Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 79
Filter
1.
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
2.
J Cardiovasc Pharmacol ; 78(6): 819-825, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34524259

ABSTRACT

ABSTRACT: The energy used by the heart is generated mainly by the metabolism of fatty acids and glucose. Trimetazidine (TMZ) inhibits fatty acid metabolism and is used for the treatment of heart diseases such as heart failure. 3-Bromopyruvate (3-BrPA) can suppress glucose metabolism, and it is considered a promising candidate agent for tumor therapy. Because TMZ and 3-BrPA can separately inhibit the 2 main cardiac energy sources, it is necessary to investigate the effects of 3-BrPA combined with TMZ on the heart. Forty male Wistar rats were randomly divided into 4 groups: a control group, a TMZ group, a 3-BrPA group, and a 3-BrPA + TMZ group. Weight was recorded every day, and echocardiography was performed 14 days later. Heart function, the levels of adenosine triphosphate, oxidative stress-related factors (ROS, glutathione, oxidized glutathione, malondialdehyde, superoxide dismutase and total antioxidant capacity), and apoptosis in heart tissues were assessed to evaluate the effects of 3-BrPA and TMZ on the heart. In our study, no obvious changes occurred in the 3-BrPA group or the TMZ group compared with the control group. The combination of 3-BrPA and TMZ worsened heart function, decreased adenosine triphosphate levels, and increased oxidative stress and myocardial apoptosis. In conclusion, 3-BrPA and TMZ are not recommended for concurrent use.


Subject(s)
Apoptosis/drug effects , Cardiovascular Agents/toxicity , Enzyme Inhibitors/toxicity , Heart Diseases/chemically induced , Myocytes, Cardiac/drug effects , Oxidative Stress/drug effects , Pyruvates/toxicity , Trimetazidine/toxicity , Adenosine Triphosphate/metabolism , Animals , Cardiotoxicity , Energy Metabolism/drug effects , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Male , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Rats, Wistar , Signal Transduction , Ventricular Function, Left/drug effects
3.
Catheter Cardiovasc Interv ; 95(2): 319-328, 2020 02.
Article in English | MEDLINE | ID: mdl-31696642

ABSTRACT

BACKGROUND: The diameter of balloons or stents is selected according to the estimated reference vessel diameter and do not adapt to the vessel anatomy. The aim of the present preclinical studies was to investigate a novel, vessel anatomy adjusting hypercompliant drug-coated balloon catheter (HCDCB). METHODS: Hypercompliant balloon membranes were coated in a constricted state with high drug density. Drug adherence was investigated in vitro, transfer to the porcine peripheral arteries and longitudinal distribution in vivo. In young domestic swine, neointimal proliferation was induced by vessel overstretch and continuous irritation by permanent stents. Uncoated hypercompliant balloons (HCB), and standard uncoated balloons and drug-coated balloons (DCB) served as controls. Efficacy was assessed by angiography, optical coherence tomography (OCT), and histomorphometry. RESULTS: HCDCB lost 18.0 ± 3.9% of dose during in vitro simulated delivery to the lesion. Drug transfer to the vessel wall was 13.9 ± 6.4% and drug concentration was 1,044 ± 529 ng/mg tissue. Four weeks after treatment, the histomorphometric neointimal area was smaller with HCDCB versus uncoated HCB (2.39 ± 0.55 mm2 vs. 3.26 ± 0.72 mm2 , p = .038) and area stenosis (OCT) was less (11.6 ± 6.9% vs. 24.7 ± 9.7%, p = .022). No premature death occurred and no in-life clinical symptoms or treatment-associated thrombi were observed. CONCLUSIONS: HCDCB were found to inhibit excessive neointimal proliferation. Balloon adaption to different vessel diameters and shapes may provide drug-delivery in irregular lumen and facilitate balloon selection.


Subject(s)
Angioplasty, Balloon/instrumentation , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible , Iliac Artery , Paclitaxel/administration & dosage , Vascular Access Devices , Angiography , Angioplasty, Balloon/adverse effects , Animals , Cardiovascular Agents/toxicity , Cell Proliferation , Equipment Design , Iliac Artery/diagnostic imaging , Iliac Artery/pathology , Neointima , Paclitaxel/toxicity , Sus scrofa , Time Factors , Tomography, Optical Coherence
4.
Drug Chem Toxicol ; 43(6): 553-559, 2020 Nov.
Article in English | MEDLINE | ID: mdl-30239237

ABSTRACT

Digoxin is a cardiac glycoside derived from the common foxglove digitalis purpurea and has been available for several centuries as a medicinal agent. Despite extensive patient experience over many years, there remains some controversy regarding the possibility that digoxin might have a deleterious effect on survival. This study was constructed to assess trends in digoxin toxicity research using well-established qualitative and quantitative bibliometric indicators. The current study is based on publications that have been indexed in Scopus. Articles referring to the subject of digoxin toxicity between 1849 and 2015 were assessed according to the document type, publication language, countries/territories, institutions, journal, impact factors, total number of citations, h-index, average number of citations per publication, and international collaborations. There were 2900 publications that included 2542 (87.7%) original research articles, while 5.3% were reviews and 4.6% letters. The country of origin was the USA in 849 publications, Germany in 241, the UK in 150, and France in 143. The USA and the UK had the highest number of international collaborations. The average number of citations per publications related to digoxin toxicity was 8.1, and the h-index was 59. The USA and Canada had the highest h-indices by country at 46 and 22, respectively. This study presents the first bibliometric analysis on digoxin toxicity publications. The USA was the most important contributors to digoxin toxicity literature with the greatest international collaboration, largest number of articles and highest h-index, followed by Germany and the UK. There has been a trend towards reduced publication numbers related to digoxin toxicity at global level, although it is still an important issue and we present the current research themes related to digoxin toxicity that were identified.


Subject(s)
Biomedical Research/trends , Cardiovascular Agents/toxicity , Digitoxin/toxicity , Internationality , Toxicology/trends , Animals , Bibliometrics , Humans , Risk Assessment , Toxicity Tests/trends
5.
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
6.
J Vasc Interv Radiol ; 29(7): 1041-1049.e3, 2018 07.
Article in English | MEDLINE | ID: mdl-29754850

ABSTRACT

PURPOSE: To compare the drug effect in treated vessels and downstream effects in distal skeletal muscle of drug-coated balloons (DCBs) and drug-eluting stents (DESs) in a healthy preclinical swine model. MATERIALS AND METHODS: Four groups of treated iliofemoral arteries (percutaneous transluminal angioplasty [PTA]+DES, DCB+DES, DCB+bare metal stent [BMS], and DCB alone) of 12 healthy swine were assessed, with euthanasia at 30 days. Biological drug effect was evaluated using smooth muscle cell (SMC) loss score according to both depth and circumference as well as a neointimal fibrin and medial proteoglycan scores which were compared between the 4 groups. Vascular and skeletal muscle changes in regions downstream from the treated site were also assessed histologically for evidence of emboli. RESULTS: DESs showed greater medial SMC loss in the treated arteries irrespective of preceding DCB or PTA treatment in terms of depth (DCB+DES vs PTA+DES vs DCB+BMS vs DCB alone; median, 4.0 mm vs 3.8 mm vs 3.0 mm vs 2.2 mm; P = .009) and circumference (4.0 mm vs 3.5 mm vs 2.0 mm vs 1.2 mm, respectively; P = .007). Sections of skeletal muscles downstream from the treated arteries showed arteriolar changes of fibrinoid necrosis consistent with paclitaxel effect exclusively in the DCB groups (DCB+BMS, 26.9% of sections; DCB+DES, 14.3%; DCB alone, 19.2%; PTA+DES, 0%; P = .02). CONCLUSIONS: In the treated arteries, irrespective of preceding DCB treatment or PTA, DES treatment showed maximum drug effects vs DCB alone or in combination with BMS placement, and there was no detrimental toxic effect in DCB-treated iliofemoral arteries before DES treatment compared with PTA before DES treatment. Downstream vascular changes were exclusively seen in groups treated with DCBs.


Subject(s)
Angioplasty, Balloon/instrumentation , Cardiovascular Agents/administration & dosage , Coated Materials, Biocompatible , Drug-Eluting Stents , Femoral Artery/drug effects , Muscle, Skeletal/blood supply , Paclitaxel/administration & dosage , Popliteal Artery/drug effects , Vascular Access Devices , Angioplasty, Balloon/adverse effects , Animals , Cardiovascular Agents/toxicity , Femoral Artery/metabolism , Femoral Artery/pathology , Fibrin/metabolism , Models, Animal , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Neointima , Paclitaxel/toxicity , Popliteal Artery/metabolism , Popliteal Artery/pathology , Proteoglycans/metabolism , Sus scrofa , Time Factors
7.
Biochim Biophys Acta ; 1863(7 Pt B): 1728-48, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26524115

ABSTRACT

Cardiomyocytes from human pluripotent stem cells (hPSCs-CMs) could revolutionise biomedicine. Global burden of heart failure will soon reach USD $90bn, while unexpected cardiotoxicity underlies 28% of drug withdrawals. Advances in hPSC isolation, Cas9/CRISPR genome engineering and hPSC-CM differentiation have improved patient care, progressed drugs to clinic and opened a new era in safety pharmacology. Nevertheless, predictive cardiotoxicity using hPSC-CMs contrasts from failure to almost total success. Since this likely relates to cell immaturity, efforts are underway to use biochemical and biophysical cues to improve many of the ~30 structural and functional properties of hPSC-CMs towards those seen in adult CMs. Other developments needed for widespread hPSC-CM utility include subtype specification, cost reduction of large scale differentiation and elimination of the phenotyping bottleneck. This review will consider these factors in the evolution of hPSC-CM technologies, as well as their integration into high content industrial platforms that assess structure, mitochondrial function, electrophysiology, calcium transients and contractility. This article is part of a Special Issue entitled: Cardiomyocyte Biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Biomedical Research/methods , Cardiovascular Agents/pharmacology , Cell Lineage , Drug Discovery/methods , Heart Diseases/drug therapy , High-Throughput Screening Assays , Induced Pluripotent Stem Cells/physiology , Myocytes, Cardiac/physiology , Toxicity Tests/methods , Cardiovascular Agents/toxicity , Cell Differentiation , Cell Proliferation , Cells, Cultured , Genotype , Heart Diseases/chemically induced , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Phenotype , Risk Assessment
8.
Biochim Biophys Acta ; 1863(7 Pt B): 1717-27, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26952934

ABSTRACT

Cardiac drug discovery is hampered by the reliance on non-human animal and cellular models with inadequate throughput and physiological fidelity to accurately identify new targets and test novel therapeutic strategies. Similarly, adverse drug effects on the heart are challenging to model, contributing to costly failure of drugs during development and even after market launch. Human induced pluripotent stem cell derived cardiac tissue represents a potentially powerful means to model aspects of heart physiology relevant to disease and adverse drug effects, providing both the human context and throughput needed to improve the efficiency of drug development. Here we review emerging technologies for high throughput measurements of cardiomyocyte physiology, and comment on the promises and challenges of using iPSC-derived cardiomyocytes to model disease and introduce the human context into early stages of drug discovery. This article is part of a Special Issue entitled: Cardiomyocyte biology: Integration of Developmental and Environmental Cues in the Heart edited by Marcus Schaub and Hughes Abriel.


Subject(s)
Cardiovascular Agents/pharmacology , Drug Discovery/methods , Heart Diseases/drug therapy , High-Throughput Screening Assays , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Animals , Cardiovascular Agents/toxicity , Cell Differentiation/drug effects , Cell Lineage , Cell Proliferation/drug effects , Cells, Cultured , Genetic Predisposition to Disease , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Humans , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/pathology , Myocardial Contraction/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Risk Assessment
9.
J Vasc Interv Radiol ; 27(11): 1676-1685.e2, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27641674

ABSTRACT

PURPOSE: Different carrier excipients unique to individual drug-coated balloons (DCBs) may influence embolic safety characteristics in peripheral vascular territories through embolization of released particulates. A comparator study of IN.PACT Admiral vs Lutonix 035 balloons in healthy swine was therefore performed to assess which balloon produces more downstream emboli. MATERIALS AND METHODS: Single or overlapping 80-mm IN.PACT and Lutonix 035 DCBs were assessed in the femoral arteries of 21 swine with 28- and 90-day follow-up, with standard balloon angioplasty as a control. Histologic analysis of arterial wall and downstream skeletal muscle and coronary band was performed. This analysis was supported by an analytic measurement of paclitaxel levels. RESULTS: IN.PACT DCBs demonstrated a more pronounced change in medial wall composition, characterized by a paclitaxel-induced loss of medial smooth muscle cells accompanied by increased proteoglycans. The percentage of sections with arterioles exhibiting paclitaxel-associated fibrinoid necrosis in downstream tissues was higher at 90 days with overlapping IN.PACT DBCs compared with Lutonix 035 DCBs (46.2% [interquartile range, 19.2-57.7] vs 0.0% [0.0-11.5]; P = .01), with similar trends noted for 28-day single and overlapping DCBs. Drug analysis in parallel tissues further confirmed higher paclitaxel concentrations in nontarget tissues for IN.PACT than Lutonix 035 balloons for single and overlapping configurations at both time points. Rare embolic crystalline material was observed in downstream tissues, but only for IN.PACT balloons. CONCLUSIONS: There was more fibrinoid necrosis in tissues treated with IN.PACT DCBs compared with Lutonix DCBs, suggesting increased emboli debris with higher paclitaxel levels.


Subject(s)
Angioplasty, Balloon/instrumentation , Cardiovascular Agents/toxicity , Coated Materials, Biocompatible , Coronary Vessels/drug effects , Embolism/etiology , Femoral Artery/drug effects , Muscle, Skeletal/blood supply , Paclitaxel/toxicity , Vascular Access Devices , Angioplasty, Balloon/adverse effects , Animals , Arterioles/drug effects , Arterioles/pathology , Cardiovascular Agents/administration & dosage , Coronary Vessels/pathology , Embolism/pathology , Equipment Design , Femoral Artery/pathology , Fibrosis , Models, Animal , Necrosis , Neointima , Paclitaxel/administration & dosage , Sus scrofa , Time Factors
10.
Arch Toxicol ; 90(8): 1803-16, 2016 Aug.
Article in English | MEDLINE | ID: mdl-26676948

ABSTRACT

Cardiac toxicity represents one of the leading causes of drug failure along different stages of drug development. Multiple very successful pharmaceuticals had to be pulled from the market or labeled with strict usage warnings due to adverse cardiac effects. In order to protect clinical trial participants and patients, the International Conference on Harmonization published guidelines to recommend that all new drugs to be tested preclinically for hERG (Kv11.1) channel sensitivity before submitting for regulatory reviews. However, extensive studies have demonstrated that measurement of hERG activity has limitations due to the multiple molecular targets of drug compound through which it may mitigate or abolish a potential arrhythmia, and therefore, a model measuring multiple ion channel effects is likely to be more predictive. Several phenotypic rapid-throughput methods have been developed to predict the potential cardiac toxic compounds in the early stages of drug development using embryonic stem cells- or human induced pluripotent stem cell-derived cardiomyocytes. These rapid-throughput methods include microelectrode array-based field potential assay, impedance-based or Ca(2+) dynamics-based cardiomyocytes contractility assays. This review aims to discuss advantages and limitations of these phenotypic assays for cardiac toxicity assessment.


Subject(s)
Cardiovascular Agents/toxicity , Drug Evaluation, Preclinical , Embryonic Stem Cells/drug effects , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Action Potentials/drug effects , Animals , Cardiotoxicity , Cells, Cultured , ERG1 Potassium Channel/antagonists & inhibitors , High-Throughput Screening Assays , Humans , Microelectrodes , Safety-Based Drug Withdrawals
11.
Cardiol Young ; 26(3): 493-8, 2016 Mar.
Article in English | MEDLINE | ID: mdl-25912244

ABSTRACT

BACKGROUND: Serum digoxin levels have limited utility for determining digoxin toxicity in adults. Paediatric data assessing the utility of monitoring serum digoxin concentration are scarce. We sought to determine whether serum digoxin concentrations are associated with signs and symptoms of digoxin toxicity in children. METHODS: We carried out a retrospective review of patients 2 ng/ml). RESULTS: There were 87 patients who met study criteria (male 46%, mean age 8.4 years). CHD was present in 67.8% and electrocardiograms were performed in 72.4% of the patients. The most common indication for digoxin toxicity was heart failure symptoms (61.5%). Toxic serum digoxin concentrations were present in 6.9% of patients (mean 2.6 ng/ml). Symptoms associated with digoxin toxicity occurred in 48.4%, with nausea/vomiting as the most common symptom (36.4%), followed by tachycardia (29.5%). Compared with those without toxic serum digoxin concentrations, significantly more patients with toxic serum digoxin concentrations were female (p=0.02). The presence of electrocardiogram abnormalities and/or signs and symptoms of digoxin toxicity was not significantly different between patients with and without serum digoxin concentrations (p>0.05). CONCLUSION: Serum digoxin concentrations in children are not strongly associated with signs and symptoms of digoxin toxicity.


Subject(s)
Arrhythmias, Cardiac/chemically induced , Cardiovascular Agents/blood , Cardiovascular Agents/toxicity , Digoxin/blood , Digoxin/toxicity , Heart Failure/chemically induced , Adolescent , Age Factors , Child , Child, Preschool , Electrocardiography , Female , Humans , Infant , Male , Nausea , Retrospective Studies , Texas , Vomiting
12.
Acta Pharmacol Sin ; 36(5): 627-43, 2015 May.
Article in English | MEDLINE | ID: mdl-25891082

ABSTRACT

AIM: Tanshinol is an important catechol in the antianginal herb Salvia miltiorrhiza roots (Danshen). This study aimed to characterize tanshinol methylation. METHODS: Metabolites of tanshinol were analyzed by liquid chromatography/mass spectrometry. Metabolism was assessed in vitro with rat and human enzymes. The major metabolites were synthesized for studying their interactions with drug metabolizing enzymes and transporters and their vasodilatory properties. Dose-related tanshinol methylation and its influences on tanshinol pharmacokinetics were also studied in rats. RESULTS: Methylation, preferentially in the 3-hydroxyl group, was the major metabolic pathway of tanshinol. In rats, tanshinol also underwent considerable 3-O-sulfation, which appeared to be poor in human liver. These metabolites were mainly eliminated via renal excretion, which involved tubular secretion mainly by organic anion transporter (OAT) 1. The methylated metabolites had no vasodilatory activity. Entacapone-impaired methylation did not considerably increase systemic exposure to tanshinol in rats. The saturation of tanshinol methylation in rat liver could be predicted from the Michaelis constant of tanshinol for catechol-O-methyltransferase (COMT). Tanshinol had low affinity for human COMT and OATs; its methylated metabolites also had low affinity for the transporters. Tanshinol and its major human metabolite (3-O-methyltanshinol) exhibited negligible inhibitory activities against human cytochrome P450 enzymes, organic anion transporting polypeptides 1B1/1B3, multidrug resistance protein 1, multidrug resistance-associated protein 2, and breast cancer resistance protein. CONCLUSION: Tanshinol is mainly metabolized via methylation. Tanshinol and its major human metabolite have low potential for pharmacokinetic interactions with synthetic antianginal agents. This study will help define the risk of hyperhomocysteinemia related to tanshinol methylation.


Subject(s)
Caffeic Acids/pharmacokinetics , Cardiovascular Agents/pharmacokinetics , Drugs, Chinese Herbal/pharmacokinetics , Liver/enzymology , Salvia miltiorrhiza/chemistry , Administration, Oral , Animals , Biotransformation , Caffeic Acids/administration & dosage , Caffeic Acids/isolation & purification , Caffeic Acids/toxicity , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/isolation & purification , Cardiovascular Agents/toxicity , Catechol O-Methyltransferase/metabolism , Chromatography, Liquid , Cytochrome P-450 Enzyme System/metabolism , Drugs, Chinese Herbal/administration & dosage , Drugs, Chinese Herbal/isolation & purification , Drugs, Chinese Herbal/toxicity , Herb-Drug Interactions , Humans , Injections, Intravenous , Kidney Tubules/metabolism , Male , Mass Spectrometry , Membrane Transport Proteins/metabolism , Methylation , Microsomes, Liver/enzymology , Organic Anion Transport Protein 1/metabolism , Phytotherapy , Plant Roots , Plants, Medicinal , Rats, Sprague-Dawley , Renal Elimination , Sulfates/metabolism
13.
Xenobiotica ; 45(1): 3-9, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25050791

ABSTRACT

1. Perhexiline, a chiral anti-anginal agent, may be useful to develop new cardiovascular therapies, despite its potential hepatotoxicity. 2. This study compared Dark Agouti (DA) and Sprague-Dawley (SD) rats, as models of perhexiline's metabolism and hepatotoxicity in humans. Rats (n = 4/group) received vehicle or 200 mg/kg/d of racemic perhexiline maleate for 8 weeks. Plasma and liver samples were collected to determine concentrations of perhexiline and its metabolites, hepatic function and histology. 3. Median (range) plasma and liver perhexiline concentrations in SD rats were 0.09 (0.04-0.13) mg/L and 5.42 (0.92-8.22) ng/mg, respectively. In comparison, DA rats showed higher (p < 0.05) plasma 0.50 (0.16-1.13) mg/L and liver 24.5 (9.40-54.7) ng/mg perhexiline concentrations, respectively, 2.5- and 3.7-fold higher cis-OH-perhexiline concentrations, respectively (p < 0.05), and lower plasma metabolic ratio (0.89 versus 1.55, p < 0.05). In both strains, the (+):(-) enantiomer ratio was 2:1. Perhexiline increased plasma LDH concentrations in DA rats (p < 0.05), but had no effect on plasma biochemistry in SD rats. Liver histology revealed lower glycogen content in perhexiline-treated SD rats (p < 0.05), but no effects on lipid content in either strain. 4. DA rats appeared more similar to humans with respect to plasma perhexiline concentrations, metabolic ratio, enantioselective disposition and biochemical changes suggestive of perhexiline-induced toxicity.


Subject(s)
Alcohol Oxidoreductases/metabolism , Cardiovascular Agents/metabolism , Liver/drug effects , Models, Animal , Perhexiline/metabolism , Rats, Sprague-Dawley , Animals , Cardiovascular Agents/toxicity , Cytochrome P450 Family 2 , Female , Liver/metabolism , Liver/ultrastructure , Perhexiline/toxicity
14.
Int J Mol Sci ; 16(12): 29345-56, 2015 Dec 09.
Article in English | MEDLINE | ID: mdl-26690140

ABSTRACT

Our previous study indicated that the combination of salvianolic acid B (SalB) and ginsenoside Rg1 (Rg1), the main components of Salvia miltiorrhizae and Panax notoginseng, improves myocardium structure and ventricular function in rats with ischemia/reperfusion injury. The present study aimed to determine the safety of the combined SalB and Rg1 (SalB-Rg1) in mice. The safety of SalB-Rg1 was evaluated through acute toxicity and repeated-dose toxicity. In the acute toxicity study, the up and down procedure was carried out firstly, and then, the Bliss method was applied. In the toxicity study for seven-day repeated treatment of SalB-Rg1, forty Kunming mice were randomly divided into four groups. The intravenous median lethal dose (LD50) of the SalB-Rg1 combination was 1747 mg/kg using the Bliss method. For both the acute toxicity study and the seven-day repeated toxicity study, SalB-Rg1 did not induce significant abnormality on brain, heart, kidney, liver and lung structure at any dose based on H&E stain. There were no significant changes related to the SalB-Rg1 toxicity detected on biochemical parameters for two kinds of toxicity studies. The LD50 in mice was 1747 mg/kg, which was more than one hundred times higher than the effective dose. Both studies of acute toxicity and seven-day repeated dose toxicity indicated the safety of the SalB-Rg1 combination.


Subject(s)
Benzofurans/toxicity , Cardiovascular Agents/toxicity , Ginsenosides/toxicity , Animals , Drug Combinations , Drug Evaluation, Preclinical , Female , Lethal Dose 50 , Male , Mice
15.
Biotechnol Lett ; 36(5): 1089-94, 2014 May.
Article in English | MEDLINE | ID: mdl-24563288

ABSTRACT

A mini-microscope-based system for multisite detection of cardiovascular toxicity was developed. The mini-microscope consisted of an image sensor and lens module extracted from an inexpensive webcam. The flipped lens module enabled cells to be magnified and monitored during testing. The portability and compactness of this system enables short-term and potential long-term experimentation inside a conventional incubator. The toxicity test results demonstrated that the normalized beating rates of cardiac muscle cells selected from multiple regions increased over time when treated with 100 nM isoprenaline. The presented system could be a promising cost-effective cell-based testing tool for discovering and screening drugs.


Subject(s)
Cardiovascular Agents/toxicity , Microscopy/methods , Myocytes, Cardiac/drug effects , Optical Imaging/methods , Animals , Cells, Cultured , Chick Embryo , Isoproterenol/toxicity , Microscopy/instrumentation , Myocytes, Cardiac/cytology , Myocytes, Cardiac/physiology , Optical Imaging/instrumentation
16.
Catheter Cardiovasc Interv ; 81(4): 698-708, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22581717

ABSTRACT

OBJECTIVE: The aim of this study was to assess the efficacy of stent-based delivery of succinobucol alone and in combination with rapamycin in a porcine coronary model. BACKGROUND: Current drugs and polymers used to coat coronary stents remain suboptimal in terms of long term efficacy and safety. Succinobucol is a novel derivative of probucol with improved antioxidant and anti-inflammatory properties. METHODS: Polymer-free Yukon stents were coated with 1% succinobucol (SucES), 2% rapamycin (RES), or 1% succinobucol plus 2% rapamycin solutions (SucRES) and compared with a bare metal stent (BMS). RESULTS: The in vivo release profile of SucES indicated drug release up to 28 days (60% drug released at 7 days); 41 stents (BMS, n = 11; SucES, n =10; RES, n = 10; SucRES, n = 10) were implanted in the coronary arteries of 17 pigs. After 28 days, mean neointimal thickness was 0.31 ± 0.14 mm for BMS, 0.51 ± 0.14 mm for SucES, 0.19 ± 0.11 mm for RES, and 0.36 ± 0.17 mm for SucRES (P < 0.05 for SucES vs. BMS). SucES increased inflammation and fibrin deposition compared with BMS (P < 0.05), whereas RES reduced inflammation compared with BMS (P < 0.05). CONCLUSION: In this model, stent-based delivery of 1% succinobucol using a polymer-free stent platform increased neointimal formation and inflammation following coronary stenting.


Subject(s)
Cardiovascular Agents/toxicity , Coronary Vessels/drug effects , Drug-Eluting Stents , Inflammation/chemically induced , Percutaneous Coronary Intervention/instrumentation , Probucol/analogs & derivatives , Animals , Cardiovascular Agents/administration & dosage , Cardiovascular Agents/pharmacokinetics , Cattle , Cell Survival/drug effects , Cells, Cultured , Coronary Vessels/metabolism , Coronary Vessels/pathology , Dose-Response Relationship, Drug , Drug Therapy, Combination , Endothelial Cells/drug effects , Endothelial Cells/pathology , Fibrin/metabolism , Inflammation/pathology , Male , Metals , Models, Animal , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/pathology , Neointima , Percutaneous Coronary Intervention/adverse effects , Probucol/administration & dosage , Probucol/pharmacokinetics , Probucol/toxicity , Prosthesis Design , Sirolimus/administration & dosage , Swine
17.
Am J Physiol Heart Circ Physiol ; 302(7): H1466-80, 2012 Apr 01.
Article in English | MEDLINE | ID: mdl-22198175

ABSTRACT

Cell lines expressing ion channels (IC) and the advent of plate-based electrophysiology device have enabled a molecular understanding of the action potential (AP) as a means of early QT assessment. We sought to develop an in silico AP (isAP) model that provides an assessment of the effect of a compound on the myocyte AP duration (APD) using concentration-effect curve data from a panel of five ICs (hNav1.5, hCav1.2, hKv4.3/hKChIP2.2, hKv7.1/hminK, hKv11.1). A test set of 53 compounds was selected to cover a range of selective and mixed IC modulators that were tested for their effects on optically measured APD. A threshold of >10% change in APD at 90% repolarization (APD(90)) was used to signify an effect at the top test concentration. To capture the variations observed in left ventricular midmyocardial myocyte APD data from 19 different dogs, the isAP model was calibrated to produce an ensemble of 19 model variants that could capture the shape and form of the APs and also quantitatively replicate dofetilide- and diltiazem-induced APD(90) changes. Provided with IC panel data only, the isAP model was then used, blinded, to predict APD(90) changes greater than 10%. At a simulated concentration of 30 µM and based on a criterion that six of the variants had to agree, isAP prediction was scored as showing greater than 80% predictivity of compound activity. Thus, early in drug discovery, the isAP model allows integrating separate IC data and is amenable to the throughput required for use as a virtual screen.


Subject(s)
Action Potentials/physiology , Cardiovascular Agents/pharmacology , Cardiovascular Agents/toxicity , Computer Simulation , Drug-Related Side Effects and Adverse Reactions , Heart/physiology , Animals , Calcium Channel Blockers/pharmacology , Calcium Channels, L-Type/drug effects , Calibration , Dogs , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical , ERG1 Potassium Channel , Electrodes, Implanted , Electrophysiological Phenomena , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Female , Fluorescence , Fluorescent Dyes , Myocytes, Cardiac/physiology , Potassium Channel Blockers/pharmacology , Risk Assessment , Threshold Limit Values
18.
Inhal Toxicol ; 24(12): 790-7, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23033993

ABSTRACT

CONTEXT: Exposure to single pollutants e.g. particulate matter (PM) is associated with adverse health effects, but it does not represent a real world scenario that usually involves multiple pollutants. OBJECTIVES: Determine if simultaneous exposure to PM and NO2 results in synergistic interactions. MATERIALS AND METHODS: Healthy young volunteers were exposed to clean air, nitrogen dioxide (NO2, 0.5 ppm), concentrated fine particles from Chapel Hill air (PM(2.5)CAPs, 89.5 ± 10.7 µg/m³), or NO2+PM(2.5)CAPs for 2 h. Each subject performed intermittent exercise during the exposure. Parameters of heart rate variability (HRV), changes in repolarization, peripheral blood endpoints and lung function were measured before and 1 and 18 h after exposure. Bronchoalveolar lavage (BAL) was performed 18 h after exposure. RESULTS: NO2 exposure alone increased cholesterol and HDL 18 h after exposure, decreased high frequency component of HRV one and 18 h after exposure, decreased QT variability index 1 h after exposure, and increased LDH in BAL fluid. The only significant change with PM(2.5)CAPs was an increase in HDL 1 h after exposure, likely due to the low concentrations of PM(2.5)CAPs in the exposure chamber. Exposure to both NO2 and PM(2.5)CAPs increased BAL α1-antitrypsin, mean t wave amplitude, the low frequency components of HRV and the LF/HF ratio. These changes were not observed following exposure to NO2 or PM(2.5)CAPs alone, suggesting possible interactions between the two pollutants. DISCUSSION AND CONCLUSIONS: NO2 exposure may produce and enhance acute cardiovascular effects of PM(2.5)CAPs. Assessment of health effects by ambient PM should consider its interactions with gaseous copollutants.


Subject(s)
Air Pollutants/toxicity , Cardiovascular Agents/toxicity , Cardiovascular System/drug effects , Nitrogen Dioxide/toxicity , Particulate Matter/toxicity , Adult , Arrhythmias, Cardiac/chemically induced , Atmosphere Exposure Chambers , Bronchoalveolar Lavage Fluid/chemistry , Cardiovascular System/metabolism , Cholesterol/blood , Drug Synergism , Female , Heart Rate/drug effects , Humans , Hypercholesterolemia/chemically induced , Lactate Dehydrogenases/metabolism , Male , North Carolina , Young Adult , alpha 1-Antitrypsin/metabolism
19.
Inhal Toxicol ; 24(12): 783-9, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23033992

ABSTRACT

PURPOSE: To determine the effect of pre-exercise exposure to diesel exhaust (DE) on 20-km cycling performance, pulmonary function, and cardio-respiratory variables during exercise. METHODS: Eight endurance-trained males participated in the study. Test days consisted of a 60-min exposure to either filtered air (FA) or DE, followed by a 20 km cycling time trial. Exposures to DE were at a concentration of 300 µg/m³ of PM(2.5). Forced expiratory volume in 1 s (FEV1) and forced vital capacity (FVC) were measured before and after exposure, and after exercise. Oxygen consumption (VO2) and carbon dioxide production (VCO2), minute ventilation (V(E)), tidal volume (V(T)), breathing frequency (F(B)), heart rate and oxyhemoglobin saturation (SpO2), were collected during the time trials. The effect of condition on time trial duration, an order effect, and mean cardio-respiratory variables were each analysed using paired T-tests. Repeated-measures ANOVA were used to assess the effect of DE exposure on pulmonary function. RESULTS: There was a main effect of condition (FA vs. DE) on the change in FEV1 from baseline, and in exercise heart rate. Post hoc tests revealed that exercise-induced bronchodilation was significantly attenuated following DE compared to FA. There were no main effects of condition on 20 km cycling performance, or VO2, VCO2, V(E), V(T), F(B) and SpO2 during a 20 km time trial. CONCLUSION: A 60-min exposure to DE prior to exercise significantly attenuated exercise-induced bronchodilation and significantly increased heart rate during exercise. Pre-exercise exposure to diesel exhaust did not significantly impair 20 km cycling time trial performance.


Subject(s)
Athletic Performance , Bronchoconstrictor Agents/toxicity , Cardiovascular Agents/toxicity , Cardiovascular System/drug effects , Exercise , Respiratory System/drug effects , Vehicle Emissions/toxicity , Adult , Bicycling , Carbon Dioxide/metabolism , Cardiovascular System/metabolism , Cross-Over Studies , Heart Rate/drug effects , Humans , Male , Oxygen Consumption/drug effects , Oxyhemoglobins/analysis , Particulate Matter/toxicity , Respiratory Physiological Phenomena/drug effects , Respiratory System/metabolism , Single-Blind Method , Young Adult
20.
Cardiovasc Res ; 118(1): 20-36, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33757124

ABSTRACT

Manifestations of cardiovascular diseases (CVDs) in a patient or a population differ based on inherent biological makeup, lifestyle, and exposure to environmental risk factors. These variables mean that therapeutic interventions may not provide the same benefit to every patient. In the context of CVDs, human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CMs) offer an opportunity to model CVDs in a patient-specific manner. From a pharmacological perspective, iPSC-CM models can serve as go/no-go tests to evaluate drug safety. To develop personalized therapies for early diagnosis and treatment, human-relevant disease models are essential. Hence, to implement and leverage the utility of iPSC-CMs for large-scale treatment or drug discovery, it is critical to (i) carefully evaluate the relevant limitations of iPSC-CM differentiations, (ii) establish quality standards for defining the state of cell maturity, and (iii) employ techniques that allow scalability and throughput with minimal batch-to-batch variability. In this review, we briefly describe progress made with iPSC-CMs in disease modelling and pharmacological testing, as well as current iPSC-CM maturation techniques. Finally, we discuss current platforms for large-scale manufacturing of iPSC-CMs that will enable high-throughput drug screening applications.


Subject(s)
Biomedical Research , Cardiology , Cardiovascular Agents/pharmacology , Cardiovascular Diseases/drug therapy , Cell Differentiation , Cell Proliferation , Drug Discovery , Induced Pluripotent Stem Cells/drug effects , Myocytes, Cardiac/drug effects , Cardiotoxicity , Cardiovascular Agents/toxicity , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/metabolism , Cardiovascular Diseases/pathology , Cell Culture Techniques, Three Dimensional , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Clinical Decision-Making , Humans , Induced Pluripotent Stem Cells/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Phenotype , Risk Assessment , Toxicity Tests
SELECTION OF CITATIONS
SEARCH DETAIL