Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Biomolecules ; 14(2)2024 Feb 07.
Artigo em Inglês | MEDLINE | ID: mdl-38397436

RESUMO

Serum biomarkers represent a reproducible, sensitive, minimally invasive and inexpensive method to explore possible adverse cardiovascular effects of antineoplastic treatments. They are useful tools in risk stratification, the early detection of cardiotoxicity and the follow-up and prognostic assessment of cancer patients. In this literature review, we aim at describing the current state of knowledge on the meaning and the usefulness of cardiovascular biomarkers in patients with cancer; analyzing the intricate relationship between cancer and cardiovascular disease (especially HF) and how this affects cardiovascular and tumor biomarkers; exploring the role of cardiovascular biomarkers in the risk stratification and in the identification of chemotherapy-induced cardiotoxicity; and providing a summary of the novel potential biomarkers in this clinical setting.


Assuntos
Antineoplásicos , Neoplasias , Humanos , Cardiotoxicidade/etiologia , Cardiotoxicidade/diagnóstico , Cardio-Oncologia , Antineoplásicos/efeitos adversos , Neoplasias/complicações , Neoplasias/tratamento farmacológico , Neoplasias/induzido quimicamente , Biomarcadores , Biomarcadores Tumorais
2.
Curr Probl Cardiol ; 49(3): 102229, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38154703

RESUMO

In recent years, important advances have been made in the field of Cardio-Oncology. The 2022 ESC Guidelines on Cardio-Oncology proposed a baseline cardiovascular risk stratification for cancer patients and preventive strategies in patients at high and very-high risk of cardiotoxicity. Cardiovascular toxic effects of anti-cancer drugs are being extensively studied; surveillance programs have been proposed, based on the baseline cardiovascular risk. On the other hand, there is little data on Cardio-Oncological management of patients at high and very-high cardiovascular risk with previous cardiovascular diseases. For example, little is known about management of cancer patients with heart failure with reduced ejection fraction (HFrEF), patients with a recent myocardial infarction or other cardiovascular diseases; when to resume anti-cancer drugs after a cardiovascular toxic event. Collaboration between Cardiologists and Oncologists and multidisciplinary team evaluations are certainly essential to decide the best therapeutic strategy for cancer patients, to treat cancer while saving the heart. Therefore, in the present review, we attempt to provide a useful guide to clinicians in treating patients with high and very-high risk of cardiotoxicity by enucleating main questions and answering them based on the evidence available as well as expert opinion and our clinical experience.


Assuntos
Antineoplásicos , Insuficiência Cardíaca , Neoplasias , Disfunção Ventricular Esquerda , Humanos , Insuficiência Cardíaca/terapia , Insuficiência Cardíaca/tratamento farmacológico , Cardiotoxicidade/etiologia , Cardiotoxicidade/prevenção & controle , Volume Sistólico , Neoplasias/tratamento farmacológico , Neoplasias/induzido quimicamente , Antineoplásicos/efeitos adversos , Disfunção Ventricular Esquerda/induzido quimicamente
3.
G Ital Cardiol (Rome) ; 23(7): 516-522, 2022 Jul.
Artigo em Italiano | MEDLINE | ID: mdl-35771017

RESUMO

BACKGROUND: In the last decades the population undergoing non-cardiac surgery has become more numerous and complex. Cardiovascular perioperative complications represent at least one third of the perioperative deaths. Despite the pivotal role of the cardiologist in the perioperative management, current guidelines are often hardly useful in different settings of clinical practice. Local clinical protocols contribute to fill these gaps, to define the role of each specialist in the perioperative context and to achieve the best medical outcome. METHODS: This single-center retrospective study analyzes the background of 33 463 preoperatory cardiologic visits, the adherence to scientific evidence in our institute and the impact of the implementation of a shared clinical protocol (CP) in terms of reduction of inappropriate requests of cardiological evaluations. RESULTS: Among all the patients, the mean age was 59 ± 18 years, 52.8% were male. Hypertension was the most prevalent disease followed by diabetes, chronic coronary syndrome and atrial fibrillation. The "low-risk surgery" category was the most represented (56.2%) and the vast majority of patients (70.1%) was totally free from predictors of perioperative cardiovascular events. After the introduction of the CP, the number of inappropriate evaluations decreased by 32%, mainly in the low-risk category. However, despite the overall reduction, almost two thirds of the evaluations were still deemed inappropriate according to the CP, mostly (82.9%) in the low-risk category and to a lesser degree (55%) in the moderate/high-risk category. CONCLUSIONS: The inappropriate use of the resources resulted in disappointing organizational performance, poor assistance quality and a huge number of inappropriate preoperatory evaluations. The implementation of a CP, developed on the basis of the local needs, is a useful tool to enhance the organizational standards for the cardiological evaluation of patients undergoing non-cardiac surgery. Regular verifications, a widespread knowledge of the guidelines and a more efficient system of management and surveillance may improve the appropriateness of these evaluations.


Assuntos
Fibrilação Atrial , Cardiologia , Cardiopatias , Adulto , Idoso , Feminino , Cardiopatias/cirurgia , Humanos , Masculino , Pessoa de Meia-Idade , Cuidados Pré-Operatórios , Estudos Retrospectivos , Medição de Risco
4.
J Clin Med ; 11(6)2022 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-35330019

RESUMO

A multicenter, cross-sectional observational study (Italian GENder Differences in Awareness of Cardiovascular risk, IGENDA study) was carried out to evaluate the perception and knowledge of cardiovascular risk among Italian women. An anonymous questionnaire was completed by 4454 women (44.3 ± 14.1 years). The 70% of respondents correctly identified cardiovascular disease (CVD) as the leading cause of death. More than half of respondents quoted cancer as the greatest current and future health problem of women of same age. Sixty percent of interviewed women considered CVD as an almost exclusively male condition. Although respondents showed a good knowledge of the major cardiovascular risk factors, the presence of cardiovascular risk factors was not associated with higher odds of identifying CVD as the biggest cause of death. Less than 10% of respondents perceived themselves as being at high CVD risk, and the increased CVD risk perception was associated with ageing, higher frequency of cardiovascular risk factors and disease, and a poorer self-rated health status. The findings of this study highlight the low perception of cardiovascular risk in Italian women and suggest an urgent need to enhance knowledge and perception of CVD risk in women as a real health problem and not just as a as a life-threatening threat.

5.
Metabolites ; 12(2)2022 01 27.
Artigo em Inglês | MEDLINE | ID: mdl-35208197

RESUMO

Traditional cardiovascular (CV) risk factors (RFs) and coronary artery disease (CAD) do not always show a direct correlation. We investigated the metabolic differences in a cohort of patients with a high CV risk profile who developed, or did not develop, among those enrolled in the Coronary Atherosclerosis in Outlier Subjects: Protective and Novel Individual Risk Factors Evaluation (CAPIRE) study. We studied 112 subjects with a high CV risk profile, subdividing them according to the presence (CAD/High-RFs) or absence of CAD (No-CAD/High-RFs), assessed by computed tomography angiography. The metabolic differences between the two groups were identified by gas chromatography-mass spectrometry. Characteristic patterns and specific metabolites emerged for each of the two phenotypic groups: high concentrations of pyruvic acid, pipecolic acid, p-cresol, 3-aminoisobutyric acid, isoleucine, glyceric acid, lactic acid, sucrose, phosphoric acid, trimethylamine-N-oxide, 3-hydroxy-3-methylglutaric acid, erythritol, 3-hydroxybutyric acid, glucose, leucine, and glutamic acid; and low concentrations of cholesterol, hypoxanthine, glycerol-3-P, and cysteine in the CAD/High-RFs group vs the No-CAD/High-RFs group. Our results show the existence of different metabolic profiles between patients who develop CAD and those who do not, despite comparable high CV risk profiles. A specific cluster of metabolites, rather than a single marker, appears to be able to identify novel predisposing or protective mechanisms towards CAD beyond classic CVRFs.

7.
Eur Heart J Cardiovasc Imaging ; 22(4): 406-415, 2021 03 22.
Artigo em Inglês | MEDLINE | ID: mdl-33432333

RESUMO

AIMS: In breast cancer (BC) patients treated with anthracyclines-based therapies, we aim at assessing whether adjuvant drugs impact cardiac function differently and whether their cardiotoxicity has a regional pattern. METHODS AND RESULTS: In a multicentre study, 146 BC patients (56 ± 11 years) were prospectively enrolled and divided into three groups according to the received treatments: AC/EC-Group (doxorubicin or epirubicin + cyclophosphamide), AC/EC/Tax-Group (AC/EC + taxanes), FEC/Tax-Group (fluorouracil + EC + taxanes). Fifty-six patients of the total cohort also received trastuzumab. Left ventricular ejection fraction (LVEF) and global longitudinal strain (GLS) were calculated before starting chemotherapy (T0), at 3 months (T3), at 6 (T6), and 12 months (T12). A ≥10% drop of EF, while remaining within the normal range, was reached at T6 in 25.3% of patients from the whole cohort with an early decrease only in FEC/Tax-Group (P = 0.04). A ≥15% GLS reduction was observed in many more (61.6%) patients. GLS decreased early both in the whole population (P < 0.001) and in the subgroups. The FEC-Tax Group showed the worst GLS at T6. Trastuzumab further worsened GLS at T12 (P = 0.031). A significant reduction of GLS was observed in all LV segments and was more relevant in the anterior septum and apex. CONCLUSIONS: The decrease of GLS is more precocious and pronounced in BC patients who received FEC + taxanes. Cardiac function further worsens after 6 months of adjuvant trastuzumab. All LV segments are damaged, with the anterior septum and the apex showing the greatest impairments.


Assuntos
Neoplasias da Mama , Cardiologia , Preparações Farmacêuticas , Disfunção Ventricular Esquerda , Antraciclinas/efeitos adversos , Neoplasias da Mama/tratamento farmacológico , Cardiotoxicidade/diagnóstico por imagem , Ecocardiografia , Feminino , Humanos , Itália , Volume Sistólico , Trastuzumab/efeitos adversos , Disfunção Ventricular Esquerda/induzido quimicamente , Disfunção Ventricular Esquerda/diagnóstico por imagem , Função Ventricular Esquerda
8.
Antioxid Redox Signal ; 34(7): 572-588, 2021 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-32151144

RESUMO

Significance: Chemotherapy-induced cardiotoxicity (CTX) has been associated with redox signaling imbalance. In fact, redox reactions are crucial for normal heart physiology, whereas excessive oxidative stress can cause cardiomyocyte structural damage. Recent Advances: An antioxidant approach as a cardioprotective strategy in this setting has shown encouraging results in preventing anticancer drug-induced CTX. Critical Issues: In fact, traditional heart failure drugs as well as many other compounds and nonpharmacological strategies, with a partial effect in reducing oxidative stress, have been shown to counterbalance chemotherapy-induced CTX in this setting to some extent. Future Directions: Given the various pathways of toxicity involved in different chemotherapeutic schemes, interactions with redox balance need to be fine-tuned and a personalized cardioprotective approach seems to be required.


Assuntos
Antioxidantes/farmacologia , Cardiotônicos/farmacologia , Cardiopatias/tratamento farmacológico , Insuficiência Cardíaca/tratamento farmacológico , Antineoplásicos/efeitos adversos , Cardiopatias/induzido quimicamente , Cardiopatias/metabolismo , Insuficiência Cardíaca/induzido quimicamente , Insuficiência Cardíaca/metabolismo , Humanos , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo
9.
Antioxidants (Basel) ; 9(7)2020 Jul 21.
Artigo em Inglês | MEDLINE | ID: mdl-32708201

RESUMO

Metabolic syndrome (MetS) is a well established risk factor for cardiovascular (CV) diseases. In addition, several studies indicate that MetS correlates with the increased risk of cancer in adults. The mechanisms linking MetS and cancer are not fully understood. Several risk factors involved in MetS are also cancer risk factors, such as the consumption of high calorie-food or high fat intake, low fibre intake, and sedentary lifestyle. Other common aspects of both cancer and MetS are oxidative stress and inflammation. In addition, some anticancer treatments can induce cardiotoxicity, including, for instance, left ventricular (LV) dysfunction and heart failure (HF), endothelial dysfunction and hypertension. In this review, we analyse several aspects of MetS, cancer and cardiotoxicity from anticancer drugs. In particular, we focus on oxidative stress in ageing, cancer and CV diseases, and we analyse the connections among CV risk factors, cancer and cardiotoxicity from anticancer drugs.

10.
J Cardiovasc Echogr ; 30(Suppl 1): S11-S16, 2020 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-32566461

RESUMO

Cardiovascular complications during chemotherapy and radiotherapy are becoming an increasing problem because many patients with cancer are treated with agents that exert significant vascular toxicity. Coronary heart disease in patients with cancer presents particular challenges, which directly impact the management of both the coronary disease and malignancy. Several chemotherapeutic agents have been shown to trigger ischemic heart disease, and as it has happened for myocardial cardiotoxicity, more attention should be dedicated to improving early recognition and prevention of cardiac vascular toxicity. Cardiac imaging could facilitate early detection of vascular toxicity, but a thorough risk stratification should always be performed to identify patients at higher risk of vascular impairment.

11.
J Clin Med ; 9(4)2020 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-32326001

RESUMO

BACKGROUND: Cardiovascular adverse events (CV-AEs) are considered critical complications in chronic myeloid leukemia (CML) patients treated with second- and third-generation tyrosine kinase inhibitors (TKIs). The aim of our study was to assess the correlation between metabolic profiles and CV-AEs in CML patients treated with TKIs. METHODS: We investigated 39 adult CML patients in chronic-phase (mean age 49 years, range 24-70 years), with no comorbidities evidenced at baseline, who were consecutively identified with CML and treated with imatinib, nilotinib, dasatinib, and ponatinib. All patients performed Gas-Chromatography-Mass-Spectrometry-based metabolomic analysis and were divided into two groups (with and without CV-AEs). RESULTS: Ten CV-AEs were documented. Seven CV-AEs were rated as 3 according to the Common Toxicity Criteria, and one patient died of a dissecting aneurysm of the aorta. The patients' samples were clearly separated into two groups after analysis and the main discriminant metabolites were tyrosine, lysine, glutamic acid, ornithine, 2-piperdinecarboxylic acid, citric acid, proline, phenylalanine, threonine, mannitol, leucine, serine, creatine, alanine, and 4-hydroxyproline, which were more abundant in the CV-AE group. Conversely, myristic acid, oxalic acid, arabitol, 4-deoxy rithronic acid, ribose, and elaidic acid were less represented in the CV-AE group. CONCLUSIONS: CML patients with CV-AEs show a different metabolic profile, suggesting probable mechanisms of endothelial damage.

12.
J Clin Med ; 9(3)2020 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-32197359

RESUMO

Ponatinib, a third-generation tyrosine kinase inhibitor (TKI), is the only approved TKI that is effective against T315I mutations in patients with chronic myeloid leukemia (CML). Specific activation of Notch signaling in CML cells by ponatinib can be considered as the "on-target effect" on the tumor and represents a therapeutic approach for CML. Nevertheless, ponatinib-induced vascular toxicity remains a serious concern, with underlying mechanisms being poorly understood. We aimed to determine the mechanisms of ponatinib-induced vascular toxicity, defining associated signaling pathways and identifying potential rescue strategies. We exposed human umbilical endothelial cells (HUVECs) to ponatinib or vehicle in the presence or absence of the neutralizing factor anti-Notch-1 antibody for exposure times of 0-72 h. Label-free proteomics and network analysis showed that protein cargo of HUVECs treated with ponatinib triggered apoptosis and inhibited vasculature development. We validated the proteomic data showing the inhibition of matrigel tube formation, an up-regulation of cleaved caspase-3 and a downregulation of phosphorylated AKT and phosphorylated eNOS. We delineated the signaling of ponatinib-induced vascular toxicity, demonstrating that ponatinib inhibits endothelial survival, reduces angiogenesis and induces endothelial senescence and apoptosis via the Notch-1 pathway. Ponatinib induced endothelial toxicity in vitro. Hyperactivation of Notch-1 in the vessels can lead to abnormal vascular development and vascular dysfunction. By hyperactivating Notch-1 in the vessels, ponatinib exerts an "on-target off tumor effect", which leads to deleterious effects and may explain the drug's vasculotoxicity. Selective blockade of Notch-1 prevented ponatinib-induced vascular toxicity.

13.
Int J Mol Sci ; 20(19)2019 Oct 04.
Artigo em Inglês | MEDLINE | ID: mdl-31590338

RESUMO

Despite advances in supportive and protective therapy for myocardial function, cardiovascular diseases due to antineoplastic therapy-primarily cardiomyopathy associated with contractile dysfunction-remain a major cause of morbidity and mortality. Because of the limitations associated with current therapies, investigators are searching for alternative strategies that can timely recognise cardiovascular damage-thus permitting a quick therapeutic approach-or prevent the development of the disease. Damage to the heart can result from both traditional chemotherapeutic agents, such as anthracyclines, and new targeted therapies, such as tyrosine kinase inhibitors. In recent years, metabolomics has proved to be a practical tool to highlight fundamental changes in the metabolic state in several pathological conditions. In this article, we present the state-of-the-art technology with regard to the metabolic mechanisms underlying cardiotoxicity and cardioprotection.


Assuntos
Antineoplásicos/toxicidade , Insuficiência Cardíaca/induzido quimicamente , Metaboloma , Metabolômica/métodos , Animais , Cardiotônicos/uso terapêutico , Cardiotoxicidade , Descoberta de Drogas/métodos , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Humanos
14.
J Cardiovasc Med (Hagerstown) ; 19(7): 315-323, 2018 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-29708912

RESUMO

: Chemotherapy-induced cardiotoxicity (CTX) remains a determining factor for the quality of life and mortality of patients treated with potentially cardiotoxic drugs. Considerable advances have been made in this field with increase in awareness regarding chemotherapy-induced CTX, which has changed the treatment approach to include cardiovascular risk among the first factors to be evaluated before therapy. Moreover, a better understanding of the pathophysiology of chemotherapy-induced CTX has also facilitated early identification of patients at risk with the help of new imaging technologies. The newly developed imaging tools in cardio-oncology have led to the introduction of novel parameters for evaluation of myocardial function. This, together with a renewed standardization of measurements, has increased the adherence to monitoring protocols. With respect to treatment and prevention, researchers have started focusing attention on the development of new strategies as well as new cardioprotective agents that will play a crucial role in the prevention of CTX in the near future.


Assuntos
Antraciclinas/efeitos adversos , Antineoplásicos/efeitos adversos , Cardiotoxicidade/prevenção & controle , Doenças Cardiovasculares/induzido quimicamente , Doenças Cardiovasculares/prevenção & controle , Cardiotônicos/uso terapêutico , Cardiotoxicidade/fisiopatologia , Monitoramento de Medicamentos , Ecocardiografia , Eletrocardiografia , Humanos , Neoplasias/tratamento farmacológico , Qualidade de Vida , Ensaios Clínicos Controlados Aleatórios como Assunto , Volume Sistólico
15.
Med Res Rev ; 38(5): 1447-1468, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29283446

RESUMO

Cancer immunotherapy has become a well-established treatment option for some cancers after the development of a family of drugs targeting the so-called immune checkpoints, such as CTLA4 and PD-1 with PD-L1. These co-receptors/ligands inhibit the activation of T-cell, thus preventing an excessive inflammatory response. Tumors exploit these pathways to induce immune tolerance to themselves. Thus, the main effect of checkpoint-blocking drugs is to awake an immune response primarily directed against cancer cells. Nonetheless, as the immune response elicited by these drugs is not completely tumor-specific, their use may actually cause several adverse effects, including adverse cardiovascular effects. In this review, we will discuss the principles and potentiality of immunotherapy for cancer treatment, the experimental and clinical data on the role of CTLA4 and PD-1 with PD-L1 as immune-checkpoints in the cancer environment and in the cardiovascular system, and strategies aimed at preventing possible cardiovascular adverse effects of immune-checkpoint blockers.


Assuntos
Doenças Cardiovasculares/etiologia , Imunoterapia/efeitos adversos , Neoplasias/terapia , Animais , Humanos , Fatores de Risco , Pesquisa Translacional Biomédica
16.
Curr Drug Targets ; 18(6): 719-723, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-27033187

RESUMO

Novel antineoplastic therapies have greatly improved cancer survival; nevertheless they are bringing in new forms of cardiomyopathy, that can often limit proper cancer treatments. Novel cardioprotective therapies are therefore needed, for improving clinical outcomes in cancer patients. In order to test novel therapeutic strategies, there is an increasing need for appropriate experimental models of chemotherapy-induced cardiomyopathy. Induced pluripotent stem (iPS) cell- and human embryonic stem cell (hESC )-derived cardiomyocytes may be used as alternative in vitro models for studying mechanisms that underly chemotherapy-induced cardiomyopathy. In this review we discuss the use of iPS- and hESC-derived cardiomyocytes for evaluating additional pharmacological targets and for predicting chemotherapy-induced cardiotoxicity.


Assuntos
Cardiotoxicidade , Modelos Biológicos , Miócitos Cardíacos/efeitos dos fármacos , Células-Tronco Pluripotentes/citologia , Antineoplásicos/efeitos adversos , Células Cultivadas , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Técnicas In Vitro , Miócitos Cardíacos/citologia , Células-Tronco Pluripotentes/efeitos dos fármacos
17.
J Cardiovasc Med (Hagerstown) ; 17 Suppl 1 Special issue on Cardiotoxicity from Antiblastic Drugs and Cardioprotection: e12-e18, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27755238

RESUMO

The risk and mechanism of chemotherapy-induced cardiotoxicity (CTX) vary depending on the type and intensity of the anticancer regimen. Myriad chemotherapeutic drugs produce adverse cardiovascular effects such as arterial hypertension, heart failure, and thromboembolic events. Among the numerous classes of these drugs, anthracyclines have been studied most extensively because of their overt cardiovascular effects and the high associated incidence of heart failure. However, CTX might also be caused by other types of chemotherapeutic agents, including alkylating agents (cyclophosphamide, ifosfamide), platinum agents, antimetabolites (5-fluorouracil, capecitabine), antibiotics (mitoxantrone, mitomycin, bleomycin), and antimicrotubule agents (taxanes). Here, we review the incidence, clinical impact, and potential mechanisms of CTX associated with nonanthracycline chemotherapy used for cancer patients. The published data support a marked increase in CTX risk, particularly with certain drugs such as 5-fluorouracil and cisplatin. Each anticancer regimen is associated with distinct modes of heart damage, both symptomatic and asymptomatic. However, the underlying mechanisms of CTX have been established only in a few cases, and only few nonanthracycline chemotherapeutics (mitoxantrone, mitomycin, ifosfamide) act through a recognizable mechanism and show a predictable dose dependence. Lastly, nonanthracycline chemotherapy can induce both chronic lesions, such as systolic dysfunction, and acute lesions, such as the ischemia that occurs within hours or days after treatment. An increased understanding of the incidence, mechanisms, and potential therapeutic targets of CTX induced by various nonanthracycline chemotherapeutic agents is clearly required.


Assuntos
Antibacterianos/efeitos adversos , Antineoplásicos Alquilantes/efeitos adversos , Cardiotoxicidade/fisiopatologia , Neoplasias/tratamento farmacológico , Antraciclinas/efeitos adversos , Cardiotoxicidade/etiologia , Fluoruracila/efeitos adversos , Insuficiência Cardíaca/induzido quimicamente , Humanos , Estresse Oxidativo/efeitos dos fármacos , Platina/efeitos adversos , Volume Sistólico , Taxoides/efeitos adversos , Disfunção Ventricular Esquerda/induzido quimicamente
18.
J Cardiovasc Med (Hagerstown) ; 17 Suppl 1 Special issue on Cardiotoxicity from Antiblastic Drugs and Cardioprotection: e55-e63, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27755243

RESUMO

Notwithstanding the steady progress in survival rates of children and adolescents suffering from cancer, the benefits associated with chemotherapy do not come without risks involving multiple organs and systems, including the cardiovascular apparatus. Anthracyclines-often administered in combination with radiation therapy and/or surgery-are the most used chemotherapeutic compounds in order to treat tumours and blood malignancies even in paediatric age. Being an important side-effect of anthracyclines, carduitoxicity may limit their efficacy during the treatment and induce long-term sequelae, observed even many years after therapy completion. The purpose of this review was to perform an overview about all the possible strategies to prevent and/or limit the anthracyclines adverse side-effects for the cardiovascular system in childhood cancer survivors.


Assuntos
Antraciclinas/efeitos adversos , Antibióticos Antineoplásicos/efeitos adversos , Biomarcadores/sangue , Cardiotoxicidade/prevenção & controle , Insuficiência Cardíaca/induzido quimicamente , Neoplasias/tratamento farmacológico , Adolescente , Animais , Cardiotônicos/uso terapêutico , Cardiotoxicidade/diagnóstico por imagem , Criança , Ecocardiografia/métodos , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Ratos , Taxa de Sobrevida
19.
J Cardiovasc Med (Hagerstown) ; 17 Suppl 1 Special issue on Cardiotoxicity from Antiblastic Drugs and Cardioprotection: e76-e83, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27755245

RESUMO

Despite advances in supportive and protective therapy for myocardial function, heart failure caused by various clinical conditions, including cardiomyopathy due to antineoplastic therapy, remains a major cause of morbidity and mortality. Because of the limitations associated with current therapies, investigators have been searching for alternative treatments that can effectively repair the damaged heart and permanently restore its function. Damage to the heart can result from both traditional chemotherapeutic agents, such as anthracyclines, and new targeted therapies, such as trastuzumab. Because of this unresolved issue, investigators are searching for alternative therapeutic strategies. In this article, we present state-of-the-art technology with regard to the genomic and epigenetic mechanisms underlying cardiotoxicity and cardioprotection, the role of anticancer in influencing the redox (reduction/oxidation) balance and the function of stem cells in the repair/regeneration of the adult heart. These findings, although not immediately transferable to clinical applications, form the basis for the development of personalized medicine based on the prevention of cardiotoxicity with the use of genetic testing. Proteomics, metabolomics and investigations on reactive oxygen species-dependent pathways, particularly those that interact with the production of NO and energy metabolism, appear to be promising for the identification of early markers of cardiotoxicity and for the development of cardioprotective agents. Finally, autologous cardiac stem and progenitor cells may represent future contributions in the field of myocardial protection and recovery in the context of antiblastic therapy.


Assuntos
Antraciclinas/efeitos adversos , Antibióticos Antineoplásicos/efeitos adversos , Cardiomiopatias/terapia , Cardiotônicos/uso terapêutico , Cardiotoxicidade/fisiopatologia , Animais , Cardiomiopatias/induzido quimicamente , Cardiotoxicidade/genética , Coração/efeitos dos fármacos , Humanos , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Ratos , Espécies Reativas de Oxigênio/metabolismo , Transplante de Células-Tronco , Células-Tronco/efeitos dos fármacos
20.
J Cardiovasc Med (Hagerstown) ; 17 Suppl 1: S12-8, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27183520

RESUMO

The risk and mechanism of chemotherapy-induced cardiotoxicity (CTX) vary depending on the type and intensity of the anticancer regimen. Myriad chemotherapeutic drugs produce adverse cardiovascular effects such as arterial hypertension, heart failure, and thromboembolic events. Among the numerous classes of these drugs, anthracyclines have been studied most extensively because of their overt cardiovascular effects and the high associated incidence of heart failure. However, CTX might also be caused by other types of chemotherapeutic agents, including alkylating agents (cyclophosphamide, ifosfamide), platinum agents, antimetabolites (5-fluorouracil, capecitabine), antibiotics (mitoxantrone, mitomycin, bleomycin), and antimicrotubule agents (taxanes). Here, we review the incidence, clinical impact, and potential mechanisms of CTX associated with nonanthracycline chemotherapy used for cancer patients. The published data support a marked increase in CTX risk, particularly with certain drugs such as 5-fluorouracil and cisplatin. Each anticancer regimen is associated with distinct modes of heart damage, both symptomatic and asymptomatic. However, the underlying mechanisms of CTX have been established only in a few cases, and only few nonanthracycline chemotherapeutics (mitoxantrone, mitomycin, ifosfamide) act through a recognizable mechanism and show a predictable dose dependence. Lastly, nonanthracycline chemotherapy can induce both chronic lesions, such as systolic dysfunction, and acute lesions, such as the ischemia that occurs within hours or days after treatment. An increased understanding of the incidence, mechanisms, and potential therapeutic targets of CTX induced by various nonanthracycline chemotherapeutic agents is clearly required.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Doenças Cardiovasculares/induzido quimicamente , Coração/efeitos dos fármacos , Coração/fisiopatologia , Cardiotoxicidade/fisiopatologia , Doenças Cardiovasculares/prevenção & controle , Humanos , Neoplasias/tratamento farmacológico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA