeting one protein in the sarcomere can be effective in treating patients with genetic variants in other sarcomeric proteins, as well as in patients with non-sarcomere-based disease.
Graphical Abstract
Variants in genes encoding sarcomeric proteins can cause hypertrophic or dilated cardiomyopathy. In this Review, the authors discuss therapeutic strategies to target the cardiac sarcomere, focusing on four small molecules that have been developed that inhibit or activate the myosin motor protein to decrease or increase contractile force, respectively.
Introduction
In 1990, the first genetic variant linked to hypertrophic cardiomyopathy (HCM) was discovered in MYH7, which encodes cardiac myosin heavy chain-β1. Since then, HCM has been called a ‘disease of the sarcomere’ because hundreds of variants in genes encoding cardiac myosin and other sarcomeric proteins have been linked to the disease. Similarly, variants in genes encoding sarcomeric proteins were implicated in genetic dilated cardiomyopathy (DCM) in 20002. In a major advance, genetic variants in TTN, encoding titin, were identified in 30% of patients with genetic DCM3. Compared with HCM, DCM can be caused by variants in a wider range of genes. In addition to variants in genes encoding sarcomeric proteins, variants in genes encoding non-sarcomeric proteins (including nuclear envelope proteins, transcription cofactors and cytoskeletal proteins) can cause DCM4. The mechanisms of how variants in genes encoding sarcomeric proteins cause disease have been studied in detail5, and some of these findings have been informative in defining and refining how small molecules that target the sarcomere are likely to change the course of clinical disease.
Clinically, HCM is characterized by a reduced left ventricular cavity (and thickened left ventricular walls), with hyperdynamic contraction, diastolic dysfunction and increased energy consumption6,7. Surgical interventions in patients with HCM include implantation of a cardioverter–defibrillator, heart transplantation and, in those with obstructive HCM, surgical myectomy8. In some patients with obstructive HCM, alcohol septal ablation can be used instead of surgical myectomy9. Medical management for patients with HCM includes the use of β-blockers, Ca2+-channel blockers and, in those with symptomatic, obstructive HCM, the antiarrhythmic drug disopyramide because of its negative inotropic effects10. Despite medical and surgical management, HCM is a progressive disease, with atrial fibrillation and heart failure occurring in 20% of patients by the age of 50–70 years6.
By contrast, DCM is characterized by thinned ventricular walls and reduced contractility7. Treatments for patients with DCM include symptom management and approaches to preserve myocardial function. Effective medical therapies include mineralocorticoid-receptor antagonists, neprilysin inhibitors and sodium–glucose cotransporter 2 inhibitors, and cardiac resynchronization therapy can be used in a subset of patients7,11. When medical management is insufficient to prevent disease progression, patients can develop heart failure and require heart transplantation or implantation of a left ventricular assist device. Therefore, an unmet need exists to modulate cardiac contractility directly without systemic effects on other organs; particularly in the setting of cardiomyopathy caused by variants in genes encoding sarcomeric proteins. Although the distinct clinical phenotypes of HCM and DCM require different therapeutic strategies, drug development for both has converged on the modulation of cardiac myosin function12.
Manipulating the myosin motor directly (either increasing or reducing its function) has been the subject of intense interest over the past decade, culminating in several successful clinical trials for patients with HCM or DCM. The strategy to identify myosin modulators was to screen for molecules that bind to cardiac myosin specifically, alter its ATPase activity in vitro, and increase or decrease contractility in vivo. To ensure cardiac selectivity, candidate compounds were screened against non-muscle, smooth muscle or fast-twitch skeletal myosins. Of note, although many sarcomeric protein isoforms in the heart are different from those in skeletal muscle, the MYH7 gene product (known as the myosin heavy chain-β or slow-twitch isoform) is also expressed in type I fibres in skeletal muscle. Therefore, small molecules that modulate myosin heavy chain-β in the heart should also modulate myosin in slow-twitch skeletal muscle, given that it is the same protein13,14.
In this Review, we discuss therapeutic strategies to target the cardiac sarcomere. Progress has also been made in therapeutic approaches to modulate the skeletal muscle sarcomere12, but that is beyond the scope of this article. We discuss how targeting one protein in the sarcomere can be effective in treating patients with genetic variants in other sarcomeric proteins, as well as in patients with genetic variants in non-sarcomeric proteins.
The cardiac sarcomere: an overview
The two main filaments in sarcomeres are the Ca2+-dependent, regulatory thin filaments and the myosin-based, force-generating thick filaments15 (Fig. 1). The thin filament is composed primarily of five proteins: three that form the troponin complex (troponin C, the Ca2+-binding subunit; troponin I, the inhibitory subunit; and troponin T, the tropomyosin-binding subunit), tropomyosin and actin. These proteins work together in a highly coordinated fashion to transmit the Ca2+ status of the myofilament, which, in turn, regulates interactions between the thin and thick filaments16. Parallel to the thin filament, the thick filament is comprised of myosin molecules, which couple the chemical energy of ATP hydrolysis to mechanical movement (shortening) of the sarcomere. Importantly, myosin ATPase activity is highly coordinated with Ca2+ activation of the thin filament to ensure actomyosin interactions and force generation.
Fig. 1 ∣. The cardiac sarcomere and its components.
The cardiac sarcomere is made up primarily of regulatory thin filaments, force-generating thick filaments (myosin), regulatory myosin-binding protein C and tension-sensing titin. Variants in the genes encoding these proteins are linked to various genetic cardiomyopathies, such as hypertrophic cardiomyopathy (HCM) and dilated cardiomyopathy (DCM), which are typically characterized by a unique molecular mechanism of disease. Therefore, these proteins are potential druggable targets for treating cardiomyopathies.
The ATPase activity of myosin is defined by three distinct rates: actin-activated activity (fast), the disordered relaxed (DRX) state (~100-fold slower than actin-activated) and the super-relaxed (SRX) state (~10-fold slower than DRX) (Fig. 2). Myosin heads that are not bound to actin exist in a dynamic equilibrium between the DRX and SRX states17-19. The DRX state is defined by the basal ATPase activity of myosin, in which the myosin heads are released from the thick filament backbone and available for interaction with actin. The SRX state is defined by an ultra-slow, energy-conserving ATPase activity of myosin20-22. This reduction in ATPase activity is, in part, achieved by a structural conformation of the myosin molecule, termed the interacting heads motif (IHM), in which the motor domain is folded back onto the thick filament axis, sequestering the myosin in an auto-inhibited state23-26. From electron microscopy images, 3D models of the IHM were mapped to mouse ventricular thick filaments, suggesting the existence of the IHM structural state in cardiac tissue, although the images were low resolution19. Another study measured the IHM in bovine cardiac heavy meromyosin via FRET (fluorescence resonance energy transfer), providing higher-resolution structural confirmation that the IHM structure does exist in cardiac muscle26. However, the researchers note that perturbations to the SRX state did not produce concomitant alterations to the IHM population, suggesting that the SRX state in cardiac muscle is not always synonymous with IHM; instead, additional energy-conserving myosin conformation(s) are likely to exist27,28.
Fig. 2 ∣. The functional states of myosin.
a ∣ Myosin molecules exist in an equilibrium between three distinct ATP-utilization states: the actin-bound state (green), the disordered relaxed (DRX) state (orange) and the super-relaxed (SRX) state (red). The SRX state can be divided into the folded back, interacting heads motif (IHM) structure or by another, undefined structural state. These states are defined by specific myosin ATPase activities, in which the activity in the actin-bound state is ~100-fold faster than in the DRX state and nearly 1,000-fold faster than in the SRX state83. These differences in ATPase activity govern ATP utilization of the sarcomere and the heart. b ∣ The simulated ATPase activity for each state is shown as a function of time. The black curve depicts the myosin ATPase activity of cardiac muscle based on the physiological distribution of myosin heads between the three states. Adapted with permission from REF.139.
In addition to the thin and thick filaments, cardiac sarcomeres contain titin and myosin-binding protein C (MyBP-C), two large proteins that regulate the function of the sarcomere (Fig. 1). Titin, the largest protein in the human proteome, spans the half sarcomere and is crucial in maintaining sarcomere structure throughout muscle contraction and relaxation29. MyBP-C has been shown to interact with both the thin and thick filaments and titin. MyBP-C has been postulated to have a primary role in the regulation of myosin–actin cross-bridge formation28,30-33, but the exact mechanisms of how MyBP-C regulates sarcomere function are incompletely understood. Interestingly, MyBP-C has been shown to regulate the SRX state in both in vitro systems and animal models28,34-37. In skeletal muscle, the population of SRX myosin heads was higher in the so-called C-zone (where MyBP-C resides) than in the other thick filament zones35. Loss of MyBP-C led to destabilization of the SRX population in mouse isolated left ventricular cardiomyocytes34, and protein kinase A phosphorylation of MyBP-C destabilized the SRX population in mouse hearts38, suggesting that transient recruitment of myosin heads out of the SRX state is, in part, regulated by MyBP-C.
Therapies for different aetiologies
When targeting the sarcomere to treat inherited cardiomyopathies, it is important to consider the different mechanisms of pathogenesis for each target. Given that the vast majority of genetic cardiomyopathies are inherited in an autosomal dominant fashion, therapeutic strategies for recessively inherited cardiomyopathies are not discussed in this Review. In addition, different strategies might need to be developed if the disease is the result of haploinsufficiency caused by either loss of function (via a reduction in the amount of functional protein) or gain of function (via a dominant-negative polypeptide mechanism). Finally, genome-wide association studies have identified a growing number of cardiomyopathy-associated variants in genes encoding non-sarcomeric proteins39. Patients with these genetic variants were included in the clinical trials discussed below; however, the mechanism of action for sarcomere-targeted therapies in patients with variants in non-sarcomeric proteins is not yet well understood.
Most variants in MYH7 (the second most common cause of HCM) are autosomal dominant and largely result in single amino acid changes40. Cardiomyopathy is caused by the mutant protein interfering with the function of the wild-type protein. HCM-causing variants in genes encoding most other sarcomeric proteins, such as the troponin subunits and tropomyosin isoforms, also fall into this gain-of-function category, which is unsurprising in a structure such as the sarcomere, in which many different proteins assemble into structures that function together.
By contrast, the majority of variants in MYBPC3 (the most common cause of genetic HCM) cause frameshift mutations that result in a reduction in the total quantity of functional protein41. The reduced levels of functional protein could be due to a reduction in the amount of mRNA owing to nonsense-mediated decay of the mutant mRNA or, theoretically, to the production of truncated, inactive protein from the mRNA carrying the frameshift mutation. This mechanism of pathogenesis would be haploinsufficiency caused by a loss-of-function mechanism. For a long time, MYBPC3 mRNA that carried a frameshift mutation was proposed to encode a truncated protein that functioned as a poison polypeptide, which would be a gain-of-function mechanism42. However, no convincing data indicate that truncated forms of MyBP-C are translated from the mRNA carrying the frameshift mutation, and haploinsufficiency caused by loss of function is now widely considered to be the mechanism of most disease caused by MYBPC3 variants. Analyses of cardiac samples from patients with MYBPC3 variants suggest that the wild-type allele can compensate to some extent; the amount of wild-type MyBP-C protein was ~70% of normal levels, rather than the predicted 50%41. This finding is still consistent with a mechanism of haploinsufficiency if, for example, a threshold of >70% MyBP-C expression needs to be exceeded to have a normally functioning heart. Using induced pluripotent stem cell-derived cardiomyocytes from individuals who were heterozygous for MyBP-C-truncating variants, the same researchers found that the compensation by the wild-type allele was sufficient to achieve no decrease in total MyBP-C protein levels and no contractile deficits43. Furthermore, the compensation seemed to be the result of decreased degradation of the wild-type MyBP-C protein43. However, these observations in cardiomyocytes derived from induced pluripotent stem cells in vitro might not be relevant to an intact heart in vivo.
Importantly, the hypothesis that destabilization of the SRX state of myosin is a primary molecular driver of the hyperdynamic contractile phenotype seen in patients with HCM has been investigated44. Biophysical and biochemical studies have demonstrated that single-nucleotide variants in MYH7 can increase the DRX state of myosin, leading to increased contractility and energy expenditure in the heart22,37,45-50. Therefore, regulation of the SRX state is a potential mechanistic avenue for myosin-targeted small molecules (discussed below).
Variants in the same gene (such as MYH7 or TNNI3) can lead to either HCM or DCM51. The mechanisms whereby two different variants in the same gene lead to two different diseases are not well understood, although the most logical hypothesis is that different variants result in different amino acid substitutions, which affect the protein function differently. However, disease phenotype could also be affected by genetic background. For example, variants in LTBP4 can modify phenotypes in patients with Duchenne muscular dystrophy52. Furthermore, a missense variant in TTN has been shown to modify the phenotype of patients with a variant in LMNA53.
The discovery that TTN variants are common causes of genetic DCM has led to important discoveries not only about the pathophysiology of DCM but also about titin biology3,54. Approximately 30% of DCM can be attributed to variants in TTN3. The vast majority of these variants are inherited in an autosomal dominant manner, and many of them are called truncating variants because they have the potential to produce a truncated titin protein if transcribed and translated55. However, in contrast to disease-causing variants in MYBPC3, multiple mechanisms of pathogenesis are associated with TTN variants, including both haploinsufficiency and truncated titin polypeptides as well as variants that alter post-translational modifications of titin55. In 2021, definitive proof of the presence of truncated titin polypeptides in patients with DCM was reported56.
Sarcomeric contractile activators
Small molecules targeting the cardiac sarcomere to increase contractility have largely been developed to treat idiopathic heart failure but could, in principle, ameliorate inherited cardiomyopathies characterized by poorly contracting hearts. These activators directly increase the Ca2+ sensitivity of the thin filament or increase the ATPase activity of the myosin motor. In this section, we discuss the preclinical and clinical data that are available for compounds that increase sarcomeric contractility and cardiac function.
Ca2+ sensitizers: targeting thin filaments
Therapy to increase cardiac contractility in hypocontractile diseases (such as heart failure) has for a long time substantially relied on three types of inotropic agent: digitalis-derived cardiotropic glycosides, the oldest inotropic drugs, which inhibit the sodium–potassium ATPase pump, resulting in the accumulation of Na+ that, in turn, reverses the sodium–calcium exchanger and leads to increased Ca2+ influx in systole; β-adrenergic agonists, such as dopamine or dobutamine; and phosphodiesterase inhibitors, which lead to increased levels of cAMP, thereby avoiding desensitization and downregulation of cardiac β-adrenergic receptors. All these agents rely on a generalized disruption of Ca2+ homeostasis in cardiomyocytes to increase contractility57. However, the long-term use of β-adrenergic agonists or phosphodiesterase inhibitors has adverse effects, including maladaptive cardiac remodelling, probably resulting from the chronic activation of protein kinase A and Ca2+/calmodulin-dependent protein kinase II signalling. In the 1980s, a new strategy was proposed, aimed at directly increasing the Ca2+ sensitivity of the sarcomere by targeting the myofilaments58-62. Independent of the mechanism of action, Ca2+ sensitizers induce a leftward shift in the curve describing the relationship between contractile force and Ca2+ concentration, leading to increased systolic force generation for any given cytoplasmic concentration of Ca2+. Conversely, Ca2+ sensitizers inherently impair diastole by slowing relaxation when the cytoplasmic Ca2+ concentration decreases. Levosimendan, a Ca2+ sensitizer that does not prolong relaxation61,62, stabilizes the Ca2+-bound troponin C conformation and increases Ca2+ sensitivity63-65. Additionally, levosimendan increases intracellular concentrations of cAMP66 via a potent and selective inhibition of phosphodiesterase type 360,67,68 Therefore, levosimendan is likely to act via a combination of increasing Ca2+ sensitivity by binding to cardiac troponin C and ‘classic’ phosphodiesterase inhibition that increases Ca2+ sensitivity without impairing relaxation. Numerous clinical trials have been conducted to investigate the effects of levosimendan in patients with heart failure. Initial findings from the LIDO trial69 suggested short-term beneficial effects of levosimendan in patients with acutely decompensated chronic heart failure. However, levosimendan had no significant effect on all-cause mortality or other secondary outcomes (cardiovascular mortality or number of days alive and out of hospital) in patients with chronic heart failure in the SURVIVE trial69,70 or in patients with heart failure undergoing cardiac surgery in the CHEETAH71, LEVO-CTS72 and LICORN73 trials (Table 1). Furthermore, in the REVIVE trial74, levosimendan treatment was associated with an increased risk of adverse cardiac events, including increased heart rate and incidence of cardiac arrhythmia and frequent occurrence of hypotension in patients with acutely decompensated chronic heart failure. The ESC heart failure guidelines recommend levosimendan treatment in the acute management of patients with hypotension and/or symptoms of hypoperfusion75; however, levosimendan is not approved for use in the treatment of heart failure in the USA.
Table 1 ∣.
Clinical trial results of sarcomere-targeted small molecules
| Small molecule | Sarcomeric target |
Molecular mechanism |
Contractile effect |
Clinical trial | Clinical effects | Refs |
|---|---|---|---|---|---|---|
| Sarcomeric contractile activators | ||||||
| Levosimendan | Troponin C (and PDE3) | Stabilizes troponin I–troponin C interaction | Increased owing to increased Ca2+ sensitivity | SURVIVE, REVIVE, CHEETAH, LEVO-CTS | Improved haemodynamic profile, no effect on cardiac relaxation; increased incidence of cardiac arrhythmia, no improvement in heart failure outcomes or mortality | 59-74 |
| CK-136 (AMG-594) | Troponin complex | Increases Ca2+ sensitivity | Increased owing to increased Ca2+ sensitivity | – | Not available | 76,77 |
| Omecamtiv mecarbil | Myosin heavy chain-β | Stabilizes the pre- powerstroke state of myosin | Increased owing to increased actomyosin interactions | GALACTIC-HF | Modest reduction in the rate of heart failure events and cardiac death; greatest improvement in function in patients with a left ventricular ejection fraction ≤28% | 92,94-95 |
| COSMIC-HF | Improved left ventricular systolic ejection time, stroke volume, end-diastolic diameter and volume, and heart rate; reduced plasma NT-proBNP level | 89, 90 | ||||
| Danicamtiv (MYK-491) | Myosin heavy chain-β | Unpublished | Increased | Phase II trial ongoing, recruiting participants with dilated cardiomyopat hy | Results not available | 99,100 |
| Sarcomeric contractile inhibitors | ||||||
| Mavacamten (MYK-461) | Myosin heavy chain-β | Stabilizes the super-relaxed (SRX) state of myosin | Decreased owing to reduced myosin head availability | EXPLORER-HCM | Improved peak oxygen consumption and NYHA functional class; reduced left ventricular outflow tract obstruction | 118-120 |
| PIONEER-HCM | Improved post-exercise peak left ventricular outflow tract gradient | 117 | ||||
| MAVERICK-HCM | Decreased risk of adverse events, decreased plasma NT pro-BNP and cardiac troponin I levels | 116,122 | ||||
| Aficamten (CK-274) | Myosin heavy chain-β | Slows phosphate release from myosin | Decreased owing to stabilization of weak actin-binding myosin conformation | REDWOOD-HCM | Results not available | 135 |
NT-proBNP, N-terminal pro-B-type natriuretic peptide; PDE3, phosphodiesterase type 3.
In March 2020, Amgen and Cytokinetics presented preclinical data on CK-136 (formerly referred to as AMG-594) showing that CK-136 is a selective activator of the cardiac troponin complex, increasing sarcomere Ca2+ sensitivity and resulting in an increased number of myosin–actin cross-bridges and therefore higher contractile force76. Preliminary data from studies using rat isolated ventricular myocytes indicated that CK-136 increased cellular shortening, with no changes in diastolic Ca2+ concentration, systolic Ca2+ transient amplitude and time to 75% decline of the Ca2+ transient76. The researchers did not report on relaxation velocity. A subsequent study showed that TA1, a CK-136 derivative, improved cardiac contractile performance (developed pressure) in healthy rats but did not alter heart rate, diastolic pressure or energetic expenditure77. The researchers suggest that by directly targeting the sarcomere, TA1 can increase Ca2+ sensitivity and contractility without off-target effects such as increased cAMP signalling and Ca2+ cycling, which exacerbate the energetic deficits reported with other inotropes (such as dobutamine). A phase I clinical study was completed by Amgen, but no results have been made available (Table 1).
Myosin activators
As an alternative to sensitizing the regulatory proteins to Ca2+, increased cardiac contractility can also be achieved by directly activating cardiac myosin (Fig. 3). In 2010, omecamtiv mecarbil was discovered by high-throughput screening as the first selective, small-molecule activator of cardiac myosin heavy chain-α and cardiac myosin heavy chain-β78, but not of other striated-muscle myosins13. Omecamtiv mecarbil was initially described as a direct myosin activator that accelerates the actin-activated rate of phosphate release and structurally primes the myosin molecule for interaction with actin13,79-82. Paradoxically, omecamtiv mecarbil actually inhibits the working stroke of myosin82 and is predicted to stabilize the SRX state of myosin83. Although structural studies suggest that omecamtiv mecarbil does not induce the IHM state of myosin25, omecamtiv mecarbil does stabilize the pre-powerstroke state of myosin84, which has been associated with the SRX state46. Therefore, omecamtiv mecarbil might stabilize a structurally distinct SRX state, although the direct effects of omecamtiv mecarbil on the SRX state have not been measured.
Fig. 3 ∣. The molecular mechanisms of myosin modulation by targeted small molecules.
The myosin heavy chain has a primary role in muscle contraction through the formation of force-generating cross-bridges with actin thin filaments. The steps in the chemomechanical ATPase cycle of myosin are depicted in the context of the basic structural changes, energetics and their role in force production. Small molecules have been developed that target cardiac myosin. Aficamten, blebbistatin and mavacamten all decrease the rate of inorganic phosphate (Pi) release from myosin, leading to reduced ATPase activity of myosin and contractile force production79-82,108,127,128. Additionally, mavacamten increases the population of myosin heads in the autoinhibited, super-relaxed (SRX) state46,47, whereas blebbistatin, and probably also aficamten, exert their inhibitory effects in an SRX-independent mechanism in cardiac muscle49,129. Danicamtiv and omecamtiv mecarbil both increase the rate of Pi release from myosin, thereby increasing the ATPase activity and contractile force79-82,97,98. Danicamtiv also increases the number of myosin heads available for actomyosin cross-bridge formation97, which contributes to the increase in contractile force98. DRX, disordered relaxed state of myosin.
Despite these inhibitor-like molecular mechanisms, the positive inotropic effects of omecamtiv mecarbil have been explained by simulating the cooperative activation of the thin filament82. By prolonging the actomyosin attachment of a small population of non-force-generating myosins, omecamtiv mecarbil activates the thin filament to recruit more drug-free myosins for cross-bridge cycles85, with the net result of increasing the number of myosin heads in a force-generating state. This mechanism is highly dependent on the concentration of the drug: higher doses slowed relaxation to the point of ischaemia, as also demonstrated in preclinical and clinical studies13,86-88. Indeed, at higher doses, omecamtiv mecarbil increased myocardial oxygen consumption in a pig model with left ventricular dysfunction86, but at a lower dose, no changes in myocardial oxygen consumption were observed in rat and dog models of heart failure13,87. Additionally, adverse effects were observed at higher concentrations of omecamtiv mecarbil in the first-in-human study, with signs of myocardial ischaemia due to prolonged contraction88.
In the COSMIC-HF trial89, which assessed the maximum concentration of omecamtiv mecarbil in the plasma as the primary end point, patients with heart failure with reduced ejection fraction had significant improvements in all the secondary end points of left ventricular systolic ejection time, stroke volume and left ventricular end-systolic diameter, as well as a reduced plasma level of N-terminal pro-B-type natriuretic peptide (NT-proBNP). In the follow-up COSMIC-HF study90, patients with heart failure with reduced ejection fraction who were treated with omecamtiv mecarbil also had improved right ventricular structure and function compared with the placebo group. Right ventricular dysfunction commonly results from elevated left ventricular pressure in patients with heart failure and is associated with poor clinical outcomes91. To date, one phase III trial of omecamtiv mecarbil has been completed (the GALACTIC-HF trial92) and another is ongoing (the METEORIC-HF trial93) (Table 1). Although the results of the GALACTIC-HF trial94 showed an absolute reduction in cardiac events or cardiovascular death of only 2.1% with omecamtiv mecarbil treatment, no evidence of an increased risk of myocardial ischaemic events, ventricular arrhythmias, or cardiovascular or all-cause death was found, confirming the safety of the drug dosage in humans. Omecamtiv mecarbil also seemed to be safe in conjunction with other standard therapies for heart failure, given that all the patients in the GALACTIC-HF study were receiving standard-of-care treatment. Importantly, the GALACTIC-HF study92,94 indicated that omecamtiv mecarbil was of greater benefit in patients with a left ventricular ejection fraction ≤28%. Additionally, a post-hoc analysis of the GALACTIC-HF data identified a clinically significant reduction in the composite end point of time to first cardiovascular event or cardiovascular death among patients classified with severe heart failure95. Therefore, omecamtiv mecarbil might hold promise for patients diagnosed with substantially reduced systolic function and severe heart failure, for whom current treatment options are limited.
A promising novel cardiac myotrope is danicamtiv, formerly known as MYK-491. Danicamtiv also targets cardiac myosin and selectively increases cardiac actomyosin activity by increasing phosphate release rates and myosin head availability96-98. In vivo, danicamtiv had little effect on left ventricular diastolic stiffness, relaxation and Ca2+ homeostasis in an experimental model of heart failure in dogs and in a phase IIa study in patients with heart failure with reduced ejection fraction96,99. Danicamtiv also improved left atrial volume and function99, an effect that warrants further investigation and which could be relevant in patients with atrial fibrillation associated with heart failure with reduced ejection fraction. Omecamtiv mecarbil and danicamtiv were directly compared in human engineered heart tissues from induced pluripotent stem cell-derived cardiomyocytes cultured onto decellularized porcine myocardial slices98. Although the two small molecules had similar effects on the engineered tissues, such as a dose- dependent positive inotropy associated with changes in contraction kinetics, danicamtiv seemed to improve systolic contraction at a smaller lusitropic cost than that with omecamtiv mecarbil98. Currently, patients are being recruited for an exploratory phase II clinical trial designed to test the use of danicamtiv to treat patients with DCM resulting from variants in either MYH7 or TTN100 (Table 1).
Sarcomeric contractile inhibitors
In this section, we discuss the efforts being made to develop small molecules to inhibit the thin filament and preclinical and clinical data on the molecular mechanisms of myosin-targeted inhibitors.
Thin filament inhibitors
A possible strategy to counteract hypercontractility is to desensitize thin filaments to Ca2+. In cardiomyocytes, myofilament Ca2+ sensitivity is reduced by β-adrenergic signalling via protein kinase A phosphorylation of troponin I at serine 23 and serine 24101,102. This mechanism is important to attenuate the myofilament force response to the overall positive inotropic effect of β-adrenergic signalling. β-Blockers have been used for five decades to treat a variety of cardiac diseases, including obstructive HCM103.
Furthermore, molecules developed to inhibit calmodulin104-106 could theoretically be used to reduce myofilament Ca2+ sensitivity owing to the structural homology between calmodulin and cardiac troponin C. However, different pharmacological classes of calmodulin antagonist have various effects on cardiac contractility; some increase contraction (such as bepridil), whereas others reduce contraction (such as W7)105. Currently, no molecules that selectively reduce Ca2+ sensitivity by acting directly on the thin filaments are clinically available, but some preclinical efforts are being made107.
Myosin inhibitors
Mavacamten (previously known as MYK-461) is a small molecule identified by MyoKardia (acquired by Bristol Myers Squibb) in a screening, with the objective to reduce sarcomere contractility in HCM14. This small molecule binds to myosin and decreases the rate of inorganic phosphate release, stabilizing the SRX conformation of myosin46,47,108 (Fig. 3). Interestingly, in skeletal muscle, mavacamten increased the SRX population of myosin molecules that were not located in the MyBP-C zone35, suggesting a spatially distinct effect of this pharmacological intervention in the sarcomere.
Mavacamten reduced myosin ATPase activity in mouse and bovine cardiac myofibrils, and reduced fractional shortening with no changes on Ca2+ transients in rat cardiomyocytes14 In vivo preclinical studies on mouse, cat, dog and minipig models of HCM showed that mavacamten treatment reduced fractional shortening, cardiac fibrosis and cardiomyocyte disarray14,46,109,110. Importantly, mavacamten has been shown to reduce diastolic stiffness, probably owing to stabilization of the SRX state of myosin, while still maintaining the length-dependent increase in Ca2+ sensitivity (the Frank–Starling relationship) in response to sarcomere stretch111,112. Mavacamten has also been shown to increase the rate of ADP release from myosin, ATP attachment to myosin and the kinetics of cross-bridge detachment in mouse cardiac strips112 and human tissues111,113, all of which contribute to reduced diastolic stiffness.
Preclinical studies on the use of a myosin inhibitor in the setting of non-myosin-based disease are very promising. For example, mavacamten can reverse the increase in active myosin motors caused by genetic variants in either MYH7 or MYBPC3110,114, despite the discrete localization of mavacamten outside of the C-zone of the sarcomere35. Mavacamten also decreased maximum contractile force and Ca2+ sensitivity in a mouse model of HCM caused by a point mutation in Myl2, encoding the ventricular/cardiac muscle isoform of the myosin regulatory light chain 2113. Mavacamten also partially reversed the increased myofilament Ca2+ sensitivity and dysregulation of Ca2+ flux in cardiomyocyte models of HCM-causing variants in TNNI3 and TNNT2, encoding cardiac troponin I and troponin T, respectively115.
In the phase II MAVERICK-HCM trial116, mavacamten was well tolerated in patients with non-obstructive HCM and was associated with significant reductions in plasma NT-proBNP and cardiac troponin I levels compared with placebo. Patients with obstructive HCM also had a reduction in symptoms with mavacamten in the open-label, phase II PIONEER-HCM trial117 and the phase III EXPLORER-HCM trial118. In the EXPLORER-HCM trial119, the primary end point (a ≥1.5 ml/kg/min increase in peak oxygen consumption and an improvement by at least one NYHA class or a ≥3.0 ml/kg/min increase in peak oxygen consumption with no worsening in NYHA class) occurred in 37% of patients in the mavacamten group and 17% of patients in the placebo group (P < 0.0005) (Table 1). One of the more promising aspects of this trial was that patients with known variants in genes encoding sarcomeric proteins as well as those without known variants responded well to mavacamten119. A subset of patients in the phase III EXPLORER-HCM trial also underwent cardiac MRI, showing that mavacamten treatment significantly reduced left ventricular mass, left atrial mass index and left ventricular wall thickness, with no changes in cardiac fibrosis compared with placebo120. These structural changes correlated with significant reductions in the plasma levels of NT-proBNP and high-sensitivity cardiac troponin I120. A long-term extension study of the safety of mavacamten (the MAVA-LTE trial121) is ongoing in patients with HCM who have completed the MAVERICK-HCM or EXPLORER-HCM trials. Preliminary, interim results on the MAVERICK-HCM cohort indicate that long-term treatment with mavacamten (data presented through to week 48) is generally well tolerated, with only mild or moderate adverse events122. These patients had a significant reduction in plasma NT-proBNP level (by 67% at week 48) and improved left ventricular relaxation and diastolic function122. A potential disadvantage of mavacamten is its slow clearance and half-life of ~9 days123, because these parameters increase the risk of drug toxicity, interactions with other drugs and longer times to achieve steady-state concentrations in the plasma.
Other small molecules (MYK-581 and MYK-224) with similarities to MYK-461 (that is, mavacamten) have been described. Preliminary preclinical data on MYK-581 have been presented at conferences124,125, and a phase I study of MYK-224 is ongoing according to the Australian New Zealand Clinical Trials Registry126.
A preclinical comparison of mavacamten and blebbistatin (a non-specific inhibitor of myosin II) revealed that these two small molecules work via different mechanisms. Blebbistatin has been shown to bind near to the phosphate-binding site in the myosin motor, inhibiting phosphate release and slowing myosin ATPase activity127,128 (Fig. 3). Unlike mavacamten, blebbistatin has minimal effect on the myosin SRX population in skeletal or cardiac myosin83. Instead, blebbistatin is likely to stabilize a relaxed state of myosin that is structurally distinct from the IHM24, but that still results in ultra-slow myosin ATPase activity.
Another myosin inhibitor has been developed by Cytokinetics called aficamten (also known as CK-274 or CK-3773274). Aficamten was shown to reduce myosin ATPase activity in bovine cardiac myofibrils and to reduce contractility in rat primary cardiomyocytes129. Although the effects of aficamten on the SRX state of myosin have not been reported, aficamten has been shown to bind to the ATP-binding pocket of myosin129, in a similar manner to that of blebbistatin. Therefore, aficamten is likely to stabilize a myosin off-state that is structurally distinct from the closed state induced by mavacamten83. Despite the predicted unique molecular mechanisms of inhibition, preliminary data indicate that aficamten reduces cardiac contractility in vivo in healthy rats and beagle dogs, similar to the effects of mavacamten130,131 (Fig. 3), as well as in a transgenic mouse model of HCM carrying the R403Q variant in myosin132. A detailed assessment of aficamten reported faster pharmacokinetics than those of mavacamten, with a predicted half-life in humans of 2.8 days, an actual half-life of 3.4 days and no substantial effect on cytochrome P450129,133, suggesting safety and tolerability of the drug for therapeutic interventions. Currently, the phase II REDWOOD-HCM trial134 is recruiting patients with outflow-tract obstruction HCM to assess the incidence of adverse events during dosing of aficamten (Table 1). Preliminary results presented at a conference and in a press release from the company135 report that aficamten treatment significantly reduced resting left ventricular outflow tract gradients and the plasma NT-proBNP level compared with placebo. Aficamten was also generally well tolerated, with no serious adverse events reported135. A phase III trial of aficamten is expected to start soon.
Conclusions
In the past 20 years, major scientific advances have been made in understanding the pathophysiology and genetics of cardiovascular disease. However, drug development for these diseases has been comparatively stagnant, in large part owing to the high costs and risks associated with the clinical management of cardiovascular disease136. Although druggable targets have been identified in the cardiac sarcomere for direct modulation of contractile function in these diseases, many unknowns remain in the field of sarcomere pharmacology. One major question, given the vast number of variants in such a large number of genes and several mechanisms of pathogenesis, is whether one or a small number of small molecules targeting sarcomeric proteins will be effective in a heterogeneous spectrum of diseases. Of note, mavacamten was effective in patients with HCM regardless of whether they had an identified variant in a gene encoding a sarcomeric protein119. Consistent with these results is a report that the proteomes of patients with HCM and variants in different genes encoding sarcomeric proteins converge downstream of the primary genetic variant137. Clinical data from patients receiving aficamten treatment will help to refine the question of whether mechanistically distinct small molecules, such as the myosin inhibitors aficamten and mavacamten, have more or less benefit in particular subgroups of patients with HCM.
A phase II clinical trial of danicamtiv has recruited patients with DCM and genetic variants in either MYH7 or TTN. The preclinical development of compounds targeting MyBP-C138 and research on their efficacy in patients with MYBPC3 or other gene variants will continue to explore the interactions between sarcomeric proteins and their direct versus indirect modulation in clinical management. Although the developmental and tissue-specific expression patterns of sarcomeric proteins complicate the development of compounds targeted to the sarcomere, the studies discussed in this Review highlight the benefit of directly modulating contractile function in cardiac muscle, supporting myofilament-targeted compounds as the future of muscle-based therapeutics.
Key points.
Variants in genes encoding sarcomeric proteins are a leading cause of cardiomyopathies, characterized by protein-specific molecular mechanisms of disease.
Sarcomeric proteins can be targeted by small molecules to directly modulate contractile function in cardiac muscle.
Small molecules that are targeted to the myosin heavy chain modulate enzymatic activity and/or availability of the motor, leading to an increase or decrease in force production.
Small molecules that target the myosin heavy chain, such as mavacamten and aficamten, can act via distinct molecular mechanisms that lead to altered myosin function.
Sarcomere pharmacology suggests that small molecules targeting a specific sarcomeric protein will be effective even in patients with a causal variant in a gene encoding another sarcomeric protein.
Footnotes
Competing interests
S.J.L. is currently a part-time employee of Edgewise Therapeutics. L.A.L. is on the scientific advisory board and has a sponsored research agreement with Bristol Myers Squibb/MyoKardia.
References
- 1.Geisterfer-Lowrance AA, et al. A molecular basis for familial hypertrophic cardiomyopathy: a beta cardiac myosin heavy chain gene missense mutation. Cell 62, 999–1006 (1990). [DOI] [PubMed] [Google Scholar]
- 2.Kamisago M et al. Mutations in sarcomere protein genes as a cause of dilated cardiomyopathy. N Engl J Med 343, 1688–1696 (2000). [DOI] [PubMed] [Google Scholar]
- 3.Herman D et al. Truncations of titin causing dilated cardiomyopathy. N Engl J Med 366, 619–628 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.McNally E & Mestroni L Dilated Cardiomyopathy: Genetic Determinants and Mechanisms. Circ Res 121, 731–748 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Spudich J Three perspectives on the molecular basis of hypercontractility caused by hypertrophic cardiomyopathy mutations. Pflugers Arch 471, 701–717 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ho C et al. Genotype and Lifetime Burden of Disease in Hypertrophic Cardiomyopathy: Insights from the Sarcomeric Human Cardiomyopathy Registry (SHaRe). Circulation 138, 1387–1398 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Repetti G, Toepfer C, Seidman J & Seidman C Novel Therapies for Prevention and Early Treatment of Cardiomyopathies. Circ Res 124, 1536–1550 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Said S, Dearani J, Ommen S & Schaff H Surgical treatment of hypertrophic cardiomyopathy. Expert Rev Cardiovasc Ther 11, 617–627 (2013). [DOI] [PubMed] [Google Scholar]
- 9.Mateo J & Gimeno J Alcohol septal ablation in hypertrophic cardiomyopathy. Glob Cardiol Sci Pract 2018, 30 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Verlinden N & Coons J Disopyramide for Hypertrophic Cardiomyopathy: A Pragmatic Reappraisal of an Old Drug. Pharmacotherapy 35, 1164–1172 (2015). [DOI] [PubMed] [Google Scholar]
- 11.Cardoso R et al. SGLT2 inhibitors decrease cardiovascular death and heart failure hospitalizations in patients with heart failure: A systematic review and meta-analysis. EClinicalMedicine 36, 100933 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Alsulami K & Marston S Small Molecules acting on Myofilaments as Treatments for Heart and Skeletal Muscle Diseases. Int J Mol Sci 21, 9599 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Malik FI et al. Cardiac myosin activation: a potential therapeutic approach for systolic heart failure. Science 331, 1439–1443 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Green EM et al. A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice. Science 351, 617–621 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Price H The skeletal muscle fiber in the light of electron microscope studies: A review. Am J Med 35, 589–605 (1963). [DOI] [PubMed] [Google Scholar]
- 16.Gordon A, Homsher E & Regnier M Regulation of Contraction in Striated Muscle. Physiol Rev 80, 853–924 (2000). [DOI] [PubMed] [Google Scholar]
- 17.Woodhead JL et al. Atomic model of a myosin filament in the relaxed state. Nature 436, 1195–1199 (2005). [DOI] [PubMed] [Google Scholar]
- 18.Jung H, Komatsu S, Ikebe M & Craig R Head-head and head-tail interaction: a general mechanism for switching off myosin II activity in cells. Mol Biol Cell 19, 3234–3242 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Zoghbi M, Woodhead J, Moss R & Craig R Three-dimensional structure of vertebrate cardiac muscle myosin filaments. Proc Natl Acad Sci USA 105, 2386–2390 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Stewart M, Franks-Skiba K, Chen S & Cooke R Myosin ATP turnover rate is a mechanism involved in thermogenesis in resting skeletal muscle fibers. Proc Natl Acad Sci USA 107, 430–435 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Hooijman P, Stewart M & Cooke R A New State of Cardiac Myosin with Very Slow ATP Turnover: A Potential Cardioprotective Mechanism in the Heart. Biophys J 100, 1969–1976 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Toepfer C et al. Myosin Sequestration Regulates Sarcomere Function, Cardiomyocyte Energetics, and Metabolism, Informing the Pathogenesis of Hypertrophic Cardiomyopathy. Circulation 141, 828–842 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Alamo L et al. Conserved Intramolecular Interactions Maintain Myosin Interacting- Heads Motifs Explaining Tarantula Muscle Super-Relaxed State Structural Basis. J Mol Biol 428, 1142–1164 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Ma W, Gong H & Irving T Myosin Head Configurations in Resting and Contracting Murine Skeletal Muscle. Int J Mol Sci 19, 2643 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Robert-Paganin J, Auguin D & Houdusse A Hypertrophic cardiomyopathy disease results from disparate impairments of cardiac myosin function and auto-inhibition. Nat Commun 9, 4019 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Chu S, Muretta J & Thomas D Direct detection of the myosin super-relaxed state and interacting-heads motif in solution. J Biol Chem 297, 101157 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Knupp C, Morris E & Squire J The Interacting Head Motif Structure Does Not Explain the X-Ray Diffraction Patterns in Relaxed Vertebrate (Bony Fish) Skeletal Muscle and Insect (Lethocerus) Flight Muscle. Biology (Basel) 8, 67 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Brunello E et al. Myosin filament-based regulation of the dynamics of contraction in heart muscle. Proc Natl Acad Sci USA 117, 8177–8186 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Crocini C & Gotthardt M Cardiac sarcomere mechanics in health and disease. Biophysical Reviews 13, 637–652 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Rahmanseresht S et al. The N terminus of myosin-binding protein C extends toward actin filaments in intact cardiac muscle. J Gen Physiol 153, e202012726 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Inchingolo A, Previs S, Previs M, Warshaw D & Kad N Revealing the mechanism of how cardiac myosin-binding protein C N-terminal fragments sensitize thin filaments for myosin binding. Proc Natl Acad Sci USA 116, 6828–6835 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Li A et al. Skeletal MyBP-C isoforms tune the molecular contractility of divergent skeletal muscle systems. Proc Natl Acad Sci USA 116, 21882–21892 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Moss R, Fitzsimons D & Ralphe J Cardiac MyBP-C regulates the rate and force of contraction in mammalian myocardium. Circ Res 116, 183–192 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.McNamara J et al. Ablation of cardiac myosin binding protein-C disrupts the super-relaxed state of myosin in murine cardiomyocytes. J Mol Cell Cardiol 94, 65–71 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Nelson S, Li A, Beck-Previs S, Kennedy G & Warshaw D Imaging ATP Consumption in Resting Skeletal Muscle: One Molecule at a Time. Biophys J 119, 1050–1055 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Kensler R, Craig R & Moss R Phosphorylation of cardiac myosin binding protein C releases myosin heads from the surface of cardiac thick filaments. Proc Natl Acad Sci USA 114, E1355–EE1364 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Sarkar S et al. The hypertrophic cardiomyopathy mutations R403Q and R663H increase the number of myosin heads available to interact with actin. Sci Adv 6, eaax0069 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.McNamara J, Singh R & Sadayappan S Cardiac myosin binding protein-C phosphorylation regulates the super-relaxed state of myosin. Proc Natl Acad Sci USA 116, 11731–11736 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Walsh R, Offerhaus J, Tadros R & Bezzina C Minor hypertrophic cardiomyopathy genes, major insights into the genetics of cardiomyopathies. Nat Rev Cardiol 10.1038/s41569-021-00608-2 (2021). [DOI] [PubMed] [Google Scholar]
- 40.Marian A & Braunwald E Hypertrophic Cardiomyopathy: Genetics, Pathogenesis, Clinical Manifestations, Diagnosis, and Therapy. Circ Res 121, 749–770 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Glazier A, Thompson A & Day S Allelic imbalance and haploinsufficiency in MYBPC3-linked hypertrophic cardiomyopathy. Pflugers Arch 471, 781–793 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Yang Q et al. A mouse model of myosin binding protein C human familial hypertrophic cardiomyopathy. J Clin Invest 102, 1292–1300 (1998). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Helms A et al. Effects of MYBPC3 loss-of-function mutations preceding hypertrophic cardiomyopathy. JCI Insight 5, e133782 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Schmid M & Toepfer C Cardiac myosin super relaxation (SRX): a perspective on fundamental biology, human disease and therapeutics. Biol Open 10, bio.057646 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.McNamara J et al. MYBPC3 mutations are associated with a reduced super-relaxed state in patients with hypertrophic cardiomyopathy. PLoS One 12, e0180064 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Anderson R 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 USA 115, E8143–E8152 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Rohde J, Roopnarine O, Thomas D & Muretta J Mavacamten stabilizes an autoinhibited state of two-headed cardiac myosin. Proc Natl Acad Sci USA 115, E7486–E7494 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Adhikari A et al. β-Cardiac myosin hypertrophic cardiomyopathy mutations release sequestered heads and increase enzymatic activity. Nat Commun 10, 2685 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Gollapudi S, Yu M, Gan Q & Nag S Synthetic thick filaments: A new avenue for better understanding the myosin super-relaxed state in healthy, diseased, and mavacamten-treated cardiac systems. J Biol Chem 296, 100114 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Alamo L et al. Effects of myosin variants on interacting-heads motif explain distinct hypertrophic and dilated cardiomyopathy phenotypes. eLife 6, e24634 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Dellefave L & McNally E The genetics of dilated cardiomyopathy. Curr Opin Cardiol 25, 198–204 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ceco E et al. Targeting latent TGFβ release in muscular dystrophy. Sci Transl Med 6, 259ra144 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Roncarati R et al. Doubly heterozygous LMNA and TIN mutations revealed by exome sequencing in a severe form of dilated cardiomyopathy. Eur J Hum Genet 21, 1105–1111 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Gerull B et al. Mutations of TTN, encoding the giant muscle filament titin, cause familial dilated cardiomyopathy. Nat Genet 30, 201–204 (2002). [DOI] [PubMed] [Google Scholar]
- 55.Ware J & Cook S Role of titin in cardiomyopathy: from DNA variants to patient stratification. Nat Rev Cardiol 15, 241–252 (2017). [DOI] [PubMed] [Google Scholar]
- 56.McAfee Q et al. Truncated titin proteins in dilated cardiomyopathy. Sci Transl Med 13, eabd7287 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Nagy L, Pollesello P & Papp Z Inotropes and inodilators for acute heart failure: sarcomere active drugs in focus. J Cardiovasc Pharmacol 64, 199–208 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Herzig JW, Feile K & Ruegg JC Activating effects of AR-L 115 BS on the Ca2+ sensitive force, stiffness and unloaded shortening velocity (Vmax) in isolated contractile structures from mammalian heart muscle. Arzneimittel-Forschung 31, 188–191 (1981). [PubMed] [Google Scholar]
- 59.Solaro RJ & Ruegg JC Stimulation of Ca++ binding and ATPase activity of dog cardiac myofibrils by AR-L 115BS, a novel cardiotonic agent. Circ Res 51, 290–294 (1982). [DOI] [PubMed] [Google Scholar]
- 60.Raasmaja A et al. Biochemical properties of OR-1259—a positive inotropic and vasodilatory compound with an antiarrhythmic effect. Adv Exp Med Biol 311, 423 (1992). [DOI] [PubMed] [Google Scholar]
- 61.Haikala H et al. Cardiac troponin C as a target protein for a novel calcium sensitizing drug, levosimendan. J Mol Cell Cardiol 27, 1859–1866 (1995). [DOI] [PubMed] [Google Scholar]
- 62.Pollesello P et al. Binding of a new Ca2+ sensitizer, levosimendan, to recombinant human cardiac troponin C. A molecular modelling, fluorescence probe, and proton nuclear magnetic resonance study. J Biol Chem 269, 28584–28590 (1994). [PubMed] [Google Scholar]
- 63.Sorsa T, Pollesello P & Solaro R The contractile apparatus as a target for drugs against heart failure: interaction of levosimendan, a calcium sensitiser, with cardiac troponin c. Mol Cell Biochem 266, 87–107 (2004). [DOI] [PubMed] [Google Scholar]
- 64.Sorsa T et al. Binding of levosimendan, a calcium sensitizer, to cardiac troponin C. J Biol Chem 276, 9337–9343 (2001). [DOI] [PubMed] [Google Scholar]
- 65.Haikala H & Linden I Mechanisms of action of calcium-sensitizing drugs. J Cardiovasc Pharmacol 26 (Suppl. 1), S10–S19 (1995). [PubMed] [Google Scholar]
- 66.Boknik P et al. Mechanisms of the contractile effects of levosimendan in the mammalian heart. J Pharmacol Exp Ther 280, 277–283 (1997). [PubMed] [Google Scholar]
- 67.Szilagyi S et al. The effects of levosimendan and OR-1896 on isolated hearts, myocyte-sized preparations and phosphodiesterase enzymes of the guinea pig. Eur J Pharmacol 486, 67–74 (2004). [DOI] [PubMed] [Google Scholar]
- 68.Orstavik O et al. The inotropic effect of the active metabolite of levosimendan, OR-1896, is mediated through inhibition of PDE3 in rat ventricular myocardium. PLoS One 10, e0115547 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Follath F et al. Efficacy and safety of intravenous levosimendan compared with dobutamine in severe low-output heart failure (the LIDO study): a randomised double-blind trial. Lancet 360, 196–202 (2002). [DOI] [PubMed] [Google Scholar]
- 70.Mebazaa A et al. Levosimendan vs dobutamine for patients with acute decompensated heart failure: the SURVIVE randomized trial. JAMA 297, 1883–1891 (2007). [DOI] [PubMed] [Google Scholar]
- 71.Landoni G et al. Levosimendan for Hemodynamic Support after Cardiac Surgery. N Engl J Med 376, 2021–2031 (2017). [DOI] [PubMed] [Google Scholar]
- 72.Mehta RH et al. Levosimendan in Patients with Left Ventricular Dysfunction Undergoing Cardiac Surgery. N Engl J Med 376, 2032–2042 (2017). [DOI] [PubMed] [Google Scholar]
- 73.Cholley B et al. Effect of Levosimendan on Low Cardiac Output Syndrome in Patients With Low Ejection Fraction Undergoing Coronary Artery Bypass Grafting With Cardiopulmonary Bypass: The LICORN Randomized Clinical Trial. JAMA 318, 548–556 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Packer M et al. Effect of levosimendan on the short-term clinical course of patients with acutely decompensated heart failure. JACC Heart Fail 1, 103–111 (2013). [DOI] [PubMed] [Google Scholar]
- 75.Harjola V et al. Organ dysfunction, injury and failure in acute heart failure: from pathophysiology to diagnosis and management. A review on behalf of the Acute Heart Failure Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur J Heart Fail 19, 821–836 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Reagan JD et al. The novel myotrope, AMG 594, is a small-molecule cardiac troponin activator that increases cardiac contractility in vitro and in vivo [abstract]. Presented at the Keystone Symposium on Charting a New Course for Heart Failure: From Discovery to Data. https://cytokinetics.com/wp-content/uploads/2020/03/Keystone_HF_2020_AMG594.pdf (2020). [Google Scholar]
- 77.He H et al. A Novel Small Molecule Troponin Activator Increases Cardiac Contractile Function Without Negative Impact on Energetics or Diastolic Function [abstract]. Circ Heart Fail 144, A9199 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Morgan BP et al. Discovery of omecamtiv mecarbil the first, selective, small molecule activator of cardiac Myosin. ACS Med Chem Lett 1, 472–477 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Liu Y, White HD, Belknap B, Winkelmann DA & Forgacs E Omecamtiv Mecarbil modulates the kinetic and motile properties of porcine beta-cardiac myosin. Biochemistry 54, 1963–1975 (2015). [DOI] [PubMed] [Google Scholar]
- 80.Rohde J, Thomas D & Muretta J Heart failure drug changes the mechanoenzymology of the cardiac myosin powerstroke. Proc Natl Acad Sci USA 114, E1796–E1804 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Swenson AM et al. Omecamtiv Mecarbil Enhances the Duty Ratio of Human beta- Cardiac Myosin Resulting in Increased Calcium Sensitivity and Slowed Force Development in Cardiac Muscle. JBiol Chem 292, 3768–3778 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Woody MS et al. Positive cardiac inotrope omecamtiv mecarbil activates muscle despite suppressing the myosin working stroke. Nat Commun 9, 3838 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Gollapudi S et al. Two classes of myosin inhibitors, blebbistatin, and mavacamten, stabilize β-cardiac myosin in different structural and functional states. J Mol Biol 433, 167295 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Planelles-Herrero V, Hartman J, Robert-Paganin J, Malik F & Houdusse A Mechanistic and structural basis for activation of cardiac myosin force production by omecamtiv mecarbil. Nat Commun 8, 190 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.McKillop DF & Geeves MA Regulation of the interaction between actin and myosin subfragment 1: evidence for three states of the thin filament. Biophys J 65, 693–701 (1993). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bakkehaug JP et al. Myosin Activator Omecamtiv Mecarbil Increases Myocardial Oxygen Consumption and Impairs Cardiac Efficiency Mediated by Resting Myosin ATPase Activity. Circ Heart Fail 8, 766–775 (2015). [DOI] [PubMed] [Google Scholar]
- 87.Shen YT et al. Improvement of cardiac function by a cardiac Myosin activator in conscious dogs with systolic heart failure. Circ Heart Fail 3, 522–527 (2010). [DOI] [PubMed] [Google Scholar]
- 88.Teerlink JR et al. Dose-dependent augmentation of cardiac systolic function with the selective cardiac myosin activator, omecamtiv mecarbil: a first-in-man study. Lancet 378, 667–675 (2011). [DOI] [PubMed] [Google Scholar]
- 89.Teerlink J et al. Chronic Oral Study of Myosin Activation to Increase Contractility in Heart Failure (COSMIC-HF): a phase 2, pharmacokinetic, randomised, placebo- controlled trial. Lancet 388, 2895–2903 (2016). [DOI] [PubMed] [Google Scholar]
- 90.Biering-Sørensen T et al. The effect of the cardiac myosin activator, omecamtiv mecarbil, on right ventricular structure and function in chronic systolic heart failure (COSMIC-HF). Eur J Heart Fail 23, 1052–1056 (2021). [DOI] [PubMed] [Google Scholar]
- 91.Guazzi M, Bandera F, Ozemek C, Systrom D & Arena R Cardiopulmonary Exercise Testing: What Is its Value? J Am Coll Cardiol 70, 1618–1636 (2017). [DOI] [PubMed] [Google Scholar]
- 92.Teerlink JR et al. Effect of Ejection Fraction on Clinical Outcomes in Patients Treated With Omecamtiv Mecarbil in GALACTIC-HF. J Am Coll Cardiol 78, 97–108 (2021). [DOI] [PubMed] [Google Scholar]
- 93.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03759392 (2022). [DOI] [PubMed] [Google Scholar]
- 94.Teerlink JR et al. Cardiac Myosin Activation with Omecamtiv Mecarbil in Systolic Heart Failure. N Engl J Med 384, 105–116 (2021). [DOI] [PubMed] [Google Scholar]
- 95.Felker G et al. Assessment of Omecamtiv Mecarbil for the Treatment of Patients With Severe Heart Failure. A Post Hoc Analysis of Data From the GALACTIC-HF Randomized Clinical Trial. JAMA Cardiol 7, 26–34 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Fernandes S et al. MYK-491, a Novel Small-Molecule Cardiac Myosin Activator Increases Cardiac Systolic Function and Preserves Mechanical Efficiency: Pre-Clinical in vivo and in vitro Evidence [abstract]. Circulation 140, A15707 (2019). [Google Scholar]
- 97.Bell K, Anto AR, Anderson RL, Del Rio CL & Henze M Cardiac muscle activation; the role of length dependent activation and the novel myosin activator danicamtiv. Eur Heart J 41 (Suppl. 2), ehaa946.3681 (2020). [Google Scholar]
- 98.Shen S, Sewanan LR, Jacoby DL & Campbell SG Danicamtiv Enhances Systolic Function and Frank-Starling Behavior at Minimal Diastolic Cost in Engineered Human Myocardium. J Am Heart Assoc 10, e020860 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Voors A et al. Effects of danicamtiv, a novel cardiac myosin activator, in heart failure with reduced ejection fraction: experimental data and clinical results from a phase 2a trial. Eur J Heart Fail 22, 1649–1658 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04572893 (2021). [DOI] [PubMed] [Google Scholar]
- 101.Zhang R, Zhao J & Potter JD Phosphorylation of both serine residues in cardiac troponin I is required to decrease the Ca2+ affinity of cardiac troponin C. J Biol Chem 270, 30773–30780 (1995). [DOI] [PubMed] [Google Scholar]
- 102.Kentish JC et al. Phosphorylation of troponin I by protein kinase A accelerates relaxation and crossbridge cycle kinetics in mouse ventricular muscle. Circ Res 88, 1059–1065 (2001). [DOI] [PubMed] [Google Scholar]
- 103.Ommen S et al. 2020 AHA/ACC Guideline for the Diagnosis and Treatment of Patients With Hypertrophic Cardiomyopathy. Circulation 142, e558–e631 (2020). [DOI] [PubMed] [Google Scholar]
- 104.Hidaka H et al. Calcium-regulated modulator protein interacting agents inhibit smooth muscle calcium-stimulated protein kinase and ATPase. Mol Pharmacol 17, 66–72 (1980). [PubMed] [Google Scholar]
- 105.Silver PJ, Pinto PB & Dachiw J Modulation of vascular and cardiac contractile protein regulatory mechanisms by calmodulin inhibitors and related compounds. Biochem Pharmacol 35, 2545–2551 (1986). [DOI] [PubMed] [Google Scholar]
- 106.Osawa M et al. Solution structure of calmodulin-W-7 complex: the basis of diversity in molecular recognition. J Mol Biol 276, 165–176 (1998). [DOI] [PubMed] [Google Scholar]
- 107.Thompson BR, Martindale J & Metzger JM Sarcomere neutralization in inherited cardiomyopathy: small-molecule proof-of-concept to correct hyper-Ca2+-sensitive myofilaments. Am J Physiol Heart Circ Physiol 311, H36–H43 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Kawas RF et al. A small-molecule modulator of cardiac myosin acts on multiple stages of the myosin chemomechanical cycle. J Biol Chem 292, 16571–16577 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.del Rio CL et al. In vivo Cardiac Effects of Mavacamten (MYK-461): Evidence for Negative Inotropy and Improved Compliance [abstract]. Circulation 136, A20593 (2017). [Google Scholar]
- 110.Stern J et al. A Small Molecule Inhibitor of Sarcomere Contractility Acutely Relieves Left Ventricular Outflow Tract Obstruction in Feline Hypertrophic Cardiomyopathy. PLoS One 11, e0168407 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Sewanan L, Shen S & Campbell S Mavacamten preserves length-dependent contractility and improves diastolic function in human engineered heart tissue. Am J Physiol Heart Circ Physiol 320, H1112–H1123 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Awinda P et al. Effects of mavacamten on Ca2+ sensitivity of contraction as sarcomere length varied in human myocardium. Br J Pharmacol 177, 5609–5621 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Awinda P et al. Mavacamten decreases maximal force and Ca2+ sensitivity in the N47K-myosin regulatory light chain mouse model of hypertrophic cardiomyopathy. Am J Physiol Heart Circ Physiol 320, H881–H890 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Toepfer C et al. Hypertrophic cardiomyopathy mutations in MYBPC3 dysregulate myosin. Sci Transl Med 11, eaat1199 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115.Sparrow A, Watkins H, Daniels M, Redwood C & Robinson P Mavacamten rescues increased myofilament calcium sensitivity and dysregulation of Ca2+ flux caused by thin filament hypertrophic cardiomyopathy mutations. Am J Physiol Heart Circ Physiol 318, H715–H722 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116.Ho C et al. Evaluation of Mavacamten in Symptomatic Patients With Nonobstructive Hypertrophic Cardiomyopathy. J Am Coll Cardiol 75, 2649–2660 (2020). [DOI] [PubMed] [Google Scholar]
- 117.Heitner S et al. Mavacamten Treatment for Obstructive Hypertrophic Cardiomyopathy: A Clinical Trial. Ann Intern Med 170, 741–748 (2019). [DOI] [PubMed] [Google Scholar]
- 118.Ho C et al. Study Design and Rationale of EXPLORER-HCM: Evaluation of Mavacamten in Adults With Symptomatic Obstructive Hypertrophic Cardiomyopathy. Circ Heart Fail 13, e006853 (2020). [DOI] [PubMed] [Google Scholar]
- 119.Olivotto I et al. Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (EXPLORER-HCM): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 396, 759–769 (2020). [DOI] [PubMed] [Google Scholar]
- 120.Saberi S et al. Mavacamten Favorably Impacts Cardiac Structure in Obstructive Hypertrophic Cardiomyopathy: EXPLORER-HCM Cardiac Magnetic Resonance Substudy Analysis. Circulation 143, 606–608 (2021). [DOI] [PubMed] [Google Scholar]
- 121.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03723655 (2021). [DOI] [PubMed] [Google Scholar]
- 122.Owens A et al. Long-Term Efficacy and Safety of Mavacamten in Patients with Non-Obstructive Hypertrophic Cardiomyopathy: Interim Results from the MAVERICK-LTE Cohort of the MAVA-LTE Study [abstract]. Circulation 144, A9685 (2021). [Google Scholar]
- 123.Grillo MP et al. In vitro and in vivo pharmacokinetic characterization of mavacamten, a first-in-class small molecule allosteric modulator of beta cardiac myosin. Xenobiotica 49, 718–733 (2019). [DOI] [PubMed] [Google Scholar]
- 124.del Rio CL et al. 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 [abstract]. Circulation 140, A14585 (2019). [Google Scholar]
- 125.Ferguson BS et al. Acute effects of a mavacamten-like myosin-inhibitor (MYK-581 in a feline model of obstructed hypertrophic cardiomyopathy: evidence of improved ventricular filling (beyond obstruction reprieve). Eur Heart J 41 (Suppl. 2), ehaa946.3713 (2020). [Google Scholar]
- 126.Australian New Zealand Clinical Trials Registry. https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=378908 (2021).
- 127.Allingham J, Smith R & Rayment I The structural basis of blebbistatin inhibition and specificity for myosin II. Nat Struct Mol Biol 12, 378–379 (2005). [DOI] [PubMed] [Google Scholar]
- 128.Wilson C, Naber N, Pate E & Cooke R The myosin inhibitor blebbistatin stabilizes the super-relaxed state in skeletal muscle. Biophys J 107, 1637–1646 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Chuang C et al. Discovery of Aficamten (CK-274), a Next-Generation Cardiac Myosin Inhibitor for the Treatment of Hypertrophic Cardiomyopathy. J Med Chem 64, 14142–14152 (2021). [DOI] [PubMed] [Google Scholar]
- 130.Hwee D et al. Pharmacologic Characterization of the Cardiac Myosin Inhibitor, CK-3773274: A Potential Therapeutic Approach for Hypertrophic Cardiomyopathy [abstract]. Circ Res 125, A332 (2019). [Google Scholar]
- 131.Hartman J et al. Characterization of the Cardiac Myosin Inhibitor CK-3773274: A Potential Therapeutic Approach for Hypertrophic Cardiomyopathy. Biophys J 118, 596a (2020). [Google Scholar]
- 132.Hwee D et al. The Cardiac Myosin Inhibitor, CK-3773274, Reduces Contractility in the R403q Mouse Model of Hypertrophic 9Cardiomyopathy [abstract]. Circ Res 125, A615 (2019). [Google Scholar]
- 133.Cremin P et al. In Vivo Pharmacokinetic Characterization of CK-3773274, a Novel Cardiac Myosin Inhibitor [abstract]. AAPS 304702, 887215 (2020). [Google Scholar]
- 134.US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04219826 (2021). [DOI] [PubMed] [Google Scholar]
- 135.Cytokinetics. Cytokinetics announces positive topline results of REDWOOD-HCM. https://ir.cytokinetics.com/news-releases/news-release-details/cytokinetics-announces-positive-topline-results-redwood-hcm (2021).
- 136.Fordyce C et al. Cardiovascular drug development: is it dead or just hibernating? J Am Coll Cardiol 65, 1567–1582 (2015). [DOI] [PubMed] [Google Scholar]
- 137.Tucholski T et al. Distinct hypertrophic cardiomyopathy genotypes result in convergent sarcomeric proteoform profiles revealed by top-down proteomics. Proc Natl Acad Sci USA 117, 24691–24700 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Bunch T et al. Cardiac myosin-binding protein C interaction with actin is inhibited by compounds identified in a high-throughput fluorescence lifetime screen. J Biol Chem 297, 100840 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Nag S & Trivedi D To lie or not to lie: super-relaxing with myosins. eLife 10, e63703 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]



