Skip to main content
Log in

Novel Myosin-Based Therapies in Hypertrophic Cardiomyopathy

  • Heart Failure (W Tang, Section Editor)
  • Published:
Current Treatment Options in Cardiovascular Medicine Aims and scope Submit manuscript

Abstract

Purpose of review

Hypertrophic cardiomyopathy is the most common genetic disease of myocardium and is recognized to have substantial clinical impact including sudden cardiac death, arrhythmias, and heart failure. Historically medical therapies have been used in hypertrophic cardiomyopathy (HCM) with limited prospective randomized data to support their effectiveness. In this review, we discuss a novel class of drug that inhibits myosin (myo-blocker) which was developed to precisely target the mechanism of HCM in detail.

Recent findings

We discuss and contextualize the clinical trial results for mavacamten, positioning it in the therapeutic landscape as well as presenting future questions and further developments to be made in this area.

Summary

Many sarcomeric mutations driving hypertrophic cardiomyopathy seem to have a primary effect of causing hypercontractility which may drive many secondary problems such as structural remodeling, arrhythmias, and further contractile deficits. Myo-blockers such as the first-in-class mavacamten are small molecules that specifically target myosin to decrease contractility and have been developed to precisely ameliorate the hypercontractile mechanism of HCM. Mavacamten has been shown in large, randomized, double-blind clinical trials to have benefit generally in improving symptoms, decreasing LV outflow tract obstruction, and promoting beneficial remodeling in HCM with obstructive physiology.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Similar content being viewed by others

References and Recommended Reading

Papers of particular interest, published recently, have been highlighted as: • Of importance •• Of major importance

  1. Coats CJ, Hollman A. Hypertrophic cardiomyopathy: lessons from history. Vol. 94, Heart. 2008. p. 1258–63.

  2. TEARE D. Asymmetrical hypertrophy of the heart in young adults. Br Heart J 1958 Jan;20(1):1–8.

  3. Frank S, Braunwald E. Idiopathic hypertrophic subaortic stenosis. Clinical analysis of 126 patients with emphasis on the natural history. Circulation [Internet]. 1968 [cited 2021 Feb 12];37(5):759–88. Available from: http://ahajournals.org

  4. Ommen SR, Mital S, Burke MA, Day SM, Deswal A, Elliott P, et al. 2020 AHA/ACC guideline for the diagnosis and treatment of patients with hypertrophic cardiomyopathy. Circulation [Internet]. 2020 Dec 22 [cited 2021 Feb 1];142(25):558–631. Available from: http://ahajournals.org

  5. Marian AJ, Braunwald E. Hypertrophic cardiomyopathy: genetics, pathogenesis, clinical manifestations, diagnosis, and therapy. Circ Res. 2017;121:749–70.

    Article  CAS  Google Scholar 

  6. Tardiff JC. Sarcomeric proteins and familial hypertrophic cardiomyopathy: linking mutations in structural proteins to complex cardiovascular phenotypes. Heart Fail Rev. 2005;10:237–48.

    Article  CAS  Google Scholar 

  7. Braunwald E, Maron BJ. Eugene Braunwald, MD and the early years of hypertrophic cardiomyopathy: a conversation with Dr. Barry J. Maron. Am J Cardiol [Internet]. 2012 Jun 1 [cited 2018 Oct 4];109(11):1539–47. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22425332.

  8. Geisterfer-Lowrance AA, Kass S, Tanigawa G, Vosberg HP, McKenna W, Seidman CE, et al. A molecular basis for familial hypertrophic cardiomyopathy: a beta cardiac myosin heavy chain gene missense mutation. Cell [Internet]. 1990 Sep 7 [cited 2018 Oct 4];62(5):999–1006. Available from: http://www.ncbi.nlm.nih.gov/pubmed/1975517.

  9. Maron BJ, Maron MS, Semsarian C. Genetics of hypertrophic cardiomyopathy after 20 years. J Am Coll Cardiol. 2012;60:705–15.

    Article  Google Scholar 

  10. Walsh R, Thomson KL, Ware JS, Funke BH, Woodley J, McGuire KJ, et al. Reassessment of Mendelian gene pathogenicity using 7,855 cardiomyopathy cases and 60,706 reference samples. Genet Med [Internet]. 2017 Feb 17 [cited 2018 Jul 24];19(2):192–203. Available from: http://www.nature.com/doifinder/10.1038/gim.2016.90

  11. Alfares AA, Kelly MA, McDermott G, Funke BH, Lebo MS, Baxter SB, et al. Results of clinical genetic testing of 2,912 probands with hypertrophic cardiomyopathy: expanded panels offer limited additional sensitivity. Genet Med. 2015;17:880–8.

    Article  Google Scholar 

  12. •• Ho CY, Day SM, Ashley EA, Michels M, Pereira AC, Jacoby D, et al. Genotype and lifetime burden of disease in hypertrophic cardiomyopathy insights from the sarcomeric human cardiomyopathy registry (SHaRe). Circulation [Internet]. 2018 [cited 2021 Feb 7];138(14):1387–98. Available from: https://pubmed.ncbi.nlm.nih.gov/30297972/. In this paper, the most modern comprehensive perspective of HCM is reported from the largest database of HCM patients every gathered, with interesting insights in outcomes, phenotypes, and genetics.

  13. Harper AR, Goel A, Grace C, Thomson KL, Petersen SE, Xu X, et al. Common genetic variants and modifiable risk factors underpin hypertrophic cardiomyopathy susceptibility and expressivity. Nat Genet [Internet]. 2021 Feb 1 [cited 2021 Feb 20];58(10):135–42. Available from: https://doi.org/10.1038/s41588-020-00764-0

  14. Tadros R, Francis C, Xu X, Vermeer AMC, Harper AR, Huurman R, et al. Shared genetic pathways contribute to risk of hypertrophic and dilated cardiomyopathies with opposite directions of effect. Nat Genet [Internet]. 2021 Jan 25 [cited 2021 Feb 8];28(2):128–34. Available from: www.nature.com/naturegenetics128

  15. Marstrand P, Han L, Day SM, Olivotto I, Ashley EA, Michels M, et al. Hypertrophic cardiomyopathy with left ventricular systolic dysfunction: insights from the SHaRe Registry. Circulation [Internet]. 2020 Apr 28 [cited 2021 Feb 8];1371–83. Available from: http://ahajournals.org

  16. Czepluch FS, Wollnik B, Hasenfuß G. Genetic determinants of heart failure: facts and numbers. ESC Hear Fail [Internet]. 2018 Jun [cited 2018 Oct 4];5(3):211–7. Available from: http://www.ncbi.nlm.nih.gov/pubmed/29457878.

  17. Sen-Chowdhry S, Jacoby D, Moon JC, McKenna WJ. Update on hypertrophic cardiomyopathy and a guide to the guidelines [Internet]. Vol. 13, Nature Reviews Cardiology. Nature Publishing Group; 2016 [cited 2021 Feb 1]. p. 651–75. Available from: www.nature.com/nrcardio

  18. Witjas-Paalberends ER, Güclü A, Germans T, Knaapen P, Harms HJ, Vermeer AMC, et al. Gene-specific increase in the energetic cost of contraction in hypertrophic cardiomyopathy caused by thick filament mutations. Cardiovasc Res. 2014 Jul 15;103(2):248–57.

  19. Fumagalli C, De Gregorio MG, Zampieri M, Fedele E, Tomberli A, Chiriatti C, et al. Targeted medical therapies for hypertrophic cardiomyopathy [Internet]. Vol. 22, Current Cardiology Reports. Springer; 2020 [cited 2021 Feb 1]. Available from: https://pubmed.ncbi.nlm.nih.gov/31993794/

  20. Van Der Velden J, Ho CY, Tardiff JC, Olivotto I, Knollmann BC, Carrier L. Research priorities in sarcomeric cardiomyopathies. Cardiovascular Research. 2015.

  21. Maass A, Leinwand LA. Animal models of hypertrophic cardiomyopathy. Curr Opin Cardiol [Internet]. 2000 May [cited 2018 Sep 28];15(3):189–96. Available from: http://www.ncbi.nlm.nih.gov/pubmed/10952427.

  22. Sheikh F, Chen J. Mouse models for cardiomyopathy research. Prog Pediatr Cardiol [Internet]. 2007 Nov 1 [cited 2018 Sep 28];24(1):27–34. Available from: https://www.sciencedirect.com/science/article/pii/S105898130700080X

  23. Geisterfer-Lowrance AA, Christe M, Conner DA, Ingwall JS, Schoen FJ, Seidman CE, et al. A mouse model of familial hypertrophic cardiomyopathy. Science [Internet]. 1996 May 3 [cited 2018 Sep 28];272(5262):731–4. Available from: http://www.ncbi.nlm.nih.gov/pubmed/8614836.

  24. Redwood CS, Moolman-Smook JC, Watkins H. Properties of mutant contractile proteins that cause hypertrophic cardiomyopathy. Cardiovasc Res [Internet]. 1999 Oct 1 [cited 2018 Oct 3];44(1):20–36. Available from: https://academic.oup.com/cardiovascres/article/44/1/20/275023#3360858

  25. Sequeira V, Wijnker PJM, Nijenkamp LLAM, Kuster DWD, Najafi A, Witjas-Paalberends ER, et al. Perturbed length-dependent activation in human hypertrophic cardiomyopathy with missense sarcomeric gene mutations. Circ Res. 2013;

  26. Ashrafian H, Redwood C, Blair E, Watkins H. Hypertrophic cardiomyopathy:a paradigm for myocardial energy depletion. Trends Genet [Internet]. 2003 May 1 [cited 2018 Jul 8];19(5):263–8. Available from: https://www.sciencedirect.com/science/article/pii/S0168952503000817

  27. Ujfalusi Z, Vera CD, Mijailovich SM, Svicevic M, Yu EC, Kawana M, et al. Dilated cardiomyopathy myosin mutants have reduced force-generating capacity. J Biol Chem [Internet]. 2018 Jun 8 [cited 2018 Jul 6];293(23):9017–29. Available from: http://www.ncbi.nlm.nih.gov/pubmed/29666183.

  28. Van Der Velden J, Witjas-Paalberends ER, Stienen GJM, Dos Remedios C, Ten Cate FJ, Ho CY, et al. Increased energy utilization for force generation in human familial hypertrophic cardiomyopathy caused by sarcomere gene mutations. Eur Hear J. 2013;

  29. Coppini R, Ho CY, Ashley E, Day S, Ferrantini C, Girolami F, et al. Clinical phenotype and outcome of hypertrophic cardiomyopathy associated with thin-filament gene mutations. J Am Coll Cardiol [Internet]. 2014 Dec 23 [cited 2018 Jul 24];64(24):2589–600. Available from: http://linkinghub.elsevier.com/retrieve/pii/S0735109714066364

  30. Van Dijk SJ, Paalberends ; E Rosalie, Najafi A, Michels M, Sadayappan S, Carrier ; Lucie, et al. Contractile dysfunction irrespective of the mutant protein in human hypertrophic cardiomyopathy with normal systolic function. 2011 [cited 2018 Oct 3]; Available from: http://circheartfailure.ahajournals.org

  31. Crilley JG, Boehm EA, Blair E, Rajagopalan B, Blamire AM, Styles P, et al. Hypertrophic cardiomyopathy due to sarcomeric gene mutations is characterized by impaired energy metabolism irrespective of the degree of hypertrophy. J Am Coll Cardiol. 2003;

  32. Cohn R, Thakar K, Lowe A, Ladha F, Pettinato A, Meredith E, et al. A contraction stress model of hypertrophic cardiomyopathy due to thick filament sarcomere mutations. bioRxiv [Internet]. 2018 Sep 4 [cited 2018 Sep 17];408294. Available from: https://www.biorxiv.org/content/early/2018/09/04/408294

  33. Spudich JA, Aksel T, Bartholomew SR, Nag S, Kawana M, Yu EC, et al. Effects of hypertrophic and dilated cardiomyopathy mutations on power output by human β-cardiac myosin. J Exp Biol. 2016;

  34. Witjas-Paalberends ER, Güclü A, Germans T, Knaapen P, Harms HJ, Vermeer AMC, et al. Gene-specific increase in the energetic cost of contraction in hypertrophic cardiomyopathy caused by thick filament mutations. Cardiovasc Res. 2014;

  35. Bai F, Wang L, Kawai M. A study of tropomyosin’s role in cardiac function and disease using thin-filament reconstituted myocardium. Journal of Muscle Research and Cell Motility. 2013.

  36. Ferrantini C, Belus A, Piroddi N, Scellini B, Tesi C, Poggesi C. Mechanical and energetic consequences of HCM-causing mutations. Journal of Cardiovascular Translational Research. 2009.

  37. Lopes LR, Elliott PM. A straightforward guide to the sarcomeric basis of cardiomyopathies. Heart. 2014.

  38. Fraysse B, Weinberger F, Bardswell SC, Cuello F, Vignier N, Geertz B, et al. Increased myofilament Ca2+ sensitivity and diastolic dysfunction as early consequences of Mybpc3 mutation in heterozygous knock-in mice. J Mol Cell Cardiol [Internet]. 2012 Jun [cited 2018 Nov 15];52(6):1299–307. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22465693.

  39. Cohn R, Thakar K, Lowe A, Ladha FA, Pettinato AM, Romano R, et al. A contraction stress model of hypertrophic cardiomyopathy due to sarcomere mutations. Stem Cell Reports. 2019 Jan 8;12(1):71–83.

  40. Eschenhagen T, Carrier L. Cardiomyopathy phenotypes in human-induced pluripotent stem cell-derived cardiomyocytes—a systematic review. Pflügers Arch - Eur J Physiol [Internet]. 2018 Oct 15 [cited 2019 Jan 24];1–14. Available from: http://link.springer.com/10.1007/s00424-018-2214-0

  41. Sewanan LR, Campbell SG. Modelling sarcomeric cardiomyopathies with human cardiomyocytes derived from induced pluripotent stem cells. J Physiol. 2019.

  42. Sewanan LR, Schwan J, Kluger J, Park J, Jacoby DL, Qyang Y, et al. Extracellular matrix from hypertrophic myocardium provokes impaired twitch dynamics in healthy Cardiomyocytes. JACC Basic to Transl Sci. 2019 Aug;4(4):495–505.

  43. Wu H, Yang H, Rhee J-W, Zhang JZ, Lam CK, Sallam K, et al. Modelling diastolic dysfunction in induced pluripotent stem cell-derived cardiomyocytes from hypertrophic cardiomyopathy patients. Eur Heart J [Internet]. 2019 Jun 20 [cited 2019 Nov 10]; Available from: https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehz326/5521143

  44. Yotti R, Seidman CE, Seidman JG. Advances in the genetic basis and pathogenesis of sarcomere cardiomyopathies. Annu Rev Genomics Hum Genet [Internet]. 2019 Aug 31 [cited 2021 Feb 1];20(1):129–53. Available from: https://www.annualreviews.org/doi/10.1146/annurev-genom-083118-015306

  45. Mosqueira D, Smith JGW, Bhagwan JR, Denning C. Modeling hypertrophic cardiomyopathy: mechanistic insights and pharmacological intervention. Trends in Molecular Medicine. Elsevier Ltd; 2019.

  46. Mosqueira D, Mannhardt I, Bhagwan JR, Lis-Slimak K, Katili P, Scott E, et al. CRISPR/Cas9 editing in human pluripotent stem cell-cardiomyocytes highlights arrhythmias, hypocontractility, and energy depletion as potential therapeutic targets for hypertrophic cardiomyopathy. Eur Heart J [Internet]. 2018 May 8 [cited 2018 Sep 17]; Available from: https://academic.oup.com/eurheartj/advance-article/doi/10.1093/eurheartj/ehy249/4993867

  47. •• Heitner SB, Jacoby D, Lester SJ, Owens A, Wang A, Zhang D, et al. Mavacamten treatment for obstructive hypertrophic cardiomyopathy a clinical trial. Ann Intern Med. 2019 Jun 4;170(11):741–8. In this paper, the phase 2 trial of mavacamten in humans is detailed, including a wealth of interesting mechanistic detail in human that motivate its further usage.

  48. • Green EM, Wakimoto H, Anderson RL, Evanchik MJ, Gorham JM, Harrison BC, et al. A small-molecule inhibitor of sarcomere contractility suppresses hypertrophic cardiomyopathy in mice. Science [Internet]. 2016 Feb 5 [cited 2018 Dec 5];351(6273):617–21. Available from: http://www.sciencemag.org/cgi/doi/10.1126/science.aad3456. In this paper, the preclinical mouse data for mavacamten is reported, which really show that the drug is targeted to HCM hypercontractile mutations in a way that was not possible before.

  49. Kawas RF, Anderson RL, Bartholomew Ingle SR, Song Y, Sran AS, Rodriguez HM. A small-molecule modulator of cardiac myosin acts on multiple stages of the myosin chemomechanical cycle. J Biol Chem. 2017;292(40):16571–7.

    Article  CAS  Google Scholar 

  50. Sewanan LR, Moore JR, Lehman W, Campbell SG. Predicting effects of tropomyosin mutations on cardiac muscle contraction through myofilament modeling. Front Physiol. 2016;7(OCT).

  51. Campbell KS, Janssen PML, Campbell SG. Force-dependent recruitment from the myosin off state contributes to length-dependent activation. Biophys J. 2018 Aug 7;115(3):543–53.

  52. Campbell SG, Lionetti FV, Campbell KS, McCulloch AD. Coupling of adjacent tropomyosins enhances cross-bridge-mediated cooperative activation in a markov model of the cardiac thin filament. Biophys J. 2010;98(10):2254–64.

    Article  CAS  Google Scholar 

  53. Hooijman P, Stewart MA, Cooke R. A new state of cardiac myosin with very slow ATP turnover: a potential cardioprotective mechanism in the heart. Biophys J. 2011 Apr 20;100(8):1969–76.

  54. Anderson RL, Trivedi D V., Sarkar SS, Henze M, Ma W, Gong H, et al. Deciphering the super relaxed state of human β-cardiac myosin and the mode of action of mavacamten from myosin molecules to muscle fibers. Proc Natl Acad Sci U S A [Internet]. 2018 Aug 28 [cited 2021 Feb 1];115(35):E8143–52. Available from: /pmc/articles/PMC6126717/?report=abstract.

  55. Rohde JA, Roopnarine O, Thomas DD, Muretta JM. Mavacamten stabilizes an autoinhibited state of two-headed cardiac myosin. Proc Natl Acad Sci U S A. 2018 Aug 7;115(32):E7486–94.

  56. Spudich JA. The myosin mesa and a possible unifying hypothesis for the molecular basis of human hypertrophic cardiomyopathy. Biochem Soc Trans. 2015 Feb 1;43:64–72.

  57. Ait-Mou Y, Hsu K, Farman GP, Kumar M, Greaser ML, Irving TC, et al. Titin strain contributes to the frank-starling law of the heart by structural rearrangements of both thin- and thick-filament proteins. Proc Natl Acad Sci U S A [Internet]. 2016 Feb 23 [cited 2020 Nov 19];113(8):2306–11. Available from: www.pnas.org/cgi/doi/10.1073/pnas.1516732113

  58. Anderson RL, Trivedi DV, Sarkar SS, Henze M, Ma W, Gong H, et al. Deciphering the super relaxed state of human β-cardiac myosin and the mode of action of mavacamten from myosin molecules to muscle fibers. Proc Natl Acad Sci U S A. 2018 Aug 28;115(35):E8143–52.

  59. Sewanan LR, Shen S, Campbell SG. Mavacamten preserved length-dependent contractility and improved diastolic function in human engineered heart tissue. Am J Physiol Circ Physiol [Internet]. 2021 Jan 15 [cited 2021 Feb 1]; Available from: https://journals.physiology.org/doi/abs/10.1152/ajpheart.00325.2020

  60. Awinda PO, Watanabe M, Bishaw YM, Huckabee AM, Agonias KB, Kazmierczak K, et al. Mavacamten decreases maximal force and Ca 2+ −sensitivity in the N47K-myosin regulatory light chain mouse model of hypertrophic cardiomyopathy. Am J Physiol Circ Physiol [Internet]. 2020 Dec 18 [cited 2021 Feb 1]; Available from: https://journals.physiology.org/doi/abs/10.1152/ajpheart.00345.2020

  61. Toepfer CN, Wakimoto H, Garfinkel AC, McDonough B, Liao D, Jiang J, et al. Hypertrophic cardiomyopathy mutations in MYBPC3 dysregulate myosin. Sci Transl Med [Internet]. 2019 Jan 23 [cited 2021 Feb 1];11(476). Available from: https://www.pmc.com/articles/PMC7184965/?report=abstract.

  62. Janssen PML. Kinetics of cardiac muscle contraction and relaxation are linked and determined by properties of the cardiac sarcomere. Am J Physiol Circ Physiol [Internet]. 2010 Oct [cited 2018 Oct 4];299(4):H1092–9. Available from: http://www.physiology.org/doi/10.1152/ajpheart.00417.2010

  63. Davis J, Davis LC, Correll RN, Makarewich CA, Schwanekamp JA, Moussavi-Harami F, et al. A tension-based model distinguishes hypertrophic versus dilated cardiomyopathy. Cell. 2016 May 19;165(5):1147–59.

  64. Bytyçi I, Nistri S, Mörner S, Henein MY. Alcohol septal ablation versus septal myectomy treatment of obstructive hypertrophic cardiomyopathy: a systematic review and meta-analysis. J Clin Med [Internet]. 2020 Sep 23 [cited 2021 Feb 8];9(10):3062. Available from: https://pubmed.ncbi.nlm.nih.gov/32977442/

  65. •• Olivotto I, Oreziak A, Barriales-Villa R, Abraham TP, Masri A, Garcia-Pavia P, et al. Mavacamten for treatment of symptomatic obstructive hypertrophic cardiomyopathy (EXPLORER-HCM): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet. 2020 Sep 12;396(10253):759–69. In this paper, the definitive phase 3 trial results for mavacamten in oHCM are reported, which show its efficacy in a fairly unselective oHCM population.

  66. Ryall KA, Holland DO, Delaney KA, Kraeutler MJ, Parker AJ, Saucerman JJ. Network reconstruction and systems analysis of cardiac myocyte hypertrophy signaling. J Biol Chem. 2012 Dec 7;287(50):42259–68.

  67. Tan PM, Buchholz KS, Omens JH, McCulloch AD, Saucerman JJ. Predictive model identifies key network regulators of cardiomyocyte mechano-signaling. PLoS Comput Biol. 2017 Nov;1:13(11).

  68. Saberi S, Cardim N, Yamani MH, Schulz-Menger J, Li W, Florea V, et al. Mavacamten favorably impacts cardiac structure in obstructive hypertrophic cardiomyopathy: EXPLORER-HCM CMR substudy analysis. Circulation [Internet]. 2020 Nov 15 [cited 2021 Feb 1]; Available from: http://ahajournals.org

  69. Vikhorev PG, Vikhoreva NN. Cardiomyopathies and related changes in contractility of human heart muscle, vol. 19. MDPI AG: International Journal of Molecular Sciences; 2018.

    Google Scholar 

  70. Camacho P, Fan H, Liu Z, He JQ. Small mammalian animal models of heart disease. Vol. 6, American Journal of Cardiovascular Disease. E-Century Publishing Corporation; 2016. p. 70–80.

  71. Lynn ML, Lehman SJ, Tardiff JC. Biophysical derangements in genetic cardiomyopathies. Vol. 14, Heart Failure Clinics. Elsevier Inc.; 2018. p. 147–59.

  72. Sewanan LR, Campbell SG. Modelling sarcomeric cardiomyopathies with human cardiomyocytes derived from induced pluripotent stem cells. J Physiol. 2020;598(14).

  73. Ho CY, Mealiffe ME, Bach RG, Bhattacharya M, Choudhury L, Edelberg JM, et al. Evaluation of mavacamten in symptomatic patients with nonobstructive hypertrophic cardiomyopathy. J Am Coll Cardiol [Internet]. 2020 Jun 2 [cited 2021 Feb 1];75(21):2649–60. Available from: https://pubmed.ncbi.nlm.nih.gov/32466879/

  74. Wu H, Yang H, Rhee J-W, Zhang JZ, Lam CK, Sallam K, et al. Modelling diastolic dysfunction in induced pluripotent stem cell-derived cardiomyocytes from hypertrophic cardiomyopathy patients. Eur Heart J. 2019 Jun 20;

  75. Gaffin RD, Peña JR, Alves MSL, Dias FAL, Chowdhury SAK, Heinrich LS, et al. Long-term rescue of a familial hypertrophic cardiomyopathy caused by a mutation in the thin filament protein, tropomyosin, via modulation of a calcium cycling protein. J Mol Cell Cardiol. 2011 Nov;51(5):812–20.

  76. Viswanathan MC, Schmidt W, Rynkiewicz MJ, Agarwal K, Gao J, Katz J, et al. Distortion of the actin A-triad results in contractile disinhibition and cardiomyopathy. Cell Rep. 2017 Sep 12;20(11):2612–25.

  77. Bai F, Weis A, Takeda AK, Chase PB, Kawai M. Enhanced active cross-bridges during diastole: molecular pathogenesis of tropomyosin’s HCM mutations. Biophys J. 2011 Feb 16;100(4):1014–23.

  78. Ferrantini C, Coppini R, Pioner JM, Gentile F, Tosi B, Mazzoni L, et al. Pathogenesis of hypertrophic cardiomyopathy is mutation rather than disease specific: a comparison of the cardiac troponin T E163R and R92Q mouse models. J Am Heart Assoc [Internet]. 2017 Jul 11 [cited 2018 Dec 5];6(7). Available from: http://www.ncbi.nlm.nih.gov/pubmed/28735292.

  79. Ho CY, Day SM, Colan SD, Russell MW, Towbin JA, Sherrid M V, et al. The burden of early phenotypes and the influence of wall thickness in hypertrophic cardiomyopathy mutation carriers: findings from the HCMNet study. JAMA Cardiol [Internet]. 2017 Apr 1 [cited 2018 Dec 5];2(4):419–28. Available from: http://cardiology.jamanetwork.com/article.aspx?doi=10.1001/jamacardio.2016.5670

  80. Alsulami K, Marston S. Small molecules acting on myofilaments as treatments for heart and skeletal muscle diseases. Int J Mol Sci [Internet]. 2020 Dec 16 [cited 2021 Feb 12];21(24):9599. Available from: https://www.mdpi.com/1422-0067/21/24/9599

  81. Robertson LA, Armas DR, Robbie E, Osmukhina A, Li H, Malik FI, et al. A first in human study of the selective cardiac myosin inhibitor, CK-3773274. J Card Fail [Internet]. 2019 Aug 1 [cited 2021 Feb 1];25(8):S79–80. Available from: http://www.onlinejcf.com/article/S1071916419310000/fulltext

Download references

Funding

This work was supported by following sources of funding: P.D. Soros Fellowship for New Americans, National Institute of Health/National Institute of General Medical Sciences Medical Scientist Training Program Grant (T32GM007205), and American Heart Association Predoctoral Fellowship to LRS.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Daniel L. Jacoby MD.

Ethics declarations

Ethics Approval and Consent to Participate

This article does not contain any studies with human or animal subjects performed by any of the authors.

Conflict of Interest

Lorenzo R. Sewanan declares that he has no conflict of interest.

Daniel L. Jacoby has received personal fees from MyoKardia/BMS and Cytokinetics. Daniel L. Jacoby has a financial stake in Propria, LLC, and is a partial holder of a patent for the use of engineered heart tissue.

Additional information

Publisher’s Note

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

This article is part of the Topical Collection on Heart Failure

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sewanan, L.R., Jacoby, D.L. Novel Myosin-Based Therapies in Hypertrophic Cardiomyopathy. Curr Treat Options Cardio Med 23, 46 (2021). https://doi.org/10.1007/s11936-021-00921-6

Download citation

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1007/s11936-021-00921-6

Keywords

Navigation