Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
J Med Chem ; 64(19): 14142-14152, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34606259

ABSTRACT

Hypercontractility of the cardiac sarcomere may be essential for the underlying pathological hypertrophy and fibrosis in genetic hypertrophic cardiomyopathies. Aficamten (CK-274) is a novel cardiac myosin inhibitor that was discovered from the optimization of indoline compound 1. The important advancement of the optimization was discovery of an Indane analogue (12) with a less restrictive structure-activity relationship that allowed for the rapid improvement of drug-like properties. Aficamten was designed to provide a predicted human half-life (t1/2) appropriate for once a day (qd) dosing, to reach steady state within two weeks, to have no substantial cytochrome P450 induction or inhibition, and to have a wide therapeutic window in vivo with a clear pharmacokinetic/pharmacodynamic relationship. In a phase I clinical trial, aficamten demonstrated a human t1/2 similar to predictions and was able to reach steady state concentration within the desired two-week window.


Subject(s)
Cardiac Myosins/drug effects , Cardiomyopathy, Hypertrophic/drug therapy , Drug Discovery , Dose-Response Relationship, Drug , Humans , Molecular Structure , Structure-Activity Relationship
2.
J Biol Chem ; 296: 100471, 2021.
Article in English | MEDLINE | ID: mdl-33639160

ABSTRACT

Actin-myosin mediated contractile forces are crucial for many cellular functions, including cell motility, cytokinesis, and muscle contraction. We determined the effects of ten actin-binding compounds on the interaction of cardiac myosin subfragment 1 (S1) with pyrene-labeled F-actin (PFA). These compounds, previously identified from a small-molecule high-throughput screen (HTS), perturb the structural dynamics of actin and the steady-state actin-activated myosin ATPase activity. However, the mechanisms underpinning these perturbations remain unclear. Here we further characterize them by measuring their effects on PFA fluorescence, which is decreased specifically by the strong binding of myosin to actin. We measured these effects under equilibrium and steady-state conditions, and under transient conditions, in stopped-flow experiments following addition of ATP to S1-bound PFA. We observed that these compounds affect early steps of the myosin ATPase cycle to different extents. They increased the association equilibrium constant K1 for the formation of the strongly bound collision complex, indicating increased ATP affinity for actin-bound myosin, and decreased the rate constant k+2 for subsequent isomerization to the weakly bound ternary complex, thus slowing the strong-to-weak transition that actin-myosin interaction undergoes early in the ATPase cycle. The compounds' effects on actin structure allosterically inhibit the kinetics of the actin-myosin interaction in ways that may be desirable for treatment of hypercontractile forms of cardiomyopathy. This work helps to elucidate the mechanisms of action for these compounds, several of which are currently used therapeutically, and sets the stage for future HTS campaigns that aim to discover new drugs for treatment of heart failure.


Subject(s)
Actins/chemistry , Actins/metabolism , Cardiac Myosins/metabolism , Actins/drug effects , Adenosine Triphosphatases/drug effects , Adenosine Triphosphatases/metabolism , Animals , Cardiac Myosins/drug effects , Cardiac Myosins/physiology , Cattle , Fluorescence , High-Throughput Screening Assays/methods , Kinetics , Muscle Contraction/physiology , Myosin Subfragments/drug effects , Myosin Subfragments/metabolism , Myosins/drug effects , Myosins/metabolism , Physics , Protein Binding , Pyrenes/chemistry , Rabbits , Small Molecule Libraries/pharmacology
3.
N Engl J Med ; 384(2): 105-116, 2021 01 14.
Article in English | MEDLINE | ID: mdl-33185990

ABSTRACT

BACKGROUND: The selective cardiac myosin activator omecamtiv mecarbil has been shown to improve cardiac function in patients with heart failure with a reduced ejection fraction. Its effect on cardiovascular outcomes is unknown. METHODS: We randomly assigned 8256 patients (inpatients and outpatients) with symptomatic chronic heart failure and an ejection fraction of 35% or less to receive omecamtiv mecarbil (using pharmacokinetic-guided doses of 25 mg, 37.5 mg, or 50 mg twice daily) or placebo, in addition to standard heart-failure therapy. The primary outcome was a composite of a first heart-failure event (hospitalization or urgent visit for heart failure) or death from cardiovascular causes. RESULTS: During a median of 21.8 months, a primary-outcome event occurred in 1523 of 4120 patients (37.0%) in the omecamtiv mecarbil group and in 1607 of 4112 patients (39.1%) in the placebo group (hazard ratio, 0.92; 95% confidence interval [CI], 0.86 to 0.99; P = 0.03). A total of 808 patients (19.6%) and 798 patients (19.4%), respectively, died from cardiovascular causes (hazard ratio, 1.01; 95% CI, 0.92 to 1.11). There was no significant difference between groups in the change from baseline on the Kansas City Cardiomyopathy Questionnaire total symptom score. At week 24, the change from baseline for the median N-terminal pro-B-type natriuretic peptide level was 10% lower in the omecamtiv mecarbil group than in the placebo group; the median cardiac troponin I level was 4 ng per liter higher. The frequency of cardiac ischemic and ventricular arrhythmia events was similar in the two groups. CONCLUSIONS: Among patients with heart failure and a reduced ejection, those who received omecamtiv mecarbil had a lower incidence of a composite of a heart-failure event or death from cardiovascular causes than those who received placebo. (Funded by Amgen and others; GALACTIC-HF ClinicalTrials.gov number, NCT02929329; EudraCT number, 2016-002299-28.).


Subject(s)
Cardiac Myosins/metabolism , Cardiotonic Agents/therapeutic use , Heart Failure, Systolic/drug therapy , Urea/analogs & derivatives , Aged , Aged, 80 and over , Cardiac Myosins/drug effects , Cardiotonic Agents/adverse effects , Cardiotonic Agents/pharmacology , Cardiovascular Diseases/mortality , Female , Heart Failure, Systolic/metabolism , Heart Failure, Systolic/physiopathology , Humans , Male , Middle Aged , Myocardial Contraction/drug effects , Stroke Volume , Urea/adverse effects , Urea/pharmacology , Urea/therapeutic use
4.
Bioorg Med Chem ; 28(22): 115742, 2020 11 15.
Article in English | MEDLINE | ID: mdl-33007555

ABSTRACT

To identify novel potent cardiac myosin activator, a series of diphenylalkylisoxazol-5-amine compounds 4-7 have been synthesized and evaluated for cardiac myosin ATPase activation. Among the 37 compounds, 4a (CMA at 10 µM = 81.6%), 4w (CMA at 10 µM = 71.2%) and 6b (CMA at 10 µM = 67.4%) showed potent cardiac myosin activation at a single concentration of 10 µM. These results suggested that the introduction of the amino-isoxazole ring as a bioisostere for urea group is acceptable for the cardiac myosin activation. Additional structure-activity relationship (SAR) studies were conducted. Para substitution (-Cl, -OCH3, -SO2N(CH3)2) to the phenyl rings or replacement of a phenyl ring with a heterocycle (pyridine, piperidine and tetrahydropyran) appeared to attenuate cardiac myosin activation at 10 µM. Additional hydrogen bonding acceptor next to the amino group of the isoxazoles did not enhance the activity. The potent isoxazole compounds showed selectivity for cardiac myosin activation over skeletal and smooth muscle myosin, and therefore these potent and selective isoxazole compounds could be considered as a new series of cardiac myosin ATPase activators for the treatment of systolic heart failure.


Subject(s)
Adenosine Triphosphatases/metabolism , Amines/pharmacology , Cardiac Myosins/drug effects , Isoxazoles/pharmacology , Amines/chemical synthesis , Amines/chemistry , Cardiac Myosins/metabolism , Dose-Response Relationship, Drug , Humans , Isoxazoles/chemical synthesis , Isoxazoles/chemistry , Molecular Structure , Structure-Activity Relationship
5.
Nat Commun ; 11(1): 3405, 2020 07 07.
Article in English | MEDLINE | ID: mdl-32636378

ABSTRACT

Omecamtiv mecarbil (OM) is a putative positive inotropic tool for treatment of systolic heart dysfunction, based on the finding that in vivo it increases the ejection fraction and in vitro it prolongs the actin-bond life time of the cardiac and slow-skeletal muscle isoforms of myosin. OM action in situ, however, is still poorly understood as the enhanced Ca2+-sensitivity of the myofilaments is at odds with the reduction of force and rate of force development observed at saturating Ca2+. Here we show, by combining fast sarcomere-level mechanics and ATPase measurements in single slow demembranated fibres from rabbit soleus, that the depressant effect of OM on the force per attached motor is reversed, without effect on the ATPase rate, by physiological concentrations of inorganic phosphate (Pi) (1-10 mM). This mechanism could underpin an energetically efficient reduction of systolic tension cost in OM-treated patients, whenever [Pi] increases with heart-beat frequency.


Subject(s)
Cardiac Myosins/drug effects , Myocardial Contraction/drug effects , Myosins/metabolism , Phosphates/pharmacology , Urea/analogs & derivatives , Adenosine Triphosphatases/metabolism , Animals , Calcium/metabolism , Drug Synergism , Male , Muscle, Skeletal/metabolism , Rabbits , Sarcomeres/metabolism , Stress, Mechanical , Urea/pharmacology
6.
Bioorg Med Chem ; 27(18): 4110-4123, 2019 09 15.
Article in English | MEDLINE | ID: mdl-31378598

ABSTRACT

The sulfonamidophenylethylamide analogues were explored for finding novel and potent cardiac myosin activators. Among them, N-(4-(N,N-dimethylsulfamoyl)phenethyl-N-methyl-5-phenylpentanamide (13, CMA at 10 µM = 48.5%; FS = 26.21%; EF = 15.28%) and its isomer, 4-(4-(N,N-dimethylsulfamoyl)phenyl-N-methyl-N-(3-phenylpropyl)butanamide (27, CMA at 10 µM = 55.0%; FS = 24.69%; EF = 14.08%) proved to be efficient cardiac myosin activators both in in vitro and in vivo studies. Compounds 13 (88.2 + 3.1% at 5 µM) and 27 (46.5 + 2.8% at 5 µM) showed positive inotropic effect in isolated rat ventricular myocytes. The potent compounds 13 and 27 were highly selective for cardiac myosin over skeletal and smooth muscle myosin, and therefore these potent and selective amide derivatives could be considered a new class of cardiac myosin activators for the treatment of systolic heart failure.


Subject(s)
Amides/therapeutic use , Cardiac Myosins/drug effects , Amides/pharmacology , Humans , Structure-Activity Relationship
7.
Bioorg Med Chem Lett ; 28(14): 2369-2374, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29937058

ABSTRACT

To explore novel cardiac myosin activator, a series of diphenylalkyl substituted 1,3,4-oxadiazoles and 1,2,4-oxadiazoles have been prepared and tested for cardiac myosin ATPase activation in vitro. In all cases, three carbon spacer between the oxadiazole core and one of the phenyl ring was considered crucial. In case of 1,3,4-oxadiazole, zero to two carbon spacer between oxadiazole core and other phenyl ring are favorable. Phenyl ring can be replaced by cyclohexyl moiety. In case of 1,2,4-oxadiazole, zero or one carbon spacer between the oxadiazole and other phenyl ring are favorable. Introduction of hydrogen bonding donor (NH) group at the 2nd position of the 1,3,4-oxadiazole enhances the activity. Substitutions on either of the phenyl rings or change of phenyl ring to other heterocycle are not tolerated for both the oxadiazoles. The prepared oxadiazoles showed selective activation for cardiac muscle over smooth and skeleton muscles.


Subject(s)
Adenosine Triphosphatases/metabolism , Cardiac Myosins/drug effects , Oxadiazoles/pharmacology , Cardiac Myosins/metabolism , Dose-Response Relationship, Drug , Humans , Hydrogen Bonding , Molecular Structure , Oxadiazoles/chemical synthesis , Oxadiazoles/chemistry , Structure-Activity Relationship
8.
Arthritis Rheumatol ; 70(11): 1879-1889, 2018 11.
Article in English | MEDLINE | ID: mdl-29781582

ABSTRACT

OBJECTIVE: Immune complex (IC) deposition activates polymorphonuclear neutrophils (PMNs), increases vascular permeability, and leads to organ damage in systemic lupus erythematosus and rheumatoid arthritis. The bioactive lipid sphingosine 1-phosphate (S1P), acting via S1P receptor 1 (S1P1 ), is a key regulator of endothelial cell (EC) barrier function. This study was undertaken to investigate whether augmenting EC integrity via S1P1 signaling attenuates inflammatory injury mediated by ICs. METHODS: In vitro barrier function was assessed in human umbilical vein endothelial cells (HUVECs) by electrical cell-substrate impedance sensing. Phosphorylation of myosin light chain 2 (p-MLC-2) and VE-cadherin staining in HUVECs were assessed by immunofluorescence. A reverse Arthus reaction (RAR) was induced in the skin and lungs of mice with S1P1 deleted from ECs (S1P1 EC-knockout [ECKO] mice) and mice treated with S1P1 agonists and antagonists. RESULTS: S1P1 agonists prevented loss of barrier function in HUVECs treated with IC-activated PMNs. S1P1 ECKO and wild-type (WT) mice treated with S1P1 antagonists had amplified RAR, whereas specific S1P1 agonists attenuated skin and lung RAR in WT mice. ApoM-Fc, a novel S1P chaperone, mitigated EC cell barrier dysfunction induced by activated PMNs in vitro and attenuated lung RAR. Expression levels of p-MLC-2 and disruption of VE-cadherin, each representing manifestations of cell contraction and destabilization of adherens junctions, respectively, that were induced by activated PMNs, were markedly reduced by treatment with S1P1 agonists and ApoM-Fc. CONCLUSION: Our findings indicate that S1P1 signaling in ECs modulates vascular responses to IC deposition. S1P1 agonists and ApoM-Fc enhance the EC barrier, limit leukocyte escape from capillaries, and provide protection against inflammatory injury. The S1P/S1P1 axis is a newly identified target to attenuate tissue responses to IC deposition and mitigate end-organ damage.


Subject(s)
Antigen-Antibody Complex/metabolism , Capillary Permeability/genetics , Endothelial Cells/metabolism , Receptors, Lysosphingolipid/genetics , Adherens Junctions/drug effects , Adherens Junctions/metabolism , Anilides/pharmacology , Animals , Antigens, CD/drug effects , Antigens, CD/metabolism , Apolipoproteins M/pharmacology , Arthus Reaction , Cadherins/drug effects , Cadherins/metabolism , Capillary Permeability/drug effects , Cardiac Myosins/drug effects , Cardiac Myosins/metabolism , Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells , Humans , Indans/pharmacology , Lung/blood supply , Lung/drug effects , Lung/metabolism , Lysophospholipids/pharmacology , Mice , Mice, Knockout , Myosin Light Chains/drug effects , Myosin Light Chains/metabolism , Organophosphonates/pharmacology , Oxadiazoles/pharmacology , Receptors, Lysosphingolipid/agonists , Receptors, Lysosphingolipid/antagonists & inhibitors , Receptors, Lysosphingolipid/metabolism , Skin/blood supply , Skin/drug effects , Skin/metabolism , Sphingosine/analogs & derivatives , Sphingosine/pharmacology , Sphingosine-1-Phosphate Receptors , Thiophenes/pharmacology
9.
Eur J Med Chem ; 143: 1869-1887, 2018 Jan 01.
Article in English | MEDLINE | ID: mdl-29224951

ABSTRACT

To optimize the lead urea scaffold 1 and 2 as selective cardiac myosin ATPase activator, a series of urea derivatives have been synthesized to explore its structure activity relationship. Among them N,N-dimethyl-4-(2-(3-(3-phenylpropyl)ureido)ethyl)benzenesulfonamide (13, CMA = 91.6%, FS = 17.62%; EF = 11.55%), N,N-dimethyl-4-(2-(1-methyl-3-(3-phenylpropyl)ureido)ethyl)benzene sulfonamide (40, CMA = 52.3%, FS = 38.96%; EF = 24.19%) and N,N-dimethyl-4-(2-(3-methyl-3-(3-phenylpropyl)ureido)ethyl)benzenesulfonamide (41, CMA = 47.6%, FS = 23.19%; EF = 15.47%) proved to be efficient to activate the cardiac myosin in vitro and in vivo. Further the % change in ventricular cell contractility at 5 µM of 13 (47.9 ± 3.2), 40 (45.5 ± 2.4) and 41 (63.5 ± 2.2) showed positive inotropic effect in isolated rat ventricular myocytes. The potent compounds 13, 40, 41 were highly selective for cardiac myosin over skeletal and smooth muscle myosin, thus proving them these new urea derivatives is a novel scaffold for discovery of cardiac myosin activators for the treatment of systolic heart failure.


Subject(s)
Cardiac Myosins/drug effects , Drug Design , Urea/pharmacology , Animals , Cardiac Myosins/metabolism , Dose-Response Relationship, Drug , Male , Molecular Structure , Rats , Rats, Sprague-Dawley , Structure-Activity Relationship , Urea/analogs & derivatives , Urea/chemistry
10.
PLoS Comput Biol ; 13(11): e1005826, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29108014

ABSTRACT

New promising avenues for the pharmacological treatment of skeletal and heart muscle diseases rely on direct sarcomeric modulators, which are molecules that can directly bind to sarcomeric proteins and either inhibit or enhance their activity. A recent breakthrough has been the discovery of the myosin activator omecamtiv mecarbil (OM), which has been shown to increase the power output of the cardiac muscle and is currently in clinical trials for the treatment of heart failure. While the overall effect of OM on the mechano-chemical cycle of myosin is to increase the fraction of myosin molecules in the sarcomere that are strongly bound to actin, the molecular basis of its action is still not completely clear. We present here a Molecular Dynamics study of the motor domain of human cardiac myosin bound to OM, where the effects of the drug on the dynamical properties of the protein are investigated for the first time with atomistic resolution. We found that OM has a double effect on myosin dynamics, inducing a) an increased coupling of the motions of the converter and lever arm subdomains to the rest of the protein and b) a rewiring of the network of dynamic correlations, which produces preferential communication pathways between the OM binding site and distant functional regions. The location of the residues responsible for these effects suggests possible strategies for the future development of improved drugs and the targeting of specific cardiomyopathy-related mutations.


Subject(s)
Cardiac Myosins/metabolism , Molecular Dynamics Simulation , Protein Conformation/drug effects , Urea/analogs & derivatives , Allosteric Regulation , Allosteric Site , Cardiac Myosins/chemistry , Cardiac Myosins/drug effects , Crystallography, X-Ray , Humans , Urea/pharmacology
11.
Nat Commun ; 8(1): 190, 2017 08 04.
Article in English | MEDLINE | ID: mdl-28775348

ABSTRACT

Omecamtiv mecarbil is a selective, small-molecule activator of cardiac myosin that is being developed as a potential treatment for heart failure with reduced ejection fraction. Here we determine the crystal structure of cardiac myosin in the pre-powerstroke state, the most relevant state suggested by kinetic studies, both with (2.45 Å) and without (3.10 Å) omecamtiv mecarbil bound. Omecamtiv mecarbil does not change the motor mechanism nor does it influence myosin structure. Instead, omecamtiv mecarbil binds to an allosteric site that stabilizes the lever arm in a primed position resulting in accumulation of cardiac myosin in the primed state prior to onset of cardiac contraction, thus increasing the number of heads that can bind to the actin filament and undergo a powerstroke once the cardiac cycle starts. The mechanism of action of omecamtiv mecarbil also provides insights into uncovering how force is generated by molecular motors.Omecamtiv mecarbil (OM) is a cardiac myosin activator that is currently in clinical trials for heart failure treatment. Here, the authors give insights into its mode of action and present the crystal structure of OM bound to bovine cardiac myosin, which shows that OM stabilizes the pre-powerstroke state of myosin.


Subject(s)
Cardiac Myosins/chemistry , Urea/analogs & derivatives , Animals , Binding Sites , Cardiac Myosins/drug effects , Cattle , Crystallization , Protein Conformation , Urea/pharmacology
12.
Proc Natl Acad Sci U S A ; 114(10): E1796-E1804, 2017 03 07.
Article in English | MEDLINE | ID: mdl-28223517

ABSTRACT

Omecamtiv mecarbil (OM), a putative heart failure therapeutic, increases cardiac contractility. We hypothesize that it does this by changing the structural kinetics of the myosin powerstroke. We tested this directly by performing transient time-resolved FRET on a ventricular cardiac myosin biosensor. Our results demonstrate that OM stabilizes myosin's prepowerstroke structural state, supporting previous measurements showing that the drug shifts the equilibrium constant for myosin-catalyzed ATP hydrolysis toward the posthydrolysis biochemical state. OM slowed the actin-induced powerstroke, despite a twofold increase in the rate constant for actin-activated phosphate release, the biochemical step in myosin's ATPase cycle associated with force generation and the conversion of chemical energy into mechanical work. We conclude that OM alters the energetics of cardiac myosin's mechanical cycle, causing the powerstroke to occur after myosin weakly binds to actin and releases phosphate. We discuss the physiological implications for these changes.


Subject(s)
Cardiac Myosins/drug effects , Heart Failure/physiopathology , Myosins/drug effects , Urea/analogs & derivatives , Animals , Biosensing Techniques , Cardiac Myosins/chemistry , Cardiac Myosins/isolation & purification , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/chemistry , Cattle , Chickens , Heart Failure/drug therapy , Humans , Kinetics , Myocardial Contraction/drug effects , Myocardium/enzymology , Myocardium/pathology , Myosins/chemistry , Phosphates/chemistry , Phosphates/metabolism , Rabbits , Urea/administration & dosage , Urea/chemistry
13.
Curr Cardiol Rep ; 18(10): 100, 2016 10.
Article in English | MEDLINE | ID: mdl-27568794

ABSTRACT

Despite the rising prevalence of HF, new evidence-based novel therapies for patients with worsening HF remain lacking, e.g., safe inotropic therapies. Traditional inotropes increase contractility by altering intracellular calcium flux, a pathway that may be responsible for the multitude of adverse effects associated with current options. Omecamtiv mecarbil, a direct myosin activator, increases contractility through a distinct pathway by increasing the proportion of myosin heads that are bound to actin in a high-affinity state. Phase II clinical trials in patients with chronic HF with this agent seem promising. A phase III trial investigating this therapy has not yet been pursued to date.


Subject(s)
Cardiac Myosins/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Heart Failure, Systolic/drug therapy , Heart Failure, Systolic/pathology , Myocardial Contraction/drug effects , Animals , Cardiac Myosins/metabolism , Disease Models, Animal , Humans , Randomized Controlled Trials as Topic , Stroke Volume , Urea/analogs & derivatives , Urea/pharmacology , Urea/therapeutic use
14.
Expert Opin Investig Drugs ; 25(1): 117-27, 2016.
Article in English | MEDLINE | ID: mdl-26587768

ABSTRACT

INTRODUCTION: Current available inotropic agents increase cardiac contractility, but are associated with myocardial ischemia, arrhythmias, and mortality. A novel selective cardiac myosin activator, omecamtiv mecarbil (CK-1827452/ AMG-423) is a small molecule that activates the sarcomere proteins directly, resulting in prolonged systolic ejection time and increased cardiac contractility. AREAS COVERED: This paper discusses the chemistry, pharmacokinetics, clinical efficacy and safety of omecamtiv mecarbil. Omecamtiv mecarbil represents a novel therapeutic approach to directly improve cardiac function and is therefore proposed as a potential new treatment of patients with systolic heart failure. The authors review results of previous studies investigating the effect of omecamtiv mecarbil in heart failure animal models, healthy volunteers, and patients with acute and chronic systolic heart failure. EXPERT OPINION: Results of phase I and phase II studies demonstrate that omecamtiv mecarbil is safe and well tolerated both as an intravenous and oral formulation. In healthy volunteers and chronic systolic heart failure patients, administration of omecamtiv mecarbil resulted in a concentration-dependent increase of left ventricular ejection time, ejection fraction, fractional shortening, and stroke volume. The first results of a double-blind, randomized, placebo-controlled phase IIb dose-finding study with the oral formulation of omecamtiv mecarbil demonstrated beneficial effects on cardiac function and N-terminal pro-brain natriuretic peptide levels. This study will provide essential dosing information for the requisite phase III trials which will investigate whether the beneficial effects of omecamtiv mecarbil translate into improved clinical outcomes.


Subject(s)
Cardiac Myosins/drug effects , Heart Failure, Systolic/drug therapy , Urea/analogs & derivatives , Animals , Cardiac Myosins/metabolism , Cardiotonic Agents/adverse effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Disease Models, Animal , Heart Failure, Systolic/physiopathology , Humans , Myocardial Contraction/drug effects , Randomized Controlled Trials as Topic , Urea/adverse effects , Urea/pharmacology , Urea/therapeutic use
15.
Nat Commun ; 6: 7974, 2015 Aug 06.
Article in English | MEDLINE | ID: mdl-26246073

ABSTRACT

Omecamtiv Mecarbil (OM) is a small molecule allosteric effector of cardiac myosin that is in clinical trials for treatment of systolic heart failure. A detailed kinetic analysis of cardiac myosin has shown that the drug accelerates phosphate release by shifting the equilibrium of the hydrolysis step towards products, leading to a faster transition from weak to strong actin-bound states. The structure of the human ß-cardiac motor domain (cMD) with OM bound reveals a single OM-binding site nestled in a narrow cleft separating two domains of the human cMD where it interacts with the key residues that couple lever arm movement to the nucleotide state. In addition, OM induces allosteric changes in three strands of the ß-sheet that provides the communication link between the actin-binding interface and the nucleotide pocket. The OM-binding interactions and allosteric changes form the structural basis for the kinetic and mechanical tuning of cardiac myosin.


Subject(s)
Cardiac Myosins/drug effects , Urea/analogs & derivatives , Allosteric Regulation , Allosteric Site , Animals , Cardiac Myosins/chemistry , Cell Line , Green Fluorescent Proteins , Humans , Mice , Protein Structure, Tertiary , Urea/pharmacology
16.
Oncol Rep ; 32(4): 1473-80, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25051015

ABSTRACT

4-Amino-2-trifluoromethyl-phenyl retinate (ATPR) is a novel all-trans retinoic acid (ATRA) derivative which was reported to have a superior antitumor effect in breast cancer cells. However, little is known about its antitumor effects on human gastric cancer cells and the mechanisms have not been fully elucidated. The results of the present study suggest that in the human gastric carcinoma cell line BGC-823, ATPR plays a more effective role than ATRA at the same dose in inhibiting proliferation, migration and inducing differentiation after the same treatment time. Furthermore, we investigated the preliminary mechanism of ATPR's anti­migration effect. Immunofluorescence assay demonstrated that claudin-18 positioned from cytoplasm to cell surface following ATPR stimuli. Real-time quantitative RT-PCR and western blot analyses showed that ATPR had significant effects on downregulation of the phosphorylation level of myosin light chain II (MLC II) by suppressing myosin light chain kinase (MLCK) and Rho-associated coiled-coil containing kinase (ROCK), as well as its regulation in the protein expression of RARα and RARß. Moreover, ATPR increased the activity of myosin phosphatase by inhibiting ROCK. Consequently, ATPR showed more promising antitumor effects than ATRA in BGC-823 in vitro, and it may conduct its anti-migration effects by decreasing the phosphorylation level of MLC II, as well as by regulating MLCK and ROCK as downstream target genes.


Subject(s)
Antineoplastic Agents/pharmacology , Cardiac Myosins/drug effects , Cell Movement/drug effects , Myosin Light Chains/drug effects , Retinoids/pharmacology , Stomach Neoplasms/metabolism , Cardiac Myosins/metabolism , Cell Line, Tumor , Down-Regulation , Humans , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/drug effects , Myosin-Light-Chain Kinase/metabolism , Phosphorylation/drug effects , Receptors, Retinoic Acid/drug effects , Receptors, Retinoic Acid/metabolism , Retinoic Acid Receptor alpha , rho-Associated Kinases/drug effects , rho-Associated Kinases/metabolism
17.
Cardiol Rev ; 21(3): 155-9, 2013.
Article in English | MEDLINE | ID: mdl-23018669

ABSTRACT

Systolic heart failure remains a leading cause of death and disability, and available pharmacologic treatments for heart failure are limited in both safety and effectiveness. Existing drugs focus on diverse mechanisms related to the pathophysiology of heart failure, yet none directly target the central feature of systolic heart failure, decreased cardiac contractility. Cardiac myosin activators, specifically omecamtiv mecarbil (formerly CK-1827452), directly activate the enzymatic pathway within the cardiac myocyte leading to ventricular contraction. This unique inotropic agent has been shown in preclinical and clinical studies to be effective in improving cardiac contractility by increasing systolic ejection time without the unwanted effects of the currently available indirect inotropic drugs. Cardiac myosin activators show great promise and may prove to be a safer and more effective therapeutic approach for the treatment of systolic heart failure.


Subject(s)
Cardiac Myosins/drug effects , Cardiotonic Agents/therapeutic use , Heart Failure, Systolic/drug therapy , Urea/analogs & derivatives , Animals , Cardiotonic Agents/pharmacology , Clinical Trials, Phase I as Topic , Disease Models, Animal , Dogs , Drug Evaluation , Humans , Randomized Controlled Trials as Topic , Rats , Rats, Sprague-Dawley , Urea/pharmacology , Urea/therapeutic use
18.
Eur Heart J ; 32(15): 1838-45, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21388993

ABSTRACT

Intrinsic inotropic stimulation of the heart is central to the regulation of cardiovascular function, and exogenous inotropic therapies have been used clinically for decades. Unfortunately, current inotropic drugs have consistently failed to show beneficial effects beyond short-term haemodynamic improvement in patients with heart failure. To address these limitations, new agents targeting novel mechanisms are being developed: (i) istaroxime has been developed as a non-glycoside inhibitor of the sodium-potassium-ATPase with additional stimulatory effects on the sarcoplasmic reticulum calcium pump (SERCA) and has shown lusitropic and inotropic properties in experimental and early clinical studies; (ii) from a mechanistic point of view, the cardiac myosin activators, directly activating the acto-myosin cross-bridges, are most appealing with improved cardiac performance in both animal and early clinical studies; (iii) gene therapy approaches have been successfully employed to increase myocardial SERCA2a; (iv) nitroxyl donors have been developed and have shown evidence of positive lusitropic and inotropic, as well as potent vasodilatory effects in early animal studies; (v) the ryanodine receptor stabilizers reduce pathological leak of calcium from the sarcoplasmic reticulum with initial promising pre-clinical results; and finally, (vi) metabolic energy modulation may represent a promising means to improve contractile performance of the heart. There is an urgent clinical need for agents that improve cardiac performance with a favourable safety profile. These current novel approaches to improving cardiac function provide the hope that such agents may soon be available.


Subject(s)
Cardiotonic Agents/therapeutic use , Heart Failure/drug therapy , Technology, Pharmaceutical/trends , Calcium Channels, L-Type/drug effects , Cardiac Myosins/drug effects , Drug Design , Energy Metabolism/drug effects , Forecasting , Genetic Therapy/trends , Heart Conduction System/drug effects , Heart Conduction System/physiology , Humans , Myocardial Contraction/drug effects , Sarcoplasmic Reticulum/drug effects
19.
Circ Heart Fail ; 3(4): 522-7, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20498236

ABSTRACT

BACKGROUND: Therapy for chronic systolic heart failure (sHF) has improved over the past 2 decades, but the armamentarium of drugs is limited and consequently sHF remains a leading cause of death and disability. In this investigation, we examined the effects of a novel cardiac myosin activator, omecamtiv mecarbil (formerly CK-1827452) in 2 different models of heart failure. METHODS AND RESULTS: Two different models of sHF were used: (1) pacing-induced sHF after myocardial infarction (MI-sHF) and (2) pacing-induced sHF after 1 year of chronic pressure overload left ventricular hypertrophy (LVH-sHF). Omecamtiv mecarbil increased systolic function in sHF dogs, chronically instrumented to measure LV pressure, wall thickness, and cardiac output. Omecamtiv mecarbil, infused for 24 hours, induced a sustained increase without desensitization (P<0.05) in wall thickening (25+/-6.2%), stroke volume (44+/-6.5%) and cardiac output (22+/-2.8%), and decreased heart rate (15+/-3.0%). The major differences between the effect of omecamtiv mecarbil on cardiac function and the effect induced by a catecholamine, for example, dobutamine, is that omecamtiv mecarbil did not increase LV dP/dt but rather increased LV systolic ejection time by 26+/-2.9% in sHF. Another key difference is that myocardial O(2) consumption (MVO(2)), which increases with catecholamines, was not significantly affected by omecamtiv mecarbil. CONCLUSIONS: These results demonstrate that chronic infusion of the cardiac myosin activator, omecamtiv mecarbil, improves LV function in sHF without the limitations of progressive desensitization and increased MVO(2.) This unique profile may provide a new therapeutic approach for patients with sHF.


Subject(s)
Cardiac Myosins/drug effects , Heart Failure, Systolic/drug therapy , Myocardial Contraction/drug effects , Ventricular Function, Left/drug effects , Analysis of Variance , Animals , Cardiac Myosins/metabolism , Consciousness , Disease Models, Animal , Dobutamine/pharmacology , Dogs , Drug Administration Schedule , Female , Heart Failure, Systolic/physiopathology , Heart Function Tests , Infusions, Intravenous , Male , Myocardial Infarction/drug therapy , Myocardial Infarction/physiopathology , Oxygen Consumption/drug effects , Probability , Random Allocation , Stroke Volume/drug effects , Treatment Outcome , Urea/analogs & derivatives , Urea/pharmacology , Ventricular Remodeling/drug effects
20.
Heart Fail Rev ; 14(4): 289-98, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19234787

ABSTRACT

Decreased systolic function is a central factor in the pathogenesis of heart failure, yet there are no safe medical therapies to improve cardiac function in patients. Currently available inotropes, such as dobutamine and milrinone, increase cardiac contractility at the expense of increased intracellular concentrations of calcium and cAMP, contributing to increased heart rate, hypotension, arrhythmias, and mortality. These adverse effects are inextricably linked to their inotropic mechanism of action. A new class of pharmacologic agents, cardiac myosin activators, directly targets the kinetics of the myosin head. In vitro studies have demonstrated that these agents increase the rate of effective myosin cross-bridge formation, increasing the duration and amount of myocyte contraction, and inhibit non-productive consumption of ATP, potentially improving myocyte energy utilization, with no effect on intracellular calcium or cAMP. Animal models have shown that this novel mechanism increases the systolic ejection time, resulting in improved stroke volume, fractional shortening, and hemodynamics with no effect on myocardial oxygen demand, culminating in significant increases in cardiac efficiency. A first-in-human study in healthy volunteers with the lead cardiac myosin activator, CK-1827452, as well as preliminary results from a study in patients with stable chronic heart failure, have extended these findings to humans, demonstrating significant increases in systolic ejection time, fractional shortening, stroke volume, and cardiac output. These studies suggest that cardiac myosin activators offer the promise of a safe and effective treatment for heart failure. A program of clinical studies are being planned to test whether CK-1827452 will fulfill that promise.


Subject(s)
Cardiac Myosins/drug effects , Heart Failure/drug therapy , Urea/analogs & derivatives , Animals , Cardiac Myosins/physiology , Cardiotonic Agents/therapeutic use , Clinical Trials, Phase I as Topic , Clinical Trials, Phase II as Topic , Humans , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Urea/administration & dosage , Urea/pharmacology , Urea/therapeutic use , Ventricular Function, Left/drug effects , Ventricular Function, Left/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...