Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 466
Filter
Add more filters

Publication year range
1.
Proc Natl Acad Sci U S A ; 118(52)2021 12 28.
Article in English | MEDLINE | ID: mdl-34930837

ABSTRACT

The particulate guanylyl cyclase A receptor (GC-A), via activation by its endogenous ligands atrial natriuretic peptide (ANP) and b-type natriuretic peptide (BNP), possesses beneficial biological properties such as blood pressure regulation, natriuresis, suppression of adverse remodeling, inhibition of the renin-angiotensin-aldosterone system, and favorable metabolic actions through the generation of its second messenger cyclic guanosine monophosphate (cGMP). Thus, the GC-A represents an important molecular therapeutic target for cardiovascular disease and its associated risk factors. However, a small molecule that is orally bioavailable and directly targets the GC-A to potentiate cGMP has yet to be discovered. Here, we performed a cell-based high-throughput screening campaign of the NIH Molecular Libraries Small Molecule Repository, and we successfully identified small molecule GC-A positive allosteric modulator (PAM) scaffolds. Further medicinal chemistry structure-activity relationship efforts of the lead scaffold resulted in the development of a GC-A PAM, MCUF-651, which enhanced ANP-mediated cGMP generation in human cardiac, renal, and fat cells and inhibited cardiomyocyte hypertrophy in vitro. Further, binding analysis confirmed MCUF-651 binds to GC-A and selectively enhances the binding of ANP to GC-A. Moreover, MCUF-651 is orally bioavailable in mice and enhances the ability of endogenous ANP and BNP, found in the plasma of normal subjects and patients with hypertension or heart failure, to generate GC-A-mediated cGMP ex vivo. In this work, we report the discovery and development of an oral, small molecule GC-A PAM that holds great potential as a therapeutic for cardiovascular, renal, and metabolic diseases.


Subject(s)
Cardiovascular Agents , Cardiovascular Diseases/metabolism , Cyclic GMP/metabolism , Natriuretic Peptides/metabolism , Receptors, Atrial Natriuretic Factor , Aged , Allosteric Regulation , Animals , Cardiovascular Agents/chemistry , Cardiovascular Agents/metabolism , Cardiovascular Agents/pharmacokinetics , Cardiovascular Agents/pharmacology , Cells, Cultured , Female , HEK293 Cells , High-Throughput Screening Assays , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged , Myocytes, Cardiac/metabolism , Receptors, Atrial Natriuretic Factor/chemistry , Receptors, Atrial Natriuretic Factor/drug effects , Receptors, Atrial Natriuretic Factor/metabolism
2.
J Pharmacol Exp Ther ; 377(2): 218-231, 2021 05.
Article in English | MEDLINE | ID: mdl-33648939

ABSTRACT

Cardiovascular adverse effects in drug development are a major source of compound attrition. Characterization of blood pressure (BP), heart rate (HR), stroke volume (SV), and QT-interval prolongation are therefore necessary in early discovery. It is, however, common practice to analyze these effects independently of each other. High-resolution time courses are collected via telemetric techniques, but only low-resolution data are analyzed and reported. This ignores codependencies among responses (HR, BP, SV, and QT-interval) and separation of system (turnover properties) and drug-specific properties (potencies, efficacies). An analysis of drug exposure-time and high-resolution response-time data of HR and mean arterial blood pressure was performed after acute oral dosing of ivabradine, sildenafil, dofetilide, and pimobendan in Han-Wistar rats. All data were modeled jointly, including different compounds and exposure and response time courses, using a nonlinear mixed-effects approach. Estimated fractional turnover rates [h-1, relative standard error (%RSE) within parentheses] were 9.45 (15), 30.7 (7.8), 3.8 (13), and 0.115 (1.7) for QT, HR, total peripheral resistance, and SV, respectively. Potencies (nM, %RSE within parentheses) were IC 50 = 475 (11), IC 50 = 4.01 (5.4), EC 50 = 50.6 (93), and IC 50 = 47.8 (16), and efficacies (%RSE within parentheses) were I max = 0.944 (1.7), Imax = 1.00 (1.3), E max = 0.195 (9.9), and Imax = 0.745 (4.6) for ivabradine, sildenafil, dofetilide, and pimobendan. Hill parameters were estimated with good precision and below unity, indicating a shallow concentration-response relationship. An equilibrium concentration-biomarker response relationship was predicted and displayed graphically. This analysis demonstrates the utility of a model-based approach integrating data from different studies and compounds for refined preclinical safety margin assessment. SIGNIFICANCE STATEMENT: A model-based approach was proposed utilizing biomarker data on heart rate, blood pressure, and QT-interval. A pharmacodynamic model was developed to improve assessment of high-resolution telemetric cardiovascular safety data driven by different drugs (ivabradine, sildenafil, dofetilide, and pimobondan), wherein system- (turnover rates) and drug-specific parameters (e.g., potencies and efficacies) were sought. The model-predicted equilibrium concentration-biomarker response relationships and was used for safety assessment (predictions of 20% effective concentration, for example) of heart rate, blood pressure, and QT-interval.


Subject(s)
Biomarkers, Pharmacological/blood , Blood Pressure , Cardiovascular Agents/toxicity , Heart Rate , Animals , Cardiotoxicity/blood , Cardiotoxicity/etiology , Cardiotoxicity/physiopathology , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/pharmacokinetics , Ivabradine/administration & dosage , Ivabradine/pharmacokinetics , Ivabradine/toxicity , Male , Phenethylamines/administration & dosage , Phenethylamines/pharmacokinetics , Phenethylamines/toxicity , Pyridazines/administration & dosage , Pyridazines/pharmacokinetics , Pyridazines/toxicity , Rats , Rats, Wistar , Sildenafil Citrate/administration & dosage , Sildenafil Citrate/pharmacokinetics , Sildenafil Citrate/toxicity , Sulfonamides/administration & dosage , Sulfonamides/pharmacokinetics , Sulfonamides/toxicity
3.
Drug Metab Dispos ; 49(8): 619-628, 2021 08.
Article in English | MEDLINE | ID: mdl-34011533

ABSTRACT

Omecamtiv mecarbil (OM) is a novel cardiac myosin activator that is currently in clinical development for the treatment of heart failure. The absorption and disposition of [14C]OM (60 µCi) were studied after a single intravenous infusion (35 mg over 1 hour) or oral solution dose (35 mg) in 14 healthy male subjects. Mean recovery of the administered [14C]OM dose was 85.1% and 86.5% over 336 hours for the intravenous and oral routes, respectively. After intravenous dosing, 47.8% and 37.3% of the dose was recovered in urine and feces, respectively; after oral dosing, 48.6% and 38.0% was recovered in urine and feces, respectively. Unchanged OM accounted for a minor percentage of radioactivity in urine (mean 7.7% of dose) and feces (mean 4.1% of dose) across all subjects. The major metabolites recovered in urine and feces were M3 (decarbamoylation product) and sequential metabolite M4 (lactam of M3), which accounted for means of 26.5% and 11.6% of the administered dose, respectively. The CYP4 family of enzymes was primarily responsible for the formation of M3 based on in vitro studies. Other metabolic pathways accounted for 14.9% of the administered dose. In pooled plasma, OM, M3, and M4 accounted for 83.8%, 6.0%, and 3.3% of the total [14C]OM-related materials. No other plasma metabolites constituted more than 3% of the administered dose. The bioavailability for OM solution was 93.5% after rapid and extensive absorption. SIGNIFICANCE STATEMENT: This study characterized the absorption and disposition of OM, a novel small molecule being developed for the treatment of heart failure. OM was primarily cleared through metabolism by the CYP4 family through oxidative cleavage of a terminal carbamate moiety that resembles hydrolysis.


Subject(s)
Cytochrome P450 Family 4/metabolism , Heart Failure/drug therapy , Hepatobiliary Elimination/physiology , Intestinal Absorption/physiology , Renal Elimination/physiology , Urea/analogs & derivatives , Administration, Intravenous , Administration, Oral , Adult , Biological Availability , Biotransformation , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/pharmacokinetics , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Urea/administration & dosage , Urea/pharmacokinetics
4.
Bioorg Med Chem Lett ; 50: 128325, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34403724

ABSTRACT

Heart failure (HF) treatment remains a critical unmet medical need. Studies in normal healthy volunteers and HF patients have shown that [Pyr1]apelin-13, the endogenous ligand for the APJ receptor, improves cardiac function. However, the short half-life of [Pyr1]apelin-13 and the need for intravenous administration have limited the therapeutic potential for chronic use. We sought to identify potent, small-molecule APJ agonists with improved pharmaceutical properties to enable oral dosing in clinical studies. In this manuscript, we describe the identification of a series of pyrimidinone sulfones as a structurally differentiated series to the clinical lead (compound 1). Optimization of the sulfone series for potency, metabolic stability and oral bioavailability led to the identification of compound 22, which showed comparable APJ potency to [Pyr1]apelin-13 and exhibited an acceptable pharmacokinetic profile to advance to the acute hemodynamic rat model.


Subject(s)
Apelin Receptors/agonists , Cardiovascular Agents/pharmacology , Cardiovascular Agents/pharmacokinetics , Intercellular Signaling Peptides and Proteins/pharmacology , Animals , Area Under Curve , Cardiovascular Agents/chemical synthesis , Drug Design , Half-Life , Humans , Intercellular Signaling Peptides and Proteins/chemistry , Macaca fascicularis , Molecular Structure , Pyrimidinones/chemistry , Pyrimidinones/pharmacology , Rats , Structure-Activity Relationship
5.
Cardiovasc Drugs Ther ; 35(3): 441-454, 2021 06.
Article in English | MEDLINE | ID: mdl-32424652

ABSTRACT

PURPOSE: Major depressive disorder (MDD) and anxiety disorders (AD) are both highly prevalent among individuals with arrhythmia, ischemic heart disease, heart failure, hypertension, and dyslipidemia. There should be increased support for MDD and AD diagnosis and treatment in individuals with cardiac diseases, because treatment rates have been low. However, cardiac-psychiatric drug interaction can make pharmacologic treatment challenging. METHODS: The objective of the present systematic review was to investigate cardiac-psychiatric drug interactions in three different widely used pharmacological databases (Micromedex, Up to Date, and ClinicalKey). RESULTS: Among 4914 cardiac-psychiatric drug combinations, 293 significant interactions were found (6.0%). When a problematic interaction is detected, it may be easier to find an alternative cardiac medication (32.6% presented some interaction) than a psychiatric one (76.9%). Antiarrhythmics are the major class of concern. The most common problems produced by these interactions are related to cardiotoxicity (QT prolongation, torsades de pointes, cardiac arrest), increased exposure of cytochrome P450 2D6 (CYP2D6) substrates, or reduced renal clearance of organic cation transporter 2 (OCT2) substrates and include hypertensive crisis, increased risk of bleeding, myopathy, and/or rhabdomyolysis. CONCLUSION: Unfortunately, there is considerable inconsistency among the databases searched, such that a clinician's discretion and clinical experience remain invaluable tools for the management of patients with comorbidities present in psychiatric and cardiac disorders. The possibility of an interaction should be considered. With a multidisciplinary approach, particularly involving a pharmacist, the prescriber should be alerted to the possibility of an interaction. MDD and AD pharmacologic treatment in cardiac patients could be implemented safely both by cardiologists and psychiatrists. TRIAL REGISTRATION: PROSPERO Systematic Review Registration Number: CRD42018100424.


Subject(s)
Antipsychotic Agents/pharmacology , Cardiovascular Agents/pharmacology , Cardiovascular Diseases/drug therapy , Databases, Pharmaceutical/statistics & numerical data , Depressive Disorder, Major/drug therapy , Antipsychotic Agents/adverse effects , Antipsychotic Agents/pharmacokinetics , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Cardiovascular Diseases/epidemiology , Cytochrome P-450 CYP2D6/drug effects , Depressive Disorder, Major/epidemiology , Drug Interactions , Humans , Metabolic Clearance Rate , Organic Cation Transporter 2/drug effects
6.
Cardiovasc Drugs Ther ; 35(5): 1045-1056, 2021 10.
Article in English | MEDLINE | ID: mdl-33411112

ABSTRACT

Ivabradine is a unique agent that is distinct from beta-blockers and calcium channel blockers as it reduces heart rate without affecting myocardial contractility or vascular tone. Ivabradine is a use-dependent inhibitor targeting the sinoatrial node. It is approved for use in the United States as an adjunct therapy for heart rate reduction in patients with heart failure with reduced ejection fraction. In this scenario, ivabradine has demonstrated improved clinical outcomes due to reduction in heart failure readmissions. However, there has been conflicting evidence from prospective studies and randomized controlled trials for its use in stable ischemic heart disease regarding efficacy in symptom reduction and mortality benefit. Ivabradine may also play a role in the treatment of patients with inappropriate sinus tachycardia, who often cannot tolerate beta-blockers and/or calcium channel blockers. In this review, we highlight the evidence for the nuances of using ivabradine in heart failure, stable ischemic heart disease, and inappropriate sinus tachycardia to raise awareness for its vital role in the treatment of select populations.


Subject(s)
Cardiovascular Agents/pharmacology , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Ivabradine/pharmacology , Ivabradine/therapeutic use , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Humans , Ivabradine/adverse effects , Ivabradine/pharmacokinetics , Myocardial Ischemia/drug therapy , Prospective Studies , Randomized Controlled Trials as Topic , Stroke Volume/drug effects , Tachycardia, Sinus/drug therapy
7.
Biomed Chromatogr ; 35(7): e5095, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33607700

ABSTRACT

A sensitive and robust method has been developed using an ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) assay to quantify Tat-K13, a novel interfering peptide for the treatment of ischemic stroke, in human plasma. Automated solid-phase extraction on a Waters Oasis WCX (30 µm, 10 mg) 96-well plate was used to extract Tat-K13 from human plasma and the extracts were separated on a Waters Acquity CSH column (2.1 × 50 mm i.d., 1.7 µm) with a gradient elution method by mobile phase A (nonafluoropentanoic acid-acetic acid-water, 1:2:1000, v/v/v) and B (nonafluoropentanoic acid-acetic acid-water-acetonitrile, 1:2:100:900, v/v/v/v). The method was fully validated following international bioanalytical guidelines and showed good linearity from 2.10 to 1,050 ng/ml. The method was successfully applied to investigate the clinical pharmacokinetics of Tat-K13 in health volunteers. Rapid elimination of Tat-K13 from the body was observed, with half-life ranging from 0.26 to 0.78 h across different dose levels. The exposure of Tat-K13 was approximately dose-dependent in terms of the area under the concentration-time curve and peak concentration.


Subject(s)
Cardiovascular Agents , Chromatography, High Pressure Liquid/methods , Ischemic Stroke/drug therapy , Peptides , Tandem Mass Spectrometry/methods , Adolescent , Adult , Cardiovascular Agents/blood , Cardiovascular Agents/pharmacokinetics , Cardiovascular Agents/therapeutic use , Humans , Middle Aged , Peptides/blood , Peptides/pharmacokinetics , Peptides/therapeutic use , Reproducibility of Results , Young Adult
8.
Catheter Cardiovasc Interv ; 95(7): 1277-1284, 2020 06 01.
Article in English | MEDLINE | ID: mdl-32105408

ABSTRACT

AIMS: Peripheral arteries are constantly exposed to deformation (elongation, twisting, shortening, compression) making bioresorbable scaffolds (BRS) a potentially attractive therapeutic alternative to metallic stents. We conducted a long-term pilot preclinical study of a novel sirolimus-eluting BRS in peripheral arteries. METHODS AND RESULTS: Fourteen BRS were deployed in iliofemoral arteries of seven healthy Yucatan miniswine and examined with imaging, pharmacokinetic, histopathologic, and polymer degradation techniques at 0, 30, 90, 180 days, 1, 2, and 3.3 years. Angiographic late luminal loss remained unchanged at 30 and 180 days but significantly decreased from 1 to 3.3 years. optical coherence tomography (OCT) showed late increase in lumen area (1 year: 14.70 ± 3.58 mm2 , 2 years 22.04 ± 3.81 mm2 , and 3.3 years 23.45 ± 7.07 mm2 ; p < .05) primarily due to scaffold area enlargement between 1 and 3.3 years, while there was no difference in the percent area stenosis at all time points. Histologic evidence of scaffold degradation was observed starting at 2 years, with minimal inflammatory reaction. At 3.3 years, BRS struts were rarely discernible by OCT, confirmed by a nearly complete polymer degradation by molecular weight analysis. CONCLUSIONS: In this pilot study, novel sirolimus-eluting BRS showed promising acute and chronic performance in the iliofemoral arteries of Yucatan miniswine.


Subject(s)
Absorbable Implants , Angioplasty, Balloon/instrumentation , Cardiovascular Agents/administration & dosage , Femoral Artery/drug effects , Iliac Artery/drug effects , Sirolimus/administration & dosage , Angioplasty, Balloon/adverse effects , Animals , Cardiovascular Agents/pharmacokinetics , Equipment Design , Femoral Artery/diagnostic imaging , Femoral Artery/pathology , Iliac Artery/diagnostic imaging , Iliac Artery/pathology , Materials Testing , Models, Animal , Pilot Projects , Sirolimus/pharmacokinetics , Swine , Swine, Miniature , Time Factors
9.
Catheter Cardiovasc Interv ; 96(2): E119-E128, 2020 08.
Article in English | MEDLINE | ID: mdl-31659805

ABSTRACT

OBJECTIVES: The aim of this study is to improve local-drug delivery efficiency and tissue absorption using the ultrasound (US)-responsible drug coating based on a newly developed US-controlled paclitaxel release balloon. BACKGROUND: Low availability of the drug coating remains a major concern of the current drug coated balloon (DCB). The goal of this study is to develop a method to use an US-responsible paclitaxel-loaded microcapsules (PM) as the main content of balloon drug coating to enhance bioavailability of DCB. METHODS: An US-controlled paclitaxel release balloon is designed and fabricated based on the US-responsible paclitaxel-loaded poly (lactic-co-glycolic acid) (PLGA) microcapsules. Rapid exchange percutaneous transluminal coronary angioplasty (PTCA) balloon catheters were coated with the PM. The deployment processes of the paclitaxel-loaded microcapsules coated balloons (PMCB) under US, PMCB without US and a homogenous matrix of paclitaxel and iopromide coated balloon (PICB) were then placed in healthy and stent implanted porcine coronary arteries. RESULTS: In vitro release assay demonstrated an ability of US (1 MHz, 1.22 W/cm2 , 1 minute) to affect the release kinetics of paclitaxel from PM by inducing a 76 ± 5.4% increase in the rate of release. The paclitaxel content in target vessels are 203 ± 37 µg/g for PMCB under US, 85 ± 23 µg/g for PMCB without US, and 107 ± 31 µg/g for PICB 1-hr post-surgery. The availability of the drug for the PMCB reaches 27% under US. CONCLUSIONS: The US-controlled paclitaxel release balloon significantly improved the drug content of the target vessels in the porcine model.


Subject(s)
Angioplasty, Balloon, Coronary/instrumentation , Cardiac Catheters , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible , Coronary Vessels/metabolism , Paclitaxel/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Ultrasonic Waves , Animals , Cardiovascular Agents/chemistry , Cardiovascular Agents/pharmacokinetics , Delayed-Action Preparations , Drug Carriers , Drug Liberation , Male , Paclitaxel/chemistry , Paclitaxel/pharmacokinetics , Solubility , Sus scrofa
10.
Circ Res ; 122(9): 1176-1190, 2018 04 27.
Article in English | MEDLINE | ID: mdl-29700066

ABSTRACT

This review will provide an overview of the principles of pharmacogenomics from basic discovery to implementation, encompassing application of tools of contemporary genome science to the field (including areas of apparent divergence from disease-based genomics), a summary of lessons learned from the extensively studied drugs clopidogrel and warfarin, the current status of implementing pharmacogenetic testing in practice, the role of genomics and related tools in the drug development process, and a summary of future opportunities and challenges.


Subject(s)
Cardiovascular Agents/pharmacokinetics , Genomics , Pharmacogenetics , Precision Medicine/methods , Biological Variation, Individual , Biotransformation/genetics , Cardiovascular Agents/adverse effects , Cardiovascular Agents/therapeutic use , Drug Development , Drug-Related Side Effects and Adverse Reactions/ethnology , Drug-Related Side Effects and Adverse Reactions/genetics , Drug-Related Side Effects and Adverse Reactions/prevention & control , Forecasting , Genetic Association Studies , Genetic Predisposition to Disease , Genetic Testing , Genetic Variation , Genotyping Techniques , Human Genome Project , Humans , Randomized Controlled Trials as Topic/methods , Risk Assessment , Sample Size
11.
Circ Res ; 123(8): 986-995, 2018 09 28.
Article in English | MEDLINE | ID: mdl-30355033

ABSTRACT

RATIONALE: Metformin has been demonstrated to decrease infarct size (IS) and prevent postinfarction left ventricular (LV) remodeling in rodents when given intravenously at the time of reperfusion. It remains unclear whether similar cardioprotection can be achieved in a large animal model. OBJECTIVE: The objective of this study was to determine whether intravascular infusion of metformin at the time of reperfusion reduces myocardial IS in a porcine model of acute myocardial infarction. METHODS AND RESULTS: In a blinded and randomized preclinical study, closed-chest swine (n=20) were subjected to a 60-minute left anterior descending coronary artery occlusion to produce myocardial infarction. Contrast-enhanced computed tomography was performed during left anterior descending coronary artery occlusion to assess the ischemic area-at-risk. Animals were randomized to receive either metformin or vehicle as an initial intravenous bolus (5 mg/kg) 8 minutes before reperfusion, followed by a 15-minute left coronary artery infusion (1 mg/kg per minute) commencing with the onset of reperfusion. Echocardiography and computed tomographic imaging of LV function were performed 1 week later, at which time the heart was removed for postmortem pathological analysis of area-at-risk and IS (triphenyltetrazolium chloride). Baseline variables including hemodynamics and LV function were similar between groups. Peak circulating metformin concentrations of 374±35 µmol/L were achieved 15 minutes after reperfusion. There was no difference between the area-at-risk as a percent of LV mass by computed tomography (vehicle: 20.7%±1.1% versus metformin: 19.7%±1.3%; P=0.59) or postmortem pathology (22.4%±1.2% versus 20.2%±1.2%; P=0.21). IS relative to area-at-risk averaged 44.5%±5.0% in vehicle-treated versus 38.2%±6.8% in metformin-treated animals ( P=0.46). There was no difference in global function 7 days after myocardial infarction as assessed by echocardiography or computed tomographic ejection fraction (56.2%±2.6% versus 56.3%±2.4%; P=0.98). CONCLUSIONS: In contrast to rodent hearts, postconditioning with high-dose metformin administered immediately before reperfusion does not reduce IS or improve LV function 7 days after myocardial infarction in swine. These results reinforce the importance of rigorously testing therapies in large animal models to facilitate clinical translation of novel cardioprotective therapies.


Subject(s)
Cardiovascular Agents/administration & dosage , Metformin/administration & dosage , Myocardial Infarction/drug therapy , Myocardium/pathology , Stroke Volume/drug effects , Ventricular Function, Left/drug effects , Ventricular Remodeling/drug effects , Animals , Cardiovascular Agents/pharmacokinetics , Disease Models, Animal , Drug Administration Schedule , Echocardiography , Infusions, Intra-Arterial , Injections, Intravenous , Metformin/pharmacokinetics , Multidetector Computed Tomography , Myocardial Infarction/diagnostic imaging , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Species Specificity , Sus scrofa , Time Factors
12.
Pharmacol Res ; 159: 105031, 2020 09.
Article in English | MEDLINE | ID: mdl-32562816

ABSTRACT

Thrombosis initiated by abnormal platelet aggregation is a pivotal pathological event that precedes most cases of cardiovascular diseases (CVD). Recently, growing evidence indicates that platelet could be a potential target for CVD prevention. However, as the conventional antithrombotic management strategy, applications of current antiplatelet agents are somewhat limited by their various side effects, such as bleeding risk and drug resistance. Hence, efforts have been made to search for agents as complementary therapies. Ginsenoside, the principal active component extracted from Panax ginseng, has gained much attention for its regulations on multiple crucial events of platelet aggregation. From structural characteristics to clinical applications, this review anatomized the intrinsic structure-function relationship of antiplatelet potency of ginsenosides, and the involved signal pathways were specifically summarized. Additionally, the emphasis was placed on clinical studies that investigate the antithrombotic efficacy of ginsenosides in the treatment of CVD. Further, a broad overview of approaches for improving the bioavailability of ginsenosides was concluded. Limitations and prospects of current studies were also discussed. This study may provide some new insights into the systematic understanding of ginsenosides in CVD treatment and lay a foundation for future research.


Subject(s)
Blood Platelets/drug effects , Cardiovascular Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Ginsenosides/therapeutic use , Muscle, Smooth, Vascular/drug effects , Neointima , Platelet Aggregation Inhibitors/therapeutic use , Platelet Aggregation/drug effects , Vascular Remodeling/drug effects , Animals , Biological Availability , Blood Platelets/metabolism , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Cardiovascular Diseases/blood , Cardiovascular Diseases/pathology , Ginsenosides/adverse effects , Ginsenosides/pharmacokinetics , Humans , Molecular Structure , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Platelet Aggregation Inhibitors/adverse effects , Platelet Aggregation Inhibitors/pharmacokinetics , Signal Transduction , Structure-Activity Relationship
13.
J Cardiovasc Pharmacol ; 76(2): 189-196, 2020 08.
Article in English | MEDLINE | ID: mdl-32769849

ABSTRACT

OBJECTIVE: The natural molecule α-lipoic acid has been shown to be partially cytoprotective through antioxidant and antiapoptotic mechanisms. To obtain an initial assessment of the safety and potential efficacy of a synthetic derivative, CMX-2043, in preventing ischemic complications of percutaneous coronary intervention (PCI) we conducted the Subjects Undergoing PCI and Perioperative Reperfusion Treatment (SUPPORT-1) trial, the first patient experience with this agent. METHODS AND RESULTS: SUPPORT-1 was a phase 2a, 6-center, international, placebo-controlled, randomized, double-blind trial. A total of 142 patients were randomized to receive a single intravenous bolus dose of drug or placebo administered 15-60 minutes before PCI. Cardiac biomarker assessments included serial measurements of creatine kinase myocardial band (CK-MB) at 6, 12, 18, and 24 hours after PCI and a single measurement of troponin T (TnT) at 24 hours. Peak concentrations of CK-MB and TnT were significantly reduced in the 2.4 mg/kg group compared with placebo (P = 0.05 and 0.03, respectively). No subject administered 2.4 mg/kg of CMX-2043 had an increase of CK-MB to ≥3X upper limit of normal versus 16% for placebo (P = 0.02); 16% of the 2.4-mg/kg dose group developed an elevation of TnT to ≥3X upper limit of normal versus 39% in the placebo group (P = 0.05). No drug-related serious adverse events were observed in any group. CONCLUSION: These data suggest that CMX-2043 may reduce PCI periprocedural myonecrosis and support further clinical evaluation of this novel agent for its potential cytoprotective effects.


Subject(s)
Angioplasty, Balloon, Coronary , Cardiovascular Agents/therapeutic use , Coronary Artery Disease/therapy , Dipeptides/therapeutic use , Myocytes, Cardiac/drug effects , Thioctic Acid/analogs & derivatives , Aged , Angioplasty, Balloon, Coronary/adverse effects , Biomarkers/blood , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Coronary Artery Disease/diagnostic imaging , Creatine Kinase, MB Form/blood , Dipeptides/adverse effects , Dipeptides/pharmacokinetics , Double-Blind Method , Female , Humans , India , Male , Middle Aged , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Prospective Studies , Thioctic Acid/adverse effects , Thioctic Acid/pharmacokinetics , Thioctic Acid/therapeutic use , Time Factors , Treatment Outcome , Troponin T/blood , United States
14.
Arch Pharm (Weinheim) ; 353(12): e2000146, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32886393

ABSTRACT

The natural isomers of resveratrol, cis- and trans-resveratrol, are natural phenolic substances synthetized via the shikimate pathway and found in many sources, including grapes, peanuts, blackberries, pistachios, cacao, cranberries, and jackfruits. They have functional and pharmacological properties such as anticarcinogenic, antidiabetic, anti-inflammatory, and cardioprotective activities. The aim of this article is to review the data published on resveratrol and its isomers, and their biosynthesis in plants, food sources, health and toxic effects, and the excretion of their metabolites. Due to its contribution to the promotion of human health, it is convenient to gather more knowledge about its functional properties, food sources, and the interactions with the human body during the processes of eating, digestion, absorption, biotransformation, and excretion, to combine this information to improve the understanding of these substances.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Cardiovascular Agents/pharmacology , Food , Hypoglycemic Agents/pharmacology , Plants/metabolism , Resveratrol/pharmacology , Animals , Anti-Inflammatory Agents/metabolism , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/toxicity , Antineoplastic Agents, Phytogenic/metabolism , Antineoplastic Agents, Phytogenic/pharmacokinetics , Antineoplastic Agents, Phytogenic/toxicity , Biological Availability , Biotransformation , Cardiovascular Agents/metabolism , Cardiovascular Agents/pharmacokinetics , Cardiovascular Agents/toxicity , Drug Elimination Routes , Gastrointestinal Absorption , Humans , Hypoglycemic Agents/metabolism , Hypoglycemic Agents/pharmacokinetics , Hypoglycemic Agents/toxicity , Isomerism , Resveratrol/metabolism , Resveratrol/pharmacokinetics , Resveratrol/toxicity
15.
Bull Exp Biol Med ; 168(4): 465-469, 2020 Feb.
Article in English | MEDLINE | ID: mdl-32146624

ABSTRACT

We studied pharmacokinetics and bioavailability of verapamil, propranolol, and ethacizine in healthy volunteers after single oral administration under normal conditions and on the second day of simulated antiorthostatic hypokinesia modeling some effects of microgravity. Under conditions of antiorthostatic hypokinesia, a tendency to a decrease in half-elimination period, mean retention time, and volume of distribution and an increase in the rate of absorption, ratio of maximum concentrations, and relative rate of absorption of verapamil and propranolol were revealed. For ethacizine, a statistically significant increase in the time of attaining maximum concentration and volume of distribution and a decrease in the maximum concentration, rate of absorption, ratio of maximum concentrations, and relative rate of absorption under conditions of antiorthostatic hypokinesia were found.


Subject(s)
Cardiovascular Agents/pharmacokinetics , Hypokinesia/blood , Phenothiazines/pharmacokinetics , Propranolol/pharmacokinetics , Verapamil/pharmacokinetics , Weightlessness Simulation/methods , Adult , Area Under Curve , Biological Availability , Cardiovascular Agents/blood , Half-Life , Humans , Hypokinesia/drug therapy , Male , Middle Aged , Phenothiazines/blood , Propranolol/blood , Verapamil/blood
16.
Pak J Pharm Sci ; 33(6): 2543-2546, 2020 Nov.
Article in English | MEDLINE | ID: mdl-33867328

ABSTRACT

In this work, the aim of our study was to assess whether sesamin could influence the pharmacokinetics of ivabradine and its active metabolite N-desmethylivabradine in rats. At the begining, 12 healthy male Sprague-Dawley rats were randomly divided into two groups: The rats were received an oral administration of 1.0mg/kg ivabradine alone (the control group), and the rats were given 1.0mg/kg ivabradine co-administered with 50mg/kg sesamin by gavage (the test group). After that, blood samples were collected from the tail vein of rats, and ultra-performance liquid chromatography tandem mass spectrometry (UPLC-MS/MS) were used for determing the plasma concentrations of ivabradine and N-desmethylivabradine in rats. Finally, the pharmacokinetic parameters were estimated using DAS 2.0 software. As the results, the pharmacokinetic parameters (t1/2, Cmax, AUC (0-t) and AUC (0-oo)) of ivabradine in the control group were significantly lower than those in the test group (P<0.05). Moreover, sesamin significantly decreased t1/2, Cmax, AUC(0-t) and AUC(0-oo) of N-desmethylivabradine when compared to the control. These results demonstrated that sesamin increases plasma concentration of ivabradine and decreases N-desmethylivabradine conversely. Hence, our data indicated sesamin could influence the pharmacokinetic profile of ivabradine in rats, which might cause food-drug interaction in humans.


Subject(s)
Dioxoles/pharmacology , Ivabradine/pharmacokinetics , Lignans/pharmacology , Administration, Oral , Animals , Area Under Curve , Cardiovascular Agents/blood , Cardiovascular Agents/metabolism , Cardiovascular Agents/pharmacokinetics , Chromatography, High Pressure Liquid , Dioxoles/administration & dosage , Dioxoles/pharmacokinetics , Ivabradine/blood , Ivabradine/metabolism , Lignans/administration & dosage , Lignans/pharmacokinetics , Male , Rats, Sprague-Dawley , Tandem Mass Spectrometry
17.
Br J Clin Pharmacol ; 85(1): 20-36, 2019 01.
Article in English | MEDLINE | ID: mdl-30194701

ABSTRACT

Prescribing in heart failure (HF), a common disease state that predominantly affects the older population, is often a challenging task because of the dynamic nature of the condition, requiring frequent monitoring and medication review, the presence of various comorbidities, and the frailty phenotype of many patients. The significant alterations in various organs and tissues occurring in HF, particularly the reduced cardiac output with peripheral hypoperfusion and the structural and functional changes of the gastrointestinal tract, liver and kidney, might affect the pharmacokinetics of several drugs. This review critically appraises the results of published studies investigating the pharmacokinetics of currently marketed cardiovascular and selected non-cardiovascular drugs in HF patients and control groups, identifies gaps in the current knowledge, and suggests avenues for future research in this complex patient population.


Subject(s)
Cardiovascular Agents/pharmacokinetics , Heart Failure/physiopathology , Area Under Curve , Biological Availability , Cardiac Output/physiology , Comorbidity , Half-Life , Heart/physiopathology , Heart Failure/drug therapy , Heart Failure/epidemiology , Humans , Kidney/physiopathology , Liver/physiopathology , Metabolic Clearance Rate/physiology
18.
J Cardiovasc Pharmacol ; 74(4): 297-305, 2019 10.
Article in English | MEDLINE | ID: mdl-31356538

ABSTRACT

The NLRP3 inflammasome is an intracellular, multimeric protein complex that initiates a potent inflammatory response to danger signals. After acute myocardial infarction, NLRP3 inflammasome-dependent inflammation promotes adverse left ventricular remodeling and recurrent atherosclerotic events. Selective and nonselective inhibitors of the NLRP3 inflammasome or its downstream effectors (interleukin-1ß and interleukin-18) may prevent adverse left ventricular remodeling and recurrent atherosclerotic events. In this review, we highlight strategies to inhibit NLRP3 inflammasome activity and their potential roles in the management of acute myocardial infarction.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Cardiovascular Agents/therapeutic use , Inflammasomes/antagonists & inhibitors , Inflammation Mediators/antagonists & inhibitors , Inflammation/drug therapy , Myocardial Infarction/drug therapy , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/pharmacokinetics , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Humans , Inflammasomes/immunology , Inflammasomes/metabolism , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Inflammation Mediators/immunology , Inflammation Mediators/metabolism , Molecular Targeted Therapy , Myocardial Infarction/immunology , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/immunology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction
19.
Ren Fail ; 41(1): 118-125, 2019 Nov.
Article in English | MEDLINE | ID: mdl-30909832

ABSTRACT

PURPOSE: Ranolazine is a novel anti-angina treatment approved in the United States for chronic stable angina. Ranolazine pharmacokinetics have not been studied previously in patients who receive maintenance hemodialysis. This study describes the pharmacokinetics of ranolazine and three major metabolites (CVT-2738, CVT-2512, CVT-2514) in patients receiving thrice weekly hemodialysis. METHODS: Eight participants receiving maintenance hemodialysis completed this prospective, open-label study (study identifier NCT01435174 at Clinicaltrials.gov). Three participants received a single tablet of ranolazine 500 mg (followed by an interim analysis), and five received 2 tablets of ranolazine 500 mg. Blood samples were collected over 65 h to determine the pharmacokinetic characteristics during and between hemodialysis sessions. Non-compartmental analysis was used to determine the individual pharmacokinetic parameters. RESULTS: Ranolazine off-hemodialysis elimination phase half-lives were 3.6 and 3.9 h for 500 mg and 1000 mg doses, respectively. The time to maximum concentration ranged from 2 to 18 hours and the average maximum concentration was 0.65 ± 0.27 mcg/mL and 1.18 ± 0.48 mcg/mL for ranolazine 500 mg and 1000 mg dose, respectively. The mean hemodialysis percent reduction ratio for the ranolazine 500 mg dose was 52.3 ± 8.1% and for the ranolazine 1000 mg dose was 69.2 ± 37.6%. CONCLUSIONS: Data on ranolazine dosing in patients receiving maintenance hemodialysis is almost non-existent. Given the extent of pharmacokinetic variability observed with the 500 mg and 1000 mg oral doses of ranolazine, neither can be recommended as a starting dose in patients receiving maintenance hemodialysis. Guided by the information gained form this study about the extent of hemodialytic drug clearance, further multi-dose clinical trials of ranolazine are needed to optimize therapeutic outcomes in this patient population.


Subject(s)
Cardiovascular Agents/pharmacokinetics , Kidney Failure, Chronic/therapy , Ranolazine/pharmacokinetics , Renal Dialysis , Administration, Oral , Adult , Angina, Stable/drug therapy , Area Under Curve , Biological Variation, Population , Cardiovascular Agents/administration & dosage , Female , Humans , Kidney Failure, Chronic/blood , Male , Middle Aged , Pilot Projects , Prospective Studies , Ranolazine/administration & dosage , Tablets , Young Adult
20.
Int J Psychiatry Clin Pract ; 23(2): 134-148, 2019 Jun.
Article in English | MEDLINE | ID: mdl-30707042

ABSTRACT

Objectives: The present paper aimed to assist physicians in the accurate choice among second-generation agents (SGAs) for patients with cardiovascular disease (CVD). Methods: We reviewed the published pharmacokinetic (PK) and pharmacodynamic (PD) clinical data that report potential -or absence of- drug interactions between second-generation agents (SGAs) and CVD drugs most commonly used in cardiology, including antiplatelet drugs and anticoagulants, statins, beta-blockers, angiotensin-converting enzyme inhibitors, calcium channel blockers, diuretics and the antiarrhythmic drugs amiodarone and digoxin. We also reviewed the cardiovascular safety profile that has been published for each class of SGAs and side effects reported by patients with CVD. Results: Most relevant PK/PD data about SGAs and CVD drugs are based on small studies or detailed case reports. In many cases, the drug interactions are at most assessed in healthy volunteers so that the clinical relevance of findings needs further investigation in patients with CVD. Case reports of serious, sometimes fatal reactions due to concomitant administration of certain drugs require careful consideration. The major cardiac side effects of SGAs include HR increase, postural hypotension and slight prolongation of the intraventricular conduction time and QT interval. On normal dosage of antidepressants, both advanced heart block and ventricular arrhythmias could occur in patients with severe heart disease, together with clinically important loss of myocardial contractile force. Conclusions: Data reported in the present review should help physicians about their decision-making processes that govern SGAs use in CVD patients.


Subject(s)
Antidepressive Agents, Second-Generation/therapeutic use , Cardiovascular Agents/therapeutic use , Depressive Disorder/drug therapy , Drug Interactions , Heart Diseases/drug therapy , Antidepressive Agents, Second-Generation/adverse effects , Antidepressive Agents, Second-Generation/pharmacokinetics , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Comorbidity , Depressive Disorder/epidemiology , Heart Diseases/epidemiology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL