Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 100
Filter
1.
Clin Transl Sci ; 17(5): e13828, 2024 May.
Article in English | MEDLINE | ID: mdl-38783568

ABSTRACT

As a treatment for relapsed or refractory multiple myeloma (MM), carfilzomib has been associated with a significant risk of cardiovascular adverse events (CVAE). The goals of our study were to evaluate the metabolomic profile of MM patients to identify those at high risk prior to carfilzomib treatment and to explore the mechanisms of carfilzomib-CVAE to inform potential strategies to protect patients from this cardiotoxicity. Global metabolomic profiling was performed on the baseline and post-baseline plasma samples of 60 MM patients treated with carfilzomib-based therapy, including 31 who experienced CVAE, in a prospective cohort study. Baseline metabolites and post-baseline/baseline metabolite ratios that differ between the CVAE and no-CVAE patients were identified using unadjusted and adjusted methods. A baseline metabolomic risk score was created to stratify patients. We observed a lower abundance of tauroursodeoxycholic acid (T-UDCA) in CVAE patients at baseline (odds ratio [OR] = 0.47, 95% confidence interval [CI] = 0.21-0.94, p = 0.044) compared with the no-CVAE patients. A metabolite risk score was able to stratify patients into three risk groups. The area under the receiver-operating curve of the model with clinical predictors and metabolite risk score was 0.93. Glycochenodeoxycholic acid (OR = 0.56, 95% CI = 0.31-0.87, p = 0.023) was significantly lower in post-baseline/baseline ratios of CVAE patients compared with no-CVAE patients. Following metabolomic analysis, we created a baseline metabolite risk score that can stratify MM patients into different risk groups. The result also provided intriguing clues about the mechanism of carfilzomib-CVAE and potential cardioprotective strategies.


Subject(s)
Cardiotoxicity , Metabolomics , Multiple Myeloma , Oligopeptides , Humans , Multiple Myeloma/drug therapy , Multiple Myeloma/blood , Oligopeptides/adverse effects , Male , Female , Aged , Middle Aged , Cardiotoxicity/etiology , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Metabolomics/methods , Prospective Studies , Metabolome/drug effects , Aged, 80 and over , Risk Factors
2.
J Transl Med ; 22(1): 398, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38685030

ABSTRACT

BACKGROUND: We aimed to compare the changes in blood metabolomes and cardiac parameters following doxorubicin treatment in HER2-positive and HER2-negative breast cancer patients. Additionally, the potential roles of changes in blood metabolomes as severity and prognostic markers of doxorubicin-induced cardiotoxicity were determined. METHODS: HER2-positive (n = 37) and HER2-negative (n = 37) breast cancer patients were enrolled. Cardiac function assessment and blood collection were performed at baseline and 2 weeks after completion of doxorubicin treatment in all patients, as well as at three months after completion of doxorubicin treatment in HER2-negative breast cancer patients. Blood obtained at all three-time points was processed for measuring cardiac injury biomarkers. Blood obtained at baseline and 2 weeks after completion of doxorubicin treatment were also processed for measuring systemic oxidative stress and 85 metabolome levels. RESULTS: Cardiac injury and systolic dysfunction 2 weeks after completion of doxorubicin treatment were comparable between these two groups of patients. However, only HER2-negative breast cancer patients exhibited increased systemic oxidative stress and cardiac autonomic dysfunction at this time point. Moreover, 33 and 29 blood metabolomes were altered at 2 weeks after completion of doxorubicin treatment in HER2-positive and HER2-negative breast cancer patients, respectively. The changes in most of these metabolomes were correlated with the changes in cardiac parameters, both at 2 weeks and 3 months after completion of doxorubicin treatment. CONCLUSIONS: The changes in blood metabolomes following doxorubicin treatment were dependent on HER2 status, and these changes might serve as severity and prognostic markers of doxorubicin-induced cardiotoxicity. TRIAL REGISTRATION: The study was conducted under ethical approval from the Institutional Review Board of the Faculty of Medicine, Chiang Mai University (Registration number: MED-2563-07001; Date: April 28, 2020). The study also complied with the Declaration of Helsinki.


Subject(s)
Breast Neoplasms , Cardiotoxicity , Doxorubicin , Metabolome , Receptor, ErbB-2 , Humans , Breast Neoplasms/drug therapy , Breast Neoplasms/blood , Female , Doxorubicin/adverse effects , Receptor, ErbB-2/metabolism , Receptor, ErbB-2/blood , Middle Aged , Prognosis , Cardiotoxicity/blood , Oxidative Stress/drug effects , Biomarkers/blood , Biomarkers/metabolism , Adult
3.
Balkan Med J ; 40(2): 82-92, 2023 03 08.
Article in English | MEDLINE | ID: mdl-36883738

ABSTRACT

In cardiooncology practice, "early cardiotoxicity" refers to an emerging subclinical myocardial dysfunction/injury in response to certain chemotherapeutic regimens. This condition can progress to overt cardiotoxicity in time and hence warrants proper and timely diagnostic and preventive strategies. Current diagnostic strategies for "early cardiotoxicity" are largely based on conventional biomarkers and certain echocardiographic indices. However, a significant gap still exists in this setting, warranting further strategies to improve diagnosis and overall prognosis in cancer survivors. Copeptin (surrogate marker of the arginine vasopressine axis) might serve as a promising adjunctive guide for the timely detection, risk stratification, and management of early cardiotoxicity on top of conventional strategies largely due to its multifaceted pathophysiological implications in the clinical setting. This work aims to focus on serum copeptin as a marker of "early cardiotoxicity" and its general clinical implications in patients with cancer.


Subject(s)
Antineoplastic Agents , Cardiotoxicity , Neoplasms , Humans , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Arginine , Biomarkers/blood , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Cardiotoxicity/etiology , Echocardiography , Glycopeptides/blood , Heart Injuries/blood , Heart Injuries/chemically induced , Heart Injuries/diagnosis , Neoplasms/blood , Neoplasms/drug therapy
4.
Biomolecules ; 12(12)2022 11 30.
Article in English | MEDLINE | ID: mdl-36551214

ABSTRACT

Galectin-3 and myeloperoxidase (MPO) are novel biomarkers in the field of cardio-oncology, but conflicting results have been reported. Hence, a meta-analysis was performed to assess the monitoring value of galectin-3 and MPO in cancer-therapy-related cardiotoxicity. PubMed, Cochrane, Web of Science, Embase, CNKI databases and ClinicalTrials.gov were queried. According to the predefined inclusion and exclusion criteria, eight studies with 1979 patients were included in this meta-analysis. The examination of the study's heterogeneity (I2), quality assessment and statistical analysis were performed by two reviewers. No significant differences in galectin-3 levels were noted before and after treatment (WMD = -0.10, 90% CI -6.06-5.85, I2: 99%), and a weaker relationship was observed between galectin-3 evaluations and cancer-therapy-related cardiotoxicity (HR = 1.39, 90% CI 0.97-1.98, I2: 0%). However, MPO levels were increased in patients post-treatment (SMD = 0.58, 90% CI 0.35-0.80, I2: 56%), and an increased risk of cardiotoxicity was associated with early pre-post MPO assessments (HR = 1.16, 90% CI 1.02-1.32, I2: 21%). Surprisingly, the MPO levels were a more effective indicator of the response to tumor treatment compared with the TnI (SMD = 2.46, 90% CI -0.26-5.19, I2: 96%) and NT-proBNP levels (SMD = 1.08, 90% CI -0.82-2.98, I2: 96%). In conclusion, our meta-analysis suggests that MPO may rep-resent a potential biomarker for the early detection of cardiotoxicity in current cardio-oncology practice, but the monitoring value of galectin-3 requires further study.


Subject(s)
Antineoplastic Agents , Cardiotoxicity , Galectin 3 , Neoplasms , Peroxidase , Humans , Biomarkers/blood , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Cardiotoxicity/etiology , Galectin 3/blood , Neoplasms/drug therapy , Peroxidase/blood
5.
Eur J Cancer ; 157: 383-390, 2021 11.
Article in English | MEDLINE | ID: mdl-34571335

ABSTRACT

AIM: Despite unprecedented results of anti-programmed death protein (ligand) 1 (PD-(L)1) immune checkpoint inhibitor in the oncology's armamentarium, immune-related adverse events (irAEs) represent a therapeutic hurdle. Currently, there is no consensual recommendation on a routinely monitored biomarker to early detect irAE. Biological markers such as serum creatine phosphokinase (CPK) are commonly used to measure muscular tissue injury. The potential of routine serum CPK monitoring to predict cardiac or neuromuscular irAE in patients treated with immunotherapy remains unknown. METHODS: In this retrospective study between January 2016 and December 2018 at Gustave Roussy Cancer Campus, 1151 cancer patients treated with anti-PD-(L)1 immunotherapy were systematically monitored with serum CPK measurements before each immunotherapy cycle. We considered significant CPK increases according to Common Terminology Criteria for Adverse Events v5.0 (CTCAEV5) of grade ≥2 severity. Comparisons were performed in patients with immune-related CPK (ir-CPK) elevations symptomatic versus asymptomatic. RESULTS: Overall, 53 of 1151 (4.6%) patients showed a CPK increase. Elevations of CPK were deemed to be immunotherapy-related in 31 of 1151 (2.7%) patients. Among them, 12 of 31 (38.7%) patients experienced symptomatic cardiac or neuromuscular irAE, whereas the other 19 of 31 (61.3%) patients remained asymptomatic. In patients with symptomatic irAE, the mean ir-CPK level was higher compared with asymptomatic patients (1271 versus 771 UI/L, P value = 0.02). In the asymptomatic group, all patients experienced a spontaneous resolution of the ir-CPK increase, and none required medical intervention. CONCLUSION: Most patients with immune-related CPK increase remained asymptomatic. The CPK serum increase did not alter the clinical management of asymptomatic patients. The results of this study did not support a significant clinical interest for a systematic routine CPK monitoring in patients amenable to anti-PD-(L)1 immunotherapy.


Subject(s)
Cardiotoxicity/diagnosis , Creatine Kinase/blood , Immune Checkpoint Inhibitors/adverse effects , Neoplasms/drug therapy , Neuromuscular Diseases/diagnosis , B7-H1 Antigen/antagonists & inhibitors , Biomarkers/blood , Cardiotoxicity/blood , Cardiotoxicity/immunology , Feasibility Studies , Female , Humans , Male , Memory, Episodic , Middle Aged , Neoplasms/blood , Neuromuscular Diseases/blood , Neuromuscular Diseases/chemically induced , Neuromuscular Diseases/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Retrospective Studies
6.
Curr Treat Options Oncol ; 22(6): 46, 2021 04 17.
Article in English | MEDLINE | ID: mdl-33866434

ABSTRACT

OPINION STATEMENT: Improvements in cancer survival have led to the emergence of cardiovascular disease as an important determinant of adverse outcome in survivors. Cancer therapeutics-related cardiac dysfunction is the most well-known form of cardiotoxicity. However, newer cancer therapies bring a broader range of cardiotoxicities. The optimal method to identify patients at risk of these complications is unclear, but circulating biomarkers comprise one possible approach. Troponins and natriuretic peptides have garnered the broadest evidence base for cardiotoxicity risk prediction, but other markers are being investigated. In this review, we explore evidence for circulating biomarkers in cardiotoxicity prediction associated with cancer therapies.


Subject(s)
Antineoplastic Agents/adverse effects , Biomarkers/blood , Cardiotoxicity/etiology , Cardiotoxicity/blood , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy, Adoptive/adverse effects , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Receptors, Chimeric Antigen/immunology , Troponin/blood , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Sci Rep ; 11(1): 7954, 2021 04 12.
Article in English | MEDLINE | ID: mdl-33846495

ABSTRACT

Cardiotoxic manifestation associated with breast cancer treatment by anthracycline regimen increases patients' susceptibility to myocardial injury, reduction in left ventricular ejection fraction and complications associated with heart failure. There is currently no standardized, minimally invasive, cost effective and clinically verified procedure to monitor cardiotoxicity post-anthracycline therapy initiation, and to detect early onset of irreversible cardiovascular complications. This study aims to create a panel of novel biomarkers and circulating miRNAs associated with cardiotoxicity, further assessing their correlation with cardiac injury specific markers, troponin I and T, and demonstrate the development of cardiac dysfunction in breast cancer patients. Blood obtained from West Virginian females clinically diagnosed with breast cancer and receiving anthracyclines showed upregulated level of biomarkers and circulating miRNAs after 3 and 6 months of chemotherapy initiation with increased levels of cardiac troponin I and T. These biomarkers and miRNAs significantly correlated with elevated troponins. Following 6 months of anthracycline-regimens, 23% of the patient population showed cardiotoxicity with reduced left ventricular ejection fraction. Our results support the clinical application of plasma biomarkers and circulating miRNAs to develop a panel for early diagnosis of chemotherapy related cardiac dysfunction which will enable early detection of disease progression and management of irreversible cardiac damage.


Subject(s)
Anthracyclines/adverse effects , Anthracyclines/therapeutic use , Biomarkers/metabolism , Breast Neoplasms/drug therapy , Cardiotoxicity/diagnosis , Biomarkers/blood , Breast Neoplasms/blood , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/genetics , Cardiotoxicity/blood , Cardiotoxicity/diagnostic imaging , Cardiotoxicity/genetics , Electrocardiography , Female , Gene Expression Regulation , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Troponin I/genetics , Troponin I/metabolism , Troponin T/genetics , Troponin T/metabolism , West Virginia
8.
Curr Probl Cardiol ; 46(6): 100818, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33756178

ABSTRACT

In recent years, the cancer survival of patients has improved thanks to advances in the pharmacological field. In many guidelines, cardiotoxicity induced by anticancer drugs was defined as a reduction from baseline in the left ventricular ejection fraction (LVEF) assessed by echocardiography. It is known that LVEF is not a sensible parameter in the detection of cardiotoxicity. Therefore, a decrease from baseline in the global longitudinal strain (GLS) or troponins elevation is used to detect subclinical cardiotoxicity. LVEF and GLS as well as the increase in some biomarkers are influenced by loading conditions that are frequent during chemotherapy. Other parameters not influenced by loading conditions should be used in the early diagnosis of cardiotoxicity. The aim of this review is to delineate the role of current strategies used in the early diagnosis of cardiotoxicity and to identify new strategies that could have greater application in the future in cardioncology.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxicity , Echocardiography/methods , Ventricular Dysfunction, Left , Antineoplastic Agents/pharmacology , Biomarkers/blood , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Cardiotoxicity/diagnostic imaging , Cardiotoxicity/etiology , Early Diagnosis , Humans , Magnetic Resonance Imaging , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Stroke Volume/drug effects , Troponin/blood , Ventricular Dysfunction, Left/blood , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/diagnostic imaging , Ventricular Function, Left/drug effects
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.
Eur Rev Med Pharmacol Sci ; 25(24): 7806-7822, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34982442

ABSTRACT

OBJECTIVE: Mitoxantrone (MTX)- induced cardiotoxicity is a clinical concern that is limiting its use. The aim of this paper, therefore, was to investigate the subchronic administration of MTX plus nonspecific/specific inhibitors of CYP450/2E1, to assess the extent of oxidative-induced injury by measuring levels of oxidative cardiac and injury biomarkers in mice and to evaluate the effects of CYP2E1 on caspase 3 activity and nuclear factor erythroid 2-related factor-2 (NRF-2). MATERIALS AND METHODS: Mice (n = 32) were divided into four treatment groups of eight: control, MTX, MTX + 4-methlypyrazole (4MP) and MTX + disulfiram (Disf). After 6 weeks of treatments, blood and heart samples were collected. RESULTS: Liquid chromatography-mass spectrometry (LCMS) analysis of MTX-treated plasma samples revealed several metabolites with different retention times. Cardiac antioxidant enzymes and creatine kinase (CK) levels were not significantly different among the groups. However, cardiac troponin and caspase 3 activity were significantly raised, with increased CYP2E1 expressions and reduced NRF-2 expression. Tissue damage was observed in all the treatment groups, including MTX, leading to the conclusion that MTX-induced cardiotoxicity was mediated by CYP2E1 activity, which initiated caspase 3 production, and decreased NRF-2 expression. CONCLUSIONS: Therefore, agents that inhibit CPY2E1 expression might attenuate MTX-induced cardiotoxicity by increasing NRF-2 expression.


Subject(s)
Antineoplastic Agents/toxicity , Cardiotoxicity/drug therapy , Cytochrome P-450 CYP2E1 Inhibitors/therapeutic use , Disulfiram/therapeutic use , Fomepizole/therapeutic use , Mitoxantrone/toxicity , Animals , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Cardiotoxicity/blood , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Caspase 3/metabolism , Cytochrome P-450 CYP2E1/metabolism , Cytochrome P-450 CYP2E1 Inhibitors/pharmacology , Disulfiram/pharmacology , Female , Fomepizole/pharmacology , Male , Mice, Inbred BALB C , Mitoxantrone/blood , Mitoxantrone/pharmacokinetics , Myocardium/metabolism , Myocardium/pathology , NF-E2-Related Factor 2/metabolism , Troponin I/metabolism
11.
Strahlenther Onkol ; 197(1): 39-47, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32813034

ABSTRACT

PURPOSE: To assess acute cardiac toxicity caused by intraoperative radiotherapy (IORT) with low-energy x­rays for early breast cancer. METHODS: We prospectively analyzed pre- and postoperative troponin I and NT-proBNP in 94 women who underwent breast-conserving surgery between 2013 and 2017 at the Department of Gynecology and Obstetrics of the University Medical Center Mannheim, Germany. Thirty-nine women received IORT using low-energy x­rays during breast-conserving surgery while 55 patients without IORT formed the control group. Demographic and surgical parameters as well as cardiac markers were evaluated. RESULTS: There were no significant differences concerning age and side of breast cancer between the groups. Furthermore, no significant difference between the troponin I assays of the IORT and control groups could be found (preoperatively: 0.017 ± 0.006 ng/ml vs. 0.018 ± 0.008 ng/ml; p = 0.5105; postoperatively: 0.019 ± 0.012 ng/ml vs. 0.018 ± 0.010 ng/ml; p = 0.6225). N­terminal fragment of B­type natriuretic peptide (NT-proBNP) was significantly higher in the control group 24 h after surgery (preoperatively: 158.154 ± 169.427 pg/ml vs. 162.109 ± 147.343 pg/ml; p = 0.56; postoperatively: 168.846 ± 160.227 pg/ml vs. 232.527 ± 188.957 pg/ml; p = 0.0279). CONCLUSION: Troponin I levels as a marker of acute cardiac toxicity did not show any significant differences in patients who received IORT during breast-conserving surgery compared to those who did not.


Subject(s)
Breast Neoplasms/radiotherapy , Cardiomyopathies/etiology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Radiotherapy, Adjuvant/adverse effects , Troponin I/blood , Aged , Biomarkers , Breast Neoplasms/blood , Breast Neoplasms/surgery , Cardiomyopathies/blood , Cardiotoxicity/blood , Cardiotoxicity/etiology , Female , Humans , Intraoperative Care , Mastectomy, Segmental , Middle Aged , Prospective Studies
12.
Biomed Res Int ; 2020: 3189672, 2020.
Article in English | MEDLINE | ID: mdl-33274202

ABSTRACT

Diabetes mellitus has developed into one of the debilitating diseases disturbing the health of many people living with cardiovascular diseases in modern times. The root of Ageratum conyzoides was investigated for its effects on alloxan-induced diabetic Wistar rats' cardiac tissues. Thirty-two (32) Wistar rats weighing between 180 and 190 g were randomly divided into four groups. The animals in groups B-D were induced with a single dose of 150 mg/kg body weight of alloxan (ALX) intraperitoneally. They were confirmed hyperglycemic after 72 hours of induction and then sustained in hyperglycemic condition for 2 weeks. Animals in groups C and D received AC intervention, as stated above, for four weeks. The body weight of the experimental animals and blood collection for glucose estimation were taken weekly for six weeks using appropriate instruments. Biochemical assays for lipid profile, antioxidant enzymatic, and nonenzymatic markers were carried out. Histopathological changes in the cardiac tissues were also studied. Administration of 150 mg/kg of ALX to experimental rats induced diabetes and significantly reduced the body weights, significantly (p < 0.05) increased the glucose level, triglyceride (TG), total cholesterol (TC), and low-density lipoprotein (LDL) levels, and decreased the levels of high-density lipoprotein (HDL) and antioxidant enzymatic markers such as catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase (GPx) while the antioxidant nonenzymatic marker such as malondialdehyde (MDA) level was significantly increased. By contrast, rats given the ethanolic extract root of A. conyzoides had significantly (p < 0.05) increased the body weight gain, whereas the glucose levels significantly (p < 0.05) improved in treated diabetic rats. This extract also improved the cardiovascular system of the diabetic rats by significantly decreasing TG and LDL levels, significantly (p < 0.05) increasing the HDL level, significantly reducing the cardiac contents of CAT, SOD, and GPx, and significantly (p < 0.05) decreasing MDA. Ethanolic extract root of A. conyzoides exhibited antihyperglycemic and antihyperlipidemic activities and mitigates damage to the heart from the ALX-induced myocardial toxicity associated with type-1 diabetes.


Subject(s)
Ageratum/chemistry , Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Diabetes Mellitus, Experimental/drug therapy , Plant Extracts/therapeutic use , Plant Roots/chemistry , Administration, Oral , Animals , Antioxidants/pharmacology , Antioxidants/therapeutic use , Biomarkers/metabolism , Blood Glucose/metabolism , Body Weight/drug effects , Cardiotonic Agents/pharmacology , Cardiotoxicity/blood , Cardiotoxicity/enzymology , Cardiotoxicity/pathology , Diabetes Mellitus, Experimental/blood , Ethanol , Female , Lipids/blood , Male , Malondialdehyde/metabolism , Pancreas/drug effects , Pancreas/pathology , Plant Extracts/pharmacology , Rats, Wistar
13.
J Am Heart Assoc ; 9(23): e018143, 2020 12.
Article in English | MEDLINE | ID: mdl-33191846

ABSTRACT

Background Patients with breast cancer can be affected by cardiotoxic reactions through cancer therapies. Cardiac biomarkers, like NT-proBNP (N-terminal pro-B-type natriuretic peptide) and high-sensitivity cardiac troponin T, might have predictive value. Methods and Results Echocardiography, ECG, hemodynamic parameters, NT-proBNP and high-sensitivity cardiac troponin T were assessed in 853 patients with early-stage breast cancer randomized in the German Breast Group GeparOcto-GBG 84 phase III trial. Patients received neo-adjuvant dose-dense, dose-intensified epirubicin, paclitaxel, and cyclophosphamide (iddEPC group, n=424) or paclitaxel, non-pegylated doxorubicin, and in triple negative breast cancer, (paclitaxel, non-pegylated doxorubicin, carboplatin group, n=429) treatment for 18 weeks. Patients positive for human epidermal growth receptor 2 (n=354, 41.5%) received monoclonal antibodies on top of allocated therapy; 119 (12.9%) of all patients showed a cardiotoxic reaction during therapy (15 [1.8%] using a more strict definition). Presence of cardiotoxic reactions was irrespective of treatment allocation (P=0.31). Small but significant increases in NT-proBNP developed early in patients with a cardiotoxic reaction as compared with those without in whom NT-proBNP rose only towards the end of therapy (P=0.04). High-sensitivity cardiac troponin T rose early in both groups. Logistic regression showed that NT-proBNP (odds ratio [OR], 1.03; 95% CI, 1.008-1.055; P=0.01) and hemoglobin (OR, 1.31; 95% CI, 1.05-1.63; P=0.02) measured at 6 weeks after treatment initiation were significantly associated with cardiotoxic reactions. Conclusions NT-proBNP and hemoglobin are significantly associated with cardiotoxic reactions in patients with early-stage breast cancer undergoing dose-dense and dose-intensified chemotherapy, but high-sensitivity cardiac troponin T is not. Registration URL: http://www.clinicaltrials.gov; Unique identifier: NCT02125344.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/blood , Breast Neoplasms/drug therapy , Cardiotoxicity/epidemiology , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Troponin T/blood , Adult , Biomarkers/blood , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Cohort Studies , Echocardiography , Female , Germany , Humans , Logistic Models , Middle Aged , Odds Ratio , Prevalence , Risk Factors , Stroke Volume
14.
Oxid Med Cell Longev ; 2020: 1602816, 2020.
Article in English | MEDLINE | ID: mdl-33149803

ABSTRACT

Cardiotoxicity as an off-target effect of doxorubicin therapy is a major limiting factor for its clinical use as a choice cytotoxic agent. Seeds of Irvingia gabonensis have been reported to possess both nutritional and medicinal values which include antidiabetic, weight losing, antihyperlipidemic, and antioxidative effects. Protective effects of Irvingia gabonensis ethanol seed extract (IGESE) was investigated in doxorubicin (DOX)-mediated cardiotoxicity induced with single intraperitoneal injection of 15 mg/kg of DOX following the oral pretreatments of Wistar rats with 100-400 mg/kg/day of IGESE for 10 days, using serum cardiac enzyme markers (cardiac troponin I (cTI) and lactate dehydrogenase (LDH)), cardiac tissue oxidative stress markers (catalase (CAT), malonyldialdehyde (MDA), superoxide dismutase (SOD), glutathione-S-transferase (GST), glutathione peroxidase (GSH-Px), and reduced glutathione (GSH)), and cardiac histopathology endpoints. In addition, both qualitative and quantitative analyses to determine IGESE's secondary metabolites profile and its in vitro antioxidant activities were also conducted. Results revealed that serum cTnI and LDH were significantly elevated by the DOX treatment. Similarly, activities of tissue SOD, CAT, GST, and GSH levels were profoundly reduced, while GPx activity and MDA levels were profoundly increased by DOX treatment. These biochemical changes were associated with microthrombi formation in the DOX-treated cardiac tissues on histological examination. However, oral pretreatments with 100-400 mg/kg/day of IGESE dissolved in 5% DMSO in distilled water significantly attenuated increases in the serum cTnI and LDH, prevented significant alterations in the serum lipid profile and the tissue activities and levels of oxidative stress markers while improving cardiovascular disease risk indices and DOX-induced histopathological lesions. The in vitro antioxidant studies showed IGESE to have good antioxidant profile and contained 56 major secondary metabolites prominent among which are γ-sitosterol, Phytol, neophytadiene, stigmasterol, vitamin E, hexadecanoic acid and its ethyl ester, Phytyl palmitate, campesterol, lupeol, and squalene. Overall, both the in vitro and in vivo findings indicate that IGESE may be a promising prophylactic cardioprotective agent against DOX-induced cardiotoxicity, at least in part mediated via IGESE's antioxidant and free radical scavenging and antithrombotic mechanisms.


Subject(s)
Cardiotoxicity/drug therapy , Cellulose/chemistry , Doxorubicin/adverse effects , Plant Extracts/therapeutic use , Seeds/chemistry , Animals , Antioxidants/pharmacology , Ascorbic Acid/pharmacology , Biomarkers/metabolism , Biphenyl Compounds/chemistry , Cardiotoxicity/blood , Free Radical Scavengers/chemistry , Gas Chromatography-Mass Spectrometry , Lipids/blood , Male , Metabolome , Myocardium/pathology , Nitric Oxide/metabolism , Oxidative Stress/drug effects , Phytochemicals/analysis , Picrates/chemistry , Plant Extracts/pharmacology , Rats, Wistar , Risk Factors , Secondary Metabolism/drug effects
15.
Eur J Heart Fail ; 22(11): 1966-1983, 2020 11.
Article in English | MEDLINE | ID: mdl-33006257

ABSTRACT

Serum biomarkers are an important tool in the baseline risk assessment and diagnosis of cardiovascular disease in cancer patients receiving cardiotoxic cancer treatments. Increases in cardiac biomarkers including cardiac troponin and natriuretic peptides can be used to guide initiation of cardioprotective treatments for cancer patients during treatment and to monitor the response to cardioprotective treatments, and they also offer prognostic value. This position statement examines the role of cardiac biomarkers in the management of cancer patients. The Cardio-Oncology Study Group of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC) in collaboration with the Cardio-Oncology Council of the ESC have evaluated the current evidence for the role of cardiovascular biomarkers in cancer patients before, during and after cardiotoxic cancer therapies. The characteristics of the main two biomarkers troponin and natriuretic peptides are discussed, the link to the mechanisms of cardiovascular toxicity, and the evidence for their clinical use in surveillance during and after anthracycline chemotherapy, trastuzumab and HER2-targeted therapies, vascular endothelial growth factor inhibitors, proteasome inhibitors, immune checkpoint inhibitors, cyclophosphamide and radiotherapy. Novel surveillance clinical pathways integrating cardiac biomarkers for cancer patients receiving anthracycline chemotherapy or trastuzumab biomarkers are presented and future direction in cardio-oncology biomarker research is discussed.


Subject(s)
Antineoplastic Agents , Cardiotoxicity , Heart Failure , Neoplasms , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Biomarkers, Tumor/blood , Cardiotonic Agents/administration & dosage , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Cardiotoxicity/etiology , Heart Failure/blood , Heart Failure/chemically induced , Heart Failure/diagnosis , Humans , Neoplasms/blood , Neoplasms/drug therapy
16.
Biomed Res Int ; 2020: 4018412, 2020.
Article in English | MEDLINE | ID: mdl-32851069

ABSTRACT

Doxorubicin is an anthracycline antibiotic that is used for the treatment of various types of cancer. However, its clinical usage is limited due to its potential life-threatening adverse effects, such as cardio- and nephrotoxicities. Nonetheless, simultaneous administration of doxorubicin and antioxidants, such as those found in green tea leaves, could reduce cardiac and renal tissue damage caused by oxidative stress. The methylxanthine fraction isolated from Bancha tea leaves were tested in vitro for its antioxidant activity and in vivo for its organoprotective properties against doxorubicin-induced cardio- and nephrotoxicities in a rat model. The in vivo study was conducted on male Wistar rats divided into 6 groups. Methylxanthines were administered at high (5 mg/kg body weight) and low (1 mg/kg body weight) doses, while doxorubicin was administered at a cumulative dose of 20 mg/kg body weight. Serum creatinine, uric acid, and urea concentrations, as well as serum enzyme levels (creatinine kinase (CK), creatinine kinase MB fraction (CK-MB), aspartate aminotransferase (AST), and lactate dehydrogenase (LDH)) and electrolytes (Na+, K+, and Cl-), were analysed. In addition, histological analysis was performed to assess cardiac and renal tissue damage. The concomitant administration of Bancha methylxanthines and doxorubicin showed a dose-dependent reduction in the serum biochemical parameters, indicating a decrease in the cardiac and renal tissue damage caused by the antibiotic. Histological analysis showed that pretreatment with methylxanthines at the dose of 5 mg/kg resulted in an almost normal myocardial structure and a significant decrease in the morphological kidney changes caused by doxorubicin exposure compared with the group that received doxorubicin alone. The putative mechanism is most likely related to a reduction in the oxidative stress caused by doxorubicin.


Subject(s)
Cardiotoxicity/drug therapy , Doxorubicin/adverse effects , Kidney Diseases/drug therapy , Xanthines/pharmacology , Animals , Aspartate Aminotransferases/blood , Cardiotoxicity/blood , Cardiotoxicity/genetics , Cardiotoxicity/pathology , Creatinine/blood , Disease Models, Animal , Doxorubicin/therapeutic use , Heart/drug effects , Heart/physiopathology , Heart Diseases/chemically induced , Humans , Kidney Diseases/blood , Kidney Diseases/chemically induced , Oxidative Stress/drug effects , Plant Leaves/chemistry , Rats , Tea/chemistry , Urea/blood , Uric Acid/blood , Xanthines/chemistry
17.
Clin Chem Lab Med ; 59(1): 51-57, 2020 08 26.
Article in English | MEDLINE | ID: mdl-32845860

ABSTRACT

Drug-induced cardiotoxicity is a major clinical problem; cardiotoxic drugs may induce both cardiac dysfunction and myocardial injury. Several recent studies reported that cardiac troponins measured with high-sensitivity methods (hs-cTn) can enable the early detection of myocardial injury related to chemotherapy or abuse of drugs that are potentially cardiotoxic. Several authors have some concerns about the standard definition of cardiotoxicity, in particular, regarding the early evaluation of chemotherapy cardiotoxicity in cancer patients. Several recent studies using the hs-cTn assay indicate that myocardial injury may precede by some months or years the diagnosis of heart failure (HF) based on the evaluation of left ventricular ejection fraction (LVEF). Accordingly, hs-cTn assay should considered to be a reliable laboratory test for the early detection of asymptomatic or subclinical cardiotoxic damage in patients undergoing cancer chemotherapy. In accordance with the Fourth Universal Definition of Myocardial Infarction and also taking into account the recent experimental and clinical evidences, the definition of drug-cardiotoxicity should be updated considering the early evaluation of myocardial injury by means of hs-cTn assay. It is conceivable that the combined use of hs-cTn assay and cardiac imaging techniques for the evaluation of cardiotoxicity will significantly increase both diagnostic sensitivity and specificity, and also better prevent chemotherapy-related left ventricular (LV) dysfunction and other adverse cardiac events. However, large randomized clinical trials are needed to evaluate the cost/benefit ratio of standardized protocols for the early detection of cardiotoxicity using hs-cTn assay in patients receiving chemotherapy for malignant diseases.


Subject(s)
Cardiotoxicity/diagnosis , Troponin I/blood , Troponin T/blood , Animals , Biomarkers/blood , Cardiotoxicity/blood , Humans
18.
Eur J Pharmacol ; 882: 173229, 2020 Sep 05.
Article in English | MEDLINE | ID: mdl-32505666

ABSTRACT

Sorafenib, a tyrosine kinase inhibitor that is used in the treatment of hepatocellular and renal cell carcinoma, was reported to induce cardiotoxicity. This study aimed to investigate the potential cardioprotective effect of losartan against sorafenib-induced cardiotoxicity in rat. Sorafenib significantly reduced the left ventricular pressure, heart rate dp/dt max & dp/dt min (indexes of myocardial contractility and relaxation; respectively), and prolonged both the systolic and diastolic periods. Coadminstration of losartan significantly reversed the effects of sorafenib on heart rate, dp/dt max and dp/dt min. In addition, there was a tendency for losartan to reverse sorafenib reduction in left ventricular pressure and perfusion pressure but it did not reach statistical significance. A GC-MS non-targeted based metabolites profiling of rat plasma revealed elevated metaboites, including urea and fatty acids levels, associated with sorafenib induced cardiotoxicity. However, only glycine and lactic acid were statistically significant. Interestingly, losartan co-administration with sorafenib restored these changes, and resulted in a significantly reduced glycine, urea and some fatty acids levels namely; Cis-vaccenic acid, oleic acid, stearic acid and undecanoic acid. In addition, based on histology results, losartan coadminitration almost obviated sorafenib-induced changes in cardiac tissues. The study suggests that losartan has the potential to exert a protective effect against sorafenib-induced cardiotoxicity.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotonic Agents/therapeutic use , Cardiotoxicity/drug therapy , Losartan/therapeutic use , Protein Kinase Inhibitors/adverse effects , Sorafenib/adverse effects , Animals , Cardiotonic Agents/pharmacology , Cardiotoxicity/blood , Cardiotoxicity/metabolism , Cardiotoxicity/pathology , Heart/drug effects , Heart/physiology , Heart Rate/drug effects , Losartan/pharmacology , Male , Metabolomics , Myocardium/metabolism , Myocardium/pathology , Rats, Wistar
19.
Oncology ; 98(9): 653-660, 2020.
Article in English | MEDLINE | ID: mdl-32454480

ABSTRACT

BACKGROUND: Anthracycline is used to treat various types of cancer; however, cardiotoxicity negatively affects patient prognosis. OBJECTIVES: The aim of the present study was to investigate serial changes in levels of cardiac troponin I (TnI) and B-type natriuretic peptide (BNP) in patients treated with anthracycline-containing therapy. METHODS: 91 consecutive cancer patients planned for anthracycline treatment were enrolled and followed up for 12 months. All patients underwent echocardiography and blood sampling at baseline, 3, 6, and 12 months. RESULTS: The patients were divided into two groups based on their TnI level during the follow-up period: the elevated TnI group (TnI ≥0.03 ng/mL; n = 37) and the normal TnI group (n = 54). In the elevated TnI group, the TnI levels increased at 3 and 6 months, but they returned to within normal range at 12 months after anthracycline administration. Unlike TnI, the BNP levels began to increase after 6 months, and remained increased at 12 months. The occurrence of cancer therapeutics-related cardiac dysfunction was higher in the elevated TnI group than in the normal TnI group. When we set the cut-off value of TnI at 0.029 ng/mL, sensitivity and specificity to predict an elevated BNP level of more than 100 pg/mL were 90 and 63%, respectively. Multivariate logistic regression analysis revealed that elevated TnI was an independent predictor of elevated BNP levels. CONCLUSION: Elevated TnI was an independent predictor for the development of BNP increase. The different characteristics of TnI and BNP should be considered when managing patients treated with anthracycline-containing therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Cardiotoxicity/blood , Natriuretic Peptide, Brain/blood , Neoplasms/blood , Neoplasms/drug therapy , Troponin I/blood , Anthracyclines/administration & dosage , Anthracyclines/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cardiotoxicity/diagnostic imaging , Cardiotoxicity/etiology , Echocardiography , Female , Humans , Male , Middle Aged , Predictive Value of Tests
20.
J Vet Intern Med ; 34(3): 1260-1271, 2020 May.
Article in English | MEDLINE | ID: mdl-32255536

ABSTRACT

BACKGROUND: Long-term use of doxorubicin (DOX) is limited by cumulative dose-dependent cardiotoxicity. OBJECTIVES: Identify plasma extracellular vesicle (EV)-associated microRNAs (miRNAs) as a biomarker for cardiotoxicity in dogs by correlating changes with cardiac troponin I (cTnI) concentrations and, echocardiographic and histologic findings. ANIMALS: Prospective study of 9 client-owned dogs diagnosed with sarcoma and receiving DOX single-agent chemotherapy (total of 5 DOX treatments). Dogs with clinically relevant metastatic disease, preexisting heart disease, or breeds predisposed to cardiomyopathy were excluded. METHODS: Serum concentration of cTnI was monitored before each treatment and 1 month after the treatment completion. Echocardiography was performed before treatments 1, 3, 5, and 1 month after completion. The EV-miRNA was isolated and sequenced before treatments 1 and 3, and 1 month after completion. RESULTS: Linear mixed model analysis for repeated measurements was used to evaluate the effect of DOX. The miR-107 (P = .03) and miR-146a (P = .02) were significantly downregulated whereas miR-502 (P = .02) was upregulated. Changes in miR-502 were significant before administration of the third chemotherapeutic dose. When stratifying miRNA expression for change in left ventricular ejection fraction, upregulation of miR-181d was noted (P = .01). Serum concentration of cTnI changed significantly but only 1 month after treatment completion, and concentrations correlated with left ventricular ejection fraction and left ventricular internal dimension in diastole. CONCLUSION AND CLINICAL SIGNIFICANCE: Downregulation of miR-502 was detected before significant changes in cTnI concentrations or echocardiographic parameters. Further validation using a larger sample size will be required.


Subject(s)
Biomarkers/blood , Cardiotoxicity/veterinary , Dog Diseases/diagnosis , Doxorubicin/adverse effects , MicroRNAs/blood , Animals , Cardiotoxicity/blood , Cardiotoxicity/diagnosis , Dog Diseases/blood , Dogs , Echocardiography/veterinary , Extracellular Vesicles/drug effects , Female , Male , Neoplasms/drug therapy , Neoplasms/veterinary , Prospective Studies , Sarcoma/drug therapy , Sarcoma/veterinary , Troponin I/blood , Ventricular Function, Left/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...