Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
BMJ Case Rep ; 15(10)2022 Oct 17.
Article in English | MEDLINE | ID: mdl-36253013

ABSTRACT

5-Fluorouracil (5-FU), a known cardiotoxin, is the backbone for the treatment of colorectal cancer. It is associated with arrhythmias, myocardial infarction and sudden cardiac death. Most commonly, it is associated with coronary vasospasm secondary to direct toxic effects on vascular endothelium.A woman with metastatic colon cancer, originally treated with a 5-FU infusion as part of the FOLFIRI (Folinic acid, 5-Fluorouracil, Irinotecan) regimen, was unable to tolerate the chemotherapy due to chest pain. She was transitioned from infusional 5-FU to inferior 1-hour bolus 5-FU, in an attempt to minimise cardiotoxicity, but had disease progression. A multidisciplinary decision was made to again trial 5-FU infusion and pretreat with diltiazem. She tolerated chemotherapy without adverse events. A multidisciplinary discussion is recommended for co-management of reversible 5-FU-associated cardiotoxicity. After coronary artery disease (CAD) risk stratification and treatment, empiric treatment with calcium channel blockers and/or nitrates may allow patients with suspected coronary vasospasm, from 5-FU, to continue this vital chemotherapy.


Subject(s)
Colorectal Neoplasms , Coronary Vasospasm , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Calcium Channel Blockers/therapeutic use , Camptothecin , Cardiotoxicity/etiology , Cardiotoxins/therapeutic use , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Coronary Vasospasm/chemically induced , Coronary Vasospasm/drug therapy , Diltiazem/therapeutic use , Female , Fluorouracil , Humans , Irinotecan/therapeutic use , Leucovorin/adverse effects , Nitrates/therapeutic use
2.
Circ Res ; 128(10): 1468-1486, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33983837

ABSTRACT

Acute decompensated heart failure (ADHF) is one of the leading admission diagnoses worldwide, yet it is an entity with incompletely understood pathophysiology and limited therapeutic options. Patients admitted for ADHF have high in-hospital morbidity and mortality, as well as frequent rehospitalizations and subsequent cardiovascular death. This devastating clinical course is partly due to suboptimal medical management of ADHF with persistent congestion upon hospital discharge and inadequate predischarge initiation of life-saving guideline-directed therapies. While new drugs for the treatment of chronic HF continue to be approved, there has been no new therapy approved for ADHF in decades. This review will focus on the current limited understanding of ADHF pathophysiology, possible therapeutic targets, and current limitations in expanding available therapies in light of the unmet need among these high-risk patients.


Subject(s)
Heart Failure/drug therapy , Heart Failure/physiopathology , Acute Disease , Body Fluids/physiology , Cardio-Renal Syndrome/complications , Cardiotoxins/therapeutic use , Comorbidity , Heart Failure/diagnosis , Heart Failure/etiology , Hospitalization , Humans , Inflammation Mediators/metabolism , Myocardial Contraction/physiology , Natriuretic Peptide, Brain/metabolism , Patient Discharge , Patient Readmission , Renin-Angiotensin System/physiology , Symptom Assessment , Vascular Resistance , Vasoconstriction/physiology , Vasodilator Agents/therapeutic use
3.
J Oncol Pharm Pract ; 26(2): 434-444, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31594518

ABSTRACT

BACKGROUND: Doxorubicin is one of the most commonly prescribed and time-tested anticancer drugs. Although being considered as a first line drug in different types of cancers, the two main obstacles to doxorubicin therapy are drug-induced cardiotoxicity and drug resistance. METHOD: The study utilizes systemic reviews on publications of previous studies obtained from scholarly journal databases including PubMed, Medline, Ebsco Host, Google Scholar, and Cochrane. The study utilizes secondary information obtained from health organizations using filters and keywords to sustain information relevancy. The study utilizes information retrieved from studies captured in the peer-reviewed journals on "doxorubicin-induced cardiotoxicity" and "doxorubicin resistance." DISCUSSION AND RESULTS: The exact mechanisms of cardiotoxicity are not known; various hypotheses are studied. Doxorubicin can lead to free radical generation in various ways. The commonly proposed underlying mechanisms promoting doxorubicin resistance are the expression of multidrug resistance proteins as well as other causes. CONCLUSION: In this review, we have described the major obstacles to doxorubicin therapy, doxorubicin-induced cardiotoxicity as well as the mechanisms of cancer drug resistance and in following the treatment failures.


Subject(s)
Antibiotics, Antineoplastic/adverse effects , Cardiotoxins/adverse effects , Cardiovascular Diseases/chemically induced , Doxorubicin/adverse effects , Drug Resistance, Neoplasm/drug effects , Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/therapeutic use , Cardiotoxicity , Cardiotoxins/therapeutic use , Cardiovascular Diseases/diagnosis , Doxorubicin/therapeutic use , Drug Resistance, Neoplasm/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Neoplasms/diagnosis
4.
Clin Drug Investig ; 38(12): 1109-1123, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30327999

ABSTRACT

BACKGROUND AND OBJECTIVE: Use of the vascular endothelial growth factor receptor tyrosine kinase inhibitors (VEGFR-TKIs) has led to considerable improvements in the clinical outcome of patients with various tumor types. However, VEGFR-TKIs may be associated with increased incidence of cardiovascular toxicities. We conducted this meta-analysis to systematically review the risk of cardiovascular toxicities with VEGFR-TKIs in cancer patients. METHODS: The relevant studies of the randomized controlled trials in cancer patients treated with VEGFR-TKIs were retrieved and a systematic evaluation was conducted. EMBASE, MEDLINE, and PubMed were searched for articles published until April 2018. RESULTS: A total of 77 randomized controlled trials and 27,353 patients were included. The current meta-analysis suggests that the use of VEGFR-TKIs significantly increases the risk of developing cardiovascular toxicities, such as all-grade and high-grade hypertension, all-grade bleeding, and all-grade cardiac dysfunction. Hypertension was the most common cardiovascular toxicity. There was no significant increased risk of all-grade and high-grade thromboembolism, high-grade bleeding, and high-grade cardiac dysfunction associated with these agents. CONCLUSIONS: The available data suggest that the use of VEGFR-TKIs is associated with a significantly increased risk of cardiovascular toxicities in cancer patients. Clinicians should be aware of this risk and perform regular cardiovascular monitoring.


Subject(s)
Cardiotoxins/adverse effects , Cardiovascular Diseases/chemically induced , Neoplasms/drug therapy , Protein Kinase Inhibitors/adverse effects , Randomized Controlled Trials as Topic/methods , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Cardiotoxins/therapeutic use , Cardiovascular Diseases/enzymology , Hemorrhage/chemically induced , Hemorrhage/enzymology , Humans , Neoplasms/metabolism , Protein Kinase Inhibitors/therapeutic use , Receptors, Vascular Endothelial Growth Factor/metabolism , Risk Factors , Thromboembolism/chemically induced , Thromboembolism/enzymology
5.
Support Care Cancer ; 26(7): 2113-2123, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29671063

ABSTRACT

PURPOSE: The main purpose of this review was to synthesize evidence from existing childhood cancer survivor studies that report the effect of aerobic exercise on cardiopulmonary fitness (a marker of cardiovascular health), in survivors that were currently receiving or had been treated with a cardiotoxic agent. METHODS: Studies were identified for this review by searching both electronic databases of peer-reviewed articles, as well as various sources of gray literature. Risk of bias was qualitatively assessed in these studies using the domains outlined in the Cochrane Handbook for Systematic Reviews of Interventions. Data was analyzed quantitatively using random-effects meta-analyses and subgroup analyses in RevMan Software. RESULTS: Meta-analysis of pooled evidence from the nine included studies suggests that aerobic exercise has a statistically and clinically significant positive effect on cardiopulmonary fitness (effect estimate = 6.92%, p value = 0.02). Findings from subgroup analyses of clinical characteristics and exercise parameters were not significant. CONCLUSIONS: The findings from this review, although not directly demonstrating a cardioprotective effect, are a preliminary step towards establishing the putative cardioprotective effect of aerobic exercise against the direct cardiotoxic impact of cancer treatments. The significant positive effect estimate in favor of aerobic exercise is a small but important advancement towards the standardization of aerobic exercise in childhood cancer survivors. Further studies are necessary.


Subject(s)
Antineoplastic Agents/toxicity , Cancer Survivors , Cardiotoxins/therapeutic use , Exercise/physiology , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Cardiotoxicity/therapy , Child , Heart/physiology , Humans , Respiratory Physiological Phenomena
6.
Turk Kardiyol Dern Ars ; 45(8): 752-754, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29226898

ABSTRACT

An unconscious, 25-year-old, male patient was brought to the emergency department. The patient's electrocardiography demonstrated a wide QRS interval and first-degree atrioventricular block. He was being treated with propafenone twice daily (450 mg) for paroxysmal atrial fibrillation. The patient had consumed a substantial amount of alcohol the day before. He recovered after supportive management with sodium bicarbonate and inotropic therapy. In the presently described case, treatment resulted in quick normalization of QRS interval and stabilization of hemodynamic status.


Subject(s)
Alcohol Drinking/adverse effects , Anti-Arrhythmia Agents/adverse effects , Atrial Fibrillation/drug therapy , Atrioventricular Block , Cardiotoxins/adverse effects , Propafenone/adverse effects , Adult , Anti-Arrhythmia Agents/therapeutic use , Atrioventricular Block/chemically induced , Atrioventricular Block/therapy , Cardiotoxins/therapeutic use , Electrocardiography , Humans , Male , Propafenone/therapeutic use
7.
Cardiovasc Diabetol ; 16(1): 113, 2017 09 13.
Article in English | MEDLINE | ID: mdl-28903775

ABSTRACT

BACKGROUND: In the Saxagliptin Assessment of Vascular Outcomes Recorded in Patients with Diabetes Mellitus (SAVOR) trial in patients with type 2 diabetes mellitus (T2D) at high risk of cardiovascular (CV) disease, saxagliptin did not increase the risk for major CV adverse events. However, there was an unexpected imbalance in events of hospitalization for heart failure (hHF), one of six components of the secondary CV composite endpoint, with a greater number of events observed with saxagliptin. Here, we examined findings from nonclinical safety and clinical pharmacology studies of saxagliptin with the aim of identifying any potential signals of myocardial injury. METHODS: In vitro and in vivo (rat, dog, monkey) safety pharmacology and toxicology studies evaluating the potential effects of saxagliptin and its major active metabolite, 5-hydroxy saxagliptin, on the CV system are reviewed. In addition, results from saxagliptin clinical studies are discussed: one randomized, 2-period, double-blind, placebo-controlled single-ascending-dose study (up to 100 mg); one randomized, double-blind, placebo-controlled, sequential, multiple-ascending-high-dose study (up to 400 mg/day for 14 days); and one randomized, double-blind, 4-period, 4-treatment, cross-over thorough QTc study (up to 40 mg/day for 4 days) in healthy volunteers; as well as one randomized, placebo-controlled, sequential multiple-ascending-dose study in patients with T2D (up to 50 mg/day for 14 days). RESULTS: Neither saxagliptin nor 5-hydroxy saxagliptin affected ligand binding to receptors and ion channels (e.g. potassium channels) or action potential duration in in vitro studies. In animal toxicology studies, no changes in the cardiac conduction system, blood pressure, heart rate, contractility, heart weight, or heart histopathology were observed. In healthy participants and patients with T2D, there were no findings suggestive of myocyte injury or fluid overload. Serum chemistry abnormalities indicative of cardiac injury, nonspecific muscle damage, or fluid homeostasis changes were infrequent and balanced across treatment groups. There were no QTc changes associated with saxagliptin. No treatment-emergent adverse events suggestive of heart failure or myocardial damage were reported. CONCLUSIONS: The saxagliptin nonclinical and clinical pharmacology programs did not identify evidence of myocardial injury and/or CV harm that may have predicted or may explain the unexpected imbalance in the rate of hHF observed in SAVOR.


Subject(s)
Adamantane/analogs & derivatives , Diabetes Mellitus, Type 2/drug therapy , Dipeptides/therapeutic use , Dipeptidyl-Peptidase IV Inhibitors/therapeutic use , Adamantane/adverse effects , Adamantane/therapeutic use , Animals , Cardiotoxins/adverse effects , Cardiotoxins/therapeutic use , Cardiovascular Diseases/blood , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/epidemiology , Diabetes Mellitus, Type 2/blood , Diabetes Mellitus, Type 2/epidemiology , Dipeptides/adverse effects , Dipeptidyl-Peptidase IV Inhibitors/adverse effects , Humans , Randomized Controlled Trials as Topic/methods
8.
Int J Cardiol ; 245: 263-270, 2017 Oct 15.
Article in English | MEDLINE | ID: mdl-28735755

ABSTRACT

BACKGROUND: Preclinical studies have reported that a single treadmill session performed 24h prior to doxorubicin provides cardio-protection. We aimed to characterize the acute change in cardiac function following an initial doxorubicin treatment in humans and determine whether an exercise session performed 24h prior to treatment changes this response. METHODS: Breast cancer patients were randomized to either 30min of vigorous-intensity exercise 24h prior to the first doxorubicin treatment (n=13), or no vigorous exercise for 72h prior to treatment (control, n=11). Echocardiographically-derived left ventricular volumes, longitudinal strain, twist, E/A ratio, and circulating NT-proBNP, a marker of later cardiotoxicity, were measured before and 24-48h after the treatment. RESULTS: Following treatment in the control group, NT-proBNP, end-diastolic and stroke volumes, cardiac output, E/A ratio, strain, diastolic strain rate, twist, and untwist velocity significantly increased (all p≤0.01). Whereas systemic vascular resistance (p<0.01) decreased, and ejection fraction (p=0.02) and systolic strain rate (p<0.01) increased in the exercise group only. Relative to control, the exercise group had a significantly lower NT-proBNP (p<0.01) and a 46% risk reduction of exceeding the cut-point used to exclude acute heart failure. CONCLUSION: The first doxorubicin treatment is associated with acutely increased NT-proBNP, echocardiographic parameters of myocardial relaxation, left ventricular volume overload, and changes in longitudinal strain and twist opposite in direction to documented longer-term changes. An exercise session performed 24h prior to treatment attenuated NT-proBNP release and increased systolic function. Future investigations should verify these findings in a larger cohort and across multiple courses of doxorubicin.


Subject(s)
Breast Neoplasms/physiopathology , Breast Neoplasms/therapy , Cardiotoxins/therapeutic use , Exercise Test/trends , High-Intensity Interval Training/trends , Proof of Concept Study , Adult , Blood Pressure/drug effects , Blood Pressure/physiology , Breast Neoplasms/blood , Cardiac Output/drug effects , Cardiac Output/physiology , Cardiotoxins/adverse effects , Doxorubicin/adverse effects , Doxorubicin/therapeutic use , Exercise/physiology , Female , Humans , Middle Aged , Natriuretic Peptide, Brain/blood , Peptide Fragments/blood , Stroke Volume/drug effects , Stroke Volume/physiology
9.
Gan To Kagaku Ryoho ; 44(13): 2052-2057, 2017 Dec.
Article in Japanese | MEDLINE | ID: mdl-29361616

ABSTRACT

In Japan, cardiovascular diseases are frequent complications in cancer patients owing to the rapidly aging population and changes in the overall lifestyle. In addition, new anticancer therapies have substantially improved the prognosis of cancer patients. Cardiotoxicity, also referred to as cancer treatment-related cardiac dysfunction, has become an important cause of morbidity and mortality in cancer patients. Cardiotoxicity may consist of hypertension, arrhythmia, thromboembolism, coronary artery disease, valvular disease, and left ventricular dysfunction which may progress to heart failure. Close interactions between cardiologists and oncologists are required for the optimal care of many cancer patients. Although cardiologists are expected to assist and advise the oncologist by providing diagnostic and prognostic information regarding developing cardiotoxicity, little is known about the cardiovascular pathogenic mechanisms associated with cancer treatment. Onco-cardiology is a medical subspecialty that focuses on the diagnosis and treatment of cardiotoxicity in cancer patients. This review describes the concept of onco-cardiology, and focuses on the management of cardiotoxicity that may arise during or after cancer therapy from the standpoint of cardiology. We also discuss noninvasive diagnostic options to identify and characterize cardiotoxicity.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxins/adverse effects , Cardiovascular Diseases/chemically induced , Neoplasms/drug therapy , Antineoplastic Agents/therapeutic use , Biomarkers/blood , Cardiotoxins/therapeutic use , Cardiovascular Diseases/complications , Humans , Risk Factors
11.
Echocardiography ; 33(4): 519-26, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26992012

ABSTRACT

AIM: The combination of anthracyclines (AC) and trastuzumab (TRZ) is highly effective in patients with aggressive HER-2 + breast cancer, but has a significant risk of cardiotoxicity (CT). Trastuzumab-induced CT may be reversible. The aim of this study was to identify echocardiographic parameters associated with recovery of left ventricular ejection fraction (LVEF) in patients who developed CT after AC and TRZ treatment. METHODS AND RESULTS: Women with newly diagnosed breast cancer treated with AC followed by TRZ and monitored with serial echocardiograms were retrospectively studied. Left ventricular end-diastolic and systolic volumes, LVEF, and global longitudinal strain (GLS) were examined. Development and reversibility of CT were defined based on changes in LVEF according to the 2014 ASE/EACVI recommendations. Cox analysis was used to determine the association of echocardiographic variables with the subsequent development and reversibility of CT. Ninety-five patients underwent 5 echocardiograms or more in a 17-month (13-28 months) follow-up period. Nineteen patients (20%) developed CT. Left ventricular volumes, LVEF, and GLS measured after AC completion identified the subsequent development of CT. Of the 19 patients with CT, the LVEF partially or fully recovered in 13 (68%). GLS at the time of CT diagnosis was associated with subsequent recovery of LVEF (P = 0.004). CONCLUSION: In patients with breast cancer treated with AC and TRZ who develop CT, GLS at the time of CT diagnosis is associated with subsequent recovery of LVEF and may be useful for risk stratification and to guide treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Echocardiography/methods , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/physiopathology , Anthracyclines , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Breast Neoplasms/complications , Breast Neoplasms/metabolism , Cardiotoxins/adverse effects , Cardiotoxins/therapeutic use , Elastic Modulus/drug effects , Elasticity Imaging Techniques/methods , Female , Humans , Longitudinal Studies , Middle Aged , Receptor, ErbB-2/metabolism , Recovery of Function , Stroke Volume/drug effects , Trastuzumab/administration & dosage , Trastuzumab/adverse effects , Treatment Outcome , Ventricular Dysfunction, Left/diagnostic imaging
12.
J Nucl Cardiol ; 23(4): 824-32, 2016 08.
Article in English | MEDLINE | ID: mdl-26048264

ABSTRACT

BACKGROUND: Trastuzumab is successfully used for the treatment of HER2-positive breast cancer. Because of its association with cardiotoxicity, LVEF is monitored by MUGA, though this is a relatively late measure of cardiac function. Diastolic dysfunction (DD) is believed to be an early predictor of cardiac impairment. We evaluate the merit of MUGA-derived diastolic function parameters in the early detection of trastuzumab-induced cardiotoxicity (TIC). METHODS AND RESULTS: 77 trastuzumab-treated patients with normal baseline systolic and diastolic function were retrospectively selected (n = 77). All serial MUGA examinations were re-analyzed for systolic and diastolic function parameters. 36 patients (47%) developed SD and 45 patients (58%) DD during treatment. Both systolic and diastolic parameters significantly decreased. Of the patients with SD, 24 (67%) also developed DD. DD developed prior to systolic impairment in 54% of cases, in 42% vice versa, while time to occurrence did not differ significantly (P = .52). This also applied to the subgroup of advanced stage breast cancer patients (P = .1). CONCLUSIONS: Trastzumab-induced SD and DD can be detected by MUGA. An impairment of MUGA-derived diastolic parameters does not occur prior to SD and therefore cannot be used as earlier predictors of TIC.


Subject(s)
Breast Neoplasms/drug therapy , Coronary Angiography/methods , Radionuclide Angiography/methods , Stroke Volume/drug effects , Trastuzumab/adverse effects , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/diagnostic imaging , Antineoplastic Agents, Immunological/adverse effects , Breast Neoplasms/complications , Cardiotoxins/adverse effects , Cardiotoxins/therapeutic use , Female , Humans , Middle Aged , Reproducibility of Results , Retrospective Studies , Sensitivity and Specificity , Trastuzumab/therapeutic use , Treatment Outcome
13.
Cancer Chemother Pharmacol ; 76(4): 777-84, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26281907

ABSTRACT

PURPOSE: To characterize the cardiovascular safety profile of regorafenib in patients with advanced cancer. METHODS: Patients received regorafenib 160 mg/day for 21 days followed by a 7-day break. The primary endpoint was the change from baseline in QTcF at the regorafenib t(max) (Day 21, Cycle 1 or 2) and changes in left ventricular ejection fraction (LVEF) from baseline on Cycle 2, Day 21. Secondary objectives were pharmacokinetics, safety, anti-tumor activity and effects on electrocardiogram intervals. QT intervals were corrected using the methods of Fridericia (QTcF) and Bazett (QTcB). LVEF was assessed by multigated acquisition scanning. RESULTS: Fifty-three patients were enrolled, and all received at least one dose of regorafenib 160 mg. Twenty-five patients received regorafenib for 21 days without dose reduction. The mean change from baseline in QTcF at t(max) was (-)2 ms (90 % CI -8, 3). No patient experienced a change from baseline in QTcF > 60 ms, and two had QTcF changes between 30 and 60 ms. No patient had a QTcF or QTcB > 480 ms. In 27 patients who received at least 80 mg of regorafenib, the mean change from baseline in LVEF% ± SD was 1.7 ± 7.8. In 14 patients without a dose reduction, the mean change from baseline in LVEF% was (-)0.1 ± 8.6 at Cycle 2, Day 21. Four patients experienced a LVEF decrease between 10 and 20 %. CONCLUSION: The effects of regorafenib on the QT/QTc interval and LVEF were modest and unlikely to be of clinical significance in the setting of advanced cancer therapy.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiovascular Diseases/chemically induced , Drugs, Investigational/adverse effects , Heart Ventricles/drug effects , Neoplasms/drug therapy , Phenylurea Compounds/adverse effects , Protein Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Aged , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Cardiotoxins/adverse effects , Cardiotoxins/blood , Cardiotoxins/pharmacokinetics , Cardiotoxins/therapeutic use , Cardiovascular Diseases/physiopathology , Diarrhea/chemically induced , Diarrhea/physiopathology , Drug Eruptions/physiopathology , Drugs, Investigational/metabolism , Drugs, Investigational/pharmacokinetics , Drugs, Investigational/therapeutic use , Electrocardiography/drug effects , Female , Heart Ventricles/physiopathology , Humans , Male , Middle Aged , Mucositis/chemically induced , Mucositis/physiopathology , Neoplasms/blood , Neoplasms/metabolism , Neoplasms/physiopathology , Phenylurea Compounds/blood , Phenylurea Compounds/pharmacokinetics , Phenylurea Compounds/therapeutic use , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/pharmacokinetics , Protein Kinase Inhibitors/therapeutic use , Pyridines/blood , Pyridines/pharmacokinetics , Pyridines/therapeutic use , Severity of Illness Index , Stroke Volume/drug effects
14.
Biochim Biophys Acta ; 1848(10 Pt B): 2727-37, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25596534

ABSTRACT

Antitumor drugs have long been known to introduce a measurable risk of cardiovascular events. Cardio-Oncology is the discipline that builds on collaboration between cardiologists and oncologists and aims at screening, preventing or minimizing such a risk. Overt concern about "possible" cardiovascular toxicity might expose cancer patients to the risk of tumor undertreatment and poor oncologic outcome. Careful analysis of risk:benefit balance is therefore central to the management of patients exposed to potentially cardiotoxic drugs. Concomitant or sequential management of cardiac and cancer therapies should also be tailored to the following strengths and weaknesses: i) molecular mechanisms and clinical correlates of cardiotoxicity have been characterized to some extent for anthracyclines but not for other chemotherapeutics or new generation "targeted" drugs, ii) anthracyclines and targeted drugs cause different mechanisms of cardiotoxicity (type I versus type II), and this classification should guide strategies of primary or secondary prevention, iii) with anthracyclines and nonanthracycline chemotherapeutics, cardiovascular events may occur on treatment as well as years or decades after completing chemotherapy, iv) some patients may be predisposed to a higher risk of cardiac events but there is a lack of prospective studies that characterized optimal genetic tests and pharmacologic measures to minimize excess risk, v) clinical toxicity may be preceded by asymptomatic systolic and/or diastolic dysfunction that necessitates innovative mechanism-based pharmacologic treatment, and vi) patient-tailored pharmacologic correction of comorbidities is important for both primary and secondary prevention. Active collaboration of physicians with laboratory scientists is much needed for improving management of cardiovascular sequelae of antitumor therapy. This article is part of a Special Issue entitled: Membrane channels and transporters in cancers.


Subject(s)
Anti-Arrhythmia Agents/therapeutic use , Cardiotonic Agents/therapeutic use , Cardiovascular Diseases/drug therapy , Disease Management , Neoplasms/drug therapy , Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Cardiotoxins/therapeutic use , Cardiovascular Diseases/complications , Cardiovascular Diseases/pathology , Drug Administration Schedule , Enzyme Inhibitors/therapeutic use , Humans , Neoplasms/complications , Neoplasms/pathology , Risk Assessment
15.
Breast Cancer Res Treat ; 143(2): 219-26, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24337598

ABSTRACT

Asymptomatic cardiotoxicity following breast cancer treatment is a significant issue for many patients, as these patients typically face an increased risk of cardiovascular disease (CVD). Exercise has well established benefits to improve and maintain cardiovascular function across patients with and without CVD. However, there is a dearth of information on the effects of exercise on cardiovascular outcomes in breast cancer patients. While pre-clinical studies support the use of exercise in mitigating cardiotoxicity, only one human study has specifically investigated cardiac function following an exercise intervention during chemotherapy treatment. No significant differences were observed between groups, which highlights the unidentified role of exercise in altering the risk of cardiotoxicity in breast cancer patients. Issues such as establishing the optimal timing, type, and intensity of an exercise program before, during, or after oncologic treatment for breast cancer are unclear. CVD risk and incidence increase in breast cancer survivors post therapy, and CVD is the number one killer of women in the United States. Thus, there is an increasing need to define the efficacy of exercise as a non-pharmacologic intervention in this growing population.


Subject(s)
Antineoplastic Agents/adverse effects , Breast Neoplasms/drug therapy , Cardiotoxins/therapeutic use , Cardiovascular Diseases/prevention & control , Exercise Therapy , Animals , Anthracyclines/adverse effects , Anthracyclines/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Agents/therapeutic use , Cardiovascular Diseases/chemically induced , Cardiovascular Diseases/therapy , Exercise/physiology , Female , Humans , Quality of Life , Rats , Trastuzumab
16.
Cancer Chemother Pharmacol ; 72(1): 241-9, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23719719

ABSTRACT

PURPOSE: Brentuximab vedotin (ADCETRIS®), an antibody-drug conjugate, comprises an anti-CD30 antibody conjugated by a protease-cleavable linker to a microtubule-disrupting agent, monomethyl auristatin E (MMAE). In vitro studies showed that MMAE does not interfere with hERG K+ channels at clinically relevant concentrations. In pivotal phase 2 clinical trials in patients with relapsed or refractory Hodgkin lymphoma and systemic anaplastic large cell lymphoma, brentuximab vedotin has shown substantial efficacy and an acceptable safety profile. This phase 1 open-label study was designed to evaluate the effect of brentuximab vedotin on the duration of cardiac ventricular repolarization. METHODS: Patients with CD30-positive hematologic malignancies were treated with 1.8 mg/kg brentuximab vedotin by intravenous infusion every 3 weeks for up to 16 cycles. The primary endpoint was the change from baseline to Cycle 1 Days 2, 3, and 4 in the duration of ventricular repolarization using Fridericia's corrected QT interval (QTcF). RESULTS: There was no clinically meaningful change from baseline in the duration of ventricular repolarization as measured by QTcF in the 46 evaluable patients out of 52 total patients treated with brentuximab vedotin. There was no evidence of treatment-emergent cardiac safety abnormalities. Brentuximab vedotin was generally well tolerated with a response rate and an adverse event profile consistent with prior studies. CONCLUSION: There is no significant prolongation of the QT/QTc interval with brentuximab vedotin in patients with CD30-positive hematologic malignancies.


Subject(s)
Antineoplastic Agents/adverse effects , Heart/drug effects , Hematologic Neoplasms/drug therapy , Immunoconjugates/adverse effects , Ki-1 Antigen/metabolism , Long QT Syndrome/chemically induced , Adult , Aged , Antineoplastic Agents/blood , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Brentuximab Vedotin , Cardiotoxins/adverse effects , Cardiotoxins/therapeutic use , Drug Hypersensitivity/immunology , Electrocardiography, Ambulatory/drug effects , Female , Heart/physiopathology , Heart Ventricles/drug effects , Heart Ventricles/physiopathology , Hematologic Neoplasms/blood , Hematologic Neoplasms/metabolism , Hematologic Neoplasms/physiopathology , Hodgkin Disease/blood , Hodgkin Disease/drug therapy , Hodgkin Disease/metabolism , Hodgkin Disease/physiopathology , Humans , Immunoconjugates/blood , Immunoconjugates/pharmacokinetics , Immunoconjugates/therapeutic use , Ki-1 Antigen/blood , Long QT Syndrome/physiopathology , Male , Middle Aged , Peripheral Nervous System Diseases/chemically induced , Peripheral Nervous System Diseases/physiopathology , Severity of Illness Index , Young Adult
17.
J Pharm Sci ; 102(5): 1626-35, 2013 May.
Article in English | MEDLINE | ID: mdl-23423631

ABSTRACT

Doxorubicin is an antitumor drug commonly used against a wide spectrum of tumors. However, the clinical application of DOX is restricted by its cardiotoxicity. To reduce the cardiotoxicity, we develop an albumin-based nanocarrier via a new molecular switch method for DOX delivery. Spherically shaped DOX-loaded HSA nanoparticles (NPs-DOX) are prepared with a drug-loading capacity and particle size of 4.3% and 120.1 ± 26 nm, respectively. In vivo studies demonstrate that NPs-DOX is able to preferentially accumulate in tumor and show great tumor inhibition on H22 hepatocellular-carcinoma-bearing mice. As for the toxicity, compared with free DOX, the maximum tolerated dose of NPs-DOX is increased from 10 to over 30 mg/kg, indicating the reduced systematic toxicity. More importantly, the cardiotoxicity induced by NPs-DOX is also significantly reduced because both left ventricular ejection fraction and left ventricular fractional shortening are almost not changed and other cardiotoxicity markers such as serum creatine kinase-MB, lactate dehydrogenase, superoxide dismutase, and malonaldehyde are kept constant. The reduced cardiotoxicity of NPs-DOX is also confirmed by nonhistological changes in the heart tissue. Therefore, such albumin-based nanocarrier can be one of the most promising strategies for the delivery of DOX.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Doxorubicin/administration & dosage , Doxorubicin/adverse effects , Drug Delivery Systems , Serum Albumin/chemistry , Animals , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Cardiotoxins/administration & dosage , Cardiotoxins/adverse effects , Cardiotoxins/pharmacokinetics , Cardiotoxins/therapeutic use , Cell Line, Tumor , Doxorubicin/pharmacokinetics , Doxorubicin/therapeutic use , Drug Carriers/chemistry , Echocardiography/drug effects , Heart/drug effects , Humans , Liver Neoplasms/drug therapy , Male , Mice , Myocardium/pathology , Nanoparticles/chemistry , Particle Size
18.
J Am Coll Cardiol ; 61(3): 267-74, 2013 Jan 22.
Article in English | MEDLINE | ID: mdl-23328609

ABSTRACT

Cancer genomics has focused on the discovery of mutations and chromosomal structural rearrangements that either increase susceptibility to cancer or support the cancer phenotype. Protein kinases are the most frequently mutated genes in the cancer genome, making them attractive therapeutic targets for drug design. However, the use of some of the kinase inhibitors (KIs) has been associated with toxicities to the heart and vasculature, including acute coronary syndromes and heart failure. Herein we discuss the genetic basis of cancer, focusing on mutations in the kinase genome (kinome) that lead to tumorigenesis. This will allow an understanding of the real and potential power of modern cancer therapeutics. The underlying mechanisms that drive the cardiotoxicity of the KIs are also examined. The preclinical models for predicting cardiotoxicity, including induced pluripotent stem cells and zebrafish, are reviewed, with the hope of eventually being able to identify problematic agents before their use in patients. Finally, the use of biomarkers in the clinic is discussed, and newer strategies (i.e., metabolomics and enhanced imaging strategies) that may allow earlier and more accurate detection of cardiotoxicity are reviewed.


Subject(s)
Antineoplastic Agents/adverse effects , Cardiotoxins/therapeutic use , Heart/drug effects , Neoplasms/drug therapy , Neoplasms/genetics , Protein Kinase Inhibitors/adverse effects , Animals , Antineoplastic Agents/pharmacology , Cardiotoxins/adverse effects , DNA Mutational Analysis , DNA Repair , Drug Evaluation, Preclinical/methods , Genome, Human , Humans , Metabolomics , Neoplasms/physiopathology , Practice Guidelines as Topic , Protein Kinase Inhibitors/therapeutic use , Protein Kinases/drug effects , Signal Transduction/genetics
19.
Transplantation ; 95(3): 434-41, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23296151

ABSTRACT

BACKGROUND: Th17 responses have been suggested to participate in the pathogenesis of acute allograft rejection. RORγt is the master transcription factor that controls Th17 cell differentiation and expansion. However, little is known about the effect that antagonizing RORγt activity may have on acute cardiac allograft rejection. METHODS: A model of heterotopic murine cardiac transplantation with total allomismatch (BALB/c to B6 mice) was used. Digoxin, which was recently identified as a specific antagonist of RORγt, was injected intraperitoneally daily (40 µg) starting 1 day after cardiac transplantation. The severity of rejection was determined by histology. The mRNA expression levels of cytokines and transcription factors in the grafts were measured by quantitative real-time PCR. The proportion and number of T-cell subpopulations in the allografts and spleens were analyzed by flow cytometry. In vitro, the effect of digoxin on allogeneic responses and the interleukin (IL)-6-mediated conversion of regulatory T cells (Treg) into Th17 cells were investigated. RESULTS: Treatment with digoxin significantly prolonged cardiac allograft survival compared with dimethyl sulfoxide treatment (mean survival time, 16.5±2.2 versus 8.1±0.7 days; P<0.01). Treatment with digoxin also markedly suppressed the mRNA expression levels of IL-17A, IL-17F, and granulocyte-macrophage colony-stimulating factor, reduced the number of Th17 cells, and induced Treg expansion in the allografts. In vitro, treatment with digoxin did not inhibit the proliferation of T cells in a mixed lymphocyte reaction, but it did inhibit the IL-6-mediated conversion of Tregs into Th17 cells. CONCLUSIONS: RORγt may be a promising therapeutic target to attenuate acute cardiac allograft rejection. Digoxin therefore provides a molecular basis for the design of novel immunosuppressive agents.


Subject(s)
Digoxin/pharmacology , Digoxin/therapeutic use , Graft Rejection/prevention & control , Heart Transplantation , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Nuclear Receptor Subfamily 1, Group F, Member 3/drug effects , Animals , Cardiotoxins/pharmacology , Cardiotoxins/therapeutic use , Cell Differentiation/drug effects , Dimethyl Sulfoxide/pharmacology , Dimethyl Sulfoxide/therapeutic use , Graft Rejection/pathology , Heart Transplantation/pathology , In Vitro Techniques , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Models, Animal , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/pathology , Th17 Cells/drug effects , Th17 Cells/pathology , Transplantation, Heterotopic , Transplantation, Homologous
20.
Ann Cardiol Angeiol (Paris) ; 62(1): 28-33, 2013 Feb.
Article in French | MEDLINE | ID: mdl-22513244

ABSTRACT

OBJECTIVE: Evaluate the early anthracyclines cardiotoxicity. METHODS: It is a prospective study made on 10 months of period from October 2008 to July 2009 and on patients who contracted a solid canny tumor hospitalized or followed in their movement and who would receive chemotherapy with an anthracycline molecule. On this effect, we have used tissue Doppler of mitral ring to detect clinic infratoxicity. RESULT: Forty-five patients (43 women and 2 men) who contracted the solid cancers were included in the study. The patients were 48 of age in average ± 10.12. All our patients did not show any cardiovascular symptoms at the time of the study. Cardiothoracic and electrocardiograms were not significantly modified by the chemotherapy. The cardioecography with the use of tissue Doppler revealed as followed: (a) significant low of the ejection fraction and the pick of systolic myocardia wave (Sa) on four patients (8.8%). These concerned patients were considered as having anthracycline cardio toxicity. The factor causing this cardiotoxicity was the nature of the anthracycline, which was used: the doxorubicin. The quantity accumulated threshold of the doxorubicin that shod (where toxicity appeared was 150 mg/m(2)); (b) a low of Sa pick without that of left ventricular fraction ejection observed on five patients (11.11%). These concerned patients were considered as having potential risks to develop anthracyclines cardiotoxicity; (c) the left ventricular ejection fraction was not a good indicator the check up of the patients under chemotherapy made up with anthracyclines. CONCLUSION: The tissue Doppler not only enables to make diagnostics of early myocardia dysfunctions but it mainly allows to identify people with risks of a cardiotoxicity due to a going on chemotherapy.


Subject(s)
Anthracyclines/toxicity , Antineoplastic Agents/toxicity , Cardiomyopathies/chemically induced , Cardiomyopathies/diagnostic imaging , Cardiotoxins/toxicity , Developing Countries , Echocardiography, Doppler/methods , Image Interpretation, Computer-Assisted/methods , Neoplasms/drug therapy , Adult , Aged , Anthracyclines/therapeutic use , Antineoplastic Agents/therapeutic use , Cardiotoxins/therapeutic use , Dose-Response Relationship, Drug , Doxorubicin/therapeutic use , Doxorubicin/toxicity , Epirubicin/therapeutic use , Epirubicin/toxicity , Female , Humans , Male , Middle Aged , Mitral Valve/diagnostic imaging , Mitral Valve/drug effects , Stroke Volume/drug effects , Systole/drug effects , Togo , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/diagnostic imaging
SELECTION OF CITATIONS
SEARCH DETAIL
...