Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 474
Filter
1.
Nutrients ; 16(8)2024 Apr 19.
Article in English | MEDLINE | ID: mdl-38674903

ABSTRACT

It is widely acknowledged that the ketogenic diet (KD) has positive physiological effects as well as therapeutic benefits, particularly in the treatment of chronic diseases. Maintaining nutritional ketosis is of utmost importance in the KD, as it provides numerous health advantages such as an enhanced lipid profile, heightened insulin sensitivity, decreased blood glucose levels, and the modulation of diverse neurotransmitters. Nevertheless, the integration of the KD with pharmacotherapeutic regimens necessitates careful consideration. Due to changes in their absorption, distribution, metabolism, or elimination, the KD can impact the pharmacokinetics of various medications, including anti-diabetic, anti-epileptic, and cardiovascular drugs. Furthermore, the KD, which is characterised by the intake of meals rich in fats, has the potential to impact the pharmacokinetics of specific medications with high lipophilicity, hence enhancing their absorption and bioavailability. However, the pharmacodynamic aspects of the KD, in conjunction with various pharmaceutical interventions, can provide either advantageous or detrimental synergistic outcomes. Therefore, it is important to consider the pharmacokinetic and pharmacodynamic interactions that may arise between the KD and various drugs. This assessment is essential not only for ensuring patients' compliance with treatment but also for optimising the overall therapeutic outcome, particularly by mitigating adverse reactions. This highlights the significance and necessity of tailoring pharmacological and dietetic therapies in order to enhance the effectiveness and safety of this comprehensive approach to managing chronic diseases.


Subject(s)
Diet, Ketogenic , Food-Drug Interactions , Ketosis , Humans , Biological Availability , Cardiovascular Agents/pharmacokinetics , Chronic Disease/drug therapy , Chronic Disease/therapy , Drug Interactions , Hypoglycemic Agents/pharmacokinetics , Ketosis/metabolism
2.
Expert Opin Biol Ther ; 23(8): 727-735, 2023.
Article in English | MEDLINE | ID: mdl-36880118

ABSTRACT

BACKGROUND: Dulaglutide, a glucagon-like peptide-1 (GLP-1) receptor agonist, has been approved for improving glycemic control and reducing the risk of cardiovascular (CV) adverse events. This study compared the pharmacokinetic (PK) profiles, safety, and immunogenicity of LY05008, a biosimilar candidate, to a licensed product dulaglutide in healthy Chinese male subjects. RESEARCH DESIGN AND METHODS: In this double-blind, open-label, parallel-group study, healthy Chinese male subjects were randomized 1:1 to receive either LY05008 or dulaglutide subcutaneously. Primary study endpoints were PK parameters such as the area under the concentration-time curve (AUC) from time zero to infinity (AUC0 - ∞), AUC from time zero to the last quantifiable concentration (AUC0-t), and maximum serum concentration (Cmax). Safety and immunogenicity profiles were also included for data analysis. RESULTS: 82 subjects were randomized to receive LY05008 (n = 41) or dulaglutide (n = 41). The 90% confidence intervals (CIs) of the geometric mean ratios (GMRs) of AUC0 - ∞, AUC0-t and Cmax of LY05008 to dulaglutide were all within the bioequivalence limits of 80%-125%. Other PK parameters, safety, and immunogenicity profiles were comparable across the two treatment groups. CONCLUSION: This study demonstrated PK similarity of LY05008, a dulaglutide biosimilar, to dulaglutide in healthy Chinese male subjects, with comparable safety and immunogenicity data. TRIAL REGISTRATION: The trial is registered at the Chinese Clinical Trial Registry (Identifier No. ChiCTR2200066519).


Subject(s)
Biosimilar Pharmaceuticals , Cardiovascular Agents , Glucagon-Like Peptide-1 Receptor Agonists , Hypoglycemic Agents , Humans , Male , Biosimilar Pharmaceuticals/adverse effects , Biosimilar Pharmaceuticals/metabolism , Biosimilar Pharmaceuticals/pharmacokinetics , Double-Blind Method , East Asian People , Immunoglobulin Fc Fragments/adverse effects , Immunoglobulin Fc Fragments/pharmacology , Therapeutic Equivalency , Healthy Volunteers , Glucagon-Like Peptide-1 Receptor Agonists/adverse effects , Glucagon-Like Peptide-1 Receptor Agonists/pharmacokinetics , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Hypoglycemic Agents/adverse effects , Hypoglycemic Agents/pharmacokinetics , Glucagon-Like Peptides/analogs & derivatives
3.
J Cardiovasc Pharmacol Ther ; 27: 10742484221119559, 2022.
Article in English | MEDLINE | ID: mdl-35972237

ABSTRACT

OBJECTIVE: The purpose of this study was to determine the impact of varying inflation parameters on paclitaxel delivery and retention using a commercially available DCB. BACKGROUND: Drug-coated balloons (DCB) have become the standard treatment for peripheral artery disease. Clinical data suggest that varying DCB delivery parameters directly impact patient outcome. Differences in delivery parameters can potentially alter the retention of the drug coating on DCBs. METHODS: Harvested porcine carotid arteries were utilized in an ex vivo pulsatile flow bioreactor system. The DCBs were then deployed at a DCB-to-artery ratio of 1:1 or 1.25:1, an inflation time of 30 seconds or 1 minute and transit time of 30 seconds or 3 minutes. The amount of drug retention in arterial tissue was evaluated by pharmacokinetic analysis at 1 hour and 1 day post DCB deployment. RESULTS: Arterial paclitaxel levels were found to be less at an inflation ratio of 1:1 with 3-minute transit time as compared to 30 seconds of transit time at 1 hour (12.3 ± 1.6 ng/mg vs. 391 ± 139 ng/mg, P = .036). At 1-day, DCBs deployed at a ratio of 1:1 resulted in less drug retention as compared to 1.25:1 (61.3 ± 23.1 ng/mg vs. 404 ± 195 ng/mg, P = .013). CONCLUSION: Arterial paclitaxel retention is reduced with extended transit times and sub-optimal expansion of the balloon. Optimization of delivery parameters can serve as an effective strategy to enhance clinical DCB outcomes.


Subject(s)
Angioplasty, Balloon , Cardiovascular Agents , Peripheral Arterial Disease , Animals , Cardiovascular Agents/pharmacokinetics , Coated Materials, Biocompatible , Femoral Artery , Paclitaxel , Peripheral Arterial Disease/therapy , Pharmaceutical Preparations , Swine , Treatment Outcome
4.
BMC Complement Med Ther ; 22(1): 35, 2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35120520

ABSTRACT

BACKGROUND: The chemokine CXCL12 and its two receptors (CXCR4 and CXCR7) are involved in inflammation and hematopoietic cell trafficking. This study was designed to investigate molecular docking interactions of four popular cardiovascular-active natural compounds; curcumin, resveratrol, quercetin, and eucalyptol; with these receptors and to predict their drug-like properties. We hypothesize that these compounds can modify CXCL12/CXCR4/CXCR7 pathway offering benefits for coronary artery disease patients. METHODS: Docking analyses were carried and characterized by Molecular Environment (MOE) software. Protein Data Bank ( http://www.rcsb.org/ ) has been retrieved from protein structure generation and crystal structures of CXCR4 and CXCR7 receptors (PDB code = 3ODU and 6K3F). The active sites of these receptors were evaluated and extracted from full protein and molecular docking protocol was done for compounds against them. The presented parameters included docking scores, ligand binding efficiency, and hydrogen bonding. The pharmacokinetic/toxic properties (ADME/T) were calculated using SwissADME, ProTox-II, and Pred-hERG softwares to predict drug-like properties of the compounds. The thermochemical and molecular orbital analysis, and molecular dynamics simulations were also done. RESULTS: All compounds showed efficient interactions with the CXCR4 and CXCR7 receptors. The docking scores toward proteins 3ODU of CXCR4 and 6K3F of CXCR7 were - 7.71 and - 7.17 for curcumin, - 5.97 and - 6.03 for quercetin, - 5.68 and - 5.49 for trans-resveratrol, and - 4.88 and - 4.70 for (1 s,4 s)-eucalyptol respectively indicating that all compounds, except quercetin, have more interactions with CXCR4 than with CXCR7. The structurally and functionally important residues in the interactive sites of docked CXCR4-complex and CXCR7-complex were identified. The ADME analysis showed that the compounds have drug-like properties. Only (1 s,4 s)-Eucalyptol has potential weak cardiotoxicity. The results of thermochemical and molecular orbital analysis and molecular dynamics simulation validated outcomes of molecular docking study. CONCLUSIONS: Curcumin showed the top binding interaction against active sites of CXCR4 and CXCR7 receptors, with the best safety profile, followed by quercetin, resveratrol, and eucalyptol. All compounds demonstrated drug-like properties. Eucalyptol has promising potential because it can be used by inhalation or skin massage. To our knowledge, this is the first attempt to find binding interactions of these natural agents with CXCR4 and CXCR7 receptors and to predict their druggability.


Subject(s)
Biological Products , Cardiovascular Agents , Molecular Dynamics Simulation , Receptors, CXCR4 , Receptors, CXCR , Signal Transduction , Biological Products/pharmacokinetics , Cardiovascular Agents/pharmacokinetics , Humans , Molecular Docking Simulation
5.
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
6.
Biomed Pharmacother ; 144: 112354, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34794233

ABSTRACT

Shengxian Decotion (SXT), a well-known Traditional Chinese Medicine (TCM) formula composed of Astragali Radix, Bupleuri Radix, Cimicifugae Rhizoma, Anemarrhenae Rhizoma and Platycodonis Radix, is clinically considered as an effective formula against cardiovascular diseases. However, the exact effective substance of SXT in treating chronic heart failure (CHF) still remains unclear. In the current study, we investigated the benefit of SXT in doxorubicin (DOX)-induced CHF rats and established a UHPLC-MS/MS method to simultaneously determine 18 key compounds in a subsequent comparative pharmacokinetic study in normal and CHF rats. Histopathological studies, transmission electron microscopy, and echocardiography were applied to assess the therapeutic effect of SXT on DOX-induced CHF rats, which indicated that SXT significantly ameliorated DOX-induced CHF, similar to enalapril. In addition, we successfully established a UHPLC-MS/MS method to determine the pharmacokinetics of the components in rat plasma, which was validated with good linearity, inter-day and intra-day precisions and accuracies, matrix effects, extraction recovery, and stability values. Our results showed that only astragaloside IV showed increased plasma exposure in the CHF rats, while saikosaponin A, quercetin, timosaponin B-II, ferulic acid, isoferulic acid and formononetin decreased compared to their pharmacokinetic characteristics in the normal and CHF rats. This study demonstrates that SXT enjoys obvious therapeutic effect on DOX-induced CHF rats, and the altered metabolism of some compounds in SXT is affected by the pathological state of CHF rats. Our findings provide a better understanding of the in vivo exposure to complex compounds of SXT, supporting effective substance screening and further investigation of the therapeutic mechanism.


Subject(s)
Cardiovascular Agents/pharmacokinetics , Cardiovascular Agents/therapeutic use , Drugs, Chinese Herbal/pharmacokinetics , Drugs, Chinese Herbal/therapeutic use , Heart Failure/drug therapy , Animals , Astragalus propinquus , Chromatography, High Pressure Liquid , Chronic Disease , Electrocardiography/drug effects , Heart Failure/chemically induced , Male , Mass Spectrometry , Medicine, Chinese Traditional , Rats , Rats, Sprague-Dawley , Saponins/blood , Triterpenes/blood
7.
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
8.
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
9.
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
10.
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
11.
Clín. investig. arterioscler. (Ed. impr.) ; 33(1): 33-40, ene.-feb. 2021. tab, graf
Article in Spanish | IBECS | ID: ibc-201980

ABSTRACT

Los inhibidores del cotransportador de sodio-glucosa tipo 2 (iSGLT2) fueron incialmente desarrollados para el tratamiento de la diabetes por su actividad hipoglucemiante. Sin embargo, a la luz de los estudios clínicos más recientes, están revolucionando el abordaje de la enfermedad cardiovascular (CV) en el paciente diabético. En el año 2015, el ensayo clínico EMPA-REG OUTCOME nos demuestra por primera vez que la empagliflozina -un fármaco considerado «antidiabético»- reduce la mortalidad CV y por cualquier causa, además de eventos CV mayores, hospitalización por IC y progresión de enfermedad renal. Posteriormente, otros estudios clínicos con agentes del mismo grupo farmacológico, CANVAS, con canagliflozina y DECLARE-TIMI-58 con dapagliflozina, corroboran la exitencia de los beneficios CV asociados a la inhibición del receptor SGLT2. Los beneficios observados los sitúan más allá de simples agentes hipoglucemiantes, con un demostrado efecto cardionefroprotector en la enfermedad aterosclerótica, insuficiencia cardiaca, mortalidad total, mortalidad cardiovascular y progresión de insuficiencia renal. Actualmente ya son una realidad en pacientes diabéticos de alto y muy alto riesgo cardiovascular, mientras su evidencia en el paciente no diabético es cada vez mayor. Asistimos, por tanto, a un cambio de paradigma y posiblemente al nacimiento de una nueva especialidad, la cardio-endocrinología, con la implicación de nuevos tratamientos que deben ser considerados más que sólo fármacos antidiabéticos


The sodium-glucose co-transporter 2 inhibitors (SGLT2i) were first conceived to treat type 2 diabetes due to their hypoglycaemic effect. However, due to an increasing number of studies, SGLT2i are changing the way we treat, and understand, diabetes, and cardiovascular risk, in general. The EMPA-REG OUTCOME clinical trial, in 2015, showed for the first time that empagliflozine - a glucose lowering agent - lowers the risk of death from cardiovascular causes and death from any cause. Also, this SGLT2i lowered hospital admission for heart failure and delayed renal function worsening. From then on, other clinical trials with SGLT2i such as CANVAS (canagliflozin) and DECLARE-TIMI-58 (dapagliflozin) confirmed these positive effects. With a proven and non-related glucose-lowering effect on heart failure, overall death, cardiovascular death, and renal function, SGLT2i stands out among the rest of anti-diabetic drugs. Since its role in treating patients with heart failure and type 2 diabetes has been undoubtedly established, new studies are paving the way for non-diabetic patients as well. A potential paradigm shift is being witnessed and, probably, the dawn of a new field, cardio-endocrinology, which involves new and far-reaching pharmacological agents


Subject(s)
Humans , Sodium-Glucose Transporter 2 Inhibitors/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Cardiovascular Agents/pharmacokinetics , Heart Failure/drug therapy , Diabetes Mellitus/drug therapy
12.
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
13.
Eur J Pharm Biopharm ; 158: 72-82, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33075477

ABSTRACT

Interventional therapies such as drug-eluting stents (DES) and drug-coated balloons (DCB) have significantly improved the clinical outcomes of patients with coronary occlusions in recent years. Despite this marked improvement, ischemic cardiovascular disease remains the most common cause of death worldwide. To address this, research efforts are focused on improving the safety and efficacy of the next generation of these devices. However, current experimental methods are unable to account for the influence of atherosclerotic lesions on drug uptake and retention. Therefore, in this study, we used an integrated approach utilizing both in vitro and in silico methods to assess the performance of DCB therapy. This approach was validated against existing in vivo results before being used to numerically estimate the effect of the atheroma. A bolus release of sirolimus was observed with our coating matrix. This, coupled with the rapid saturation of specific and non-specific binding sites observed in our study, indicated that increasing the therapeutic dose coated onto the balloons might not necessarily result in greater uptake and/or retention. Additionally, our findings alluded to an optimal exposure time, dependent on the coating matrix, for the DCBs to be expanded against the vessel. Moreover, our findings suggest that a biphasic drug release profile might be beneficial for establishing and maintaining the saturation of bindings sites within severely occluded vessels. Ultimately, we have demonstrated that computational methods may be capable of assessing the efficacy of DCB therapy as well as predict the influence of atherosclerotic lesions on said efficacy.


Subject(s)
Angioplasty, Balloon, Coronary/instrumentation , Atherosclerosis/complications , Cardiovascular Agents/pharmacokinetics , Coronary Occlusion/surgery , Coronary Restenosis/prevention & control , Drug-Eluting Stents , Atherosclerosis/pathology , Cardiovascular Agents/administration & dosage , Computer Simulation , Coronary Occlusion/complications , Coronary Occlusion/pathology , Coronary Restenosis/pathology , Coronary Vessels/metabolism , Coronary Vessels/pathology , Dose-Response Relationship, Drug , Drug Liberation , Humans , Models, Cardiovascular , Sirolimus/administration & dosage , Sirolimus/pharmacokinetics , Treatment Outcome
14.
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
15.
AAPS J ; 22(6): 129, 2020 10 08.
Article in English | MEDLINE | ID: mdl-33033962

ABSTRACT

Ivabradine and its metabolite both demonstrate heart rate-reducing effect (If current inhibitors) and undergo CYP3A4 metabolism. The purpose of this study was to develop a joint parent-metabolite physiologically based pharmacokinetic (PBPK)/pharmacodynamic (PD) model to predict the PK and PD of ivabradine and its metabolite following intravenous (i.v.) or oral administration (alone or co-administered with CYP3A4 inhibitors). Firstly, a parent-metabolite disposition model was developed and optimised using individual plasma concentration-time data following i.v. administration of ivabradine or metabolite within a Bayesian framework. Secondly, the model was extended and combined with a mechanistic intestinal model to account for oral absorption and drug-drug interactions (DDIs) with CYP3A4 inhibitors (ketoconazole, grapefruit juice). Lastly, a PD model was linked to the PBPK model to relate parent and metabolite PK to heart rate (HR) reduction. The disposition model described successfully parent-metabolite PK following i.v. administration. Following integration of a gut model, the PBPK model adequately predicted plasma concentration profiles and the DDI risk (92% and 85% of predicted AUC+inhibitor/AUCcontrol and Cmax+inhibitor/Cmaxcontrol for ivabradine and metabolite within the prediction limits). Ivabradine-metabolite PBPK model was linked to PD by using the simulated unbound parent-metabolite concentrations in the heart. This approach successfully predicted the effects of both entities on HR (observed vs predicted - 7.7/- 5.9 bpm and - 15.8/- 14.0 bpm, control and ketoconazole group, respectively). This study provides a framework for PBPK/PD modelling of a parent-metabolite and can be scaled to other populations or used for investigation of untested scenarios (e.g. evaluation of DDI risk in special populations).


Subject(s)
Cardiovascular Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Ivabradine/pharmacokinetics , Models, Biological , Administration, Intravenous , Administration, Oral , Adult , Area Under Curve , Bayes Theorem , Cardiovascular Agents/administration & dosage , Computer Simulation , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Drug Interactions , Enterocytes , Female , Fruit and Vegetable Juices/adverse effects , Healthy Volunteers , Heart Rate/drug effects , Humans , Intestinal Absorption/physiology , Ivabradine/administration & dosage , Ketoconazole/administration & dosage , Ketoconazole/pharmacokinetics , Male , Tissue Distribution
16.
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
17.
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
18.
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
19.
JACC Cardiovasc Interv ; 13(12): 1391-1402, 2020 06 22.
Article in English | MEDLINE | ID: mdl-32473887

ABSTRACT

Although drug-eluting stents are still the default interventional treatment of coronary artery disease, drug-coated balloons (DCBs) represent a novel alternative therapeutic strategy in certain anatomic conditions. The effect of DCBs is based on the fast and homogenous transfer of antiproliferative drugs into the vessel wall during single balloon inflation by means of a lipophilic matrix without the use of permanent implants. Although their use is established for in-stent restenosis of both bare-metal and drug-eluting stents, recent randomized clinical data demonstrate a good efficacy and safety profile in de novo small-vessel disease and high bleeding risk. In addition, there are other emerging indications (e.g., bifurcation lesions, large-vessel disease, diabetes mellitus, acute coronary syndromes). Because the interaction among the different delivery balloon designs, doses, formulations, and release kinetics of the drugs used is important, there seems to be no "class effect" of DCBs. On the basis of the amount of recently published data, the International DCB Consensus Group provides this update of previous recommendations summarizing the historical background, technical considerations such as choice of device and implantation technique, possible indications, and future perspectives.


Subject(s)
Angioplasty, Balloon, Coronary/instrumentation , Cardiac Catheters , Cardiovascular Agents/administration & dosage , Coronary Artery Disease/therapy , Angioplasty, Balloon, Coronary/adverse effects , Angioplasty, Balloon, Coronary/mortality , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Clinical Decision-Making , Consensus , Coronary Angiography , Coronary Artery Disease/diagnostic imaging , Coronary Artery Disease/mortality , Drug-Eluting Stents , Equipment Design , Humans , Risk Factors , Treatment Outcome
20.
Biomed Pharmacother ; 128: 110308, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32480216

ABSTRACT

With the advancement of science and technology, the living standards of human beings have continuously improved, but the incidence and mortality from atherosclerosis worldwide have also increased by year. Although interventional surgery and the continuous development of new drugs have significant therapeutic effects, their side effects cannot be ignored. Polydatin, an active ingredient isolated from the natural medicine Polygonum cuspidatum, has been shown to have a prominent role in the treatment of cardiovascular diseases. Polydatin treats atherosclerosis mainly from three aspects: anti-inflammatory, regulating lipid metabolism and anti-oxidative stress. This article will review the pharmacological mechanism of polydatin in anti-atherosclerosis, the biological characteristics of Polygonum cuspidatum, the toxicology and pharmacokinetics of polydatin and will provide ideas for further research.


Subject(s)
Arteries/drug effects , Atherosclerosis/drug therapy , Cardiovascular Agents/therapeutic use , Glucosides/therapeutic use , Inflammation Mediators/antagonists & inhibitors , Lipid Metabolism/drug effects , Oxidative Stress/drug effects , Stilbenes/therapeutic use , Animals , Anti-Inflammatory Agents/therapeutic use , Anticholesteremic Agents/therapeutic use , Antioxidants/therapeutic use , Arteries/metabolism , Arteries/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Cardiovascular Agents/adverse effects , Cardiovascular Agents/pharmacokinetics , Glucosides/adverse effects , Glucosides/pharmacokinetics , Humans , Inflammation Mediators/metabolism , Plaque, Atherosclerotic , Signal Transduction , Stilbenes/adverse effects , Stilbenes/pharmacokinetics
SELECTION OF CITATIONS
SEARCH DETAIL
...