Skip to main content
Discoveries logoLink to Discoveries
. 2014 Dec 31;2(4):e33. doi: 10.15190/d.2014.25

New perspective in heart failure management: could myosin activators be the answer?

Stefan Andrei 1,*, Corina Iorgoveanu 2
PMCID: PMC6941581  PMID: 32309560

Abstract

Heart failure is a worldwide leading cause of hospital admissions. There is a critical need for new methods of management in acute heart failure. The current drug panel available in the treatment of hemodinamically unstable patients is not only limited, but it is also associated with deleterious side effects. Discoveries in the heart failure field seemed to lack an adequate answer and a change in paradigm may be necessary. The cardiac myosin activator omecamtiv mecarbil is a new therapeutic approach that improves the myocardial contractility through an innovative mechanism, avoiding the harmful effects of currently used inotropic agents. Several studies provided us with promising results, but further scientific proofs are needed.

Keywords: Heart failure, omecamtiv mecarbil, myosin activator

1. Introduction

Considerable progress has been made during the last decades in understanding, prevention and management of cardiovascular disorders (CVD), but cardiovascular-related health issues still remain the leading cause of death worldwide, accounting for almost 30% of global deaths1-3.

A variety of cardiac diseases can converge in time to heart failure (HF), a pathological condition characterized by heart’s inability to provide oxygen in response to tissular metabolic demands, with normal or increased filling pressures4. In the developed world it is estimated that 1-2% of general population have HF5. The pathophysiological mechanisms of this condition are complex and involve various adaptive interconnected neurohormonal pathways which enter a positive feedback loop with deleterious long term effects. HF may have a preserved (HFPEF) or a low ejection fraction (HFLEF), according to the pathophysiological stage. The actual therapeutical approach in chronic HFLEF is focused on neurohormonal antagonism with angiotensin enzyme inhibitors, beta-blockers and mineralocorticoid receptor antagonists, associated with preventive measures, risk factors and comorbidities management. Non surgical and surgical supportive devices play also a role4.

2. Inotropes - currently essential in acute care but avoided on long-term

A pharmacological class of drugs that is traditionally used to improve systolic function in patients with HF and to break the neurohormonal loop are the inotropes (Table 1). These agents stimulate heart contractility through various mechanisms, being particularly administered in order to control hemodynamic status in acute settings and to ameliorate symptoms in chronic patients4,6.

Table 1. Current inotropes and their mechanism of action13.

Agents that increase intracellular cAMP Agents that affect sarcolemmal ions, pumps and channels Agents that modulate the release or sensitization of proteins to intracellular calcium Agents with multiple mechanisms of action
Catecholamines (Beta Adrenergic Agonists)Natural: Adrenaline; Noradrenaline; Dopamine; Synthetic: Dobutamine; Isoprenaline; Dopexamine; Ibopamine*; Phosphodiesterase inhibitors: Milrinone; Amrinone; Enoximone* Digoxin Increased intracellular Ca release 1.Flosequinan * Increased Ca sensitization 2.Levosimendan* PDE III inhibitor/Ca sensitizer 1.Pimobendan * PDE III inhibitor/ modified rectifying K currents 2.Vesnarinone*
* not FDA approved

Cardiac muscle cells have a special design of sarcomeres, where fibers are coupled through intercalated discs which contain gap junctions that are involved in transmitting a depolarizing current along the fibers’ length7. According to the model suggested by Andrew Huxley et al., muscle contraction is achieved by myosin and actin sliding movement, a process made possible by ATP hydrolysis. A distinguishing trait of cardiac muscle is that calcium is released from both the sarcoplasmic reticulum and the extracellular matrix by a particular mechanism called calcium-induced calcium release. The action potential generates an influx of calcium through T tubules, which activates ryanodine receptors found on sarcoplasmic reticulum. The cytosolic calcium binds cTnC and subsequently tropomyosin is being shifted, releasing the myosin binding sites on actin8-11.When there is no ATP attached, myosin binds firmly to actin. ATP bound to the head of myosin leads to dissociation of myosin-actin complex by inducing a conformational change in myosin. After the ATP hydrolysis, its products -ADP and Pi- continue to be attached to the myosin, forming a very stable complex until the slow dissociation occurs. This last step is rate-limiting in the cross-bridge cycle8. During the contraction cycle only few myosins succeed at being recruited12.

Inotropic agents work either by augmenting calcium sensitization or by modulating the level of intracellular calcium, a process made possible through two main mechanisms: inhibition of the Na/Ca exchange pump or increasing the level of cAMP. The latter is achieved either by stimulation of adenylate cyclase or by inhibition of cAMP degradation via phosphodiesterase (PDE)13.

The downside of using inotropic agents in the treatment of acute heart failure is their possible adverse events. The main inotropic action mechanism is also the reason of potential development of cardiac arrhythmias14. Besides the fact that increased intracytosolic calcium is the main cause to this susceptibility to developing tachyarrhythmias, it also leads to more energy involved into calcium transport back in the sarcoplasmic reticulum through Ca-ATPase (SERCA) pumps, the aftereffect being increased oxygen demand14-16.

Another problem that should be taking into account is the possible hypotension, which is the result of increased cAMP in the peripheral vessels, leading to poor coronary perfusion15-17. Beta adrenergic agonists work by activating G protein, which has as an end-point increased levels of cAMP, through the action of adenylyl cyclase on ATP. In myocytes, this process leads to phosphorylation of L-type calcium channel and the subsequent influx of calcium activates ryanodine receptors, ultimately increasing Ca-mediated myocardial contractility. In the peripheral vasculature, activation of beta-2 adrenergic receptors leads to vasodilation, a process explained by the phosphorylation of the protein phospholamban, which makes possible the reuptake of calcium by the sarcoplasmic reticulum6,18.

Patients with decompensated heart failure, under chronic treatment with full-dose beta blockers, might not experience an adequate increase in cardiac output when dobutamine is administered. Instead, they could be treated with phosphodiesterase inhibitors (PDEI)19. On the other hand, PDEI should not be used in patients with reduced glomerular filtration rate, as milrinone is renally excreted6. It is demonstrated in OPTIME-CHF (Outcomes of Prospective Trial of Intravenous Milrinone for Exacerbations of Chronic Heart Failure ) study that the use of milrinone is not recommended in patients with decompensated heart failure not only because of tachyarrhythmias and hypotension events, but also for the fact that there is no decrease in the rate of mortality between Milrinone and placebo groups20.

Another category of inotropes, calcium sensitizer agents (prototype: Levosimendan), work by improving the binding of Ca to troponin C, independent of adrenergic pathway. For this reason, it seems to have less pro-arrhythmic side effects and does not cause increased oxygen consumption. Moreover, levosimendan appears to cause vasodilation through ATP-sensitive potassium channels, but only at high concentrations, that are not usually used in clinical practice. However, no difference in mortality has been noted between levosimendan and dobutamine. Furthermore, because of its vasodilation effect, calcium sensitizers have not been effectively studied in patients with cardiogenic shock, more clinical trials being required21.

3. Paucity of new molecules in heart failure treatment

Despite this epidemiological context, there is an important gap in developing efficient new molecular entities (NME) for HF treatment, with no Food and Drug Administration (FDA) approved inotropic agent in the last two decades22. Tolvaptan - a selective V2 vasopressin antagonist - is the only HF-related NME approved in the last 10 years but for treatment of hyponatremia, including hyponatremic HF23. According to Pierre Pollesello, some contributing factors to this situation might be the continuously evolving HF definition, the low cost for present medical therapies, the redirection of funds towards other research areas by pharmaceutical companies, the demands for expensive long term mortality trials and study recruiting issues related to the heterogeneity of HF patients24. This might also imply that we are not looking in the right direction yet and new research perspectives are needed.

4. Myosin activators are a possible answer

The discovery of a new class of inotropic drugs appeared to be a possibly feasible step form theory to practice25. A small molecule that enhances contractile motor protein myosin activity bypassing the second messenger regulation could bring new hope in HF treatment26. Theoretically, an ideal small molecule myosin activator should ameliorate the rate-limiting step in actin-myosin cycle, would recruit more myosin heads during the cardiac circle, would not have vascular effects and would not elicit calcium-related adverse reactions27. So, the overall effect would be to improve cardiac contractility in a “more hands on the rope” manner28. Several compounds have been developed and tested in preclinical studies deepening our understanding about this inotropic mechanism and about the necessary properties29,30.

5. The new hope: omecamtiv mecarbil?

A group from Cytokinetics finally succeeded in creating and refining a compound with the needed characteristics for entering the clinical trials step: CK-1827452, AMG 423 or omecamtiv mecarbil (OM)31. This new molecule is able to specifically bind at S148 heavy chain activating the cardiac myosin, but it has no effect on the smooth nor the skeletal muscle myosin32. OM appears to speed up the transition of myosin from a weak actin-binding in a strong actin-binding force-generating state, this being the rate-limiting step of actin-myosin-ATP interaction and also of myocyte contraction (Figure 1)32. In vitro, OM increased the duty cycle of acto-myosin interaction in tissue purified ventricular myosin33.

Figure 1. OM does not increase the force of myocardial contraction by altering the calcium concentration, but by acting directly at the level of sarcomere.

Figure 1

The fast change from weakly bound to the strongly bound actin-myosin configuration activates the sarcomere, increasing the force of cardiomyocyte displacement up to 8nm, compared to 5nm without myosin activator. Two stabilizing light chains - essential (ELC) and regulatory (RLC) - not only hold the neck of the myosin head in a steady position, but are also the mechanical components that are part of the moving force of myosin. These myosin light chains have the particularity of a regularly turning around in order to impel the attached actin, being important controllers of cardiac force production33.

The results obtained by using two canine models of HF were promising. The HF was induced by rapid pacing in two situations: after myocardial infarction induced by ligation of left anterior descending coronary artery and after myocardial hypertrophy caused by 1 year - long ascending aortic banding. OM was administered in 0.25 mg/kg bolus followed by continuous perfusion of 0.25 mg/kg/h during 24h. The treatment decreased the heart rate (HR), mean left atrial (LA) pressure, left ventricular (LV) end-diastolic pressure and improved LV systolic ejection time (LVSET), systolic wall thickening and stroke volume (SV) at 15 minutes, 4 hours and 24 hours of treatment and increased cardiac output at 24 hours. A reduction in vascular peripheral resistance was noted at 4 and 24 hours. No changes of LV dP/dt max, mean arterial pressure or myocardial oxygen consumption were remarked34. Multiple studies in humans were performed, they are listed in Table 2.

Table 2. Omecamtiv mecarbil – human clinical studies.

Clinicaltrials.gov identifier Study Design Status Data Results
NCT01300013 Study to Evaluate the Safety and Efficacy of IV Infusion Treatment With Omecamtiv Mecarbil in Subjects With Left Ventricular Systolic Dysfunction Hospitalized for Acute Heart Failure (ATOMIC-AHF) Phase II, Double-blind, Randomized, Placebo-controlled, Multicenter Completed 2011-2013 Not published
NCT01380223 A Pharmacokinetic and Pharmacodynamic Study of Omecamtiv Mecarbil in Healthy Volunteers Phase I, Double-Blind, Randomized, Four-Way Crossover, Placebo-Controlled, Safety study Completed 2005-2006 Teerlink et al., 201128
NCT00682565 Pharmacokinetics (PK) and Tolerability of Intravenous (IV) and Oral CK-1827452 in Patients With Ischemic Cardiomyopathy and Angina Phase II, Double-Blind, Randomized, Safety study Completed 2008 Not published
NCT00941681 Pharmacokinetics of Oral CK-1827452 in Patients with Stable Heart Failure Phase II, Open label, Randomized , Parallel Assignment, Pharmacokinetics study Completed 2009 Not published
NCT00624442 A Study of CK-1827452 Infusion in Stable Heart Failure Phase II, Double-blind, Randomized, Crossover Assignment, Safety/Efficacy Study Completed 2007-2009 Cleland et al, 201135
NCT00748579 Phase II Study of the Effect of CK-1827452 Injection on Myocardial Efficiency Phase II, Open Label, Single Group Assignment, Pharmacokinetics/ Dynamics Study Terminated 2008- 2009 Not published
NCT01786512 COSMIC-HF - Chronic Oral Study of Myosin Activation to Increase Contractility in Heart Failure Phase II, Double-blind, Randomized, Placebo-controlled, Multicenter, Dose Escalation Recruiting 2013-2015 Not published
NCT01737866 Pharmacokinetics Study of AMG 423 in Healthy Subjects and Subjects with Various Degrees of Renal Insufficiency Phase I, Open Label, Single Group Assignment, Safety Study Completed 2012-2013 Not published

The first one was realized by Teerlink et al. during 2005-200628. This phase I, single center, double-blind, randomized, dose-escalating, placebo-controlled trial aimed to evaluate the maximum tolerated dose of OM and to investigate its pharmacodynamic and pharmacokinetic properties. 34 healthy men divided in 4 cohorts (8-10 individuals each) received 6 hours continuous intravenous infusions of increasing OM doses (0.005 mg/kg/h to 1 mg/kg/h) at one week apart. The maximum tolerated dose was established at 0.5 mg/kg/h which was well supported by 16 subjects during 6 hours, the maximum concentration being 905 ng/ml. Other findings concerning the pharmacologic properties were: mean elimination half time of 17.1-23h, clearance of 132-207 mL/h/kg and mean volume of distribution of 3.7-5.2 L/kg. Serial echographic measurements showed that EF, SV, SET time and fractional shortening increased in a dose-dependent manner.

The diastolic function seemed to be less influenced, with a decrease of ratio between early maximal ventricular filling velocity and late filling velocity (E/A), due to an improvement and prolongation of atrial contraction. The limiting effect was myocardial ischemia (moderate troponin elevation but no infarction) seen at doses higher than 0.75 mg/kg/h. This was most probably generated by a shortening in diastolic coronary filling time due to systolic prolongation. In the tolerated doses range the authors noted some more frequent pain at catheter insertion site, but without other adverse reactions. No effect on HR was observed.

Cleland et al conducted the first study in stable HFLEF patients (EF<40%, steady therapy status) which enrolled 45 patients in a double-blind, randomized, crossover, dose ranging, phase II design35. Only patients with sinus rhythm, glomerular filtration rate (Modification of Diet in Renal Disease -MDRD) over 35 mL/min, no cardiovascular-related hospital admission during the last 6 weeks and no Canadian Class III or IV angina were included. Divided in 5 cohorts of 8-10 individuals each, the patients received 2h, 24h or 72h escalating infusions doses (loading + maintenance) of OM in order to achieve the previously established tolerated dose range in healthy volunteers. The data analysis confirmed the directly proportional relation between plasma concentration (pC) and systolic ejection time, fractional shortening (at pC >100ng/mL) and stroke volume (at pC>200 ng/mL). The ejection fraction increased at pC>300 ng/mL and both left ventricular systolic and diastolic volumes decreased at pC>500 ng/mL. The results persisted after 24h and 72h of infusion. Atrial contraction was also prolonged, with a decrease of E/A. Interestingly, it was noted a reduction in HR which was not the case in healthy patients, and a slight decrease in QTc. Myocardial ischemia was noticed in 3 patients - one accidentally overdosed, and two others with low drug clearance, obesity and hypertension. For the remaining patients no elevation in NT-proBNP, nor troponin was observed.

The data from ATOMIC-AHF (NCT01300013) study were presented at European Society of Cardiology Congress in 201336. This double-blind, randomized, placebo-controlled, multicenter, phase II study included 600 patients with decompensated systolic HF (EF<40%) - dyspnea at rest or with minimal effort resistant to diuretics and elevated BNP or NT pro-BNP. Patients were treated in 3 cohorts with 48h OM infusion targeting pC of 115, 230, 310 ng/mL respectively. The primary endpoint of dyspnea amelioration (7 points Likert Scale) was not achieved but a favorable trend was observed with higher doses and also a reduction in episodes of worsening of HF. These might be attributed to the lower pC achieved in this study comparative with the other studies. However, the previous findings in healthy volunteers and stable HF were replicated regarding the increase in SET, decrease in HR and safety issues (despite a slightly increase in troponin non related to pC).

Other phase I or II studies investigating the pharmacological properties of oral or IV OM in patients with ischemic cardiomyopathy, stable heart failure or with various degrees of renal failure were performed without published results yet (NCT01737866, NCT00682565, NCT00941681, NCT00748579). COSMIC-HF (NCT01786512) is an interesting study that may answer some questions about the future of OM but is still recruiting at this moment. Designed as a phase II, double-blind, randomized, placebo-controlled, multicenter, dose escalation study, it aims to evaluate an oral modified release (MR) OM formulation (BID) in HFLEF over 20 weeks of treatment.

Lots of hopes have been generated by OM, supported by the rather positive findings of current studies37. A success for OM in clinical trials might lead to a change in HF management paradigm. Despite that, there are still more questions than answers because the published studies were restricted only to a specific HF population until now. For example, it would be important to clarify the behavior of OM in patients with atrial arrhythmias or atrio-ventricular conduction abnormalities, because it seems that OM might modify atrial contractility and shorten QTc. The patients with valvular pathologies or obstructive hypertrophic cardiomyopathy should also be investigated, prolongation of SET being the marker of OM. COSMIC-HF might bring insights about OM chronic therapy and also about pharmacologic properties of oral administration.

At this moment, omecamtiv mecarbil appears to be a distinctive and unique molecule whose mechanism of action seems promising in the future therapeutic interventions. It is certain that further studies are needed to understand the pharmacodynamics and the therapeutic outcomes of this promising drug.

  • Current inotropes have serious adverse effects trough calcium- or cAMP-mediated mechanisms

  • There aren’t any inotropic drugs FDA-approved in the last two decades

  • Direct myosin activator OMECAMTIV MECARBIL emerges as an innovative therapy in heart failure by prolongation of systolic ejection time but further studies are necessary to validate its importance

Acknowledgments

The authors would like to thank Dr. Radu Chirvasuta, Cardiothoracic Intensive Care Unit, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, UK for his precious help in reviewing the manuscript.

Footnotes

Conflict of interests: The authors declare no conflict of interest.

Abbreviations

Cardiovascular disorders (CVD); heart failure (HF); ); heart failure with preserved ejection fraction (HFPEF); heart failure with low ejection fraction (HFLEF); ejection fraction (EF); adenosine triphosphate (ATP); adenosine diphosphate (ADP); inorganic phosphorus (Pi); cyclic adenosine monophosphate (cAMP); troponin C (cTnC); phosphodiesterase (PDE); phosphodiesterase inhibitors (PDEI); new molecular entities (NME); sarco/endoplasmic reticulum Ca2+-ATPase (SERCA); omecamtiv mecarbil (OM); heart rate (HR); left atrium (LA); left ventriculum (LV); systolic ejection time (SET); stroke volume (SV); essential stabilizing light chain ( ELC); regulatory stabilizing light chain (RLC); ratio between early maximal ventricular filling velocity and late filling velocity (E/A); modified release (MR); plasma concentration (pC), brain natriuretic peptide (BNP)

DISCOVERIES is a peer-reviewed, open access, online, multidisciplinary and integrative journal, publishing high impact and innovative manuscripts from all areas related to MEDICINE, BIOLOGY and CHEMISTRY

References

  • 1.Global and regional mortality from 235 causes of death for 20 age groups in 1990 and 2010: a systematic analysis for the Global Burden of Disease Study 2010. Lozano Rafael, Naghavi Mohsen, Foreman Kyle, Lim Stephen, Shibuya Kenji, Aboyans Victor, Abraham Jerry, Adair Timothy, Aggarwal Rakesh, Ahn Stephanie Y, Alvarado Miriam, Anderson H Ross, Anderson Laurie M, Andrews Kathryn G, Atkinson Charles, Baddour Larry M, Barker-Collo Suzanne, Bartels David H, Bell Michelle L, Benjamin Emelia J, Bennett Derrick, Bhalla Kavi, Bikbov Boris, Bin Abdulhak Aref, Birbeck Gretchen, Blyth Fiona, Bolliger Ian, Boufous Soufiane, Bucello Chiara, Burch Michael, Burney Peter, Carapetis Jonathan, Chen Honglei, Chou David, Chugh Sumeet S, Coffeng Luc E, Colan Steven D, Colquhoun Samantha, Colson K Ellicott, Condon John, Connor Myles D, Cooper Leslie T, Corriere Matthew, Cortinovis Monica, de Vaccaro Karen Courville, Couser William, Cowie Benjamin C, Criqui Michael H, Cross Marita, Dabhadkar Kaustubh C, Dahodwala Nabila, De Leo Diego, Degenhardt Louisa, Delossantos Allyne, Denenberg Julie, Des Jarlais Don C, Dharmaratne Samath D, Dorsey E Ray, Driscoll Tim, Duber Herbert, Ebel Beth, Erwin Patricia J, Espindola Patricia, Ezzati Majid, Feigin Valery, Flaxman Abraham D, Forouzanfar Mohammad H, Fowkes Francis Gerry R, Franklin Richard, Fransen Marlene, Freeman Michael K, Gabriel Sherine E, Gakidou Emmanuela, Gaspari Flavio, Gillum Richard F, Gonzalez-Medina Diego, Halasa Yara A, Haring Diana, Harrison James E, Havmoeller Rasmus, Hay Roderick J, Hoen Bruno, Hotez Peter J, Hoy Damian, Jacobsen Kathryn H, James Spencer L, Jasrasaria Rashmi, Jayaraman Sudha, Johns Nicole, Karthikeyan Ganesan, Kassebaum Nicholas, Keren Andre, Khoo Jon-Paul, Knowlton Lisa Marie, Kobusingye Olive, Koranteng Adofo, Krishnamurthi Rita, Lipnick Michael, Lipshultz Steven E, Ohno Summer Lockett, Mabweijano Jacqueline, MacIntyre Michael F, Mallinger Leslie, March Lyn, Marks Guy B, Marks Robin, Matsumori Akira, Matzopoulos Richard, Mayosi Bongani M, McAnulty John H, McDermott Mary M, McGrath John, Mensah George A, Merriman Tony R, Michaud Catherine, Miller Matthew, Miller Ted R, Mock Charles, Mocumbi Ana Olga, Mokdad Ali A, Moran Andrew, Mulholland Kim, Nair M Nathan, Naldi Luigi, Narayan K M Venkat, Nasseri Kiumarss, Norman Paul, O'Donnell Martin, Omer Saad B, Ortblad Katrina, Osborne Richard, Ozgediz Doruk, Pahari Bishnu, Pandian Jeyaraj Durai, Rivero Andrea Panozo, Padilla Rogelio Perez, Perez-Ruiz Fernando, Perico Norberto, Phillips David, Pierce Kelsey, Pope C Arden, Porrini Esteban, Pourmalek Farshad, Raju Murugesan, Ranganathan Dharani, Rehm Jürgen T, Rein David B, Remuzzi Guiseppe, Rivara Frederick P, Roberts Thomas, De León Felipe Rodriguez, Rosenfeld Lisa C, Rushton Lesley, Sacco Ralph L, Salomon Joshua A, Sampson Uchechukwu, Sanman Ella, Schwebel David C, Segui-Gomez Maria, Shepard Donald S, Singh David, Singleton Jessica, Sliwa Karen, Smith Emma, Steer Andrew, Taylor Jennifer A, Thomas Bernadette, Tleyjeh Imad M, Towbin Jeffrey A, Truelsen Thomas, Undurraga Eduardo A, Venketasubramanian N, Vijayakumar Lakshmi, Vos Theo, Wagner Gregory R, Wang Mengru, Wang Wenzhi, Watt Kerrianne, Weinstock Martin A, Weintraub Robert, Wilkinson James D, Woolf Anthony D, Wulf Sarah, Yeh Pon-Hsiu, Yip Paul, Zabetian Azadeh, Zheng Zhi-Jie, Lopez Alan D, Murray Christopher J L, AlMazroa Mohammad A, Memish Ziad A. Lancet (London, England) 2012;380(9859):2095–128. doi: 10.1016/S0140-6736(12)61728-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.A comparative risk assessment of burden of disease and injury attributable to 67 risk factors and risk factor clusters in 21 regions, 1990-2010: a systematic analysis for the Global Burden of Disease Study 2010. Lim Stephen S, Vos Theo, Flaxman Abraham D, Danaei Goodarz, Shibuya Kenji, Adair-Rohani Heather, Amann Markus, Anderson H Ross, Andrews Kathryn G, Aryee Martin, Atkinson Charles, Bacchus Loraine J, Bahalim Adil N, Balakrishnan Kalpana, Balmes John, Barker-Collo Suzanne, Baxter Amanda, Bell Michelle L, Blore Jed D, Blyth Fiona, Bonner Carissa, Borges Guilherme, Bourne Rupert, Boussinesq Michel, Brauer Michael, Brooks Peter, Bruce Nigel G, Brunekreef Bert, Bryan-Hancock Claire, Bucello Chiara, Buchbinder Rachelle, Bull Fiona, Burnett Richard T, Byers Tim E, Calabria Bianca, Carapetis Jonathan, Carnahan Emily, Chafe Zoe, Charlson Fiona, Chen Honglei, Chen Jian Shen, Cheng Andrew Tai-Ann, Child Jennifer Christine, Cohen Aaron, Colson K Ellicott, Cowie Benjamin C, Darby Sarah, Darling Susan, Davis Adrian, Degenhardt Louisa, Dentener Frank, Des Jarlais Don C, Devries Karen, Dherani Mukesh, Ding Eric L, Dorsey E Ray, Driscoll Tim, Edmond Karen, Ali Suad Eltahir, Engell Rebecca E, Erwin Patricia J, Fahimi Saman, Falder Gail, Farzadfar Farshad, Ferrari Alize, Finucane Mariel M, Flaxman Seth, Fowkes Francis Gerry R, Freedman Greg, Freeman Michael K, Gakidou Emmanuela, Ghosh Santu, Giovannucci Edward, Gmel Gerhard, Graham Kathryn, Grainger Rebecca, Grant Bridget, Gunnell David, Gutierrez Hialy R, Hall Wayne, Hoek Hans W, Hogan Anthony, Hosgood H Dean, Hoy Damian, Hu Howard, Hubbell Bryan J, Hutchings Sally J, Ibeanusi Sydney E, Jacklyn Gemma L, Jasrasaria Rashmi, Jonas Jost B, Kan Haidong, Kanis John A, Kassebaum Nicholas, Kawakami Norito, Khang Young-Ho, Khatibzadeh Shahab, Khoo Jon-Paul, Kok Cindy, Laden Francine, Lalloo Ratilal, Lan Qing, Lathlean Tim, Leasher Janet L, Leigh James, Li Yang, Lin John Kent, Lipshultz Steven E, London Stephanie, Lozano Rafael, Lu Yuan, Mak Joelle, Malekzadeh Reza, Mallinger Leslie, Marcenes Wagner, March Lyn, Marks Robin, Martin Randall, McGale Paul, McGrath John, Mehta Sumi, Mensah George A, Merriman Tony R, Micha Renata, Michaud Catherine, Mishra Vinod, Mohd Hanafiah Khayriyyah, Mokdad Ali A, Morawska Lidia, Mozaffarian Dariush, Murphy Tasha, Naghavi Mohsen, Neal Bruce, Nelson Paul K, Nolla Joan Miquel, Norman Rosana, Olives Casey, Omer Saad B, Orchard Jessica, Osborne Richard, Ostro Bart, Page Andrew, Pandey Kiran D, Parry Charles D H, Passmore Erin, Patra Jayadeep, Pearce Neil, Pelizzari Pamela M, Petzold Max, Phillips Michael R, Pope Dan, Pope C Arden, Powles John, Rao Mayuree, Razavi Homie, Rehfuess Eva A, Rehm Jürgen T, Ritz Beate, Rivara Frederick P, Roberts Thomas, Robinson Carolyn, Rodriguez-Portales Jose A, Romieu Isabelle, Room Robin, Rosenfeld Lisa C, Roy Ananya, Rushton Lesley, Salomon Joshua A, Sampson Uchechukwu, Sanchez-Riera Lidia, Sanman Ella, Sapkota Amir, Seedat Soraya, Shi Peilin, Shield Kevin, Shivakoti Rupak, Singh Gitanjali M, Sleet David A, Smith Emma, Smith Kirk R, Stapelberg Nicolas J C, Steenland Kyle, Stöckl Heidi, Stovner Lars Jacob, Straif Kurt, Straney Lahn, Thurston George D, Tran Jimmy H, Van Dingenen Rita, van Donkelaar Aaron, Veerman J Lennert, Vijayakumar Lakshmi, Weintraub Robert, Weissman Myrna M, White Richard A, Whiteford Harvey, Wiersma Steven T, Wilkinson James D, Williams Hywel C, Williams Warwick, Wilson Nicholas, Woolf Anthony D, Yip Paul, Zielinski Jan M, Lopez Alan D, Murray Christopher J L, Ezzati Majid, AlMazroa Mohammad A, Memish Ziad A. Lancet (London, England) 2012;380(9859):2224–60. doi: 10.1016/S0140-6736(12)61766-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Cardiovascular diseases Fact Sheet no 317. World Health Organization. 2013.
  • 4.ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. McMurray John J V, Adamopoulos Stamatis, Anker Stefan D, Auricchio Angelo, Böhm Michael, Dickstein Kenneth, Falk Volkmar, Filippatos Gerasimos, Fonseca Cândida, Gomez-Sanchez Miguel Angel, Jaarsma Tiny, Køber Lars, Lip Gregory Y H, Maggioni Aldo Pietro, Parkhomenko Alexander, Pieske Burkert M, Popescu Bogdan A, Rønnevik Per K, Rutten Frans H, Schwitter Juerg, Seferovic Petar, Stepinska Janina, Trindade Pedro T, Voors Adriaan A, Zannad Faiez, Zeiher Andreas. European heart journal. 2012;33(14):1787–847. doi: 10.1093/eurheartj/ehs104. [DOI] [PubMed] [Google Scholar]
  • 5.Clinical practice. Systolic heart failure. McMurray John J V. The New England journal of medicine. 2010;362(3):228–38. doi: 10.1056/NEJMcp0909392. [DOI] [PubMed] [Google Scholar]
  • 6.Inotropes. Francis Gary S, Bartos Jason A, Adatya Sirtaz. Journal of the American College of Cardiology. 2014;63(20):2069–2078. doi: 10.1016/j.jacc.2014.01.016. [DOI] [PubMed] [Google Scholar]
  • 7.Cell communications in the heart. Tirziu Daniela, Giordano Frank J, Simons Michael. Circulation. 2010;122(9):928–37. doi: 10.1161/CIRCULATIONAHA.108.847731. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Cooper GM. Actin, Myosin, and Cell Movement, The Cell: A Molecular Approach. 2nd edition. Sinauer Associates; 2000. [Google Scholar]
  • 9.Karp G. Cytoskeleton and cell motility, Cell and molecular biology. 6th Edition. John Wiley & Sons Inc; 2009. [Google Scholar]
  • 10.Calcium-induced release of calcium from the cardiac sarcoplasmic reticulum. Fabiato A. American Journal of Physiology-Cell Physiology. 1983;245(1):C1-C14. doi: 10.1152/ajpcell.1983.245.1.C1. [DOI] [PubMed] [Google Scholar]
  • 11.Calcium cycling in the cardiomyocyte. Choong-Chin L, Dzau VJ VJ. NAt. Rev Genet. 2004:811–825. [Google Scholar]
  • 12.Effects of cardiac myosin isoform variation on myofilament function and crossbridge kinetics in transgenic rabbits. Suzuki Takeki, Palmer Bradley M, James Jeanne, Wang Yuan, Chen Zengyi, VanBuren Peter, Maughan David W, Robbins Jeffrey, LeWinter Martin M. Circulation. Heart failure. 2009;2(4):334–41. doi: 10.1161/CIRCHEARTFAILURE.108.802298. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Rubin S, Pondok T. Inotropic therapy in clinical practice, Heart failure: pharmacologic management. Blackwell publishing; 2006. pp. 104–105. [Google Scholar]
  • 14.Electrophysiologic and proarrhythmic effects of intravenous inotropic agents. Tisdale J E, Patel R, Webb C R, Borzak S, Zarowitz B J. Progress in cardiovascular diseases. 1995;38(2):167–80. doi: 10.1016/s0033-0620(05)80005-2. [DOI] [PubMed] [Google Scholar]
  • 15.Mechanisms and use of calcium-sensitizing agents in the failing heart. Kass David A, Solaro R John. Circulation. 2006;113(2):305–15. doi: 10.1161/CIRCULATIONAHA.105.542407. [DOI] [PubMed] [Google Scholar]
  • 16.Heart failure. Krum Henry, Abraham William T. The Lancet. 2009;373(9667):941-955. doi: 10.1016/S0140-6736(09)60236-1. [DOI] [PubMed] [Google Scholar]
  • 17.A novel approach to improve cardiac performance: cardiac myosin activators. Teerlink John R. Heart failure reviews. 2009;14(4):289–98. doi: 10.1007/s10741-009-9135-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Inotropes and vasopressors: review of physiology and clinical use in cardiovascular disease. Overgaard Christopher B, Dzavík Vladimír. Circulation. 2008;118(10):1047–56. doi: 10.1161/CIRCULATIONAHA.107.728840. [DOI] [PubMed] [Google Scholar]
  • 19.Inotropes and beta-blockers: is there a need for new guidelines? Bristow M R, Shakar S F, Linseman J V, Lowes B D. Journal of cardiac failure. 2001;7(2 Suppl 1):8–12. doi: 10.1054/jcaf.2001.26655. [DOI] [PubMed] [Google Scholar]
  • 20.Short-term intravenous milrinone for acute exacerbation of chronic heart failure: a randomized controlled trial. Cuffe Michael S, Califf Robert M, Adams Kirkwood F, Benza Raymond, Bourge Robert, Colucci Wilson S, Massie Barry M, O'Connor Christopher M, Pina Ileana, Quigg Rebecca, Silver Marc A, Gheorghiade Mihai. JAMA. 2002;287(12):1541–7. doi: 10.1001/jama.287.12.1541. [DOI] [PubMed] [Google Scholar]
  • 21.Levosimendan in critical illness: a literature review. Pierrakos Charalampos, Velissaris Dimitrios, Franchi Federico, Muzzi Luigi, Karanikolas Menelaos, Scolletta Sabino. Journal of clinical medicine research. 2014;6(2):75–85. doi: 10.14740/jocmr1702w. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.FDA Approved Drugs. CenterWatch. 2014. http://www.centerwatch.com/drug-information/fda-approved-drugs/year http://www.centerwatch.com/drug-information/fda-approved-drugs/year
  • 23.2009 FDA drug approvals. Hughes Bethan. Nature Reviews Drug Discovery. 2010;9(2):89-92. doi: 10.1038/nrd3101. [DOI] [PubMed] [Google Scholar]
  • 24.Drug discovery and development for acute heart failure drugs: Are expectations too high? Pollesello Piero. International Journal of Cardiology. 2014;172(1):11-13. doi: 10.1016/j.ijcard.2013.12.136. [DOI] [PubMed] [Google Scholar]
  • 25.Cardiac myosin activation: will theory and practice coincide? Dickstein Kenneth. The Lancet. 2011;378(9792):639-641. doi: 10.1016/S0140-6736(11)61298-1. [DOI] [PubMed] [Google Scholar]
  • 26.Chemically Tuned Myosin Motors. Leinwand L. A., Moss R. L. Science. 2011;331(6023):1392-1393. doi: 10.1126/science.1204207. [DOI] [PubMed] [Google Scholar]
  • 27.A novel approach to improve cardiac performance: cardiac myosin activators. Campia U U, Nodari S, Gheorghiade M, Teerlink JR, Metra M, Zacà V. Heart Fail Rev. 2013;14:689–696. doi: 10.1007/s10741-009-9135-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Dose-dependent augmentation of cardiac systolic function with the selective cardiac myosin activator, omecamtiv mecarbil: a first-in-man study. Teerlink John R, Clarke Cyril P, Saikali Khalil G, Lee Jacqueline H, Chen Michael M, Escandon Rafael D, Elliott Lyndsey, Bee Rachel, Habibzadeh Mohammad Reza, Goldman Jonathan H, Schiller Nelson B, Malik Fady I, Wolff Andrew A. Lancet (London, England) 2011;378(9792):667–75. doi: 10.1016/S0140-6736(11)61219-1. [DOI] [PubMed] [Google Scholar]
  • 29.Cellular responses of the myosin activator CK689705 in normal and heart failure. Niu C, Cox DR, Lee KH, Sylvester S, Sueoka SH, Qian X. https://cytokinetics.com/wp-content/uploads/2015/10/ASCB2004_04_25.pdf Mol Biol Cell. 2004;15(271A) [Google Scholar]
  • 30.Dissecting temporal and spatial control of cytokinesis with a myosin II Inhibitor. Straight Aaron F, Cheung Amy, Limouze John, Chen Irene, Westwood Nick J, Sellers James R, Mitchison Timothy J. Science (New York, N.Y.) 2003;299(5613):1743–7. doi: 10.1126/science.1081412. [DOI] [PubMed] [Google Scholar]
  • 31.Discovery of omecamtiv mecarbil the first, selective, small molecule activator of cardiac Myosin. Morgan Bradley P, Muci Alexander, Lu Pu-Ping, Qian Xiangping, Tochimoto Todd, Smith Whitney W, Garard Marc, Kraynack Erica, Collibee Scott, Suehiro Ion, Tomasi Adam, Valdez S Corey, Wang Wenyue, Jiang Hong, Hartman James, Rodriguez Hector M, Kawas Raja, Sylvester Sheila, Elias Kathleen A, Godinez Guillermo, Lee Kenneth, Anderson Robert, Sueoka Sandra, Xu Donghong, Wang Zhengping, Djordjevic Nebojsa, Malik Fady I, Morgans David J. ACS medicinal chemistry letters. 2010;1(9):472–7. doi: 10.1021/ml100138q. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Cardiac myosin activation: a potential therapeutic approach for systolic heart failure. Malik Fady I, Hartman James J, Elias Kathleen A, Morgan Bradley P, Rodriguez Hector, Brejc Katjusa, Anderson Robert L, Sueoka Sandra H, Lee Kenneth H, Finer Jeffrey T, Sakowicz Roman, Baliga Ramesh, Cox David R, Garard Marc, Godinez Guillermo, Kawas Raja, Kraynack Erica, Lenzi David, Lu Pu Ping, Muci Alexander, Niu Congrong, Qian Xiangping, Pierce Daniel W, Pokrovskii Maria, Suehiro Ion, Sylvester Sheila, Tochimoto Todd, Valdez Corey, Wang Wenyue, Katori Tatsuo, Kass David A, Shen You-Tang, Vatner Stephen F, Morgans David J. Science (New York, N.Y.) 2011;331(6023):1439–43. doi: 10.1126/science.1200113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Analytical comparison of natural and pharmaceutical ventricular myosin activators. Wang Yihua, Ajtai Katalin, Burghardt Thomas P. Biochemistry. 2014;53(32):5298–306. doi: 10.1021/bi500730t. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Improvement of cardiac function by a cardiac Myosin activator in conscious dogs with systolic heart failure. Shen You-Tang, Malik Fady I, Zhao Xin, Depre Christophe, Dhar Sunil K, Abarzúa Patricio, Morgans David J, Vatner Stephen F. Circulation. Heart failure. 2010;3(4):522–7. doi: 10.1161/CIRCHEARTFAILURE.109.930321. [DOI] [PubMed] [Google Scholar]
  • 35.The effects of the cardiac myosin activator, omecamtiv mecarbil, on cardiac function in systolic heart failure: a double-blind, placebo-controlled, crossover, dose-ranging phase 2 trial. Cleland John G F, Teerlink John R, Senior Roxy, Nifontov Evgeny M, Mc Murray John J V, Lang Chim C, Tsyrlin Vitaly A, Greenberg Barry H, Mayet Jamil, Francis Darrel P, Shaburishvili Tamaz, Monaghan Mark, Saltzberg Mitchell, Neyses Ludwig, Wasserman Scott M, Lee Jacqueline H, Saikali Khalil G, Clarke Cyril P, Goldman Jonathan H, Wolff Andrew A, Malik Fady I. Lancet (London, England) 2011;378(9792):676–83. doi: 10.1016/S0140-6736(11)61126-4. [DOI] [PubMed] [Google Scholar]
  • 36.Acute Treatment With Omecamtiv Mecarbil to Increase Contractility in Acute Heart Failure: The ATOMIC-AHF Study. Teerlink John R, Felker G Michael, McMurray John J V, Ponikowski Piotr, Metra Marco, Filippatos Gerasimos S, Ezekowitz Justin A, Dickstein Kenneth, Cleland John G F, Kim Jae B, Lei Lei, Knusel Beat, Wolff Andrew A, Malik Fady I, Wasserman Scott M. Journal of the American College of Cardiology. 2016;67(12):1444–1455. doi: 10.1016/j.jacc.2016.01.031. [DOI] [PubMed] [Google Scholar]
  • 37.A new approach to inotropic therapy in the treatment of heart failure: cardiac myosin activators in treatment of HF. Garg Vinisha, Frishman William H. Cardiology in review. 2013;21(3):155–9. doi: 10.1097/CRD.0b013e318275889c. [DOI] [PubMed] [Google Scholar]

Articles from Discoveries are provided here courtesy of Applied Systems

RESOURCES