Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (40)

Search Parameters:
Keywords = cancer therapy-related cardiovascular toxicity

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
31 pages, 1201 KB  
Review
Aromatase Inhibitors as Adjuvant Therapy in Early Breast Cancer: Insights into Toxicities and Their Management
by Simone Nardin, Beatrice Ruffilli, Tommaso Lupo Landolfo, Giulia Isingrini, Ida Taglialatela, Andrea Delbarba, Francesca D’Avanzo, Valentina Rossi, Eduardo Celentano, Benedetta Conte, Matteo Nardin and Alessandra Gennari
Cancers 2025, 17(17), 2726; https://doi.org/10.3390/cancers17172726 - 22 Aug 2025
Viewed by 824
Abstract
Aromatase inhibitors (AIs), with or without gonadotropin-releasing hormone analogs, are the cornerstone of adjuvant endocrine therapy for women with hormone receptor-positive early-stage breast cancer, offering significant reductions in recurrence risk and improving long-term survival. Their use is frequently accompanied by treatment-related toxicities that [...] Read more.
Aromatase inhibitors (AIs), with or without gonadotropin-releasing hormone analogs, are the cornerstone of adjuvant endocrine therapy for women with hormone receptor-positive early-stage breast cancer, offering significant reductions in recurrence risk and improving long-term survival. Their use is frequently accompanied by treatment-related toxicities that can adversely affect patients’ quality of life (QoL) and adherence to therapy. Commonly reported side effects include vasomotor symptoms, such as hot flashes; musculoskeletal disorders, such as arthralgia and myalgia; mood disorders; and genitourinary discomfort, such as vaginal dryness and dyspareunia. Additionally, AIs are associated with a heightened risk of bone loss, leading to osteoporosis and fractures, and may have implications for cardiovascular health. Effective management of these adverse events is pivotal in maintaining treatment adherence and preserving QoL. Evidence-based strategies to address these toxicities include pharmacological interventions, such as analgesics for joint pain, bisphosphonates or denosumab for bone health, and hormonal or non-hormonal approaches for vasomotor and genitourinary symptoms. Non-pharmacological measures, including physical activity, dietary adjustments, and complementary therapies, can also help mitigate symptoms. This review examines the broad spectrum of AI-associated toxicities, discusses their clinical implications, and provides an overview of evidence-based management strategies. These insights aim to support clinicians in optimizing patient care while minimizing the toxicities of therapy. Full article
(This article belongs to the Special Issue The Role of Aromatase Inhibitors in Breast Cancer Treatment)
Show Figures

Graphical abstract

16 pages, 1248 KB  
Article
Coronary Artery Calcium Score as a Predictor of Anthracycline-Induced Cardiotoxicity: The ANTEC Study
by Anna Borowiec, Patrycja Ozdowska, Magdalena Rosinska, Agnieszka Maria Zebrowska, Sławomir Jasek, Beata Kotowicz, Joanna Waniewska, Hanna Kosela-Paterczyk, Elzbieta Lampka, Katarzyna Pogoda, Zbigniew Nowecki and Jan Walewski
Pharmaceuticals 2025, 18(8), 1102; https://doi.org/10.3390/ph18081102 - 25 Jul 2025
Viewed by 418
Abstract
Background: Many risk factors for cancer therapy-related cardiovascular toxicity overlap with risk factors for atherosclerosis. According to the ESC 2022 Cardio-Oncology Guidelines, coronary computed tomography angiography and coronary artery calcium score are not recommended as part of routine risk assessment prior to oncological [...] Read more.
Background: Many risk factors for cancer therapy-related cardiovascular toxicity overlap with risk factors for atherosclerosis. According to the ESC 2022 Cardio-Oncology Guidelines, coronary computed tomography angiography and coronary artery calcium score are not recommended as part of routine risk assessment prior to oncological treatment. The aim of this study was to prospectively assess the influence of coronary artery calcium score (CAC score) on cancer therapy-related cardiac dysfunction in patients with moderate and high risk of cardiovascular toxicity, qualified for anthracycline treatment. Methods: In all patients, risk factors were collected, laboratory tests, echocardiography with global longitudinal strain (GLS) assessment and coronary artery tomography with coronary artery calcium score were performed. A total of 80 patients were included in the study, of which 77 (96.25%) were followed for an average of 11.5 months. The mean age at baseline was 60.5 years and 72 (93.51%) were women. Results: During observation, five patients (6.49%) died, including two due to heart failure and three due to cancer progression. The majority of patients (59, 76.6%) had breast cancer, 11 (14.3%) were diagnosed with sarcoma and seven (9.1%) with lymphoma. According to the HFA-ICOS risk score, 40 patients (51.9%) were classified as moderate risk (MR), and 37 patients (48.1%) as high risk (HR) for cancer therapy-related cardiovascular toxicity. A CAC score greater than 100 was calculated in 17 (22.1%) patients and greater than 400 in three (3.9%) patients. The CAC score above zero was more common in older patients and in patients classified as high risk (p < 0.001). There was also a significant association between CAC score and hypertension, hyperlipidemia, chronic kidney disease, and the level of NT-proBNP. During 12-month follow-up, mild CTRCD occurred in 38 (49.4%) patients, moderate CTRCD was diagnosed in seven (9.1%), and severe in three (3.9%) patients. In the univariable analysis, CTRCD was more common in the high-risk group (p = 0.005) and in patients with a CAC score greater than zero (p = 0.036). In multivariable analysis, the incidence of CTRCD remains higher in the CAC score > 0 group, even after adjusting for age, hypertension, and hyperlipidemia. In this study group, the CTRCD rates increased with the HFA-ICOS risk score. Conclusions: In moderate and high-risk patients, a coronary artery calcium score greater than zero was identified as a significant risk factor for the development of cancer therapy-related cardiac dysfunction during anthracycline-based treatment. Furthermore, the HFA-ICOS risk score demonstrated good correlation with the incidence of CTRCD in this study, supporting its validity as a predictive tool in patients receiving anthracycline therapy. Full article
Show Figures

Figure 1

12 pages, 663 KB  
Article
Renal Function Deterioration in Postoperative (Adjuvant) Chemotherapy for Colon Cancer—Real-Life Data
by Aleksandra Gładyś, Sylwia Kozak, Aleksander Jerzy Owczarek, Ewa Cedrych, Zofia Irena Niemir, Stanisław Łącki-Zynzeling, Anna Chudek, Izolda Mrochen-Domin, Iwona Gisterek-Grocholska and Jerzy Chudek
Curr. Oncol. 2025, 32(6), 351; https://doi.org/10.3390/curroncol32060351 - 13 Jun 2025
Viewed by 604
Abstract
The knowledge concerning mild-to-moderate renal toxicity of adjuvant chemotherapy (CTH) in colon cancer patients is scarce. We retrospectively evaluated changes in the estimated glomerular filtration rate (eGFR) after three months of adjuvant treatment and the overall renal risk of the 6-month regimen in [...] Read more.
The knowledge concerning mild-to-moderate renal toxicity of adjuvant chemotherapy (CTH) in colon cancer patients is scarce. We retrospectively evaluated changes in the estimated glomerular filtration rate (eGFR) after three months of adjuvant treatment and the overall renal risk of the 6-month regimen in 145 patients who completed three months of therapy at three oncological centers. A decrease in eGFR of at least 1.5 mL/min/1.73 m2 after three months and 3.0 mL/min/1.73 m2 after six months was considered relevant in terms of kidney-related cardiovascular risk. Out of 114 patients who completed a 6-month regimen, kidney function deterioration occurred in 62 (54.4%) after 3 months and in 54 (47.4%) after 6 months. Age ≥ 70 years (RR = 2.66; 95% CI: 1.15–6.16) and diabetes (RR = 2.52; 95% CI: 0.98–6.45) were risk factors for kidney outcomes during the first three months of CTH. However, renal function decline during the first three months did not increase the risk of further deterioration on CTH continuation. In conclusion, older age and diabetes are factors increasing the risk of renal function deterioration during adjuvant CTH in colon cancer patients without preexisting chronic kidney disease. However, the decline during the first three months does not allow for predicting further changes under continued adjuvant therapy. Full article
Show Figures

Figure 1

28 pages, 727 KB  
Review
Potential New Applications of Sodium–Glucose Cotransporter-2 Inhibitors Across the Continuum of Cancer-Related Cardiovascular Toxicity
by Agnieszka Maria Zebrowska and Anna Borowiec
Pharmaceuticals 2025, 18(6), 857; https://doi.org/10.3390/ph18060857 - 9 Jun 2025
Viewed by 1000
Abstract
Sodium–glucose cotransporter-2 inhibitors (SGLT2i), initially developed for the management of type 2 diabetes mellitus, have demonstrated significant nephroprotective and cardioprotective effects. These benefits have led to their inclusion in heart failure (HF) management guidelines, irrespective of glycemic status and left ventricular ejection fraction [...] Read more.
Sodium–glucose cotransporter-2 inhibitors (SGLT2i), initially developed for the management of type 2 diabetes mellitus, have demonstrated significant nephroprotective and cardioprotective effects. These benefits have led to their inclusion in heart failure (HF) management guidelines, irrespective of glycemic status and left ventricular ejection fraction (LVEF). Various anticancer therapies, particularly anthracyclines, are associated with substantial cardiotoxicity risks, resulting in cancer therapy-related cardiovascular toxicity (CTR-CVT). Promising evidence from preclinical and observational studies indicates that SGLT2i may mitigate cardiotoxic effects of cancer therapy by alleviating LVEF decline, reducing HF incidence and hospitalizations, and lowering overall mortality. Moreover, improved survival has been reported in patients with various malignancies. The current review explores the potential applications of SGLT2i in the prevention of CTR-CVT, highlights their possible mechanisms of cardioprotection, discusses the published evidence, and emphasizes the need for the results from ongoing randomized controlled trials to establish SGLT2i efficacy and safety in cardio-oncology patients. Full article
Show Figures

Figure 1

22 pages, 2743 KB  
Review
SGLT2 Inhibitors in Cancer Patients: A Comprehensive Review of Clinical, Biochemical, and Therapeutic Implications in Cardio-Oncology
by Alessandra Greco, Maria Laura Canale, Vincenzo Quagliariello, Stefano Oliva, Andrea Tedeschi, Alessandro Inno, Marzia De Biasio, Irma Bisceglia, Luigi Tarantini, Nicola Maurea, Alessandro Navazio, Marco Corda, Attilio Iacovoni, Furio Colivicchi, Massimo Grimaldi and Fabrizio Oliva
Int. J. Mol. Sci. 2025, 26(10), 4780; https://doi.org/10.3390/ijms26104780 - 16 May 2025
Cited by 1 | Viewed by 1775
Abstract
Patients with active cancer and cancer survivors are at a markedly increased risk for developing cardiovascular comorbidities, including heart failure, coronary artery disease, and renal dysfunction, which are often compounded by the cardiotoxic effects of cancer therapies. This heightened cardiovascular vulnerability underscores the [...] Read more.
Patients with active cancer and cancer survivors are at a markedly increased risk for developing cardiovascular comorbidities, including heart failure, coronary artery disease, and renal dysfunction, which are often compounded by the cardiotoxic effects of cancer therapies. This heightened cardiovascular vulnerability underscores the urgent need for effective, safe, and evidence-based cardioprotective strategies to reduce both cardiovascular morbidity and mortality. Sodium-glucose cotransporter 2 inhibitors (SGLT2is), a class of drugs originally developed for the treatment of type 2 diabetes, have demonstrated significant cardiovascular and renal benefits in high-risk populations, independent of glycemic control. Among the currently available SGLT2i, such as empagliflozin, canagliflozin, dapagliflozin, and sotagliflozin, there is growing evidence supporting their role in reducing major adverse cardiovascular events (MACEs), hospitalization for heart failure, and the progression of chronic kidney disease. Recent preclinical and clinical data suggest that SGLT2is exert cardioprotective effects through multiple mechanisms, including the modulation of inflammasome activity, specifically by reducing NLRP3 inflammasome activation and MyD88-dependent signaling, which are critical drivers of cardiac inflammation and fibrosis. Moreover, SGLT2is have been shown to enhance mitochondrial viability in cardiac cells, promoting improved cellular energy metabolism and function, thus mitigating cardiotoxicity. This narrative review critically evaluates the emerging evidence on the cardiorenal protective mechanisms of SGLT2is, with a particular focus on their potential role in cardio-oncology. We explore the common pathophysiological pathways between cardiovascular dysfunction and cancer, the molecular rationale for the use of SGLT2is in cancer patients, and the potential benefits in both primary and secondary prevention of cardiovascular toxicity related to oncological treatments. The aim is to propose a therapeutic paradigm utilizing SGLT2is to reduce cardiovascular mortality, MACE, and the burden of cardiotoxicity in high-risk oncology patients, fostering an integrated approach to cardio-oncology care. Full article
Show Figures

Graphical abstract

23 pages, 3997 KB  
Review
Anticancer Mechanisms of Ginsenoside Compound K: A Review
by Yu-Po Lee, Hui-Ting Chan, Tzu-Hsuan Li, Lichieh (Julie) Chu, Sheau-Long Lee, Yu-Quan Chang and Robert YL Wang
Diseases 2025, 13(5), 143; https://doi.org/10.3390/diseases13050143 - 5 May 2025
Viewed by 1313
Abstract
Cancer, also known as malignant tumors, is formed due to abnormal mutations and the proliferation of human cells. Cancer cells not only demonstrate accelerated proliferation but also show robust invasive and metastatic potential, disseminating from a primary affected region of the body to [...] Read more.
Cancer, also known as malignant tumors, is formed due to abnormal mutations and the proliferation of human cells. Cancer cells not only demonstrate accelerated proliferation but also show robust invasive and metastatic potential, disseminating from a primary affected region of the body to multiple areas and potentially culminating in organ dysfunction or failure, thereby jeopardizing the individual’s life. The rapid growth of the biopharmaceutical market has given rise to numerous novel medicines, thereby precipitating a paradigm shift in contemporary drug development methodologies. This modification is focused on identifying methodologies that can effectively target cancerous cells while minimizing damage to normal cells. There is an increasing societal movement that supports the utilization of natural ingredients derived from plants. In recent years, traditional herbal medicine has experienced a surge in popularity within the global cancer market. In comparison with the use of more toxic chemotherapy methods, there has been an increasing focus on advanced therapies that exhibit reduced side effects. Ginsenoside compound K (CK) is derived from the natural components in ginseng through biotransformation. The utilization of CK in cancer research is a practice engaged in by numerous scientists. The underlying rationale is that CK exhibits a multitude of effects within the realm of cancer research, including but not limited to the mitigation of inflammation, the suppression of cancerous cell proliferation, and the safeguarding of cardiovascular, hepatic, and renal functions. This review methodically identifies and organizes CK-related journals according to the following key points of cancer treatment: the effects on cancer cells themselves, angiogenesis inhibition, modulation of immune response to identify cancer cells, and inflammation regulation. The intricate interplay between ginsenoside CK and cells is elucidated through a graphical representation. The present review focuses on the results of CK in in vitro tests. It is our hope that the present article will aid future studies on the results of CK in vivo tests, clarify the correlation between cellular mechanisms in vivo and in vitro tests, and assist in the development of drugs. Full article
Show Figures

Figure 1

38 pages, 8552 KB  
Review
Enzyme-Based Anti-Inflammatory Therapeutics for Inflammatory Diseases
by Kannan Badri Narayanan
Pharmaceutics 2025, 17(5), 606; https://doi.org/10.3390/pharmaceutics17050606 - 2 May 2025
Cited by 1 | Viewed by 4292
Abstract
Inflammation is a multifaceted biological response of the immune system against various harmful stimuli, including pathogens (such as bacteria and viruses), cellular damage, toxins, and natural/synthetic irritants. This protective mechanism is essential for eliminating the cause of injury, removing damaged cells, and initiating [...] Read more.
Inflammation is a multifaceted biological response of the immune system against various harmful stimuli, including pathogens (such as bacteria and viruses), cellular damage, toxins, and natural/synthetic irritants. This protective mechanism is essential for eliminating the cause of injury, removing damaged cells, and initiating the repair process. While inflammation is a fundamental component of the body’s defense and healing process, its dysregulation can lead to pathological consequences, contributing to various acute and chronic diseases, such as autoimmune disorders, cancer, metabolic syndromes, cardiovascular diseases, neurodegenerative conditions, and other systemic complications. Generally, non-steroidal anti-inflammatory drugs (NSAIDs), corticosteroids, disease-modifying anti-rheumatic drugs (DMARDs), antihistamines, biologics, and colchicine are used as pharmacological agents in the management of inflammatory diseases. However, these conventional treatments often have limitations, including adverse side effects, long-term toxicity, and drug resistance. In contrast, enzyme-based therapeutics have emerged as a promising alternative due to their high specificity, catalytic efficiency, and ability to modulate inflammatory pathways with reduced side effects. These enzymes function by scavenging reactive oxygen species (ROS), inhibiting cytokine transcription, degrading circulating cytokines, and blocking cytokine release by targeting exocytosis-related receptors. Additionally, their role in tissue repair and regeneration further enhances their therapeutic potential. Most natural anti-inflammatory enzymes belong to the oxidoreductase class, including catalase and superoxide dismutase, as well as hydrolases such as trypsin, chymotrypsin, nattokinase, bromelain, papain, serratiopeptidase, collagenase, hyaluronidase, and lysozyme. Engineered enzymes, such as Tobacco Etch Virus (TEV) protease and botulinum neurotoxin type A (BoNT/A), have also demonstrated significant potential in targeted anti-inflammatory therapies. Recent advancements in enzyme engineering, nanotechnology-based enzyme delivery, and biopharmaceutical formulations have further expanded their applicability in treating inflammatory diseases. This review provides a comprehensive overview of both natural and engineered enzymes, along with their formulations, used as anti-inflammatory therapeutics. It highlights improvements in stability, efficacy, and specificity, as well as minimized immunogenicity, while discussing their mechanisms of action and clinical applications and potential future developments in enzyme-based biomedical therapeutics. Full article
(This article belongs to the Special Issue Medical Applications of Biologic Drugs)
Show Figures

Figure 1

12 pages, 1408 KB  
Article
Interleukin-6 in Anthracycline-Related Cardiac Dysfunction: A Comparison with Myeloperoxidase and TNF-Alpha
by Anna Borowiec, Patrycja Ozdowska, Magdalena Rosinska, Agnieszka Maria Zebrowska, Sławomir Jasek, Beata Kotowicz, Agata Makowka, Joanna Waniewska, Hanna Kosela-Paterczyk, Elzbieta Lampka, Katarzyna Pogoda, Zbigniew Nowecki and Jan Walewski
Int. J. Mol. Sci. 2025, 26(9), 4071; https://doi.org/10.3390/ijms26094071 - 25 Apr 2025
Viewed by 636
Abstract
Interleukin-6, myeloperoxidase, and tumor necrosis factor alpha are proinflammatory cytokines that play a role both in cardiovascular and oncological diseases. The study aimed to prospectively investigate the clinical value of interleukin-6, myeloperoxidase, and tumor necrosis factor alpha as potential biomarkers of cancer therapy-related [...] Read more.
Interleukin-6, myeloperoxidase, and tumor necrosis factor alpha are proinflammatory cytokines that play a role both in cardiovascular and oncological diseases. The study aimed to prospectively investigate the clinical value of interleukin-6, myeloperoxidase, and tumor necrosis factor alpha as potential biomarkers of cancer therapy-related cardiac dysfunction (CTRCD) in patients receiving anthracycline treatment. A total of 80 patients were included, with 77 (96.25%) followed for an average for 11.5 months. The mean age at baseline was 60.5 years, and 72 (93.51%) were women. Clinical risk factors were documented for all patients, and laboratory assessments, including measurements of IL-6, MPO, and TNF-α, were conducted. All participants also underwent echocardiography with assessment of global longitudinal strain (GLS). In the study group, coronary CT angiography with coronary artery calcium (CAC) score calculation was performed once at the beginning of the study. During observation, any degree of CTRCD was diagnosed in 48 (62.4%) patients. Mild CTRCD occurred in 38 (49.4%) patients, moderate CTRCD was diagnosed in 7 (9.1%), and severe in 3 (3.9%). In patients with high baseline risk, IL-6 levels were significantly elevated compared to those with moderate risk (p = 0.018). Higher levels of IL-6 were found to correlate with an increased grade of CTRCD. In a multivariate repeated measures model of the biomarkers studied, only a higher level of IL-6 was significantly associated with the diagnosis of CTRCD. Among the novel biomarkers studied, we found evidence for IL-6 for its potential use in the detection of cardiac dysfunction related to cancer therapy in patients treated with anthracyclines. Full article
(This article belongs to the Special Issue Heart Failure Risk Factors and Therapies on a Molecular Basis)
Show Figures

Figure 1

21 pages, 356 KB  
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 2 | Viewed by 1745
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)
22 pages, 1598 KB  
Article
Bevacizumab—Insights from EudraVigilance Database on the Assessments of the Safety Profile of Monoclonal Antibodies Used as Targeted Cancer Treatment
by Razvan Constantin Vonica, Anca Butuca, Claudiu Morgovan, Manuela Pumnea, Remus Calin Cipaian, Adina Frum, Carmen Maximiliana Dobrea, Andreea Loredana Vonica-Tincu, Aliteia-Maria Pacnejer, Steliana Ghibu, Florina Batar and Felicia Gabriela Gligor
Pharmaceuticals 2025, 18(4), 501; https://doi.org/10.3390/ph18040501 - 30 Mar 2025
Cited by 1 | Viewed by 998
Abstract
Background/Objectives: Worldwide, colon cancer is a major cause of cancer-related mortality, with an increasing incidence influenced by genetic, environmental, and lifestyle factors. Despite advances in diagnosis and personalized treatments, challenges remain in improving patient prognosis, particularly in metastatic colorectal cancer (mCRC). Bevacizumab (BEV), [...] Read more.
Background/Objectives: Worldwide, colon cancer is a major cause of cancer-related mortality, with an increasing incidence influenced by genetic, environmental, and lifestyle factors. Despite advances in diagnosis and personalized treatments, challenges remain in improving patient prognosis, particularly in metastatic colorectal cancer (mCRC). Bevacizumab (BEV), a monoclonal antibody, is widely used in colorectal cancer treatment. This study aimed to analyze adverse events associated with BEV compared with other therapies based on data from the EudraVigilance (EV) database. Methods: A descriptive and disproportionality analysis was conducted on signals reported in the EV database related to BEV. The study included comparisons with other antineoplastic treatments, such as chemotherapy, targeted therapy, and immunotherapy. Patient demographics, severity of adverse drug reactions (ADRs), and distribution patterns were analyzed to assess the safety profile of BEV in colorectal cancer treatment. Results: The majority of the signals for BEV were from patients aged 18–64 years (39.42%) and 65–85 years (34.08%). Hypertension, thromboembolism, proteinuria, and gastrointestinal disorders have been the most frequently reported. Serious ADRs, including gastrointestinal perforations, hemorrhage, and arterial thromboembolism, were observed in 93.74% of Individual Case Safety Reports. BEV was associated with a higher likelihood of vascular and endocrine disorders compared with chemotherapy and other targeted therapies. Immunotherapy was linked to increased immunological ADRs, while BEV demonstrated fewer immune-related toxicities. Conclusions: Continuous monitoring is necessary to optimize patient management, particularly in elderly patients or those with cardiovascular comorbidities. Understanding BEV’s safety profile allows for better personalization of treatment strategies, minimizing risks while enhancing therapeutic outcomes. Full article
(This article belongs to the Section Pharmacology)
Show Figures

Figure 1

16 pages, 287 KB  
Review
Genetic Background in Patients with Cancer Therapy-Induced Cardiomyopathy
by Luca Fazzini, Nicola Campana, Stefano Cossu, Martino Deidda, Cristina Madaudo, Vincenzo Quagliariello, Nicola Maurea, Daniela Di Lisi, Giuseppina Novo, Concetta Zito and Christian Cadeddu Dessalvi
J. Clin. Med. 2025, 14(4), 1286; https://doi.org/10.3390/jcm14041286 - 15 Feb 2025
Viewed by 1347
Abstract
Emerging evidence indicates that specific genetic variants are associated with an increased risk of toxicity from anticancer treatments and cancer-related cardiovascular complications. These genetic factors influence drug metabolism, efficacy, and susceptibility to adverse effects. For cancer patients, the genetic background can have two [...] Read more.
Emerging evidence indicates that specific genetic variants are associated with an increased risk of toxicity from anticancer treatments and cancer-related cardiovascular complications. These genetic factors influence drug metabolism, efficacy, and susceptibility to adverse effects. For cancer patients, the genetic background can have two major cardiovascular implications, namely therapy-related cardiotoxicity and cancer-related cardiovascular complications. Baseline risk stratification is essential to identify higher-risk individuals and ensure they receive appropriate preventive and therapeutic interventions and more frequent follow-up. Current guidelines recommend stratification based on cardiovascular risk factors, but these factors alone cannot accurately define individual risk. Genetic background has been shown to enhance risk stratification. Beyond rare genetic variants, recent genome-wide association studies have identified single nucleotide polymorphisms implicated in cancer therapy toxicity. Despite their current limitations, polygenic risk scores are expected to play a significant role in risk stratification. This review aims to summarize the current evidence on the role of the genetic background of patients with cancer treated with potentially cardiotoxic drugs who develop cardiotoxicity, aiming to provide insights to refine risk stratification further and tailor the management of these patients. Full article
Show Figures

Graphical abstract

15 pages, 1718 KB  
Article
Cardiopulmonary Functional Profiles in Cancer Therapy-Related Cardiac Dysfunction Among Patients with Non-Hodgkin Lymphoma
by Daniela Bursacovschi, Valeriu Revenco, Maria Robu and Oleg Arnaut
Diagnostics 2025, 15(4), 417; https://doi.org/10.3390/diagnostics15040417 - 8 Feb 2025
Viewed by 1193
Abstract
Background: Non-Hodgkin lymphoma (NHL) is associated with significant cardiovascular risks due to treatment-related toxicities, including cancer therapy-related cardiac dysfunction (CTRCD). While multimodality imaging, particularly echocardiography, is pivotal in monitoring cardiac function, the prognostic role of cardiopulmonary exercise testing (CPET) in predicting CTRCD remains [...] Read more.
Background: Non-Hodgkin lymphoma (NHL) is associated with significant cardiovascular risks due to treatment-related toxicities, including cancer therapy-related cardiac dysfunction (CTRCD). While multimodality imaging, particularly echocardiography, is pivotal in monitoring cardiac function, the prognostic role of cardiopulmonary exercise testing (CPET) in predicting CTRCD remains underexplored. Methods: The prospective study enrolled 127 NHL patients, 72 men (56.7%) and 55 women (43.3%), with a median age of 62 years (range 34–83 years). The patients were assessed before initiating antitumor treatment and at six months follow-up using echocardiography and cardiopulmonary exercise testing. Results: Asymptomatic CTRCD occurred in 14.2% of NHL patients at six months of treatment. Patients with CTRCD exhibited significantly lower median work rates, volume of oxygen (VO2) at the anaerobic threshold, and O2 consumption efficiency, reflecting compromised metabolic and functional performance. Baseline peak oxygen consumption (VO2 peak) positively correlated with left ventricle ejection fraction (LVEF) at six months, while VO₂ peak < 14 mL/kg/min was negatively associated with LVEF. Conclusions: Asymptomatic CTRCD was identified in 14.2% of NHL patients at six months, with lower work rates, VO2 at the anaerobic threshold, and O2 consumption efficiency, indicating impaired performance. Baseline peak oxygen consumption correlated positively with LVEF, highlighting CPET’s potential for early CTRCD risk assessment. Full article
(This article belongs to the Special Issue Diagnosis and Prognosis of Heart Disease, 2nd Edition)
Show Figures

Figure 1

13 pages, 865 KB  
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 983
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

20 pages, 1074 KB  
Article
A Real-World Comparison of the Safety Profile for Immune Checkpoint Inhibitors in Oncology Patients
by Abdulrahman Alwhaibi, Miteb A. Alenazi, Sultan Alghadeer, Wael Mansy, Reem A. Alsaif, Nawaf E. Abualreesh, Rakan J. Alanazi, Abdullah Alroumi and Saleh A. Alanazi
J. Clin. Med. 2025, 14(2), 388; https://doi.org/10.3390/jcm14020388 - 9 Jan 2025
Cited by 1 | Viewed by 2078
Abstract
Background/Objectives: Owing to the growing use of immune checkpoint inhibitors (ICIs) in the treatment of cancer, a wide spectrum of toxicity has arisen among cancer patients. Yet, limited ICI toxicity-related research is currently conducted in our region. Methods: This is a retrospective observational [...] Read more.
Background/Objectives: Owing to the growing use of immune checkpoint inhibitors (ICIs) in the treatment of cancer, a wide spectrum of toxicity has arisen among cancer patients. Yet, limited ICI toxicity-related research is currently conducted in our region. Methods: This is a retrospective observational study conducted on adult cancer patients who received at least one cycle of ICI single therapy. Toxicity profiles of different ICI monotherapies were described and compared, and their association with different risk factors was assessed. SPSS version 28 was used for statistical analyses, and p < 0.05 was considered statistically significant. Results: A total of 428 patients were treated with anti-PD1 (nivolumab [n = 221, 51.6%] and pembrolizumab [n = 126, 29.5%]) or anti-PD-L1 (atezolizumab [n = 78, 18.2%] and durvalumab [n = 3, 0.7%]). Pneumonia was the most common complication (10%), followed by acute kidney injury (AKI; 8.2%) and hepatitis (7.9%). The proportion of hepatitis cases was significantly higher among atezolizumab compared to nivolumab-, pembrolizumab-, and durvalumab-treated patients (17.95% vs. 7.7% vs. 2.4% vs. 0.0%, respectively; p < 0.001). Gastrointestinal complication (colitis) was detected in 3.3% of patients with a significant difference between treatment groups (4.5%, 1.6%, 1.3%, and 33.3% in nivolumab, pembrolizumab, atezolizumab, and durvalumab, respectively; p = 0.008). Cardiac complications occurred in 1.2% of patients with a significant difference between treatment groups (0.5% in the nivolumab, 3.8% in the atezolizumab, 33.3% in the durvalumab, and none in the pembrolizumab groups (p < 0.001)). Musculoskeletal side effects, including both arthralgia and fatigue, were the most-reported side effects by 39.5% of patients, with significantly higher arthralgia complainers only in nivolumab (7.7%) compared to other treatment groups (0%, 2.6%, and 0% in pembrolizumab, atezolizumab, and durvalumab, respectively, p = 0.007). Hepatic, cardiovascular, hematological, respiratory, renal, gastrointestinal complications, thyroid complications, and dermatological side effects were found to occur on weeks 6, 7.5, 8, 8, 10, 10, 10.5, and 12 after treatment initiation, respectively, with no significant difference between treatment groups. Despite that, hepatitis and AKI tended to occur earlier with atezolizumab (week 2, p = 0.084) and pembrolizumab (week 2, p = 0.062), respectively, compared to their comparators. The female gender and a history of hepatitis were found to increase the odds of hepatic complication with anti-PD1 or anti-PD-L1 use [OR = 2.71; 95% CI 1.07–6.85, OR = 11.14; 95% CI 3.46–35.88, respectively]. Previous exposure to cancer therapy only was found to increase the odds of developing pneumonia among the treated patients [OR = 3.08; 95% CI 1.12–8.85]. Having hematological malignancy influenced the odds of hematological complications positively (either neutropenia or thrombocytopenia) compared to solid malignancies when patients were treated with anti-PD1 or anti-PD-L1 [OR = 17.18; 95% CI 4.06–72.71]. Finally, the female gender was found to positively associate with the odds of nausea/vomiting and fatigue secondary to anti-PD1 or anti-PD-L1 administration [OR = 2.08; 95% CI 1.34–3.21, OR = 1.65; 95% CI 1.09–2.51, respectively]. On the other hand, previous exposure to cancer therapy was found to reduce the risk of having arthralgia with anti-PD1 or anti-PD-L1 administration [OR = 0.344; 95% CI 0.121–0.974]. Conclusions: Treatment with anti-PD1 or anti-PD-L1 was associated with a spectrum of complications and side effects. Several risk factors have been identified to impact their occurrence. ICI toxicities and risk factors influencing their odds should be recognized and considered in clinical practice, as this could help in individualizing therapeutics regimens and avoiding treatment interruption. Full article
Show Figures

Figure 1

33 pages, 3174 KB  
Review
Oxidative Stress and Age-Related Tumors
by Emma Di Carlo and Carlo Sorrentino
Antioxidants 2024, 13(9), 1109; https://doi.org/10.3390/antiox13091109 - 13 Sep 2024
Cited by 31 | Viewed by 5037
Abstract
Oxidative stress is the result of the imbalance between reactive oxygen and nitrogen species (RONS), which are produced by several endogenous and exogenous processes, and antioxidant defenses consisting of exogenous and endogenous molecules that protect biological systems from free radical toxicity. Oxidative stress [...] Read more.
Oxidative stress is the result of the imbalance between reactive oxygen and nitrogen species (RONS), which are produced by several endogenous and exogenous processes, and antioxidant defenses consisting of exogenous and endogenous molecules that protect biological systems from free radical toxicity. Oxidative stress is a major factor in the aging process, contributing to the accumulation of cellular damage over time. Oxidative damage to cellular biomolecules, leads to DNA alterations, lipid peroxidation, protein oxidation, and mitochondrial dysfunction resulting in cellular senescence, immune system and tissue dysfunctions, and increased susceptibility to age-related pathologies, such as inflammatory disorders, cardiovascular and neurodegenerative diseases, diabetes, and cancer. Oxidative stress-driven DNA damage and mutations, or methylation and histone modification, which alter gene expression, are key determinants of tumor initiation, angiogenesis, metastasis, and therapy resistance. Accumulation of genetic and epigenetic damage, to which oxidative stress contributes, eventually leads to unrestrained cell proliferation, the inhibition of cell differentiation, and the evasion of cell death, providing favorable conditions for tumorigenesis. Colorectal, breast, lung, prostate, and skin cancers are the most frequent aging-associated malignancies, and oxidative stress is implicated in their pathogenesis and biological behavior. Our aim is to shed light on the molecular and cellular mechanisms that link oxidative stress, aging, and cancers, highlighting the impact of both RONS and antioxidants, provided by diet and exercise, on cellular senescence, immunity, and development of an antitumor response. The dual role of ROS as physiological regulators of cell signaling responsible for cell damage and diseases, as well as its use for anti-tumor therapeutic purposes, will also be discussed. Managing oxidative stress is crucial for promoting healthy aging and reducing the risk of age-related tumors. Full article
(This article belongs to the Special Issue Reactive Nitrogen Species (RNS) and Redox Signaling in Tumors)
Show Figures

Figure 1

Back to TopTop