Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (46)

Search Parameters:
Keywords = cardiotoxicity cardiovascular outcomes

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
20 pages, 2693 KiB  
Review
Navigating Cardiotoxicity in Immune Checkpoint Inhibitors: From Diagnosis to Long-Term Management
by Simone Nardin, Beatrice Ruffilli, Pietro Costantini, Rocco Mollace, Ida Taglialatela, Matteo Pagnesi, Mauro Chiarito, Davide Soldato, Davide Cao, Benedetta Conte, Monica Verdoia, Alessandra Gennari and Matteo Nardin
J. Cardiovasc. Dev. Dis. 2025, 12(7), 270; https://doi.org/10.3390/jcdd12070270 - 16 Jul 2025
Viewed by 535
Abstract
The advent of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, significantly improving patient outcomes across multiple malignancies. Nonetheless, these therapies are associated with immune-related adverse effects, including cardiotoxicity, which remains a critical concern. This review provides a comprehensive analysis of ICI-related cardiotoxicity, [...] Read more.
The advent of immune checkpoint inhibitors (ICIs) has revolutionized cancer treatment, significantly improving patient outcomes across multiple malignancies. Nonetheless, these therapies are associated with immune-related adverse effects, including cardiotoxicity, which remains a critical concern. This review provides a comprehensive analysis of ICI-related cardiotoxicity, encompassing its pathophysiological mechanisms, risk factors, diagnostic modalities, and management strategies. The onset of cardiotoxicity varies widely, ranging from acute myocarditis to long-term cardiovascular complications. Early identification through clinical assessment, biomarkers, and advanced imaging techniques is crucial for timely intervention. Management strategies include high-dose corticosteroids, other immunosuppressive agents, and supportive therapies, with a focus on balancing oncologic efficacy and cardiovascular safety. Additionally, rechallenging patients with ICIs following cardiotoxic events remains a complex clinical decision requiring multidisciplinary evaluation. As immunotherapy indications expand to include high-risk populations in a curative setting too, optimizing screening, prevention, and treatment strategies is essential to mitigate cardiovascular risks. A deep understanding of the molecular and clinical aspects of ICI-related cardiotoxicity will enhance patient safety and therapeutic decision-making, underscoring the need for ongoing research in this rapidly evolving field. Full article
Show Figures

Graphical abstract

15 pages, 638 KiB  
Article
Efficacy, Toxicity and Effect of Pretreatment Cardiologic Consultation on Outcomes of Ibrutinib Therapy for Chronic Lymphocytic Leukemia—A KroHem Study
by Inga Mandac Smoljanović, Igor Aurer, Nikola Bulj, Barbara Dreta, Antonija Miljak, Fran Petričević, Marija Ivić, Sandra Bašić-Kinda, Viktor Zatezalo, Sanja Madunić, Dubravka Čaržavec, Jasminka Sinčić-Petričević, Dragana Grohovac, Ozren Jakšić, Ivan Krečak, Martina Morić-Perić, Božena Coha, Petra Berneš, Neno Živković and Vlatko Pejša
Cancers 2025, 17(14), 2302; https://doi.org/10.3390/cancers17142302 - 10 Jul 2025
Viewed by 324
Abstract
Background/Objectives: Ibrutinib has revolutionized the treatment of chronic lymphocytic leukemia but has off-target side effects, most notably cardiac. In order to evaluate the efficacy and toxicity of ibrutinib treatment, risk factors for adverse outcomes and the influence of pretreatment cardiologic evaluation, KroHem collected [...] Read more.
Background/Objectives: Ibrutinib has revolutionized the treatment of chronic lymphocytic leukemia but has off-target side effects, most notably cardiac. In order to evaluate the efficacy and toxicity of ibrutinib treatment, risk factors for adverse outcomes and the influence of pretreatment cardiologic evaluation, KroHem collected data on Croatian patients with chronic lymphocytic leukemia treated with this drug. Methods: This is a retrospective survey performed in order to analyze the efficacy and toxicity of ibrutinib in a real-life setting. Patients starting therapy with ibrutinib for chronic lymphocytic leukemia between the time the drug became reimbursable in 2015 and 31 December 2021 were included, irrespective of treatment line. Results: We identified 436 patients fulfilling entry criteria; 404 (92.7%) responded to treatment. Cardiovascular side effects occurred in 25.0% of patients and hemorrhagic in 15.6%. The dose of ibrutinib was permanently reduced in 22.2% of patients. Median follow-up of the cohort was 29 months (IQR 18–41 months), estimated median overall survival 75 months (IQR 36 months–not reached), progression-free survival 54 months (IQR 24–81 months) and time on ibrutinib treatment 44 months (IQR 14–78 months). Factors significantly related to overall survival in multivariate analysis were stage, treatment line and age. Factors significantly related to progression-free survival in multivariate analysis were treatment line, age and pretreatment history or ECG finding of cardiac arrhythmia. Factors significantly related to time on ibrutinib treatment in multivariate analysis were age, pretreatment history or ECG finding of cardiac arrhythmia, and permanent dose reduction for toxicity. Sex, FISH and the presence of arterial hypertension were not independently significantly related to any of these outcomes. Pretreatment cardiologic consultation did not improve time on ibrutinib therapy, progression-free survival, overall survival, risk of stopping treatment due to cardiovascular side effects or risk of cardiovascular or sudden death, neither in the whole cohort nor in the subgroup of patients with and without pretreatment cardiac arrhythmia. Conclusions: Our analysis confirms the efficacy and tolerability of ibrutinib for the treatment of chronic lymphocytic leukemia. Patients older than 75 do significantly less well. Routine pretreatment cardiologic consultation does not improve outcomes and should not be considered part of standard pretreatment assessment without additional proof of its usefulness. Future investigations should aim at identifying predictive factors, mechanisms, and preventive strategies for reducing cardiotoxicity in chronic lymphocytic leukemia patients taking Bruton tyrosine kinase inhibitors. Full article
Show Figures

Figure 1

23 pages, 676 KiB  
Review
Cardiotoxicity in Elderly Breast Cancer Patients
by Kalliopi Keramida, Anastasia Constantinidou, Dorothea Tsekoura, Effrosyni Kampouroglou, Chrissovalantis Aidarinis, Emmanouil Saloustros, Georgia Karanasiou, Gaia Giulia Angela Sacco, Erika Matos, Andri Papakonstantinou, Manolis Tsiknakis, Cameron Brown, Athos Antoniades, Carlo Cipolla, Daniela Cardinale, Dimitrios Fotiadis, Gerasimos Filippatos and Investigators CARDIOCARE Consortium
Cancers 2025, 17(13), 2198; https://doi.org/10.3390/cancers17132198 - 30 Jun 2025
Viewed by 943
Abstract
Cardiotoxicity is a leading cause of mortality in the growing populations of elderly breast cancer (BC) patients. Breast cancer treatment in the elderly is highly challenging due to its heterogeneous nature and the lack of specific evidence, as this population is usually underrepresented [...] Read more.
Cardiotoxicity is a leading cause of mortality in the growing populations of elderly breast cancer (BC) patients. Breast cancer treatment in the elderly is highly challenging due to its heterogeneous nature and the lack of specific evidence, as this population is usually underrepresented in randomized clinical trials. Decision making requires a comprehensive approach, considering the type and stage of BC, the patient’s overall health status, life expectancy, geriatric and frailty assessment, the risk of cancer recurrence, comorbidities, cardiotoxicity risk, and the patient’s preferences. The cardiotoxic effects of BC treatments cover the whole spectrum of cardiovascular diseases: heart failure, hypertension, arrhythmias, and myocardial ischemia. Cardiotoxicity risk in these patients is defined by several factors: anticancer therapies, polypharmacy, established cardiovascular disease, comorbidities, frailty, cellular senescence, hormonal changes, and genetic predisposition. Preventive oncological and cardio-oncological strategies, as well as patients’ education, are critical for improved outcomes. Prospective clinical trials in this population are urgently needed. Full article
(This article belongs to the Section Cancer Survivorship and Quality of Life)
Show Figures

Graphical abstract

9 pages, 216 KiB  
Article
Cardiovascular Toxicities in Chimeric Antigen Receptor Therapy in Relapsed and Refractory Multiple Myeloma and Lymphoma Using FAERS Database
by Fathima Shehnaz Ayoobkhan, Suryakumar Balasubramanian, Arindam Bagga and Tarun Parvataneni
Lymphatics 2025, 3(3), 16; https://doi.org/10.3390/lymphatics3030016 - 20 Jun 2025
Viewed by 314
Abstract
Introduction: In the past decade, chimeric antigen receptor T-cell therapy (CAR-T) has revolutionized the treatment of relapsed refractory multiple myeloma (RRMM) and lymphoma, but it is associated with significant cardiovascular adverse effects. We aim to analyze the incidence, patterns, and outcomes of cardiac [...] Read more.
Introduction: In the past decade, chimeric antigen receptor T-cell therapy (CAR-T) has revolutionized the treatment of relapsed refractory multiple myeloma (RRMM) and lymphoma, but it is associated with significant cardiovascular adverse effects. We aim to analyze the incidence, patterns, and outcomes of cardiac events in RRMM and lymphoma patients undergoing CAR-T therapy utilizing the FDA Adverse Event Reporting System (FAERS) database, paving the way for future research and being more vigilant in treating high-risk populations. Methods: We conducted a retrospective post-marketing pharmacovigilance inquiry using the FDA Adverse Event Reporting System (FAERS) database and the Medical Dictionary for Regulatory Activities (MEDRA). We examined the adverse effects associated with CAR-T and TCE since their FDA approval in US and non-US populations (accessed 5 January 2024), and we analyzed the incidence of cardiac events related to six CAR-T products: Idecabtagene vicleucel, Ciltacabtagene autoleucel, Axicabtagene ciloleucel, Tisagenlecleucel, Lisocabtagene maraleucel, and Brexucabtagene autoleucel since FDA approval. Cardiotoxicities were assessed, including coronary artery disease (CAD), myocardial infarction (MI), arrhythmia, heart failure, and hypotension. Results: Out of 12,949 adverse events, we identified 675 (5.2%) cardiac events irrespective of severity. Almost 440 (65%) cardiac events were associated with cytokine release syndrome (CRS). The most common cardiotoxic event was atrial fibrillation (122), followed by the development of heart failure (113), ventricular arrhythmia (108), hypotension (87), and bradyarrhythmia (41). The mortality rate was highest among Brexucabtagene autoleucel recipients (n = 26, 2.3%), followed by Tisagenlecleucel (n = 71, 2.1%) and Lisocabtagene maraleucel (n = 10, 2.1%). Conclusions: CAR-T therapy can result in fatal adverse events due to its cardiotoxic properties. Timely monitoring, such as screening echocardiography and electrocardiograms, can help identify the at-risk population and allow for early intervention—particularly in patients with high baseline cardiovascular risk or previous exposure to cardiotoxic agents—thereby improving outcomes by enabling risk stratification and supportive management. Full article
(This article belongs to the Collection Lymphomas)
16 pages, 960 KiB  
Article
Evaluating the Necessity and Impact of Cardiac Imaging on Breast Cancer Care in Northwestern Ontario
by Hannah Shortreed, Megan Clark, Husam Abdel-Qadir, Rabail Siddiqui and Olexiy Aseyev
Cancers 2025, 17(12), 1909; https://doi.org/10.3390/cancers17121909 - 8 Jun 2025
Viewed by 596
Abstract
Introduction: Breast cancer affects over 25,000 women annually in Canada and has seen improved survival rates due to advances in screening and treatment. However, cardiotoxic therapies including anthracyclines and trastuzumab have made cardiovascular disease a leading cause of death among survivors. Baseline left [...] Read more.
Introduction: Breast cancer affects over 25,000 women annually in Canada and has seen improved survival rates due to advances in screening and treatment. However, cardiotoxic therapies including anthracyclines and trastuzumab have made cardiovascular disease a leading cause of death among survivors. Baseline left ventricular ejection fraction is a reliable predictor of heart failure, and various guidelines recommend pretreatment cardiac imaging; however, its utility is largely based on expert opinion. Methods: This retrospective cohort study analyzed 93 breast cancer patients treated at a single cancer centre in Northwestern Ontario between 2012 and 2017 to determine the yield (defined as imaging leading to clinically actionable changes in care) of imaging. Results: Patients were grouped by treatment regimen: trastuzumab-only (cohort A, n = 3, mean age = 73.55 ± 9.90), anthracycline-only (cohort B, n = 60, mean age = 58.83 ± 9.83), and combination therapy with both trastuzumab and anthracyclines (cohort C, n = 30, mean age = 59.37 ± 10.91). Due to the very small sample size in cohort A, findings are presented for qualitative insight only. Cohort B had the highest imaging yield (13.33%), while cohorts A and C showed lower yields (7.14% and 4.17%) with more frequent imaging. Predictors of higher yield varied, with cohort B identifying the most, including diabetes and coronary artery disease. Conclusions: These findings underscore the need for targeted cardiac imaging to optimize resource allocation and patient outcomes, particularly in resource-limited settings such as Northwestern Ontario. Subsequent investigations should seek to stratify proactive versus reactive interventions, evaluate outcomes, refine imaging guidelines, and gather more data on patients receiving trastuzumab. Full article
(This article belongs to the Section Cancer Therapy)
Show Figures

Graphical abstract

22 pages, 717 KiB  
Review
Lifestyle and Pharmacological Interventions to Prevent Anthracycline-Related Cardiotoxicity in Cancer Patients
by Luigi Spadafora, Francesca Maria Di Muro, Chiara Intonti, Ludovica Massa, Mauro Monelli, Roberto Franco Enrico Pedretti, Edvige Palazzo Adriano, Pasquale Guarini, Gaia Cantiello, Marco Bernardi, Federico Russo, Stefano Cacciatore, Pierre Sabouret, Michele Golino, Giuseppe Biondi Zoccai, Francesca Romana Zimatore and Laura Adelaide Dalla Vecchia
J. Cardiovasc. Dev. Dis. 2025, 12(6), 212; https://doi.org/10.3390/jcdd12060212 - 4 Jun 2025
Viewed by 1478
Abstract
Anthracyclines remain a cornerstone of cancer therapy but are associated with a significant risk of cardiotoxicity, which can lead to overt heart failure. The risk is modulated by cumulative dose, pre-existing cardiovascular disease, and patient-specific factors. As cancer survival improves, the long-term cardiovascular [...] Read more.
Anthracyclines remain a cornerstone of cancer therapy but are associated with a significant risk of cardiotoxicity, which can lead to overt heart failure. The risk is modulated by cumulative dose, pre-existing cardiovascular disease, and patient-specific factors. As cancer survival improves, the long-term cardiovascular consequences of anthracycline exposure have become a growing concern, underscoring the need for effective preventive strategies. This narrative review examines lifestyle and pharmacological interventions aimed at mitigating anthracycline-induced cardiotoxicity. Evidence suggests that structured exercise programs and antioxidant-rich diets may enhance cardiovascular resilience, while beta-blockers, renin-angiotensin system inhibitors, and dexrazoxane remain central pharmacological options. Emerging therapies, including sodium-glucose co-transporter 2 inhibitors and sacubitril/valsartan, show promise but require further investigation. A comprehensive approach that integrates lifestyle modifications with pharmacological strategies within a multidisciplinary cardio-oncology framework may provide optimal protection, improving long-term cardiovascular outcomes in cancer patients receiving anthracyclines. Full article
(This article belongs to the Section Epidemiology, Lifestyle, and Cardiovascular Health)
Show Figures

Figure 1

26 pages, 1707 KiB  
Review
Doxorubicin-Induced Cardiotoxicity and the Emerging Role of SGLT2 Inhibitors: From Glycemic Control to Cardio-Oncology
by Iacob-Daniel Goje, Greta-Ionela Goje, Valentin Laurențiu Ordodi, Valentina Gabriela Ciobotaru, Vlad Sabin Ivan, Roxana Buzaș, Oana Tunea, Florina Bojin and Daniel-Florin Lighezan
Pharmaceuticals 2025, 18(5), 681; https://doi.org/10.3390/ph18050681 - 3 May 2025
Cited by 1 | Viewed by 2510
Abstract
Cancer remains the second leading cause of death worldwide. Doxorubicin (DOX) is a cornerstone of hematologic malignancy treatment, but it is limited by its dose-dependent cardiotoxicity, leading to systolic and diastolic cardiac dysfunction and, ultimately, dilated hypokinetic cardiomyopathy. Cardio-oncology has emerged as a [...] Read more.
Cancer remains the second leading cause of death worldwide. Doxorubicin (DOX) is a cornerstone of hematologic malignancy treatment, but it is limited by its dose-dependent cardiotoxicity, leading to systolic and diastolic cardiac dysfunction and, ultimately, dilated hypokinetic cardiomyopathy. Cardio-oncology has emerged as a subspecialty addressing cardiovascular complications in cancer patients, highlighting preventive and therapeutic strategies to reduce cancer therapy-related cardiac dysfunction (CTRCD). Current approaches, including beta-blockers, renin–angiotensin system (RAS) inhibitors, and statins, offer partial cardioprotection. Sodium-glucose cotransporter-2 (SGLT2) inhibitors, initially developed for type 2 diabetes mellitus (T2DM), demonstrate pleiotropic cardioprotective effects beyond glycemic control, including reduced oxidative stress, inflammation, and myocardial remodeling. This review explores the interplay between anthracycline therapy, particularly DOX, and cardiotoxicity while evaluating SGLT2 inhibitors as novel agents in cardio-oncology. Preclinical studies suggest SGLT2 inhibitors attenuate CTRCD by preserving mitochondrial function and inhibiting apoptosis, while clinical trials highlight their efficacy in reducing heart failure (HF) hospitalizations and cardiovascular (CV) mortality. Integrating SGLT2 inhibitors into cardio-oncology protocols could revolutionize the management of CTRCD, enhancing patient outcomes in oncology and cardiovascular care. Considering the emerging evidence, SGLT2 inhibitors may provide significant benefits to patients undergoing anthracycline therapy, particularly those with elevated cardiovascular risk profiles. We recommend that future prospective, large-scale clinical trials further evaluate the efficacy and safety of these agents as cardioprotective therapy to optimize individualized treatment strategies. Full article
Show Figures

Figure 1

19 pages, 1602 KiB  
Review
Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity
by Lauren A. Ling, Asma Boukhalfa, Andrew H. Kung, Vicky K. Yang and Howard H. Chen
Pharmaceuticals 2025, 18(5), 671; https://doi.org/10.3390/ph18050671 - 1 May 2025
Viewed by 961
Abstract
Autophagy, an evolutionarily conserved process, plays an important role in cellular homeostasis and human diseases. Cardiovascular dysfunction, which presents during cancer treatment or in cancer-free individuals years after treatment, is a growing clinical challenge. Millions of cancer survivors and patients face an unpredictable [...] Read more.
Autophagy, an evolutionarily conserved process, plays an important role in cellular homeostasis and human diseases. Cardiovascular dysfunction, which presents during cancer treatment or in cancer-free individuals years after treatment, is a growing clinical challenge. Millions of cancer survivors and patients face an unpredictable risk of developing cardiotoxicity. Cardiotoxicity due to cancer treatment, as well as cancer progression, has been linked to autophagy dysregulation. Modulating autophagy has been further proposed as a therapeutic treatment for both cancer and cardiovascular disorders. The safe and effective use of autophagy modulation as a cardioprotective strategy during cancer treatment especially requires careful consideration and experimentation to minimize the impact on cancer treatment. We focus here on recent advances in targeted autophagy modulation strategies that utilize interdisciplinary approaches in biomedical sciences and are potentially translatable to treat cardiotoxicity and improve cancer treatment outcomes. This review highlights non-small molecule autophagy modulators to enhance targeted therapy, nanomedicine for autophagy modulation and monitoring, and in vitro models and future experiments needed to bring novel autophagy discoveries from basic research to clinical translation. Full article
Show Figures

Figure 1

25 pages, 1535 KiB  
Review
Risk Factors and Prevention of Cancer and CVDs: A Chicken and Egg Situation
by Maurizio Giuseppe Abrignani, Fabiana Lucà, Vincenzo Abrignani, Mariacarmela Nucara, Daniele Grosseto, Chiara Lestuzzi, Marinella Mistrangelo, Bruno Passaretti, Carmelo Massimiliano Rao and Iris Parrini
J. Clin. Med. 2025, 14(9), 3083; https://doi.org/10.3390/jcm14093083 - 29 Apr 2025
Cited by 1 | Viewed by 2080
Abstract
Cardiovascular diseases and cancer are the two primary causes of mortality worldwide. Although traditionally regarded as distinct pathologies, they share numerous pathophysiological mechanisms and risk factors, including chronic inflammation, insulin resistance, obesity, and metabolic dysregulation. Notably, several cancers have been identified as closely [...] Read more.
Cardiovascular diseases and cancer are the two primary causes of mortality worldwide. Although traditionally regarded as distinct pathologies, they share numerous pathophysiological mechanisms and risk factors, including chronic inflammation, insulin resistance, obesity, and metabolic dysregulation. Notably, several cancers have been identified as closely linked to cardiovascular diseases, including lung, breast, prostate, and colorectal cancers, as well as hematological malignancies, such as leukemia and lymphoma. Additionally, renal and pancreatic cancers exhibit a significant association with cardiovascular complications, partly due to shared risk factors and the cardiotoxic effects of cancer therapies. Addressing the overlapping risk factors through lifestyle modifications—such as regular physical activity, a balanced diet, and cessation of smoking and alcohol—has proven effective in reducing both CV and oncological morbidity and mortality. Furthermore, even in patients with established cancer, structured interventions targeting physical activity, nutritional optimization, and smoking cessation have been associated with improved outcomes. Beyond lifestyle modifications, pharmacological strategies play a crucial role in the prevention of both diseases. Several cardiovascular medications, including statins, aspirin, beta-blockers, and metformin, exhibit pleiotropic effects that extend beyond their primary indications, demonstrating potential anti-neoplastic properties in preclinical and observational studies. Recently, novel therapeutic agents have garnered attention for their possible cardioprotective and metabolic benefits. Glucagon-like peptide-1 receptor agonists (GLP-1 RAs) and sodium-glucose cotransporter-2 inhibitors (SGLT2is), initially developed for managing type 2 diabetes, have shown CV and renal protective effects, alongside emerging evidence of their role in modulating cancer-related metabolic pathways. Inclisiran, a small interfering RNA targeting PCSK9, effectively lowers LDL cholesterol and may contribute to reducing CV risk, with potential implications for tumor biology. Additionally, sacubitril/valsartan, an angiotensin receptor–neprilysin inhibitor, has revolutionized heart failure management by improving hemodynamic parameters and exerting anti-inflammatory effects that may have broader implications for chronic disease prevention. Given the intricate interplay between CVD and cancer, further research is essential to clarify the exact mechanisms linking these conditions and assessing the potential of CV therapies in cancer prevention. This review aims to examine shared risk factors, consider the role of pharmacological and lifestyle interventions, and emphasize crucial epidemiological and mechanistic insights into the intersection of CV and oncological health. Full article
Show Figures

Figure 1

19 pages, 904 KiB  
Article
Predicting Cardiovascular Risk Factors for Acute Leukemia Patients by Assessing Subclinical Atherosclerosis and Left Ventricular Function Before Chemotherapy
by Anda Gabriela Militaru, Daniel Florin Lighezan, Anca Maria Cimpean, Elena Amaricai and Marius Militaru
Life 2025, 15(5), 704; https://doi.org/10.3390/life15050704 - 27 Apr 2025
Viewed by 584
Abstract
Background: Subclinical atherosclerosis is a “silent” cardiovascular disease that can be devastating when combined with other illnesses. Its presence may affect therapy responses but can potentially worsen hematological malignancies due to most chemotherapy regimens’ cardiovascular adverse effects. Thus, cardiovascular risk factor (CVRF) assessment [...] Read more.
Background: Subclinical atherosclerosis is a “silent” cardiovascular disease that can be devastating when combined with other illnesses. Its presence may affect therapy responses but can potentially worsen hematological malignancies due to most chemotherapy regimens’ cardiovascular adverse effects. Thus, cardiovascular risk factor (CVRF) assessment is required before chemotherapy. Unfortunately, this rarely happens. Aim: we aim to examine the impact of CVRFs on hemodynamic parameters of acute leukemia (AL) patients before chemotherapy. Methods: Overall, 45 AL patients and 26 controls were included. Intima-media thickness (IMT), ankle brachial index (ABI), pulse wave velocity (PWV), and functional cardiac parameters were used. CVRFs were found in 26 AL patients (36.6%), while 19 AL (26.8%) patients lacked CVRFs. CVRFs were also found in 26 controls (36.6%). Results: Left ventricular ejection fraction (LVEF) significantly decreased for patients with CVRFs (59.26 ± 5.62) compared to those without CVRFs (64.05 ± 7.43, p < 0.05). Hypertensive and diabetic patients had a significantly higher left IMT (mm) of 0.92 ± 0.01 compared to those without them (0.76 ± 0.03, p < 0.05). Patients with acute myeloid leukemia (AML) with CVRFs had a significantly higher PWV (m/s) of 8.4 ± 0.12 compared to those without CVRFs (6.87 ± 0.66) (p < 0.05). Conclusions: AL and cardiovascular risk factors interacted before chemotherapy. To decrease cardiotoxicity, AL patients need cardiovascular risk assessment. Subclinical atherosclerosis and echocardiography help chemotherapy patients to choose a treatment regimen, predict long-term outcomes, and predict cardiovascular issues. Full article
Show Figures

Figure 1

21 pages, 356 KiB  
Review
Overview of Oncology: Drug-Induced Cardiac Toxicity
by Nilima Rajpal Kundnani, Vincenzo Passini, Iulia Stefania Carlogea, Patrick Dumitrescu, Vlad Meche, Roxana Buzas and Daniel Marius Duda-Seiman
Medicina 2025, 61(4), 709; https://doi.org/10.3390/medicina61040709 - 12 Apr 2025
Cited by 1 | Viewed by 1350
Abstract
Cancer medications can cause cardiac issues, which are difficult to treat in oncologic patients because of the risk of complications. In some cases, this may significantly impact their well-being and treatment outcomes. Overall, these complications fall under the term “drug induced cardiotoxicity”, mainly [...] Read more.
Cancer medications can cause cardiac issues, which are difficult to treat in oncologic patients because of the risk of complications. In some cases, this may significantly impact their well-being and treatment outcomes. Overall, these complications fall under the term “drug induced cardiotoxicity”, mainly due to chemotherapy drugs being specifically toxic to the heart, causing a decrease in the heart’s capacity to pump blood efficiently and leading to a reduction in the left ventricular ejection fraction (LVEF), and subsequently possibly leading to heart failure. Anthracyclines, alkylating agents, and targeted therapies for cancer hold the potential of causing harmful effects on the heart. The incidence of heart-related issues varies from patient to patient and depends on multiple factors, including the type of medication, dosage, duration of the treatment, and pre-existing heart conditions. The underlying mechanism leading to oncologic-drug-induced cardiovascular harmful effects is quite complex. One particular group of drugs, called anthracyclines, have garnered attention due to their impact on oxidative stress and their ability to cause direct harm to heart muscle cells. Reactive oxygen species (ROS) cause harm by inducing damage and programmed cell death in heart cells. Conventional biomarkers alone can only indicate some degree of damage that has already occurred and, therefore, early detection is key. Novel methods like genetic profiling are being developed to detect individuals at risk, prior to the onset of clinical symptoms. Key management strategies—including early detection, personalized medicine approaches, and the use of novel biomarkers—play a crucial role in mitigating cardiotoxicity and improving patient outcomes. Identification of generated genetic alterations and the association to an increased likelihood of cardiotoxicity will allow treatment in a more personalized approach, aiming at decreasing rates of cardiac events while maintaining high oncological efficacy. Oncology drug-induced cardiotoxicity is managed through a combination of preventive strategies and therapeutic interventions from the union of cardiac and oncological knowledge. Full article
(This article belongs to the Special Issue New Insights into Heart Failure)
28 pages, 1031 KiB  
Review
AI and Smart Devices in Cardio-Oncology: Advancements in Cardiotoxicity Prediction and Cardiovascular Monitoring
by Luiza Camelia Nechita, Dana Tutunaru, Aurel Nechita, Andreea Elena Voipan, Daniel Voipan, Ancuta Elena Tupu and Carmina Liana Musat
Diagnostics 2025, 15(6), 787; https://doi.org/10.3390/diagnostics15060787 - 20 Mar 2025
Cited by 3 | Viewed by 1565
Abstract
The increasing prevalence of cardiovascular complications in cancer patients due to cardiotoxic treatments has necessitated advanced monitoring and predictive solutions. Cardio-oncology is an evolving interdisciplinary field that addresses these challenges by integrating artificial intelligence (AI) and smart cardiac devices. This comprehensive review explores [...] Read more.
The increasing prevalence of cardiovascular complications in cancer patients due to cardiotoxic treatments has necessitated advanced monitoring and predictive solutions. Cardio-oncology is an evolving interdisciplinary field that addresses these challenges by integrating artificial intelligence (AI) and smart cardiac devices. This comprehensive review explores the integration of artificial intelligence (AI) and smart cardiac devices in cardio-oncology, highlighting their role in improving cardiovascular risk assessment and the early detection and real-time monitoring of cardiotoxicity. AI-driven techniques, including machine learning (ML) and deep learning (DL), enhance risk stratification, optimize treatment decisions, and support personalized care for oncology patients at cardiovascular risk. Wearable ECG patches, biosensors, and AI-integrated implantable devices enable continuous cardiac surveillance and predictive analytics. While these advancements offer significant potential, challenges such as data standardization, regulatory approvals, and equitable access must be addressed. Further research, clinical validation, and multidisciplinary collaboration are essential to fully integrate AI-driven solutions into cardio-oncology practices and improve patient outcomes. Full article
(This article belongs to the Special Issue AI-Assisted Diagnostics in Telemedicine and Digital Health)
Show Figures

Figure 1

20 pages, 2564 KiB  
Article
Association Between Advanced TNM Stages and Increased Risk of Cardiac Dysfunction in Patients with LVEF < 50%
by Sergiu Ioan Murg, Loredana Matiș, Andrada Florina Moldovan, Dorel Ionel Popovici, Alina Gabriela Negru, Timea Claudia Ghitea and Mircea Ioachim Popescu
Medicina 2025, 61(2), 301; https://doi.org/10.3390/medicina61020301 - 10 Feb 2025
Viewed by 1074
Abstract
Background and Objectives: Cardio-oncology addresses the growing concern of cardiovascular complications arising from cancer therapies. Although cancer treatments have greatly enhanced survival outcomes, they frequently carry substantial risks to cardiovascular health. This research examines the cardiovascular toxicity associated with HER2-targeted therapies, focusing on [...] Read more.
Background and Objectives: Cardio-oncology addresses the growing concern of cardiovascular complications arising from cancer therapies. Although cancer treatments have greatly enhanced survival outcomes, they frequently carry substantial risks to cardiovascular health. This research examines the cardiovascular toxicity associated with HER2-targeted therapies, focusing on the interconnection between tumor characteristics, including histopathological profiles and TNM classification, and the development of cardiovascular complications. The objective is to identify key correlations that inform better prevention and management strategies for cardiotoxicity in oncology patients. Materials and Methods: This retrospective study analyzed cancer patients undergoing cytostatic treatments, particularly anthracyclines, radiotherapy, and HER2-targeted therapies. Cardiac function was monitored using echocardiographic assessments, including global longitudinal strain and left ventricular ejection fraction (LVEF). Patients were stratified based on TNM cancer staging and histopathological findings to evaluate correlations between treatment regimens and cardiovascular outcomes. Results: The analysis revealed a significant association between advanced TNM stages and reduced LVEF, with patients in stage T4 showing the highest prevalence of cardiac dysfunction. Cytostatic treatments, such as anthracyclines and HER2-targeted therapies, were identified as key contributors to cardiotoxicity, particularly in advanced-stage cancer patients. These findings emphasize the importance of regular cardiac monitoring to detect early signs of cardiotoxicity, as patients with pre-existing cardiovascular risk factors demonstrated a higher prevalence of complications. Conclusions: This study highlights the need for personalized treatment approaches and tailored cardioprotective strategies to improve outcomes and enhance the quality of life for oncology patients. Future studies should prioritize developing improved strategies to reduce the cardiovascular complications linked to contemporary cancer treatments. Full article
(This article belongs to the Section Cardiology)
Show Figures

Figure 1

32 pages, 10992 KiB  
Article
Small Extracellular Vesicles from Breast Cancer Cells Induce Cardiotoxicity
by Jhon Jairo Osorio-Méndez, Luis Alberto Gómez-Grosso, Gladis Montoya-Ortiz, Susana Novoa-Herrán and Yohana Domínguez-Romero
Int. J. Mol. Sci. 2025, 26(3), 945; https://doi.org/10.3390/ijms26030945 - 23 Jan 2025
Cited by 1 | Viewed by 2020
Abstract
Cardiovascular diseases and cancer are leading global causes of morbidity and mortality, necessitating advances in diagnosis and treatment. Doxorubicin (Doxo), a potent chemotherapy drug, causes long-term heart damage due to cardiotoxicity. Small extracellular vesicles (sEVs) carry bioactive molecules—such as proteins, lipids, and nucleic [...] Read more.
Cardiovascular diseases and cancer are leading global causes of morbidity and mortality, necessitating advances in diagnosis and treatment. Doxorubicin (Doxo), a potent chemotherapy drug, causes long-term heart damage due to cardiotoxicity. Small extracellular vesicles (sEVs) carry bioactive molecules—such as proteins, lipids, and nucleic acids—that can modulate gene expression and signaling pathways in recipient cells, including cardiomyocytes. Through the delivery of cytokines, microRNAs, and growth factors, sEVs can influence cell survival, which plays a critical role in the development of cardiotoxicity. This study investigates the role of sEVs derived from breast cancer cells treated or not with Doxo and their potential to induce cardiomyocyte damage, thereby contributing to cardiotoxicity. We isolated sEVs from MCF-7 cells treated or not to Doxo using ultracentrifugation and characterized them through Nanoparticle Tracking Analysis (NTA), Scanning Electron Microscopy (SEM), and Western Blotting (WB) for the markers CD63, CD81, and TSG101. We analyzed cytokine profiles using a Multiplex Assay and Cytokine Membrane Array. We exposed Guinea pig cardiomyocytes to different concentrations of sEVs. We assessed their viability (MTT assay), shortening, reactive oxygen species (ROS–DHE dye) production, mitochondrial membrane potential (JC-1 dye), and calcium dynamics (FLUO-4 dye). We performed statistical analyses, including t-tests, ANOVA, Cohen’s d, and η2 to validate the robustness of the results. Treatment of MCF-7 cells with 0.01 μM Doxorubicin resulted in increased sEVs production, particularly after 48 h of exposure (~1.79 × 108 ± 2.77 × 107 vs. ~5.1 × 107 ± 1.28 × 107 particles/mL, n = 3, p = 0.0019). These sEVs exhibited protein profiles in the 130–25 kDa range and 93–123 nm sizes. They carried cytokines including TNF-α, IL-1β, IL-4, IFN-γ, and IL-10. Exposure of cardiomyocytes to sEVs (0.025 μg/mL to 2.5 μg/mL) from both Doxo-treated and untreated cells significantly reduced cardiomyocyte viability, shortened cell length by up to 20%, increased ROS production, and disrupted calcium homeostasis and mitochondrial membrane potential, indicating severe cellular stress and cardiotoxicity. These findings suggest that Doxo enhances sEVs production from breast cancer cells, which plays a key role in cardiotoxicity through their cytokine cargo. The study highlights the potential of these sEVs as biomarkers for early cardiotoxicity detection and as therapeutic targets to mitigate cardiovascular risks in chemotherapy patients. Future research should focus on understanding the mechanisms by which Doxorubicin-induced sEVs contribute to cardiotoxicity and exploring their diagnostic and therapeutic potential to improve patient safety and outcomes in cancer therapy. Full article
(This article belongs to the Special Issue Exosomes and Non-Coding RNA Research in Health and Disease)
Show Figures

Figure 1

13 pages, 865 KiB  
Article
Anticancer-Drug-Related Cardiotoxicity from Adjuvant Goserelin and Tamoxifen Therapy
by Olivia Manfrini, Edina Cenko, Maria Bergami, Jinsung Yoon, Jelena Kostadinovic, Darko Zdravkovic, Marija Zdravkovic and Raffaele Bugiardini
J. Clin. Med. 2025, 14(2), 484; https://doi.org/10.3390/jcm14020484 - 14 Jan 2025
Viewed by 910
Abstract
Background: Breast cancer is a prevalent malignancy with rising incidence globally. Advances in endocrine therapy have improved outcomes for premenopausal women with hormone receptor-positive breast cancer. However, these treatments may induce menopause-like states, potentially elevating cardiovascular risks, including left ventricular (LV) dysfunction. This [...] Read more.
Background: Breast cancer is a prevalent malignancy with rising incidence globally. Advances in endocrine therapy have improved outcomes for premenopausal women with hormone receptor-positive breast cancer. However, these treatments may induce menopause-like states, potentially elevating cardiovascular risks, including left ventricular (LV) dysfunction. This study aims to evaluate the impact of one year of adjuvant endocrine therapy with goserelin and tamoxifen on LV function in premenopausal breast cancer patients. Methods: The ISACS cardiovascular toxicity (NCT01218776) is a pilot multicenter registry of breast cancer patients referred to hospitals for routine surveillance, suspected, or confirmed anticancer-drug-related cardiotoxicity (ADRC). Patients may be enrolled retrospectively (1 year) and prospectively. The pilot phase focused on the available data on combined goserelin and tamoxifen therapy for breast cancer and its impact on LV disfunction at 1-year follow-up. Inverse probability of treatment weighting (IPTW) analysis of the ISACS registry was performed assigning 70 patients to combined endocrine therapy (goserelin and tamoxifen). Controls consisted of 120 patients with no adjuvant combined goserelin and tamoxifen therapy. None of the patients developed distant metastasis. Primary outcome measures were as follows: low LV function in women as defined by a left ventricular ejection fraction (LVEF) < 65% and subclinical LV dysfunction as defined by a 10-percentage point decrease in LVEF. Results: In the overall population, combined goserelin and tamoxifen therapy did not affect the mean LV function compared with controls at 3-, 6-, and 12-month follow-up (65.7 ± 2.7% versus 65.3 ± 2.1%, p value = 0.27; 65.5 ± 2.9% versus 65.1 ± 2.5%, p value = 0.34; 65.0 ± 3.2% versus 64.6 ± 3.1%, p value = 0.29, respectively). The mean LVEF reduction in patients who did or did not receive combination therapy for 12 months was small and approximately similar (1.03 ± 2.5% versus 1.16 ± 2.9%, p value = 0.73). Using IPTW analyses, there were no significant associations between combined therapy and low LV function (risk ratio [RR]: 1.75; 95% CI: 0.71–4.31) or subclinical LV dysfunction (RR: 1.50; 95% CI: 0.35–6.53) compared with controls. Conclusions: One year of endocrine therapy with goserelin and tamoxifen does not cause ADRC in patients with invasive breast cancer. Findings are independent of the severity of the disease. Results may not be definitive without replication in studies with larger sample size. Full article
(This article belongs to the Section Cardiovascular Medicine)
Show Figures

Figure 1

Back to TopTop