Overview of Oncology: Drug-Induced Cardiac Toxicity
Abstract
:1. Introduction
Importance of Recognizing Cardiotoxicity in Cancer Treatment
2. Background on Cancer Treatments: Evolution of Chemotherapy and Targeted Therapies
2.1. Patient Survival Rates and Quality of Life
2.2. Definition of Cardiotoxicity: Variability in Definitions and Clinical Implications
- Concomitant signs or symptoms of congestive heart failure CHF characterized by a reduction in LVEF of at least 5% to below 55%.
- LVEF of at least 10% decrease to below 55% in the absence of clinical signs or symptoms [22].
- A drop of LVEF by a value of >10 percentage points to a value of <50%.
- >20 percentage points absolute decrease in LVEF.
- Any LVEF declines to <50%.
2.3. Cardiotoxicity Mechanism and Types
2.3.1. Mechanisms of Cardiotoxicity
- Oxidative Stress: Cardiac myocytes are similarly damaged by oxidative damage from the generation of reactive oxygen species and free radicals by anthracyclines [24].
- Mitochondrial Dysfunction: These drugs could influence their mitochondrial function and, thus, energy production in the cardiomyocytes [24].
- DNA Damage: Anthracyclines may intercalate into DNA synthesis, causing breaks and inhibition of DNA repair mechanisms [24].
- Calcium Dysregulation: Chemotherapy with anthracyclines can affect calcium homeostasis, causing cardiac contractility [24].
- Apoptosis Induction: Programmed cell death can be triggered in cardiomyocytes by anthracyclines [24].
- Tyrosine kinase inhibitors: tyrosine kinase inhibitors can disrupt important normal cellular signaling pathways needed for cardiac function [25]. Imatinib, for example, inhibits the BCR-ABL tyrosine kinase and other kinases, including c-Abl, which play crucial roles in cardiac function. Inhibition of c-Abl can disrupt mitochondrial integrity and function, leading to cardiomyocyte apoptosis and subsequent cardiac dysfunction [26].
- Immune checkpoint inhibitors: Another notable class of antitumoral drugs, immune checkpoint inhibitors (ICIs), specifically inhibitors of anticytotoxic T-lymphocyte associated antigen 4 and anti-PD/PD-L1 (anti-programmed death receptor and receptor ligand-1), are the mainstay of treatment for many malignancies. The mechanism of immune-related adverse events is mainly related to the aberrant activity of autoreactive T cells, which leads to autoimmune inflammatory reactions affecting both on-target and off-target organs [29]. Immune-related adverse events have variable presentations, ranging from asymptomatic laboratory findings to fulminant life-threatening diseases. There are specific guidelines for treating these immune-related adverse events, depending on the severity of toxicity (graded from 1 to 4) [30].
- Anti-angiogenic drugs: another class of anti-tumoral drugs, called anti-angiogenic drugs, particularly those targeting the vascular endothelial growth factor (VEGF) pathway, have been instrumental in cancer therapy but are associated with cardiotoxic effects through various mechanisms. Inhibition of VEGF signaling can lead to hypertension, as VEGF plays a crucial role in maintaining endothelial function and nitric oxide production; its inhibition results in vasoconstriction and elevated blood pressure [32].
2.3.2. Types of Cardiotoxicity
- Arrhythmias: General chemotherapeutics can affect the complex rhythms, from benign to life-threatening [25].
- Myocardial Ischemia: For example, fluoropyrimidines can induce coronary vasospasm, resulting in ischemia [25].
- Hypertension: VEGF inhibitors have specifically been shown to exacerbate (or in some cases, cause) hypertension [25].
- Thromboembolism: Some chemotherapies are associated with an increased risk of thromboembolic events [25].
2.4. Acute vs. Chronic Cardiotoxicity
2.4.1. Acute Cardiotoxicity
2.4.2. Chronic Cardiotoxicity
2.5. Immediate Effects Versus Long-Term Consequences
2.6. Cardiotoxicity Pathways
- Oxidative Stress: Reactive oxygen species (ROS) produced by many chemotherapeutic agents (e.g., anthracyclines) have been shown to damage cellular components (proteins, lipids, DNA). Cardiomyocyte death can result from this oxidative stress and a malfunctioning mitochondrial system [24].
- Mitochondrial Dysfunction: Anthracyclines are used as drugs that directly impair mitochondrial function to reduce energy production in cardiomyocytes closing cellular dysfunction or death [24].
- DNA Damage: Anthracyclines are drugs which intercalate with DNA and create breaks in the DNA, disturbing DNA repair mechanisms. Apoptosis or senescence of cardiac cells [27].
- Calcium Homeostasis Disruption: Chemotherapeutics can alter calcium handling in cardiomyocytes, thus reducing contractility and potentially causing arrhythmias [22].
- Vascular Toxicity: Consequently, certain targeted therapies such as VEGF inhibitors may lead to endothelial dysfunction and indeed hypertension, in turn affecting cardiac function [37].
- Immune-Mediated Damage: Immune mediated myocarditis downstream of immune checkpoint inhibitors may result in severe cardiac complications [37].
2.6.1. Short-Term vs. Long-Term Consequences
2.6.2. Short Term Effects
- Acute Arrhythmias: Some drugs, such as anthracyclines, can provoke immediate rhythm disturbances, from benign to lethal [22].
- Acute Left Ventricular Dysfunction: The activity can occur shortly after or during treatment, and tends to reverse when the drug is discontinued [24].
- Myocarditis: Acute myocarditis has been particularly associated with immune checkpoint inhibitors and appears as early as weeks after treatment initiation [37].
- Acute Hypertension: Hypertension related to VEGF inhibitors can occur rapidly and may need to be managed immediately [37].
2.6.3. Long-Term Consequences
2.7. Reversible vs. Irreversible Cardiotoxicity
- Type I (Irreversible): This type had typically been linked with anthracyclines, and was thought to have caused permanent cardiac damage. Traditionally, cardiac dysfunction previously believed to be basically irreversible and having a poor prognosis [38].
- Type II (Reversible): This type was often linked to targeted therapies such as trastuzumab (anti-HER2 receptor monoclonal antibody) and was often reversible with discontinuation of the drug with recovery of cardiac function to normal [38].
2.8. Recovery in Anthracycline Induced Cardiotoxicity
- Time-Dependent Recovery: An inverse relationship between the time of heart failure therapy initiation versus improvement of left ventricular ejection fraction (LVEF) was seen in a study of 201 patients with anthracycline-induced cardiotoxicity. Recovery of the LVEF at 2 months following completion of chemotherapy was 64%, while for those beyond 6 months, recovery was significantly reduced [24,38,39].
- Early Intervention: There have been promising results (close monitoring of LVEF post-chemotherapy combined with early treatment with ACE inhibitors and beta-blockers) in LVEF after chemotherapy. When treated early, 82% of patients with cardiotoxicity achieve normalization of cardiac function in large study (n = 2625). However, only 11% of these patients regained their baseline LVEF [24,38,39], but it is likely that some degree of permanent damage remains.
2.9. Long Term Consequences of Permanent Damage
- Subclinical Damage: Despite normalization of LVEF, subclinical cardiac abnormalities may persist and continue to be at risk of future cardiovascular events [38].
2.10. Other Oncology Drugs: Reversibility
2.11. Implications for Management
2.12. Molecular Mechanisms
2.13. Role of Oxidative Stress and Inflammation
- Lipid peroxidation: Damage of cellular membranes, including those of mitochondria [24], can be caused by ROS induced lipid peroxidation.
- Iron-mediated damage: One group of drugs does not tolerate being around iron, as anthracyclines can bind to iron and generate highly reactive hydroxyl radicals in the presence of oxygen [24].
2.14. Inflammation and Cytokine Involvement in Cardiac Dysfunction
- NF-κB activation: Oncology drugs that activate NF-κB signaling induce inflammation, and may cause cardiac remodeling [23].
- Immune cell infiltration: Myocarditis was recently reported as a consequence of some immune checkpoint therapy, including immune cell infiltration into the heart (myocardium) [22].
- Cytokine-induced contractile dysfunction: Direct cardiomyocyte contractility can be directly impaired by pro-inflammatory cytokines through various mechanisms, including alterations in calcium handling [22].
2.15. Risk Factors for Cardiotoxicity
2.15.1. Patient-Specific Factors
2.15.2. Drug-Specific Factors
2.16. Clinical Manifestations
Symptoms and Diagnosis
- Arrhythmias: Cardiac rhythm disturbances can be benign or potentially even fatal. They may include atrial fibrillation, ventricular tachycardia, and QT interval prolongation [23]. There may be palpitations, dizziness, or syncope.
- Heart Failure: This is one of the most serious manifestations of cardiotoxicity. Exertional dyspnea, fatigue, peripheral edema, and reduced exercise tolerance [24] are symptoms of the disease. In severe cases patients may go as far as to develop acute pulmonary edema or cardiogenic shock.
- Decreased Cardiac Function: This can be the first sign of cardiotoxicity, with an asymptomatic descent in left ventricular ejection fraction (LVEF). Left untreated, this can progress to symptomatic heart failure [22].
- Myocardial Ischemia: Coronary vasospasm can occur, causing chest pain with chronic coronary syndrome or acute coronary syndrome in some oncology drugs, particularly fluoropyrimidines [23].
- Hypertension: It is known that certain targeted therapies, notably vascular endothelial growth factor (VEGF) inhibitors, have caused or exacerbated hypertension [21].
- Thromboembolism: Thromboembolic events such as deep vein thrombosis or pulmonary embolism [23] are increased by some chemotherapies.
- Echocardiography: the cornerstone of cardiac assessment of cancer patients. It enables the measurement of the LVEF, the most common parameter used to characterize cardiotoxicity. Cardiotoxicity is diagnosed when a LVEF (typically defined as <50% absolute decrease, usually >10 percentage points) is observed to decrease from >50% to <50% [22]. In particular, more advanced echocardiographic techniques such as global longitudinal strain (GLS) may detect subclinical cardiac dysfunction prior to a drop in LVEF [24].
- Biomarkers: Troponins (troponin I and T) are highly sensitive, specific markers of myocardial injury. When episodes of elevated troponin were associated with an increased risk of subsequent cardiac dysfunction, such episodes had been reported during cancer therapy [46]. Both cardiac stress markers (BNP and NT-pro BNP) have also been shown to have utility in predicting and diagnosing cardiotoxicity [24].
- Other Imaging Modalities: Cardiac MRI is useful for the detailed assessment of cardiac structure and function, and particularly to assess for myocarditis, a potential complication of immune checkpoint inhibitors [21]. Advanced techniques such as cardiac MRI have emerged as valuable in detecting subclinical cardiotoxicity while emerging biomarkers like specific microRNAs are under investigation for earlier detection. Cardiac function may also be assessed, and early cardiotoxicity is recorded with use of nuclear imaging techniques [47].
- Electrocardiogram (ECG): This is not specific to cardiotoxicity but can reveal arrhythmias and conduction abnormalities that may be due to cardiac injury.
- Endomyocardial Biopsy: It is rarely used, although it can offer definitive proof of cardiotoxicity treatment, particularly where the diagnosis cannot be otherwise determined [21].
2.17. Long-Term Consequences
2.17.1. Impact on Survivors’ Quality of Life
2.17.2. Increased Risk of Cardiovascular Diseases Post-Treatment
2.18. Management Strategies
2.18.1. Preventive Measures
- Echocardiography and biomarker assessment (troponin, BNP) [45] baseline cardiac evaluation before starting potentially cardiotoxic therapy.
- Left ventricular ejection fraction (LVEF) should be regularly monitored during treatment [50] with the dose depending on the specific drug and patient risks.
- Subclinical cardiac dysfunction that can be detected before a decline in LVEF [50] and use of more delicate imaging approaches, such as quantitative global longitudinal strain (GLS) echocardiography.
2.18.2. Treatment Approaches: Management of Symptomatic Patients
- For patients with reduced LVEF, the first line of treatment is with ACE inhibitors or angiotensin receptor blockers, and some evidence suggests that the drugs may also have a preventive benefit if used early in cancer treatment [38].
- The antioxidant properties of beta-blockers, particularly carvedilol, have been shown to be useful both as a preventative as well as a treatment for chemotherapy-induced cardiotoxicity [38].
- Referrals to cardiology services are made for patients with severe heart failure where temporary or permanent discontinuation of cardiotoxic agents may be necessary in collaboration with an oncology and cardiology team [50].
2.18.3. Integration of Cardio-Oncology Principles in Cancer Care
- The importance of early screening for cardiovascular risk factors before initiating cancer therapy, taking into account cardiac risk factors, including potential cardiotoxicity of planned therapies [45].
- Minimizing cardiovascular risk in cancer treatment regimes without compromising the oncological efficacy. This may use less cardiotoxic alternatives or modified dosing schedules [50].
- Cardio-protective strategies, including exercise programs and control of cardiotoxicity risk factors [38].
- The use of cardioprotective strategies, including ACE inhibitors, beta-blockers, and exercise programs.
- Facilitation of communication between oncologists and cardiologists in the creation of multidisciplinary cardio oncology teams to coordinate care for a patient during the cancer treatment journey [45].
- Survivorship care plans that include long-term cardiovascular monitoring and management strategies for cancer survivors [50], allows survivors to receive the right cancer treatment especially, long-term cardiovascular monitoring and management strategies. Long-term follow-up and survivorship care plans for cancer survivors at high risk of late-onset cardiotoxicity require the constant monitoring of patients.
2.19. Future Directions
2.19.1. Gaps in Understanding Mechanisms and Risk Factors
2.19.2. Importance of Personalized Medicine in Oncology
- Testing of patients at higher risk for cardiotoxicity, using pharmacogenomic testing.
- Individual risk-based monitoring protocols.
- Targeted cardioprotective agents use, such as dexrazoxane, and personalized cardioprotective strategies.
- Balanced oncological efficacy and cardiovascular risk for individualized cancer treatment regimens.
2.19.3. Innovative Therapies
2.19.4. Development of New Drugs with Reduced Cardiotoxic Profiles
2.19.5. Exploration of Adjunct Therapies to Mitigate Cardiac Risks
- Cardioprotective agents: Researchers are also studying other potential cardioprotective agents, such as antioxidants, iron chelators and modulators of cellular signaling pathways that have been implicated as sources of toxicity [53].
- Exercise interventions: Evidence is emerging for cardio protection associated with structured exercise programs during cancer treatment. More research is needed to find out how to best optimize exercise and who is best likely to benefit [23].
- Nutraceuticals and dietary interventions: Other studies have attempted to minimize the cardiotoxicity potential of nutraceuticals (such as coenzyme Q10 and L-carnitine). Although preliminary results are promising, larger clinical trials are required to establish efficacy and safety [54].
- Cell-based therapies: Cardiac tissue damaged by cancer treatments is being investigated to repair and regenerate through stem cell therapies and exosome-based approaches [55].
3. Discussion
4. Conclusions
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Clark, R.A.; Marin, T.S.; McCarthy, A.L.; Bradley, J.; Grover, S.; Peters, R.; Karapetis, C.S.; Atherton, J.J.; Koczwara, B. Cardiotoxicity after cancer treatment: A process map of the patient treatment journey. Cardiooncology 2019, 5, 14. [Google Scholar] [CrossRef] [PubMed]
- Han, X.; Zhou, Y.; Liu, W. Precision cardio-oncology: Understanding the cardiotoxicity of cancer therapy. NPJ Precis. Oncol. 2017, 1, 31. [Google Scholar] [CrossRef] [PubMed]
- Jaiswal, V.; Ang, S.P.; Deb, N.; Hanif, M.; Batra, N.; Kanagala, S.G.; Vojjala, N.; Rajak, K.; Roy, P.; Sharath, M.; et al. Association between Statin Use and Chemotherapy-Induced Cardiotoxicity: A Meta-Analysis. Medicina 2024, 60, 580. [Google Scholar] [CrossRef]
- Nicolazzi, M.A.; Carnicelli, A.; Fuorlo, M.; Scaldaferri, A.; Masetti, R.; Landolfi, R.; Favuzzi, A.M.R. Anthracycline and trastuzumab-induced cardiotoxicity in breast cancer. Eur. Rev. Med. Pharmacol. Sci. 2018, 22, 2175–2185. [Google Scholar] [CrossRef]
- Falzone, L.; Salomone, S.; Libra, M. Evolution of Cancer Pharmacological Treatments at the Turn of the Third Millennium. Front. Pharmacol. 2018, 9, 1300. [Google Scholar] [CrossRef] [PubMed]
- Santhosh, S.; Kumar, P.; Ramprasad, V.; Chaudhuri, A. Evolution of targeted therapies in cancer: Opportunities and challenges in the clinic. Future Oncol. 2015, 11, 279–293. [Google Scholar] [CrossRef]
- Zhao, J.Y.; Yuan, X.K.; Luo, R.Z.; Wang, L.X.; Gu, W.; Yamane, D.; Feng, H. Phospholipase A and acyltransferase 4/retinoic acid receptor responder 3 at the intersection of tumor suppression and pathogen restriction. Front. Immunol. 2023, 14, 1107239. [Google Scholar] [CrossRef]
- Nielsen, D.L.; Juhl, C.B.; Nielsen, O.H.; Chen, I.M.; Herrmann, J. Immune Checkpoint Inhibitor-Induced Cardiotoxicity: A Systematic Review and Meta-Analysis. JAMA Oncol. 2024, 10, 1390–1399. [Google Scholar] [CrossRef]
- Min, H.Y.; Lee, H.Y. Molecular targeted therapy for anticancer treatment. Exp. Mol. Med. 2022, 54, 1670–1694. [Google Scholar] [CrossRef]
- Meisel, J.L.; Venur, V.A.; Gnant, M.; Carey, L. Evolution of Targeted Therapy in Breast Cancer: Where Precision Medicine Began. Am. Soc. Clin. Oncol. Educ. Book 2018, 38, 78–86. [Google Scholar] [CrossRef]
- Al-Hasnawi, Z.; Hasan, H.M.; Abdul Azeez, J.M.; Kadhim, N.; Shimal, A.A.; Sadeq, M.H.; Mahood, N.; Al-Qara Ghuli, A.A.N.; Hussein, A.S.; Prajjwal, P.; et al. Cardioprotective strategies in the management of chemotherapy-induced cardiotoxicity: Current approaches and future directions. Ann. Med. Surg. 2024, 86, 7212–7220. [Google Scholar] [CrossRef] [PubMed]
- Kobo, O.; Khattak, S.; Lopez-Mattei, J.; Van Spall, H.G.C.; Graham, M.; Cheng, R.K.; Osman, M.; Sun, L.; Ullah, W.; Fischman, D.L.; et al. Trends in cardiovascular mortality of cancer patients in the US over two decades 1999–2019. Int. J. Clin. Pract. 2021, 75, e14841. [Google Scholar] [CrossRef]
- Hou, J.; He, Z.; Liu, T.; Chen, D.; Wang, B.; Wen, Q.; Zheng, X. Evolution of Molecular Targeted Cancer Therapy: Mechanisms of Drug Resistance and Novel Opportunities Identified by CRISPR-Cas9 Screening. Front. Oncol. 2022, 12, 755053. [Google Scholar] [CrossRef]
- Hui, D.; Paiva, C.E.; Del Fabbro, E.G.; Steer, C.; Naberhuis, J.; van de Wetering, M.; Fernández-Ortega, P.; Morita, T.; Suh, S.Y.; Bruera, E.; et al. Prognostication in advanced cancer: Update and directions for future research. Support Care Cancer 2019, 27, 1973–1984. [Google Scholar] [CrossRef] [PubMed]
- Cronin, K.A.; Scott, S.; Firth, A.U.; Sung, H.; Henley, S.J.; Sherman, R.L.; Siegel, R.L.; Anderson, R.N.; Kohler, B.A.; Benard, V.B.; et al. Annual report to the nation on the status of cancer, part 1: National cancer statistics. Cancer 2022, 128, 4251–4284. [Google Scholar] [CrossRef] [PubMed]
- Razzaghi, H.; Saraiya, M.; Thompson, T.D.; Henley, S.J.; Viens, L.; Wilson, R. Five-year relative survival for human papillomavirus-associated cancer sites. Cancer 2018, 124, 203–211. [Google Scholar] [CrossRef]
- Ahmad, T.A.; Gopal, D.P.; Chelala, C.; Dayem Ullah, A.Z.; Taylor, S.J. Multimorbidity in people living with and beyond cancer: A scoping review. Am. J. Cancer Res. 2023, 13, 4346–4365. [Google Scholar]
- Pinheiro, L.C.; Tan, X.; Olshan, A.F.; Wheeler, S.B.; Reeder-Hayes, K.E.; Samuel, C.A.; Reeve, B.B. Examining health-related quality of life patterns in women with breast cancer. Qual. Life Res. 2017, 26, 1733–1743. [Google Scholar] [CrossRef]
- Cheng, M.J.; Hourigan, C.S.; Smith, T.J. Adult Acute Myeloid Leukemia Long-term Survivors. J. Leuk. 2014, 2, 26855. [Google Scholar] [CrossRef]
- Cheung, C.; Boocock, E.; Grande, A.J.; Maddocks, M. Exercise-based interventions for cancer cachexia: A systematic review of randomised and non-randomised controlled trials. Asia Pac. J. Oncol. Nurs. 2023, 10, 100335. [Google Scholar] [CrossRef]
- Huang, W.; Xu, R.; Zhou, B.; Lin, C.; Guo, Y.; Xu, H.; Guo, X. Clinical Manifestations, Monitoring, and Prognosis: A Review of Cardiotoxicity After Antitumor Strategy. Front. Cardiovasc. Med. 2022, 9, 912329. [Google Scholar] [CrossRef] [PubMed]
- Morelli, M.B.; Bongiovanni, C.; Da Pra, S.; Miano, C.; Sacchi, F.; Lauriola, M.; D’Uva, G. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and Strategies for Cardioprotection. Front. Cardiovasc. Med. 2022, 9, 847012. [Google Scholar] [CrossRef] [PubMed]
- Kwok, C.; Nolan, M. Cardiotoxicity of anti-cancer drugs: Cellular mechanisms and clinical implications. Front. Cardiovasc. Med. 2023, 10, 1150569. [Google Scholar] [CrossRef] [PubMed]
- Cardinale, D.; Iacopo, F.; Cipolla, C.M. Cardiotoxicity of Anthracyclines. Front. Cardiovasc. Med. 2020, 7, 26. [Google Scholar] [CrossRef]
- Herrmann, J. Adverse cardiac effects of cancer therapies: Cardiotoxicity and arrhythmia. Nat. Rev. Cardiol. 2020, 17, 474–502. [Google Scholar] [CrossRef]
- Force, T.; Krause, D.S.; Van Etten, R.A. Molecular mechanisms of cardiotoxicity of tyrosine kinase inhibition. Nat. Rev. Cancer 2007, 7, 332–344. [Google Scholar] [CrossRef]
- Wang, H.; Wang, Y.; Li, J.; He, Z.; Boswell, S.A.; Chung, M.; You, F.; Han, S. Three tyrosine kinase inhibitors cause cardiotoxicity by inducing endoplasmic reticulum stress and inflammation in cardiomyocytes. BMC Med. 2023, 21, 147. [Google Scholar] [CrossRef]
- Hu, W.; Lu, S.; McAlpine, I.; Jamieson, J.D.; Lee, D.U.; Marroquin, L.D.; Heyen, J.R.; Jessen, B.A. Mechanistic Investigation of Imatinib-Induced Cardiac Toxicity and the Involvement of c-Abl Kinase. Toxicol. Sci. 2012, 129, 188–199. [Google Scholar] [CrossRef]
- Sharma, A.; Alexander, G.; Chu, J.H.; Markopoulos, A.; Maloul, G.; Ayub, M.T.; Fidler, M.J.; Okwuosa, T.M. Immune Checkpoint Inhibitors and Cardiotoxicity: A Comparative Meta-Analysis of Observational Studies and Randomized Controlled Trials. J. Am. Heart Assoc. 2024, 13, e032620. [Google Scholar] [CrossRef]
- Brahmer, J.R.; Lacchetti, C.; Schneider, B.J.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; Ernstoff, M.S.; Gardner, J.M.; Ginex, P.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2018, 36, 1714–1768. [Google Scholar] [CrossRef]
- Kong, Y.; Wang, X.; Qie, R. Immunotherapy-associated cardiovascular toxicities: Insights from preclinical and clinical studies. Front. Oncol. 2024, 14, 1347140. [Google Scholar] [CrossRef] [PubMed]
- Touyz, R.M.; Herrmann, J. Cardiotoxicity with vascular endothelial growth factor inhibitor therapy. NPJ Precis. Oncol. 2018, 2, 13. [Google Scholar] [CrossRef] [PubMed]
- Pandey, A.K.; Singhi, E.K.; Arroyo, J.P.; Ikizler, T.A.; Gould, E.R.; Brown, J.; Beckman, J.A.; Harrison, D.G.; Moslehi, J. Mechanisms of VEGF (Vascular Endothelial Growth Factor) Inhibitor—Associated Hypertension and Vascular Disease. Hypertension 2018, 71, e1–e8. [Google Scholar] [CrossRef] [PubMed]
- Kamba, T.; McDonald, D.M. Mechanisms of adverse effects of anti-VEGF therapy for cancer. Br. J. Cancer 2007, 96, 1788–1795. [Google Scholar] [CrossRef]
- Sharma, R.; Kashyap, J.; Olanrewaju, O.A.; Jabbar, A.; Someshwar, F.; Saeed, H.; Varrassi, G.; Qadeer, H.A.; Kumar, S.; Cheema, A.Y.; et al. Cardio-Oncology: Managing Cardiovascular Complications of Cancer Therapies. Cureus 2023, 15, e51038. [Google Scholar] [CrossRef]
- Neuendorff, N.R.; Loh, K.P.; Mims, A.S.; Christofyllakis, K.; Soo, W.K.; Bölükbasi, B.; Oñoro-Algar, C.; Hundley, W.G.; Klepin, H.D. Anthracycline-related cardiotoxicity in older patients with acute myeloid leukemia: A Young SIOG review paper. Blood Adv. 2020, 4, 762–775. [Google Scholar] [CrossRef]
- McGowan, J.V.; Chung, R.; Maulik, A.; Piotrowska, I.; Walker, J.M.; Yellon, D.M. Anthracycline Chemotherapy and Cardiotoxicity. Cardiovasc. Drugs Ther. 2017, 31, 63–75. [Google Scholar] [CrossRef]
- Bhagat, A.; Kleinerman, E.S. Anthracycline-Induced Cardiotoxicity: Causes, Mechanisms, and Prevention. Adv. Exp. Med. Biol. 2020, 1257, 181–192. [Google Scholar] [CrossRef]
- Gabani, M.; Castañeda, D.; Nguyen, Q.M.; Choi, S.K.; Chen, C.; Mapara, A.; Kassan, A.; Gonzalez, A.A.; Khataei, T.; Ait-Aissa, K.; et al. Association of Cardiotoxicity With Doxorubicin and Trastuzumab: A Double-Edged Sword in Chemotherapy. Cureus 2021, 13, e18194. [Google Scholar] [CrossRef]
- Lyon, A.R.; Dent, S.; Stanway, S.; Earl, H.; Brezden-Masley, C.; Cohen-Solal, A.; Tocchetti, C.G.; Moslehi, J.J.; Groarke, J.D.; Bergler-Klein, J.; et al. Baseline cardiovascular risk assessment in cancer patients scheduled to receive cardiotoxic cancer therapies: A position statement and new risk assessment tools from the Cardio-Oncology Study Group of the Heart Failure Association of the European Society of Cardiology in collaboration with the International Cardio-Oncology Society. Eur. J. Heart Fail 2020, 22, 1945–1960. [Google Scholar] [CrossRef]
- Ma, Y.; North, B.J.; Shu, J. Regulation of topoisomerase II stability and activity by ubiquitination and SUMOylation: Clinical implications for cancer chemotherapy. Mol. Biol. Rep. 2021, 48, 6589–6601. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Wei, J.; Zheng, Q.; Meng, L.; Xin, Y.; Yin, X.; Jiang, X. Radiation-induced heart disease: A review of classification, mechanism and prevention. Int. J. Biol. Sci. 2019, 15, 2128–2138. [Google Scholar] [CrossRef]
- Belzile-Dugas, E.; Eisenberg, M.J. Radiation-Induced Cardiovascular Disease: Review of an Underrecognized Pathology. J. Am. Heart Assoc. 2021, 10, e021686. [Google Scholar] [CrossRef] [PubMed]
- Adão, R.; de Keulenaer, G.; Leite-Moreira, A.; Brás-Silva, C. Cardiotoxicity associated with cancer therapy: Pathophysiology and prevention strategies. Rev. Port. Cardiol. 2013, 32, 395–409. [Google Scholar] [CrossRef]
- Albini, A.; Pennesi, G.; Donatelli, F.; Cammarota, R.; De Flora, S.; Noonan, D.M. Cardiotoxicity of anticancer drugs: The need for cardio-oncology and cardio-oncological prevention. J. Natl. Cancer Inst. 2010, 102, 14–25. [Google Scholar] [CrossRef] [PubMed]
- Murtagh, G.; Januzzi, J.L.; Scherrer-Crosbie, M.; Neilan, T.G.; Dent, S.; Ho, J.E.; Appadurai, V.; McDermott, R.; Akhter, N. Circulating Cardiovascular Biomarkers in Cancer Therapeutics-Related Cardiotoxicity: Review of Critical Challenges, Solutions, and Future Directions. J. Am. Heart Assoc. 2023, 12, e029574. [Google Scholar] [CrossRef]
- Mohamed, A.A.; Elmancy, L.Y.; Abulola, S.M.; Al-Qattan, S.A.; Mohamed Ibrahim, M.I.; Maayah, Z.H. Assessment of Native Myocardial T1 Mapping for Early Detection of Anthracycline-Induced Cardiotoxicity in Patients with Cancer: A Systematic Review and Meta-analysis. Cardiovasc. Toxicol. 2024, 24, 563–575. [Google Scholar] [CrossRef]
- Yu, A.F.; Flynn, J.R.; Moskowitz, C.S.; Scott, J.M.; Oeffinger, K.C.; Dang, C.T.; Liu, J.E.; Jones, L.W.; Steingart, R.M. Long-term Cardiopulmonary Consequences of Treatment-Induced Cardiotoxicity in Survivors of ERBB2-Positive Breast Cancer. JAMA Cardiol. 2020, 5, 309–317. [Google Scholar] [CrossRef]
- Díaz-Balboa, E.; Peña-Gil, C.; Rodríguez-Romero, B.; Cuesta-Vargas, A.I.; Lado-Baleato, O.; Martínez-Monzonís, A.; Pedreira-Pérez, M.; Palacios-Ozores, P.; López-López, R.; González-Juanatey, J.R.; et al. Exercise-based cardio-oncology rehabilitation for cardiotoxicity prevention during breast cancer chemotherapy: The ONCORE randomized controlled trial. Prog. Cardiovasc. Dis. 2024, 85, 74–81. [Google Scholar] [CrossRef]
- Trapani, D.; Zagami, P.; Nicolò, E.; Pravettoni, G.; Curigliano, G. Management of Cardiac Toxicity Induced by Chemotherapy. J. Clin. Med. 2020, 9, 2885. [Google Scholar] [CrossRef]
- Valeanu, A.; Margina, D.; Weber, D.; Stuetz, W.; Moreno-Villanueva, M.; Dollé, M.E.T.; Jansen, E.H.; Gonos, E.S.; Bernhardt, J.; Grubeck-Loebenstein, B.; et al. Development and validation of cardiometabolic risk predictive models based on LDL oxidation and candidate geromarkers from the MARK-AGE data. Mech. Ageing Dev. 2024, 222, 111987. [Google Scholar] [CrossRef] [PubMed]
- Herdiana, Y.; Wathoni, N.; Shamsuddin, S.; Muchtaridi, M. Cytotoxicity Enhancement in MCF-7 Breast Cancer Cells with Depolymerized Chitosan Delivery of α-Mangostin. Polymers 2022, 14, 3139. [Google Scholar] [CrossRef] [PubMed]
- Penna, C.; Granata, R.; Tocchetti, C.G.; Gallo, M.P.; Alloatti, G.; Pagliaro, P. Endogenous Cardioprotective Agents: Role in Pre and Postconditioning. Curr. Drug Targets 2015, 16, 843–867. [Google Scholar] [CrossRef]
- Wong, A.P.; Niedzwiecki, A.; Rath, M. Myocardial energetics and the role of micronutrients in heart failure: A critical review. Am. J. Cardiovasc. Dis. 2016, 6, 81–92. [Google Scholar]
- Alonzo, M.; AnilKumar, S.; Roman, B.; Tasnim, N.; Joddar, B. 3D Bioprinting of cardiac tissue and cardiac stem cell therapy. Transl. Res. 2019, 211, 64–83. [Google Scholar] [CrossRef] [PubMed]
Class of Chemotherapy Medication | Mechanism of Cardiotoxicity |
---|---|
Anthracyclines | Anthracyclines cause cardiotoxicity primarily via oxidative stress, mitochondrial dysfunction, and DNA damage. |
Tyrosine Kinase Inhibitors (TKIs) | TKIs disrupt normal cellular signaling and can impair mitochondrial function, leading to cardiomyocyte apoptosis. |
Immune Checkpoint Inhibitors | Recent findings have revealed that immune checkpoint inhibitors not only enhance anti-tumor immunity but also may induce myocarditis, arrhythmias, and other immune-related cardiac events. |
Anti-Angiogenic Drugs (anti-VEGF) | Inducing microvascular rarefaction, reducing myocardial capillary density, and impairing cardiac perfusion. |
Anti-Angiogenic Drugs (anti-PIGF) | Safer than anti-VEGF. |
Type of Cardiotoxicity | Description | Common Oncology Drugs Involved | Potential Long-Term Effects |
---|---|---|---|
Left Ventricular Dysfunction and Heart Failure | Progressive decline in heart’s ability to pump blood effectively. | Anthracyclines (e.g., doxorubicin), Anti-HER2 agents (trastuzumab). | Chronic heart failure, reduced ejection fraction. |
Arrhythmias | Irregular heartbeats, which may be benign or life-threatening. | Tyrosine kinase inhibitors (TKIs), Immune checkpoint inhibitors. | Increased risk of sudden cardiac events, atrial fibrillation. |
Myocardial Ischemia | Reduced blood flow to the heart muscle, leading to chest pain and infarction. | Fluoropyrimidines (e.g., 5-FU), VEGF inhibitors. | Coronary artery disease, increased risk of heart attacks. |
Hypertension | Elevated blood pressure due to vascular dysfunction. | VEGF inhibitors, certain TKIs. | Increased risk of stroke and heart failure. |
Thromboembolism | Formation of blood clots that can lead to deep vein thrombosis or pulmonary embolism. | VEGF inhibitors, Immunotherapy agents. | Pulmonary embolism, increased cardiovascular mortality. |
Pericardial Disease | Inflammation or fluid accumulation around the heart. | Radiation therapy, certain targeted therapies. | Pericardial effusion, chronic pericarditis. |
Strategy | Description | Application |
---|---|---|
Baseline Risk Assessment | Cardiovascular screening before initiating cancer therapy. | Identifying high-risk patients based on comorbidities, prior cardiac history. |
Regular Cardiac Monitoring | Routine imaging and biomarker assessment. | Echocardiography (LVEF, GLS), troponin levels, BNP/NT-proBNP. |
Cardioprotective Medications | Use of ACE inhibitors, beta-blockers, and dexrazoxane. | Preventing or mitigating drug-induced cardiotoxicity. |
Modified Drug Administration | Adjusting dosage, infusion rates, or drug combinations. | Reducing peak plasma concentrations of cardiotoxic drugs. |
Exercise and Lifestyle Interventions | Structured exercise programs and dietary interventions. | Improving cardiac resilience and mitigating treatment effects. |
Early Intervention and Treatment | Initiating heart failure treatments at the first sign of dysfunction. | Improving recovery rates and reducing progression to irreversible damage. |
Multidisciplinary Cardio-Oncology Collaboration | Close coordination between oncologists and cardiologists. | Optimizing both cancer therapy and cardiac protection. |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2025 by the authors. Published by MDPI on behalf of the Lithuanian University of Health Sciences. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Kundnani, N.R.; Passini, V.; Stefania Carlogea, I.; Dumitrescu, P.; Meche, V.; Buzas, R.; Duda-Seiman, D.M. Overview of Oncology: Drug-Induced Cardiac Toxicity. Medicina 2025, 61, 709. https://doi.org/10.3390/medicina61040709
Kundnani NR, Passini V, Stefania Carlogea I, Dumitrescu P, Meche V, Buzas R, Duda-Seiman DM. Overview of Oncology: Drug-Induced Cardiac Toxicity. Medicina. 2025; 61(4):709. https://doi.org/10.3390/medicina61040709
Chicago/Turabian StyleKundnani, Nilima Rajpal, Vincenzo Passini, Iulia Stefania Carlogea, Patrick Dumitrescu, Vlad Meche, Roxana Buzas, and Daniel Marius Duda-Seiman. 2025. "Overview of Oncology: Drug-Induced Cardiac Toxicity" Medicina 61, no. 4: 709. https://doi.org/10.3390/medicina61040709
APA StyleKundnani, N. R., Passini, V., Stefania Carlogea, I., Dumitrescu, P., Meche, V., Buzas, R., & Duda-Seiman, D. M. (2025). Overview of Oncology: Drug-Induced Cardiac Toxicity. Medicina, 61(4), 709. https://doi.org/10.3390/medicina61040709