Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 9 de 9
Filtrer
Plus de filtres










Base de données
Gamme d'année
2.
Future Sci OA ; 9(10): FSO898, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37753355

RÉSUMÉ

Aim: This meta-analysis was performed to assess the efficacy and safety of mavacamten in patients with hypertrophic cardiomyopathy. Methods & materials: A search was conducted using PubMed, Cochrane, and Scopus up to August 2022 for randomized studies reporting our pre-specified outcomes. Results: It was observed that mavacamten significantly improved New York Heart Association class (p < 0.009), Clinical Summary Score of the Kansas City Cardiomyopathy Questionnaire (p = 0.02), post-exercise left ventricular outflow tract gradient (p < 0.00001), functional end point (p = 0.05), and lowered septal reduction therapy rates (p < 0.00001). However, there were no significant differences in the ≥1 severe adverse events, ≥1 treatment-emergent adverse events, left ventricular volume index, left ventricular filling pressure, left ventricular end-diastolic volume index, and peak oxygen uptake (pVO2). Conclusion: Future large-scale trials are required to confirm our results and determine the long-term benefits and risks of mavacamten use in these patients.


Mavacamten is a recently introduced medication that relaxes the heart muscle and is indicated for patients with hypertrophic cardiomyopathy (a disease in which parts of the heart become thick and stiff). To determine the effectiveness and safety of this drug, the results of clinical trials were combined in order to produce an overall estimate. Overall, it was observed that mavacamten improved most functional parameters related to the heart and demonstrated no significant increases in the number of side effects. This suggests the effectiveness and safety of mavacamten, although further trials are needed to confirm our results.

3.
Bioanalysis ; 15(12): 695-706, 2023 Jun.
Article de Anglais | MEDLINE | ID: mdl-37254776

RÉSUMÉ

Aim: This study investigated the impact of food intake and circadian rhythms on the pharmacokinetics of mavacamten. Materials & methods: A sensitive bioanalytical method for quantifying mavacamten in rat plasma was developed and validated. This method was applied to assess the effect of chronopharmacology and food intake on the pharmacokinetics of mavacamten in rats. Results: A circadian variation at two doses resulted in significant changes in the volume of distribution, clearance and time of maximum plasma concentration of mavacamten (p < 0.05). In addition, food intake had an insignificant impact on the pharmacokinetic parameters except for the time of maximum plasma concentration (p < 0.05). Conclusion: These pharmacokinetic changes and human chronotype findings will help optimize dosing time.


Sujet(s)
Benzylamines , Uracile , Humains , Rats , Animaux , Benzylamines/pharmacologie , Cinétique
4.
Circulation ; 147(11): 850-863, 2023 03 14.
Article de Anglais | MEDLINE | ID: mdl-36335531

RÉSUMÉ

BACKGROUND: Septal reduction therapy (SRT) in patients with intractable symptoms from obstructive hypertrophic cardiomyopathy (oHCM) is associated with variable morbidity and mortality. The VALOR-HCM trial (A Study to Evaluate Mavacamten in Adults with Symptomatic Obstructive Hypertrophic Cardiomyopathy Who Are Eligible for Septal Reduction Therapy) examined the effect of mavacamten on the need for SRT through week 32 in oHCM. METHODS: A double-blind randomized placebo-controlled multicenter trial at 19 US sites included patients with oHCM on maximal tolerated medical therapy referred for SRT with left ventricular outflow tract gradient ≥50 mm Hg at rest or provocation (enrollment, July 2020-October 2021). The group initially randomized to mavacamten continued the drug for 32 weeks, and the placebo group crossed over to dose-blinded mavacamten from week 16 to week 32. Dose titrations were based on investigator-blinded echocardiographic assessment of left ventricular outflow tract gradient and left ventricular ejection fraction. The principal end point was the proportion of patients proceeding with SRT or remaining guideline eligible at 32 weeks in both treatment groups. RESULTS: From the 112 randomized patients with oHCM, 108 (mean age, 60.3 years; 50% men; 94% in New York Heart Association class III/IV) qualified for week 32 evaluation (56 in the original mavacamten group and 52 in the placebo cross-over group). After 32 weeks, 6 of 56 patients (10.7%) in the original mavacamten group and 7 of 52 patients (13.5%) in the placebo cross-over group met SRT guideline criteria or elected to undergo SRT. After 32 weeks, a sustained reduction in resting left ventricular outflow tract gradient (-33.0 mm Hg [95% CI, -41.1 to -24.9]) and Valsalva left ventricular outflow tract gradient (-43.0 mm Hg [95% CI, -52.1 to -33.9]) was observed in the original mavacamten group. A similar reduction in resting (-33.7 mm Hg [95% CI, -42.2 to -25.2]) and Valsalva (-52.9 mm Hg [95% CI, -63.2 to -42.6]) gradients was quantified in the cross-over group after 16 weeks of mavacamten. After 32 weeks, improvement by ≥1 New York Heart Association class was observed in 48 of 53 patients (90.6%) in the original mavacamten group and 35 of 50 patients (70%) after 16 weeks in the cross-over group. CONCLUSIONS: In severely symptomatic patients with oHCM, 32 weeks of mavacamten treatment showed sustained reduction in the proportion proceeding to SRT or remaining guideline eligible, with similar effects observed in patients who crossed over from placebo after 16 weeks. REGISTRATION: URL: https://www. CLINICALTRIALS: gov; Unique identifier: NCT04349072.


Sujet(s)
Cardiomyopathie hypertrophique , Fonction ventriculaire gauche , Adulte , Mâle , Humains , Adulte d'âge moyen , Femelle , Débit systolique , Cardiomyopathie hypertrophique/imagerie diagnostique , Cardiomyopathie hypertrophique/traitement médicamenteux , Benzylamines/pharmacologie
5.
Rev Port Cardiol ; 41(8): 693-703, 2022 Aug.
Article de Anglais, Portugais | MEDLINE | ID: mdl-36073269

RÉSUMÉ

Hypertrophic cardiomyopathy (HCM) is one of the most common inherited cardiac diseases, defined as a left ventricular wall thickness of ≥15 mm, in the absence of other causes of abnormal ventricular loading. A major hallmark of this disease is the presence of left ventricular outflow tract obstruction, which develops in up to three quarters of patients, referred to as obstructive hypertrophic cardiomyopathy. Current treatment is offered to symptomatic patients, based on the presence of documented left ventricular obstruction, aimed at reducing symptoms and disease progression. This is achieved through pharmacological empirical therapy, surgery, alcohol ablation and/or pacing. Mavacamten is a first-in-class allosteric inhibitor of cardiac myosin that promises to provide clinicians with targeted therapy for these patients. The aim of this review is to provide a general overview of the modern approach to the diagnosis and management of HCM, as well as to integrate all the current knowledge on mavacamten, in anticipation of a future change in the treatment algorithm of patients with HCM.

7.
Circ Res ; 124(8): 1172-1183, 2019 04 12.
Article de Anglais | MEDLINE | ID: mdl-30700234

RÉSUMÉ

RATIONALE: Human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) in combination with CRISPR/Cas9 genome editing provide unparalleled opportunities to study cardiac biology and disease. However, sarcomeres, the fundamental units of myocyte contraction, are immature and nonlinear in hiPSC-CMs, which technically challenge accurate functional interrogation of contractile parameters in beating cells. Furthermore, existing analysis methods are relatively low-throughput, indirectly assess contractility, or only assess well-aligned sarcomeres found in mature cardiac tissues. OBJECTIVE: We aimed to develop an analysis platform that directly, rapidly, and automatically tracks sarcomeres in beating cardiomyocytes. The platform should assess sarcomere content, contraction and relaxation parameters, and beat rate. METHODS AND RESULTS: We developed SarcTrack, a MatLab software that monitors fluorescently tagged sarcomeres in hiPSC-CMs. The algorithm determines sarcomere content, sarcomere length, and returns rates of sarcomere contraction and relaxation. By rapid measurement of hundreds of sarcomeres in each hiPSC-CM, SarcTrack provides large data sets for robust statistical analyses of multiple contractile parameters. We validated SarcTrack by analyzing drug-treated hiPSC-CMs, confirming the contractility effects of compounds that directly activate (CK-1827452) or inhibit (MYK-461) myosin molecules or indirectly alter contractility (verapamil and propranolol). SarcTrack analysis of hiPSC-CMs carrying a heterozygous truncation variant in the myosin-binding protein C ( MYBPC3) gene, which causes hypertrophic cardiomyopathy, recapitulated seminal disease phenotypes including cardiac hypercontractility and diminished relaxation, abnormalities that normalized with MYK-461 treatment. CONCLUSIONS: SarcTrack provides a direct and efficient method to quantitatively assess sarcomere function. By improving existing contractility analysis methods and overcoming technical challenges associated with functional evaluation of hiPSC-CMs, SarcTrack enhances translational prospects for sarcomere-regulating therapeutics and accelerates interrogation of human cardiac genetic variants.


Sujet(s)
Algorithmes , Cellules souches pluripotentes induites/physiologie , Myocytes cardiaques/physiologie , Sarcomères/physiologie , Logiciel , Benzylamines/antagonistes et inhibiteurs , Benzylamines/pharmacologie , Agents cardiovasculaires/pharmacologie , Protéines de transport/génétique , Clustered regularly interspaced short palindromic repeats , Conception assistée par ordinateur , Fluorescence , Humains , Cellules souches pluripotentes induites/effets des médicaments et des substances chimiques , Microscopie à force atomique/méthodes , Contraction myocardique , Myocytes cardiaques/effets des médicaments et des substances chimiques , Myosines/effets des médicaments et des substances chimiques , Myosines/métabolisme , Propranolol/pharmacologie , Uracile/analogues et dérivés , Uracile/antagonistes et inhibiteurs , Uracile/pharmacologie , Urée/analogues et dérivés , Urée/pharmacologie , Vérapamil/pharmacologie , Enregistrement sur magnétoscope
8.
Xenobiotica ; 49(6): 718-733, 2019 Jun.
Article de Anglais | MEDLINE | ID: mdl-30044681

RÉSUMÉ

Mavacamten is a small molecule modulator of cardiac myosin designed as an orally administered drug for the treatment of patients with hypertrophic cardiomyopathy. The current study objectives were to assess the preclinical pharmacokinetics of mavacamten for the prediction of human dosing and to establish the potential need for clinical pharmacokinetic studies characterizing drug-drug interaction potential. Mavacamten does not inhibit CYP enzymes, but at high concentrations relative to anticipated therapeutic concentrations induces CYP2B6 and CYP3A4 enzymes in vitro. Mavacamten showed high permeability and low efflux transport across Caco-2 cell membranes. In human hepatocytes, mavacamten was not a substrate for drug transporters OATP, OCT and NTCP. Mavacamten was determined to have minimal drug-drug interaction risk. In vitro mavacamten metabolite profiles included phase I- and phase II-mediated metabolism cross-species. Major pathways included aromatic hydroxylation (M1), aliphatic hydroxylation (M2); N-dealkylation (M6), and glucuronidation of the M1-metabolite (M4). Reaction phenotyping revealed CYPs 2C19 and 3A4/3A5 predominating. Mavacamten demonstrated low clearance, high volume of distribution, long terminal elimination half-life and excellent oral bioavailability cross-species. Simple four-species allometric scaling led to predicted plasma clearance, volume of distribution and half-life of 0.51 mL/min/kg, 9.5 L/kg and 9 days, respectively, in human.


Sujet(s)
Benzylamines/pharmacocinétique , Uracile/analogues et dérivés , Animaux , Benzylamines/composition chimique , Benzylamines/métabolisme , Cellules Caco-2 , Myosines cardiaques/métabolisme , Cardiomyopathie hypertrophique/traitement médicamenteux , Cytochrome P-450 enzyme system/composition chimique , Cytochrome P-450 enzyme system/métabolisme , Chiens , Interactions médicamenteuses , Hépatocytes/métabolisme , Humains , Macaca fascicularis , Mâle , Taux de clairance métabolique , Souris de lignée ICR , Microsomes du foie , Rat Sprague-Dawley , Uracile/composition chimique , Uracile/métabolisme , Uracile/pharmacocinétique
9.
J Biol Chem ; 292(40): 16571-16577, 2017 10 06.
Article de Anglais | MEDLINE | ID: mdl-28808052

RÉSUMÉ

Mavacamten, formerly known as MYK-461 is a recently discovered novel small-molecule modulator of cardiac myosin that targets the underlying sarcomere hypercontractility of hypertrophic cardiomyopathy, one of the most prevalent heritable cardiovascular disorders. Studies on isolated cells and muscle fibers as well as intact animals have shown that mavacamten inhibits sarcomere force production, thereby reducing cardiac contractility. Initial mechanistic studies have suggested that mavacamten primarily reduces the steady-state ATPase activity by inhibiting the rate of phosphate release of ß-cardiac myosin-S1, but the molecular mechanism of action of mavacamten has not been described. Here we used steady-state and presteady-state kinetic analyses to investigate the mechanism of action of mavacamten. Transient kinetic analyses revealed that mavacamten modulates multiple steps of the myosin chemomechanical cycle. In addition to decreasing the rate-limiting step of the cycle (phosphate release), mavacamten reduced the number of myosin-S1 heads that can interact with the actin thin filament during transition from the weakly to the strongly bound state without affecting the intrinsic rate. Mavacamten also decreased the rate of myosin binding to actin in the ADP-bound state and the ADP-release rate from myosin-S1 alone. We, therefore, conclude that mavacamten acts on multiple stages of the myosin chemomechanical cycle. Although the primary mechanism of mavacamten-mediated inhibition of cardiac myosin is the decrease of phosphate release from ß-cardiac myosin-S1, a secondary mechanism decreases the number of actin-binding heads transitioning from the weakly to the strongly bound state, which occurs before phosphate release and may provide an additional method to modulate myosin function.


Sujet(s)
ADP/composition chimique , Adénosine triphosphate/composition chimique , Benzylamines/composition chimique , Myosines cardiaques/composition chimique , Sous-fragments de myosine/composition chimique , Sarcomères/composition chimique , Uracile/analogues et dérivés , ADP/métabolisme , Adénosine triphosphate/métabolisme , Animaux , Myosines cardiaques/métabolisme , Cardiomégalie/métabolisme , Bovins , Sous-fragments de myosine/métabolisme , Sarcomères/métabolisme , Uracile/composition chimique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...