Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Targeting the sarcomere in inherited cardiomyopathies

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.

Key points

  • Variants in genes encoding sarcomeric proteins are a leading cause of cardiomyopathies that are 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 myosin 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.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: The cardiac sarcomere and its components.
Fig. 2: The functional states of myosin.
Fig. 3: The molecular mechanisms of myosin modulation by targeted small molecules.

Similar content being viewed by others

References

  1. Geisterfer-Lowrance, A. A. et al. A molecular basis for familial hypertrophic cardiomyopathy: a β cardiac myosin heavy chain gene missense mutation. Cell 62, 999–1006 (1990).

    Article  CAS  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).

    Article  CAS  PubMed  Google Scholar 

  3. Herman, D. et al. Truncations of titin causing dilated cardiomyopathy. N. Engl. J. Med. 366, 619–628 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. McNally, E. & Mestroni, L. Dilated cardiomyopathy: genetic determinants and mechanisms. Circ. Res. 121, 731–748 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Spudich, J. Three perspectives on the molecular basis of hypercontractility caused by hypertrophic cardiomyopathy mutations. Pflug. Arch. 471, 701–717 (2019).

    Article  CAS  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).

    Article  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Said, S., Dearani, J., Ommen, S. & Schaff, H. Surgical treatment of hypertrophic cardiomyopathy. Expert Rev. Cardiovasc. Ther. 11, 617–627 (2013).

    Article  CAS  PubMed  Google Scholar 

  9. Mateo, J. & Gimeno, J. Alcohol septal ablation in hypertrophic cardiomyopathy. Glob. Cardiol. Sci. Pract. 2018, 30 (2018).

    PubMed  PubMed Central  Google Scholar 

  10. Verlinden, N. & Coons, J. Disopyramide for hypertrophic cardiomyopathy: a pragmatic reappraisal of an old drug. Pharmacotherapy 35, 1164–1172 (2015).

    Article  CAS  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).

    Article  PubMed  PubMed Central  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).

    Article  CAS  PubMed Central  Google Scholar 

  13. Malik, F. I. et al. Cardiac myosin activation: a potential therapeutic approach for systolic heart failure. Science 331, 1439–1443 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Green, E. M. et al. A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice. Science 351, 617–621 (2016).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  Google Scholar 

  16. Gordon, A., Homsher, E. & Regnier, M. Regulation of contraction in striated muscle. Physiol. Rev. 80, 853–924 (2000).

    Article  CAS  PubMed  Google Scholar 

  17. Woodhead, J. L. et al. Atomic model of a myosin filament in the relaxed state. Nature 436, 1195–1199 (2005).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  PubMed Central  CAS  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).

    Article  PubMed  PubMed Central  CAS  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).

    Article  CAS  PubMed  PubMed Central  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 8, 67 (2019).

    Article  CAS  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Crocini, C. & Gotthardt, M. Cardiac sarcomere mechanics in health and disease. Biophys. Rev. 13, 637–652 (2021).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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–E1364 (2017).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  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. 19, 151–167 (2022).

    Article  PubMed  Google Scholar 

  40. Marian, A. & Braunwald, E. Hypertrophic cardiomyopathy: genetics, pathogenesis, clinical manifestations, diagnosis, and therapy. Circ. Res. 121, 749–770 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Glazier, A., Thompson, A. & Day, S. Allelic imbalance and haploinsufficiency in MYBPC3-linked hypertrophic cardiomyopathy. Pflug. Arch. 471, 781–793 (2019).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Helms, A. et al. Effects of MYBPC3 loss-of-function mutations preceding hypertrophic cardiomyopathy. JCI Insight 5, e133782 (2020).

    Article  PubMed Central  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).

    Article  CAS  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).

    Article  PubMed  PubMed Central  CAS  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  PubMed  PubMed Central  CAS  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).

    Article  CAS  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).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Dellefave, L. & McNally, E. The genetics of dilated cardiomyopathy. Curr. Opin. Cardiol. 25, 198–204 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  52. Ceco, E. et al. Targeting latent TGFβ release in muscular dystrophy. Sci. Transl Med. 6, 259ra144 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  53. Roncarati, R. et al. Doubly heterozygous LMNA and TTN mutations revealed by exome sequencing in a severe form of dilated cardiomyopathy. Eur. J. Hum. Genet. 21, 1105–1111 (2013).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  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).

    Article  PubMed  CAS  Google Scholar 

  56. McAfee, Q. et al. Truncated titin proteins in dilated cardiomyopathy. Sci. Transl Med. 13, eabd7287 (2021).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Herzig, J. W., Feile, K. & Ruegg, J. C. 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. Arzneimittelforschung 31, 188–191 (1981).

    CAS  PubMed  Google Scholar 

  59. Solaro, R. J. & Ruegg, J. C. 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).

    Article  CAS  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).

    Article  CAS  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).

    Article  CAS  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).

    Article  CAS  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).

    Article  CAS  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).

    Article  CAS  PubMed  Google Scholar 

  65. Haikala, H. & Linden, I. Mechanisms of action of calcium-sensitizing drugs. J. Cardiovasc. Pharmacol. 26 (Suppl. 1), 10–19 (1995).

    Article  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).

    CAS  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).

    Article  CAS  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).

    Article  PubMed  PubMed Central  CAS  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).

    Article  CAS  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).

    Article  CAS  PubMed  Google Scholar 

  71. Landoni, G. et al. Levosimendan for hemodynamic support after cardiac surgery. N. Engl. J. Med. 376, 2021–2031 (2017).

    Article  CAS  PubMed  Google Scholar 

  72. Mehta, R. H. et al. Levosimendan in patients with left ventricular dysfunction undergoing cardiac surgery. N. Engl. J. Med. 376, 2032–2042 (2017).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  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).

    Article  PubMed  Google Scholar 

  76. Reagan, J. D. et al. The novel myotrope, AMG 594, is a small-molecule cardiac troponin activator that increases cardiac contractility in vitro and in vivo. Presented at the Keystone Symposium on Charting a New Course for Heart Failure: From Discovery to Data (Amgen and Cytokinetics, 2020).

  77. He, H. et al. A novel small molecule troponin activator increases cardiac contractile function without negative impact on energetics or diastolic function. Circ. Heart Fail. 144, A9199 (2021).

    Google Scholar 

  78. Morgan, B. P. et al. Discovery of omecamtiv mecarbil the first, selective, small molecule activator of cardiac myosin. ACS Med. Chem. Lett. 1, 472–477 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Liu, Y., White, H. D., Belknap, B., Winkelmann, D. A. & Forgacs, E. Omecamtiv mecarbil modulates the kinetic and motile properties of porcine β-cardiac myosin. Biochemistry 54, 1963–1975 (2015).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Swenson, A. M. et al. Omecamtiv mecarbil enhances the duty ratio of human β-cardiac myosin resulting in increased calcium sensitivity and slowed force development in cardiac muscle. J. Biol. Chem. 292, 3768–3778 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Woody, M. S. et al. Positive cardiac inotrope omecamtiv mecarbil activates muscle despite suppressing the myosin working stroke. Nat. Commun. 9, 3838 (2018).

    Article  PubMed  PubMed Central  CAS  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  85. McKillop, D. F. & Geeves, M. A. Regulation of the interaction between actin and myosin subfragment 1: evidence for three states of the thin filament. Biophys. J. 65, 693–701 (1993).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Bakkehaug, J. P. 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).

    Article  CAS  PubMed  Google Scholar 

  87. Shen, Y. T. 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).

    Article  PubMed  Google Scholar 

  88. Teerlink, J. R. 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).

    Article  CAS  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).

    Article  CAS  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).

    Article  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).

    Article  PubMed  Google Scholar 

  92. Teerlink, J. R. 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).

    Article  CAS  PubMed  Google Scholar 

  93. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03759392 (2022).

  94. Teerlink, J. R. et al. Cardiac myosin activation with omecamtiv mecarbil in systolic heart failure. N. Engl. J. Med. 384, 105–116 (2021).

    Article  CAS  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).

    Article  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, A. R., Anderson, R. L., Del Rio, C. L. & 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).

    Article  Google Scholar 

  98. Shen, S., Sewanan, L. R., Jacoby, D. L. & Campbell, S. G. Danicamtiv enhances systolic function and Frank-Starling behavior at minimal diastolic cost in engineered human myocardium. J. Am. Heart Assoc. 10, e020860 (2021).

    Article  PubMed  PubMed Central  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).

    Article  CAS  PubMed  Google Scholar 

  100. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04572893 (2021).

  101. Zhang, R., Zhao, J. & Potter, J. D. 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).

    Article  CAS  PubMed  Google Scholar 

  102. Kentish, J. C. 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).

    Article  CAS  PubMed  Google Scholar 

  103. Ommen, S. et al. 2020 AHA/ACC guideline for the diagnosis and treatment of patients with hypertrophic cardiomyopathy: a report of the American College of Cardiology/American Heart Association Joint Committee on clinical practice guidelines. Circulation 142, e558–e631 (2020).

    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).

    CAS  PubMed  Google Scholar 

  105. Silver, P. J., Pinto, P. B. & Dachiw, J. Modulation of vascular and cardiac contractile protein regulatory mechanisms by calmodulin inhibitors and related compounds. Biochem. Pharmacol. 35, 2545–2551 (1986).

    Article  CAS  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).

    Article  CAS  PubMed  Google Scholar 

  107. Thompson, B. R., Martindale, J. & Metzger, J. M. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  108. Kawas, R. F. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. del Rio, C. L. 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).

    Article  PubMed  PubMed Central  CAS  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  PubMed  Google Scholar 

  114. Toepfer, C. et al. Hypertrophic cardiomyopathy mutations in MYBPC3 dysregulate myosin. Sci. Transl Med. 11, eaat1199 (2019).

    Article  CAS  PubMed  PubMed Central  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).

    Article  PubMed  PubMed Central  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).

    Article  CAS  PubMed  Google Scholar 

  117. Heitner, S. et al. Mavacamten treatment for obstructive hypertrophic cardiomyopathy: a clinical trial. Ann. Intern. Med. 170, 741–748 (2019).

    Article  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).

    Article  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).

    Article  CAS  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).

    Article  PubMed  Google Scholar 

  121. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT03723655 (2021).

  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).

    Article  Google Scholar 

  123. Grillo, M. P. 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).

    Article  CAS  PubMed  Google Scholar 

  124. del Rio, C. L. 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, B. S. 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).

    Article  Google Scholar 

  126. Australian New Zealand Clinical Trials Registry. ANZCTR 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).

    Article  CAS  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).

    Article  CAS  PubMed  PubMed Central  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).

    Article  CAS  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).

    Article  Google Scholar 

  132. Hwee, D. et al. The cardiac myosin inhibitor, CK-3773274, reduces contractility in the R403q mouse model of hypertrophic cardiomyopathy [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. AAPS 304702, 887215 (2020).

  134. US National Library of Medicine. ClinicalTrials.gov https://clinicaltrials.gov/ct2/show/NCT04219826 (2021).

  135. BioSpace. Cytokinetics announces positive topline results from cohort 3 of REDWOOD-HCM. BioSpace https://www.biospace.com/article/releases/cytokinetics-announces-positive-topline-results-from-cohort-3-of-redwood-hcm (2022).

  136. Cytokinetics. Cytokinetics announces positive topline results of REDWOOD-HCM. Cytokinetics https://ir.cytokinetics.com/news-releases/news-release-details/cytokinetics-announces-positive-topline-results-redwood-hcm (2021).

  137. Fordyce, C. et al. Cardiovascular drug development: is it dead or just hibernating? J. Am. Coll. Cardiol. 65, 1567–1582 (2015).

    Article  PubMed  Google Scholar 

  138. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  139. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  140. Nag, S. & Trivedi, D. To lie or not to lie: super-relaxing with myosins. eLife 10, e63703 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed substantially to all aspects of the article.

Corresponding author

Correspondence to Leslie A. Leinwand.

Ethics declarations

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. C.C. declares no competing interests.

Peer review

Peer review information

Nature Reviews Cardiology thanks Sharlene Day, Ahmad Masri and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Lehman, S.J., Crocini, C. & Leinwand, L.A. Targeting the sarcomere in inherited cardiomyopathies. Nat Rev Cardiol 19, 353–363 (2022). https://doi.org/10.1038/s41569-022-00682-0

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41569-022-00682-0

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing