Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.673
1.
Oncotarget ; 15: 355-359, 2024 Jun 03.
Article En | MEDLINE | ID: mdl-38829647

Ibrutinib was the first Bruton's tyrosine kinase (BTK) inhibitor approved for the treatment of patients with chronic lymphocytic leukemia (CLL). While producing durable responses and prolonging survival, roughly 20-25% of patients experience dose limiting side effects, mostly consisting of cardiovascular toxicities like severe hypertension and atrial fibrillation. While clinical predictors of BTK inhibitor-related cardiotoxicity have been proposed and may aid in risk stratification, there is no routine risk model used in clinical practice today to identify patients at highest risk. A recent study investigating genetic predictors of ibrutinib-related cardiotoxicity found that single nucleotide polymorphisms in KCNQ1 and GATA4 were significantly associated with cardiotoxic events. If replicated in larger studies, these biomarkers may improve risk stratification in combination with clinical factors. A clinicogenomic risk model may aid in identifying patients at highest risk of developing BTK inhibitor-related cardiotoxicity in which further risk mitigation strategies may be explored.


Agammaglobulinaemia Tyrosine Kinase , Cardiotoxicity , Leukemia, Lymphocytic, Chronic, B-Cell , Piperidines , Protein Kinase Inhibitors , Humans , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Agammaglobulinaemia Tyrosine Kinase/genetics , Protein Kinase Inhibitors/adverse effects , Cardiotoxicity/etiology , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Piperidines/adverse effects , Piperidines/therapeutic use , Adenine/analogs & derivatives , Adenine/adverse effects , Risk Assessment , Pyrimidines/adverse effects , Pyrazoles/adverse effects , Biomarkers , Polymorphism, Single Nucleotide , KCNQ1 Potassium Channel/genetics
3.
Life Sci ; 350: 122765, 2024 Aug 01.
Article En | MEDLINE | ID: mdl-38830506

BACKGROUND: Malnutrition is a common problem in developing countries, and the impact of severe malnutrition on optimal treatment outcomes of chemotherapy in pediatric cancer patients is well documented. However, despite being a more prevalent and distinct entity, moderate malnutrition is until now unexplored for its effects on treatment outcomes. AIMS: In this study we aimed to investigate the molecular basis of altered pharmacokinetics and cardiotoxicity of doxorubicin observed in early-life chronic moderate protein deficiency malnutrition. MATERIALS AND METHODS: We developed an animal model of early-life moderate protein-deficiency malnutrition and validated it using clinical samples. This model was used to study pharmacokinetic and toxicity changes and was further utilized to study the molecular changes in liver and heart to get mechanistic insights. KEY FINDINGS: Here we show that moderate protein-deficiency malnutrition in weanling rats causes changes in drug disposition in the liver by modification of hepatic ABCC3 and MRP2 transporters through the TNFα signalling axis. Furthermore, malnourished rats in repeat-dose doxorubicin toxicity study showed higher toxicity and mortality. A higher accumulation of doxorubicin in the heart was observed which was associated with alterations in cardiac metabolic pathways and increased cardiotoxicity. SIGNIFICANCE: Our findings indicate that moderate malnutrition causes increased susceptibility towards toxic side effects of chemotherapy. These results may necessitate further investigations and new guidelines on the dosing of chemotherapy in moderately malnourished pediatric cancer patients.


Cardiotoxicity , Doxorubicin , Animals , Doxorubicin/pharmacokinetics , Doxorubicin/adverse effects , Rats , Cardiotoxicity/etiology , Male , Weaning , Liver/metabolism , Protein-Energy Malnutrition/metabolism , Humans , Antibiotics, Antineoplastic/pharmacokinetics , Antibiotics, Antineoplastic/adverse effects , Antibiotics, Antineoplastic/toxicity , Female , Disease Models, Animal , Rats, Wistar
4.
Chem Res Toxicol ; 37(6): 1044-1052, 2024 Jun 17.
Article En | MEDLINE | ID: mdl-38833663

The aim of the present study was to evaluate the cardiotoxic effects of alcohol and its potential toxic mechanism on ferroptosis in mice and H9c2 cells. Mice were intragastrically treated with three different concentrations of alcohol, 7, 14, and 28%, each day for 14 days. Body weight and electrocardiography (ECG) were recorded over the 14 day period. Serum creatine kinase (CK), lactic dehydrogenase (LDH), MDA, tissue iron, and GSH levels were measured. Cardiac tissues were examined histologically, and ferroptosis was assessed. In H9c2 cardiomyocytes, cell viability, reactive oxygen species (ROS), labile iron pool (LIP), and mitochondrial membrane potential (MMP) were measured. The proteins of ferroptosis were evaluated by the western blot technique in vivo and in vitro. The results showed that serum CK, LDH, MDA, and tissue iron levels significantly increased in the alcohol treatment group in a dose-dependent manner. The content of GSH decreased after alcohol treatment. ECG and histological examinations showed that alcohol impaired cardiac function and structure. In addition, the levels of ROS and LIP increased, and MMP levels decreased after alcohol treatment. Ferrostatin-1 (Fer-1) protected cells from lipid peroxidation. Western blotting analysis showed that alcohol downregulated the expression of Nrf2, NQO1, HO-1, and GPX4. The expressions of P53 and TfR were upregulated in vivo and in vitro. Fer-1 significantly alleviated alcohol-induced ferroptosis. In conclusion, the study showed that Nrf2/NQO1-dependent ferroptosis played a vital role in the cardiotoxicity induced by alcohol.


Cardiotoxicity , Ethanol , Ferroptosis , NAD(P)H Dehydrogenase (Quinone) , NF-E2-Related Factor 2 , Animals , Ferroptosis/drug effects , NF-E2-Related Factor 2/metabolism , NAD(P)H Dehydrogenase (Quinone)/metabolism , Mice , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Male , Reactive Oxygen Species/metabolism , Rats , Mice, Inbred C57BL , Cell Survival/drug effects
5.
Rev Assoc Med Bras (1992) ; 70(suppl 1): e2024S106, 2024.
Article En | MEDLINE | ID: mdl-38865526

BACKGROUND: Chemotherapy with doxorubicin may lead to left ventricular dysfunction. There is a controversial recommendation that biomarkers can predict ventricular dysfunction, which is one of the most feared manifestations of anthracycline cardiotoxicity. OBJECTIVE: The aim of this study was to evaluate the behavior of biomarkers such as Troponin I, type B natriuretic peptide, creatine phosphokinase fraction MB, and myoglobin in predicting cardiotoxicity in a cohort of women with breast cancer undergoing chemotherapy with anthracycline. METHODS: This is an observational, prospective, longitudinal, unicentric study, which included 40 women with breast cancer, whose therapeutic proposal included treatment with doxorubicin. The protocol had a clinical follow-up of 12 months. Biomarkers such as Troponin I, type B natriuretic peptide, creatine phosphokinase fraction MB, and myoglobin were measured pre-chemotherapy and after the first, third, fourth, and sixth cycles of chemotherapy. RESULTS: There was a progressive increase in type B natriuretic peptide and myoglobin values in all chemotherapy cycles. Although creatine phosphokinase fraction MB showed a sustained increase, this increase was not statistically significant. Troponin, type B natriuretic peptide, myoglobin, and creatine phosphokinase fraction MB were the cardiotoxicity markers with the earliest changes, with a significant increase after the first chemotherapy session. However, they were not able to predict cardiotoxicity. CONCLUSION: Troponin I, type B natriuretic peptide, myoglobin, and creatine phosphokinase fraction MB are elevated during chemotherapy with doxorubicin, but they were not able to predict cardiotoxicity according to established clinical and echocardiographic criteria. The incidence of subclinical cardiotoxicity resulting from the administration of doxorubicin was 12.5%.


Biomarkers , Breast Neoplasms , Cardiotoxicity , Doxorubicin , Myoglobin , Troponin I , Humans , Female , Breast Neoplasms/drug therapy , Prospective Studies , Troponin I/blood , Doxorubicin/adverse effects , Cardiotoxicity/etiology , Middle Aged , Biomarkers/blood , Myoglobin/blood , Adult , Antibiotics, Antineoplastic/adverse effects , Natriuretic Peptide, Brain/blood , Aged , Creatine Kinase, MB Form/blood , Longitudinal Studies , Anthracyclines/adverse effects , Ventricular Dysfunction, Left/chemically induced , Predictive Value of Tests
6.
Kardiologiia ; 64(5): 33-38, 2024 May 31.
Article Ru | MEDLINE | ID: mdl-38841787

Significant advances in timely diagnosis and modern antitumor therapy have led to a considerable increase in the survival rate of cancer patients. On the other hand, the incidence of cardiovascular (CV) diseases and their complications is increasingly growing, including due to side effects of anticancer drugs. CV complications are the most common cause of non-oncological death of cancer patients. The development of polychemotherapy-induced arterial hypertension (AH) is closely associated with the use of certain groups of drugs, for example, inhibitors of vascular endothelial growth factor (iVEGF). Such AH is generally dose-dependent and reversible after interruption or termination of treatment. However, systemic AH, regardless of its genesis, is one of the key risk factors for many CV events (myocardial infarction, stroke, heart failure, arrhythmias) and kidney disease. Therefore, thorough blood pressure monitoring and its timely and adequate correction if needed are indicated when using certain groups of chemotherapy drugs. This article describes a clinical follow-up of a patient with induced AH associated with the iVEGF antitumor therapy for advanced uterine cancer with a rapid development of left ventricular myocardial dysfunction.


Hypertension , Humans , Female , Hypertension/chemically induced , Cardiotoxicity/etiology , Middle Aged , Uterine Neoplasms/drug therapy , Antineoplastic Agents/adverse effects , Ventricular Dysfunction, Left/chemically induced , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/diagnosis , Vascular Endothelial Growth Factor A/antagonists & inhibitors
7.
Cell Physiol Biochem ; 58(3): 273-287, 2024 Jun 14.
Article En | MEDLINE | ID: mdl-38881348

BACKGROUND/AIMS: Inhaled particulate air pollution is associated with cardiotoxicity with underlying mechanisms including oxidative stress and inflammation. Carnosol, commonly found in rosemary and sage, is known to possess a broad range of therapeutic properties such as antioxidant, anti-inflammatory and antiapoptotic. However, its cardioprotective effects on diesel exhaust particles (DEPs)-induced toxicity have not been studied yet. Hence, we evaluated the potential ameliorative effects of carnosol on DEPs-induced heart toxicity in mice, and the underlying mechanisms involved. METHODS: Mice were intratracheally instilled with DEPs (1 mg/kg) or saline, and 1 hour prior to instillation they were given intraperitoneally either carnosol (20 mg/kg) or saline. Twenty-four hours after the DEPs instillation, multiple parameters were evaluated in the heart by enzyme-linked immunosorbent assay, colorimetric assay, Comet assay and Western blot technique. RESULTS: Carnosol has significantly reduced the elevation in the plasma levels of lactate hydrogenase and brain natriuretic peptide induced by DEPs. Likewise, the augmented cardiac levels of proinflammatory cytokines, lipid peroxidation, and total nitric oxide in DEPs-treated groups were significantly normalized with the treatment of carnosol. Moreover, carnosol has markedly reduced the heart mitochondrial dysfunction, as well as DNA damage and apoptosis of mice treated with DEPs. Similarly, carnosol significantly reduced the elevated expressions of phosphorylated nuclear factor-кB (NF-кB) and mitogen-activated protein kinases (MAPKs) in the hearts. Furthermore, the treatment with carnosol has restored the decrease in the expression of sirtuin-1 in the hearts of mice exposed to DEPs. CONCLUSION: Carnosol significantly attenuated DEP-induced cardiotoxicity in mice by suppressing inflammation, oxidative stress, DNA damage, and apoptosis, at least partly via mechanisms involving sirtuin-1 activation and the inhibition of NF-кB and MAPKs activation.


Abietanes , Cardiotoxicity , NF-kappa B , Oxidative Stress , Vehicle Emissions , Animals , Mice , NF-kappa B/metabolism , Oxidative Stress/drug effects , Vehicle Emissions/toxicity , Abietanes/pharmacology , Abietanes/therapeutic use , Male , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Cardiotoxicity/prevention & control , Cardiotoxicity/drug therapy , Cardiotoxicity/pathology , Nitrosative Stress/drug effects , Inflammation/drug therapy , Inflammation/metabolism , Inflammation/pathology , Inflammation/chemically induced , MAP Kinase Signaling System/drug effects , Antioxidants/pharmacology , Apoptosis/drug effects , Signal Transduction/drug effects , Cardiotonic Agents/pharmacology , Cardiotonic Agents/therapeutic use , Sirtuin 1/metabolism , Sirtuin 1/genetics , DNA Damage/drug effects
8.
J Pharmacol Toxicol Methods ; 127: 107511, 2024.
Article En | MEDLINE | ID: mdl-38710237

The Health and Environmental Sciences Institute (HESI) is a nonprofit organization dedicated to resolving global health challenges through collaborative scientific efforts across academia, regulatory authorities and the private sector. Collaborative science across non-clinical disciplines offers an important keystone to accelerate the development of safer and more effective medicines. HESI works to address complex challenges by leveraging diverse subject-matter expertise across sectors offering access to resources, data and shared knowledge. In 2008, the HESI Cardiac Safety Committee (CSC) was established to improve public health by reducing unanticipated cardiovascular (CV)-related adverse effects from pharmaceuticals or chemicals. The committee continues to significantly impact the field of CV safety by bringing together experts from across sectors to address challenges of detecting and predicting adverse cardiac outcomes. Committee members have collaborated on the organization, management and publication of prospective studies, retrospective analyses, workshops, and symposia resulting in 38 peer reviewed manuscripts. Without this collaboration these manuscripts would not have been published. Through their work, the CSC is actively addressing challenges and opportunities in detecting potential cardiac failure modes using in vivo, in vitro and in silico models, with the aim of facilitating drug development and improving study design. By examining past successes and future prospects of the CSC, this manuscript sheds light on how the consortium's multifaceted approach not only addresses current challenges in detecting potential cardiac failure modes but also paves the way for enhanced drug development and study design methodologies. Further, exploring future opportunities and challenges will focus on improving the translational predictability of nonclinical evaluations and reducing reliance on animal research in CV safety assessments.


Cardiotoxicity , Humans , Animals , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Academies and Institutes , Drug Development/methods , Cardiovascular Diseases , Drug-Related Side Effects and Adverse Reactions/prevention & control
9.
BMJ ; 385: e075859, 2024 05 15.
Article En | MEDLINE | ID: mdl-38749554

In addition to conventional chemoradiation and targeted cancer therapy, the use of immune based therapies, specifically immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T cell therapy (CAR-T), has increased exponentially across a wide spectrum of cancers. This has been paralleled by recognition of off-target immune related adverse events that can affect almost any organ system including the cardiovascular system. The use of ICIs has been associated with myocarditis, a less common but highly fatal adverse effect, pericarditis and pericardial effusions, vasculitis, thromboembolism, and potentially accelerated atherosclerosis. CAR-T resulting in a systemic cytokine release syndrome has been associated with myriad cardiovascular consequences including arrhythmias, myocardial infarction, and heart failure. This review summarizes the current state of knowledge regarding adverse cardiovascular effects associated with ICIs and CAR-T.


Immune Checkpoint Inhibitors , Immunotherapy, Adoptive , Neoplasms , Humans , Neoplasms/drug therapy , Neoplasms/therapy , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Cardiovascular Diseases/chemically induced , Cardiotoxicity/etiology , Myocarditis/chemically induced , Myocarditis/therapy , Cytokine Release Syndrome/etiology , Pericarditis/chemically induced , Pericarditis/therapy
10.
Fish Shellfish Immunol ; 150: 109624, 2024 Jul.
Article En | MEDLINE | ID: mdl-38740228

Avermectin is one of the widely used anthelmintics in aquaculture and exhibits substantial toxicity to aquatic organisms. Silybin is extensively used for its anti-inflammatory, antioxidant and anti-apoptotic biological properties. Heart is essential for the survival of fish and plays a vital role in pumping blood oxygen and nutrients. Residual avermectin in water poses harm to carp. However, there is still insufficient research on whether silybin can mitigate the toxicity of avermectin to carp heart tissues. In this research, we established a model involving carp subjected to acute avermectin exposure and administered diets containing silybin to explore the potential protective effects of silybin against avermectin-induced cardiotoxicity. The results revealed that avermectin induced oxidative stress, inflammation, endoplasmic reticulum (ER) stress, mitochondrial pathway apoptosis and autophagy in the cardiac tissues of carp. Compared with the avermectin group, silybin significantly reduced ROS accumulation in cardiac tissues, restored antioxidant enzyme activity, inhibited mRNA transcript levels of pro-inflammatory-related factors, and attenuated ER stress, mitochondrial pathway apoptosis and autophagy. Protein-protein interaction (PPI) analysis demonstrated that silybin mitigated avermectin-induced cardiac oxidative stress, inflammation, ER stress, mitochondrial pathway apoptosis and autophagy. Silybin exerted anti-inflammatory effects through the Nuclear Factor kappa B (NF-κB) pathway, antioxidant effects through the Nuclear factor erythroid 2-related factor 2 (Nrf2) - Kelch-like ECH-associated protein 1 (Keap1) pathway, alleviated cardiac ER stress through the Glucose-regulated protein 78 (GRP78)/Activating Transcription Factor 6 (ATF6)/C/EBP homologous protein (CHOP) axis, suppressed apoptosis through the mitochondrial pathway, and inhibited excessive autophagy initiation through the PTEN-induced putative kinase 1 (PINK1)/Parkin RBR E3 ubiquitin protein ligase (PARKIN) signaling pathway. This study provided evidence supporting the protective effect of silybin against avermectin-induced cardiotoxicity in carp, highlighting its potential as a dietary additive to protect fish from adverse effects caused by avermectin exposure.


Apoptosis , Autophagy , Cardiotoxicity , Carps , Endoplasmic Reticulum Stress , Inflammation , Ivermectin , Oxidative Stress , Silybin , Animals , Ivermectin/analogs & derivatives , Ivermectin/toxicity , Endoplasmic Reticulum Stress/drug effects , Autophagy/drug effects , Carps/immunology , Apoptosis/drug effects , Oxidative Stress/drug effects , Cardiotoxicity/etiology , Silybin/pharmacology , Silybin/administration & dosage , Inflammation/chemically induced , Inflammation/veterinary , Fish Diseases/chemically induced , Fish Diseases/prevention & control , Fish Diseases/immunology , Mitochondria/drug effects , Anthelmintics/toxicity , Anthelmintics/pharmacology
13.
Dtsch Med Wochenschr ; 149(12): 719-723, 2024 Jun.
Article De | MEDLINE | ID: mdl-38781996

The emerging field of cardio-oncology addresses the critical need for specialized cardiovascular care in cancer patients, given the overlapping risk factors and potential cardiovascular complications of oncological therapies. In collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO), and the European Society of Cardiology (ESC), the first cardio-oncology guideline was developed and published in 2022. This guideline comprises 272 recommendations covering risk stratification before therapy initiation, monitoring during oncological treatment, and the diagnosis and treatment of therapy-associated cardiovascular side effects.A significant innovation in this guideline is the comprehensive risk stratification approach, which categorizes patients into low, moderate, and high-risk groups based on therapy-specific factors. This allows for tailored cardiovascular care during therapy, with varying frequencies of follow-up examinations depending on the patient's risk level. Notably, the guideline emphasizes the importance of interdisciplinary collaboration between oncologists and cardiologists to optimize patient outcomes.Overall, the cardio-oncology guideline represents a significant advancement in addressing the complex cardiovascular needs of cancer patients. Its comprehensive recommendations and emphasis on interdisciplinary care underscore the importance of optimizing cardiovascular health throughout the oncological treatment journey.This review provides an overview of the guidelines and updates on the risk stratification and therapy of patients with immune checkpoint inhibitor-associated myocarditis (ICIM), as well as the role of statins in protecting against anthracycline-associated cardiotoxicity.


Cardiovascular Diseases , Neoplasms , Humans , Neoplasms/complications , Neoplasms/therapy , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/etiology , Medical Oncology , Practice Guidelines as Topic , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Cardiology/standards , Risk Assessment , Risk Factors , Antineoplastic Agents/adverse effects , Antineoplastic Agents/therapeutic use , Cardio-Oncology
14.
Peptides ; 178: 171245, 2024 Aug.
Article En | MEDLINE | ID: mdl-38801993

BACKGROUND: Doxorubicin (DOX) is a highly effective and widely used cytotoxic agent with application for various malignancies, but it's clinically limited due to its cardiotoxicity Oxidative stress and inflammation were reported to take part in DOX-induced cardiotoxicity. Tirzepatide, a dual glucagon-like peptide-1 (GLP-1) and glucose-dependent insulinotropic polypeptide (GIP) receptor agonist has been approved to treat type 2 diabetes. However, its role in DOX-induced cardiotoxicity and the underlying mechanisms has not been explored. METHODS: The cardioprotective properties of Tirzepatide against DOX-induced cardiotoxicity are examined in this work both in vivo and in vitro. For four weeks, an intraperitoneal injection of 4 mg/kg DOX was used to cause cardiotoxicity in C57BL/6 mice. To ascertain the cardioprotective function and underlying mechanisms of Tirzepatide against DOX-induced cardiotoxicity, mice and H9c2 cells were treated with and without Tirzepatide. RESULTS: Tirzepatide treatment significantly inhibited DOX-induced oxidative stress, inflammation and cardiac injury. Mechanistically, PI3K/Akt signaling pathway contributes to the protective effect of Tirzepatide against DOX-induced cardiotoxicity and inhibited PI3K/Akt signaling pathway with LY294002 almost blocked its therapeutic effect. CONCLUSIONS: Collectively, Tirzepatide could alleviate DOX-induced oxidative stress, inflammation and cardiac injury via activating PI3K/Akt signaling pathway and Tirzepatide may be a novel therapeutic target for DOX-induced cardiotoxicity.


Cardiotoxicity , Doxorubicin , Inflammation , Oxidative Stress , Phosphatidylinositol 3-Kinases , Proto-Oncogene Proteins c-akt , Signal Transduction , Doxorubicin/adverse effects , Animals , Oxidative Stress/drug effects , Mice , Proto-Oncogene Proteins c-akt/metabolism , Cardiotoxicity/drug therapy , Cardiotoxicity/prevention & control , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Signal Transduction/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Inflammation/drug therapy , Inflammation/chemically induced , Inflammation/metabolism , Inflammation/pathology , Male , Mice, Inbred C57BL , Cardiotonic Agents/pharmacology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism
15.
Complement Ther Med ; 83: 103053, 2024 Aug.
Article En | MEDLINE | ID: mdl-38801910

OBJECTIVE: Shenmai injection is a classic herbal prescription, and is often recommended for the treatment of anthracycline-induced cardiotoxicity. However, the efficacy and safety of Shenmai injection for the treatment of anthracycline-induced cardiotoxicity have not been reported. MATERIALS AND METHODS: We conducted a comprehensive search of eight literature databases and two clinical trial registries, retrieving all randomized controlled trials (RCTs) related to the treatment of anthracycline-induced cardiotoxicity with Shenmai injection from the establishment of the databases to July 1, 2023. Data analysis was performed using the Meta package in RStudio and RevMan 5.4. The GRADE pro3.6.1 software was utilized for assessing the quality of evidence. RESULTS: A total of 16 RCTs including 2140 patients were included in this study. Meta-analysis showed that Shenmai injection had an advantage in improving ST-T segment changes (RR = 0.28; 95 % CI, 0.20 to 0.39; P < 0.0001) (P < 0.01), creatine kinase isoenzyme (SMD = -3.49; 95 % CI, -5.24 to -1.74; P < 0.0001), Prolonged QT interval (RR = 0.46; 95 % CI, 0.28 to 0.75; P = 0.0018), Low QRS Voltage (RR = 0.44; 95 % CI, 0.27 to 0.71; P = 0.0007), sinus tachycardia (RR = 0.41; 95 % CI, 0.28 to 0.60; P < 0.0001), atrial premature beats (RR = 0.55; 95 % CI, 0.35 to 0.87; P = 0.01), Premature Ventricular Contractions (RR = 0.39; 95 % CI, 0.26 to 0.59; P < 0.0001) and creatine kinase (SMD = -1.43; 95 % CI, -2.57 to -0.29; P < 0.0001) in patients with anthracycline-induced cardiotoxicity. advantage, which was supported by sensitivity analyses, but not in improving left ventricular ejection fraction (MD = 16.01; 95 % CI, -3.10 to 35.12; P = 0.10) and atrioventricular block (RR = 0.49; 95 % CI, 0.24 to 1.03; P = 0.06). The literature included in the study did not refer to data regarding the safety aspects of Shenmai injection, so we do not yet know the safety of Shenmai injection. The results of subgroup analyses suggested that heterogeneity was not related to the administered dose and chemotherapy regimen. The publication bias test showed no publication bias. The quality of evidence for the results ranged from "very low" to "moderate." CONCLUSION: This study suggests that Shenmai injection is effective in treating anthracycline-induced cardiotoxicity and is a potential treatment for anthracycline-induced cardiotoxicity. However, due to the poor methodological quality of the included RCTs, we recommend rigorous, high-quality, large-sample trials to confirm our findings.


Anthracyclines , Cardiotoxicity , Drug Combinations , Drugs, Chinese Herbal , Humans , Drugs, Chinese Herbal/therapeutic use , Drugs, Chinese Herbal/adverse effects , Drugs, Chinese Herbal/administration & dosage , Cardiotoxicity/etiology , Anthracyclines/adverse effects , Randomized Controlled Trials as Topic
16.
Am J Cardiol ; 222: 175-182, 2024 Jul 01.
Article En | MEDLINE | ID: mdl-38692401

Anthracyclines are pivotal in cancer treatment, yet their clinical utility is hindered by the risk of cardiotoxicity. Preclinical studies highlight the effectiveness of sodium-glucose cotransporter-2 inhibitors (SGLT2i) in mitigating anthracycline-induced cardiotoxicity. Nonetheless, the translation of these findings to clinical practice remains uncertain. This study aims to evaluate the safety and potential of SGLT2i for preventing cardiotoxicity in patients with cancer, without preexisting heart failure (HF), receiving anthracyclines therapy. Using the TriNetX Global Research Network, patients with cancer, without previous HF diagnosis, receiving anthracycline therapy were identified and classified into 2 groups based on SGLT2i usage. A 1:1 propensity score matching was used to control for baseline characteristics between the 2 groups. Patients were followed for 2 years. The primary end point was new-onset HF, and the secondary end points were HF exacerbation, new-onset arrhythmia, myocardial infarction, all-cause mortality, and all-cause hospitalization. Safety outcomes included acute renal failure and creatinine levels. A total of 79,074 patients were identified, and 1,412 were included post-matching (706 in each group). They comprised 53% females, 62% White, with a mean age of 62.5 ± 11.4 years. Over the 2-year follow-up period, patients on SGLT2i had lower rates of new-onset HF (hazard ratio 0.147, 95% confidence interval 0.073 to 0.294) and arrhythmia (hazard ratio 0.397, 95% confidence interval 0.227 to 0.692) compared with those not on SGLT2i. The incidence of all-cause mortality, myocardial infarction, all-cause hospitalization, and safety outcomes were similar between both groups. In conclusion, among patients with cancer receiving anthracycline therapy without preexisting HF, SGLT2i use demonstrates both safety and effectiveness in reducing anthracycline-induced cardiotoxicity, with a decreased incidence of new-onset HF, HF exacerbation, and arrhythmias.


Anthracyclines , Cardiotoxicity , Heart Failure , Neoplasms , Sodium-Glucose Transporter 2 Inhibitors , Humans , Female , Sodium-Glucose Transporter 2 Inhibitors/therapeutic use , Male , Anthracyclines/therapeutic use , Anthracyclines/adverse effects , Middle Aged , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Neoplasms/drug therapy , Aged , Heart Failure/chemically induced , Propensity Score , Hospitalization/statistics & numerical data , Myocardial Infarction/epidemiology , Myocardial Infarction/prevention & control
17.
Int J Biol Sci ; 20(7): 2622-2639, 2024.
Article En | MEDLINE | ID: mdl-38725840

Sorafenib is a standard first-line drug for advanced hepatocellular carcinoma, but the serious cardiotoxic effects restrict its therapeutic applicability. Here, we show that iron-dependent ferroptosis plays a vital role in sorafenib-induced cardiotoxicity. Remarkably, our in vivo and in vitro experiments demonstrated that ferroptosis inhibitor application neutralized sorafenib-induced heart injury. By analyzing transcriptome profiles of adult human sorafenib-treated cardiomyocytes, we found that Krüppel-like transcription factor 11 (KLF11) expression significantly increased after sorafenib stimulation. Mechanistically, KLF11 promoted ferroptosis by suppressing transcription of ferroptosis suppressor protein 1 (FSP1), a seminal breakthrough due to its ferroptosis-repressing properties. Moreover, FSP1 knockdown showed equivalent results to glutathione peroxidase 4 (GPX4) knockdown, and FSP1 overexpression counteracted GPX4 inhibition-induced ferroptosis to a substantial extent. Cardiac-specific overexpression of FSP1 and silencing KLF11 by an adeno-associated virus serotype 9 markedly improved cardiac dysfunction in sorafenib-treated mice. In summary, FSP1-mediated ferroptosis is a crucial mechanism for sorafenib-provoked cardiotoxicity, and targeting ferroptosis may be a promising therapeutic strategy for alleviating sorafenib-induced cardiac damage.


Cardiotoxicity , Ferroptosis , Repressor Proteins , S100 Calcium-Binding Protein A4 , Sorafenib , Animals , Humans , Male , Mice , Cardiotoxicity/metabolism , Cardiotoxicity/etiology , Ferroptosis/drug effects , Mice, Inbred C57BL , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Repressor Proteins/metabolism , Repressor Proteins/genetics , S100 Calcium-Binding Protein A4/metabolism , S100 Calcium-Binding Protein A4/genetics , Sorafenib/adverse effects
18.
G Ital Cardiol (Rome) ; 25(6): 453-463, 2024 Jun.
Article It | MEDLINE | ID: mdl-38808942

The survival of pediatric cancer patients has significantly increased thanks to the improvement of oncological treatments. Therefore, it is of utmost importance to manage short- and long-term cardiovascular complications. In pediatric cardio-oncology, there are no recognized guidelines as in adults. Several recommendations and many indications have been derived from the data obtained in the adult cancer population, resulting in greater discrepancies in the clinical management of patients. The aim of this position paper of the Italian Society of Pediatric Cardiology (SICP) is to collect the main evidence regarding the diagnosis, prevention, treatment and follow-up of cardiotoxicity in children, to provide useful indications for clinical practice, and to promote a network between pediatric centers.


Antineoplastic Agents , Cardiotoxicity , Neoplasms , Humans , Cardiotoxicity/prevention & control , Cardiotoxicity/etiology , Child , Neoplasms/drug therapy , Antineoplastic Agents/adverse effects , Antineoplastic Agents/administration & dosage , Italy , Cardiovascular Diseases/prevention & control , Cardiology , Follow-Up Studies , Heart Diseases/prevention & control , Heart Diseases/chemically induced , Heart Diseases/diagnosis , Societies, Medical
19.
Int J Mol Sci ; 25(10)2024 May 12.
Article En | MEDLINE | ID: mdl-38791311

Doxorubicin (DOX), widely used as a chemotherapeutic agent for various cancers, is limited in its clinical utility by its cardiotoxic effects. Despite its widespread use, the precise mechanisms underlying DOX-induced cardiotoxicity at the cellular and molecular levels remain unclear, hindering the development of preventive and early detection strategies. To characterize the cytotoxic effects of DOX on isolated ventricular cardiomyocytes, focusing on the expression of specific microRNAs (miRNAs) and their molecular targets associated with endogenous cardioprotective mechanisms such as the ATP-sensitive potassium channel (KATP), Sirtuin 1 (SIRT1), FOXO1, and GSK3ß. We isolated Guinea pig ventricular cardiomyocytes by retrograde perfusion and enzymatic dissociation. We assessed cell morphology, Reactive Oxygen Species (ROS) levels, intracellular calcium, and mitochondrial membrane potential using light microscopy and specific probes. We determined the miRNA expression profile using small RNAseq and validated it using stem-loop qRT-PCR. We quantified mRNA levels of some predicted and validated molecular targets using qRT-PCR and analyzed protein expression using Western blot. Exposure to 10 µM DOX resulted in cardiomyocyte shortening, increased ROS and intracellular calcium levels, mitochondrial membrane potential depolarization, and changes in specific miRNA expression. Additionally, we observed the differential expression of KATP subunits (ABCC9, KCNJ8, and KCNJ11), FOXO1, SIRT1, and GSK3ß molecules associated with endogenous cardioprotective mechanisms. Supported by miRNA gene regulatory networks and functional enrichment analysis, these findings suggest that DOX-induced cardiotoxicity disrupts biological processes associated with cardioprotective mechanisms. Further research must clarify their specific molecular changes in DOX-induced cardiac dysfunction and investigate their diagnostic biomarkers and therapeutic potential.


Cardiotoxicity , Doxorubicin , MicroRNAs , Myocytes, Cardiac , Reactive Oxygen Species , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/drug effects , Animals , Doxorubicin/adverse effects , Doxorubicin/toxicity , Cardiotoxicity/etiology , MicroRNAs/genetics , MicroRNAs/metabolism , Reactive Oxygen Species/metabolism , Guinea Pigs , Membrane Potential, Mitochondrial/drug effects , Heart Ventricles/drug effects , Heart Ventricles/metabolism , Heart Ventricles/cytology , Male , Calcium/metabolism , Gene Expression Regulation/drug effects
...