Abstract
Advances in cancer treatments have led to an increasing number of cancer survivors, but also high rates of short- and long-term cardiovascular (CV) toxicities. The number of new cancer drugs is constantly increasing, and the uncertain CV toxicities of these drugs make long-term care and monitoring difficult. Moreover, traditional type I and type II cardiotoxicities may not be applicable to all of these agents. Multidisciplinary care with expertise in oncology, cardiology and other related specialties is required to mitigate cancer therapeutics-related cardiovascular dysfunction (CTRCD).
The aim of this review is to provide an overview of the main CTRCD, risk assessment, early diagnosis, and strategies for the prevention and management of patients receiving cancer therapies. There are still unmet needs for cardio- oncology researchers with regards to early detection measures, better treatment strategies, better follow-up protocols, and better management of CTRCD. Experts in cardiology, oncology, hematology, and radio-oncology should thus work closely in an attempt to foster patient awareness and research in this field, as well as call for support from public and industrial sources to initiate pivotal clinical trials to solve these unmet needs.
Keywords: Cancer therapeutics-related cardiovascular dysfunction, Cardio-oncology, Cardiotoxicity, Chemotherapy, Radiotherapy
INTRODUCTION
The leading cause of death in Taiwan is cancer followed by cardiovascular disease (CVD),1 and thus cardio-oncology (CO) is an emerging issue in Taiwan where cancer treatment has advanced rapidly over the past decades. The previous treatment triads, namely cytotoxic chemotherapy, radiation therapy and surgery, have been expanded to include targeted and immune-based therapies.2 Thanks to the ever increasing number of advanced therapies available, an increasing number of cancer patients survive,3 however an emerging issue associated with these new cancer therapies is sides effects on the cardiovascular (CV) system, which cause different spectrums of morbidity and mortality.4 Cardiotoxicity refers to the direct harmful effects of cancer treatments on the CV system and/or the acceleration of CVDs in addition to traditional CV risk factors.4,5 The origin of CO can be traced back to July 1st, 2000, when the MD Anderson Cancer Center initiated a comprehensive program to diagnose, treat and manage all CV disorders of cancer survivors. Currently, the main focus of CO research is on the prevention and management of related CV complications caused by cancer therapy, including: 1) treatment-based, including cancer-related medications, surgery, or radiation; 2) CV symptoms/complications; and 3) baseline CV risk-based identification and management. The goals of this article are to provide an overview of the major CV adverse events related to cancer chemo- and radiation therapies.
EVOLVING CONCEPTS OF CARDIOTOXICITY
Previous studies on cardiotoxicity have focused on anthracyclines and trastuzumab. Ewer and Lippman introduced the concept of type I irreversible, and type II reversible, cardiotoxicity.6,7 Doxorubicin is the most well-known agent responsible for type I cancer therapeutics-related cardiovascular dysfunction (CTRCD). Since varying degrees of myocyte damage, including vacuolar swelling, myofibrillar disarray and cell death, can be observed in electron microscopy of myocardial biopsies, type I CTRCD is cumulative, dose-dependent and progressive. Therefore, there is a high probability of recurrent dysfunction with rechallenge, which may result in intractable heart failure or death without suitable interventions and adjustments in the chemotherapy regimen.8 The characteristic agent of type II CTRCD is trastuzumab. Since this type of agent has not been observed to directly cause cell damage in electron microscopy of myocardial biopsies, the damage is not considered to be cumulative, dose-dependent or progressive. Therefore, it is relatively safe to rechallenge with a high likelihood of near recovery in 2-4 months after interruption (reversible).8 Other anti-HER2-targeted therapies such as the monoclonal antibodies pertuzumab and trastuzumab emtansine, and the tyrosine kinase inhibitor lapatinib, appear to share this type II pattern of cardiotoxicity.7,9 However, recent arguments against the concept of type I and type II cardiotoxicities have arisen, and doxorubicin-induced cardiotoxicity is not always irreversible,10,11 while trastuzumab is not always reversible.12 The type, timing, duration, and combination of drugs as well as the patient’s genetic and comorbidity profile should all be taken into consideration when evaluating different patterns of cardiotoxicities. The speed of development of anti-cancer drugs continues to increase, and they have accounted for 27% of all new drugs in the United States since 2010, with approval of 126 cancer drugs to treat solid and hematologic tumors from 1980 through 2018 by the FDA (hyperlink: Cancer Drugs Account for Over a Quarter of All New Drug Approvals in the US - The ASCO Post). In view of this surge in new therapies, the practice of oncology and its related CTRCD are changing dramatically. Indeed, in addition to myocardial dysfunction (either cardiomyopathy, asymptomatic or symptomatic heart failure with preserved or reduced ejection fraction), CTRCD should include all kinds of toxic/side effects affecting the CV system, including hypertension, endothelial and vascular dysfunction, accelerated atherosclerosis, thrombosis and bleeding, pulmonary hypertension, pericardial disease, QT prolongation, conduction disease/arrhythmias, as well as radiation-induced CV disease.5,13-18 In addition, different anti-cancer therapies (chemotherapy, targeted therapy, hormone therapy, immunotherapy, radiation therapy, and surgery) and bone marrow transplantation have their own relevant CV concerns. Table 1 summarizes the common CTRCD. Of note, only two types of CTRCD may not fit all clinical scenarios, and further studies are expected to clarify more types of CTRCD.
Table 1. Common cancer therapeutics-related cardiovascular dysfunction.
| Common anti-cancer therapy/common cardiovascular complications |
|
|
|
|
|
|
| Anthracyclines (Doxorubicin, Idarubicin, Epirubicin) | v | v | ||||
| Alkylating agents (Cyclophosphamide, Ifosfamide) | v | v | v | |||
| Antimetabolites (Clofarabine) | v | v | ||||
| Antimicrotubule agents (Docetaxel, Paclitaxel) | v | v | v | |||
| Monoclonal antibodies (Trastuzumab, Bevacizumab, Pertuzumab) | v | v | ||||
| Protease inhibitors (Carfilzomib, Bortezomib) | v | v | v | |||
| Fluoropyrimidines (5-FU, capecitabine, gemcitabine) | v | v | v | |||
| TKIs (Sunitinib, Pazopanib, Sorafenib) | v | v | v | v | ||
| VEGF inhibitors (bevacizumab) | v | v | v | |||
| Arsenic trioxide, bortezomib, IL-2, methotrexate, mitoxantrone, rituximab, thalidomide, amsacrine, interferons | v | |||||
| Radiotherapy | v | v | v | v |
, left ventricular dysfunction/heart failure;
, hypertension and/or proteinuria;
, pericardium disease;
, arrhythmia/conduction problems;
, coronary artery disease/thrombosis;
, valve problems.
TKI, tyrosine kinase inhibitor; VEGF, vascular endothelial growth factor.
RADIOTHERAPY-RELATED CARDIOTOXICITY
Radiotherapy may cause damage to the pericardium, coronary arteries, valves, endocardium and myocardium, and symptoms can occur in the acute (< 6 months) or late phase (3-30 years).19,20 Breast cancer patients with radiotherapy have been shown to have a 30% greater risk of coronary heart disease and a 38% greater risk of cardiac death compared to those without radiotherapy.21 The CV risk has also been reported to be higher in patients receiving radiotherapy concomitantly with anthracyclines,22,23 with a 1.4-fold higher risk of heart injury in patients with left-sided breast cancer than in those with right-sided breast cancer.24 The direct CV risk of radiotherapy includes radiation volume and dose to which the heart and its substructures are irradiated,25,26 and the rate of major adverse cardiac events (MACEs, i.e., myocardial infarction, coronary revascularization or CV death) has been shown to increase linearly by 7.4% per Gray increase in mean heart dose.27 Risk mitigation is focused on reducing cardiac exposure to radiation, including displacement maneuvers such as prone positioning and deep inspiratory breath holding, custom blocks for the heart, intensity-modulated techniques, intraoperative irradiation and/or brachytherapy, and proton irradiation.25,28,29
IMMUNOTHERAPY-RELATED CARDIOTOXICITY
Immunotherapy-related cardiotoxicities have recently focused on chimeric antigen receptor T cell (CAR-T) therapy-associated cytokine release syndrome30 and immune checkpoint inhibitor (ICI)-associated myocarditis. Although the reported incidence of ICI-related myocarditis is low (0.04-1.14%), it is associated with a high mortality rate (25-50%) and has been reported to occur early after the initiation of therapy.13,31-34 While a review of 101 cases showed that 64% occurred after the first or second ICI dose,33 another study found that some cases occurred after the first ICI dose.32 In addition, combination ICI therapy has been reported to significantly increase the risk of myocarditis from 0.06% to 0.27%.13,32 Other risk factors are ill defined, but may include underlying autoimmune diseases, diabetes mellitus and pre-existing CVD.13,34-36 Although the diagnosis of myocarditis is challenging, elevated troponin and abnormal electrocardiography findings are common.13,37 During myocardial edema, cardiac magnetic resonance (CMR), with late gadolinium enhancement, may be useful for an early diagnosis, however it is only present in < 50% of those with ICI-associated myocarditis.37,38 An endomyocardial bio-psy is the gold standard for diagnosis, but is often underused due to its invasive nature, risk of complications, and a lack of expertise in many hospitals.37,39 The mechanism of myocarditis cardiotoxicity has been related to activated T cells,40 and < 50% of patients have been reported to respond to high doses of corticosteroids or immunosuppressants.13,41
BASELINE RISK ASSESSMENT, EARLY DIAGNOSIS OF CTRCD AND STRATEGIES FOR THE PREVENTION AND TREATMENT OF CARDIOTOXICITIES
Some risk factors for cardiotoxicity in oncology patients are traditional risk factors for CVD such as smoking, age, obesity, and hyperlipidemia,4,42,43 and some are newly identified genetic risk factors such as clonal hematopoiesis.44 The modifiable risk factors will accelerate CTRCD if they are not well identified and/or well controlled. For patients with symptoms or signs of current cardiac dysfunction, the guidelines recommend further assessing the risk using biomarkers such as troponins, natriuretic peptides, and the evaluation of left ventricular ejection fraction. Whenever possible, biomarker levels and imaging parameters should stay at baseline values throughout ongoing follow-up to ensure comparable information.4 Echocardiography-based strain imaging may be particularly useful as the follow-up imaging tool.45,46 A reduction in global longitudinal strain (GLS) of > 15% from baseline is generally considered to be abnormal and an early sign of left ventricular subclinical dysfunction4 and an early indicator of heart failure. CMR with T1 and T2 mapping may be particularly useful to evaluate vascular and structural cardiotoxicities. Stress echocardiography, stress CMR, computed tomography angiography (CTA) and positron emission tomography are alternative options to evaluate ischemia in patients receiving therapies that may cause vasospasms or accelerate atherosclerosis.46 The timing and frequency of follow-up will depend on various cancer treatments, cumulative anthracycline doses, delivery protocol and duration, as well as baseline CV risks.4 Patients identified as being at high risk should be referred to a CO specialist.4 The guidelines recommend measuring high-sensitivity cardiac troponins (cTnI or cTnT), B-type natriuretic peptide (BNP) and N-terminal pro-BNP (NT-proBNP) in those at high risk or undergoing cancer treatment.4,47,48 The European Society for Medical Oncology (ESMO) guidelines in 2012 suggested that patients receiving adjuvant chemotherapy should receive serial monitoring of cardiac function at baseline, 3, 6 and 9 months during treatment, and then 12 and 18 months after the initiation of treatment, which is feasible and cost-effective in a National Health Insurance setting such as in Taiwan.49 The ESMO guidelines recommend initiating cardioprotective agents [including angiotensin-converting enzyme (ACE) inhibitors, angiotensin II receptor blockers (ARBs) and/or beta-blockers] in patients receiving cardiotoxic treatment with decreased left ventricular (LV) ejection fraction, a decrease in GLS, or an elevation in cardiac troponin, with statins being considered in those with existing coronary artery disease.47 The ASCO guidelines recommend dexrazoxane, although it is currently not available in Taiwan, to prevent cardiotoxicity in patients with high-dose anthracyclines (e.g. doxorubicin ≥ 250 mg/m2).45 Guidance from the European Society of Cardiology (ESC) suggests the use of cardioprotective drugs (ACE inhibitors, beta-blockers, ARBs) in patients with pre-existing clinical heart failure or significant LV dysfunction at baseline, and the initiation of cardio-protective agents in patients with elevated troponin during treatment with high-dose anthracycline regimens.4
CANCER-ASSOCIATED THROMBOSIS
Cancer-associated thrombosis (CAT) is a common complication and is also a major cause of mortality in patients with cancer. Patients with cancer are at a four- to seven-fold higher risk of initial venous thromboembolism (VTE), a three-fold higher risk of recurrent VTE, a two-fold higher risk of anticoagulation-associated bleeding, and a 10-fold higher risk of death from VTE compared to patients without.50 In addition, patients with cancer have a two-fold higher risk of arterial thromboembolism than those without.51 Risk factors for CAT can be classified into cancer-related (e.g., primary site, histology, grade, initial period after diagnosis, etc.), treatment-related [e.g., surgery/hospitalization, chemotherapy, antiangiogenics, central venous cannulation, erythropoietin stimulating agent/transfusion-related, etc.], patient-related (e.g., age, ethnicity, comorbidities, etc.) and some important biomarkers (e.g., platelet count, leukocyte count, hemoglobin, D-dimer, etc.).52 Low-molecular-weight heparin (LMWH) has been shown to be more effective than warfarin for secondary prevention of VTE in cancer patients,53 and previous guidelines recommend LMWH over warfarin in cancer VTE.54 However, LMWH is inconvenient and painful for patients due to daily injections, and recent clinical trials (Hokusai-VTE Cancer, SELECT-D, CARAVAGGIO, and ADAM VTE) have proven that direct oral anticoagulants (DOACs) are noninferior to LMWH for CAT. Therefore, some DOACs (edoxaban, rivaroxaban, and apixaban) have become the mainstay in latest CAT treatment.55,56
UNMET NEEDS IN CARDIO-ONCOLOGY AND THE RECOMMENDED PROTOCOLS
The protocols for patient assessment and monitoring are based on expert consensus, and a nationwide trial or registry is still needed to determine which recommended protocol is most suitable. Currently there are no native clinical trials specifically designed to assess the prevention and management of adverse CV effects of cancer therapy, including the timing and choice of intervention. In addition, universal standardized definitions of cardiac endpoints in oncology trials are still lacking, and the awareness of CO for various health care professionals with regards to the long-term risks and need for follow-up still have to be advocated by societies of cardiology, hematology, oncology, and radio-oncology, and through electronic/public media to increase global patient awareness. The number of CO clinics in Taiwan is still limited, and more are needed to provide accessibility and adequate quality to more cancer patients receiving anti-cancer treatment. Only oncologists, radio-oncologists, cardiologists and other related specialties working together in multidisciplinary teams can increase the number of both cancer and CV survivors. Figure 1 summarizes our recommended protocols at baseline, subsequent follow-up, and suggested management when CTRCD events are suspected or encountered.
Figure 1.
Recommended protocol for baseline, follow-up and treatment. CTA, computed tomography angiography; CUT, compression ultrasound test; CV, cardiovascular; DOAC, direct oral anticoagulant; GLS, global longitudinal strain; HFpEF, heart failure with preserved ejection fraction; HFrEF, heart failure with reduced ejection fraction; hs-TnI (T): high sensitive troponin I or T; LMWH, low molecular weight heparin; MRI, magnetic resonance imaging; (NT-pro) BNP, B type natriuretic peptide or N terminal pro B type natriuretic peptide; RAS, renin-angiotensin system; Tx, treatment.
CARDIO-ONCOLOGY EDUCATION AND TRAINING
The JACC Cardio-Oncology Leadership Council recommends bidirectional CO fellowship training for Board-eligible or certified cardiologists/oncologists/hematologists to receive "Exposure and Basic Overview" level 1, "Advanced Clinical Experience and Knowledge" level 2, and "Cardio-oncology Fellowship" level 3 training to gain knowledge and experience of basic cancer biology, treatment principles and CV toxicities associated with solid and hematologic malignancy per se, team collaboration among rehabilitation, nurses, pharmacists, palliative care, in conjunction with the physical, psychological and social needs of the patients.57 Information on new advances in cancer treatment and their CV complications should be periodically updated through cross-talk among cardiology/oncology/radio-oncology societies in order to formulate the best domestic protocols for CTRCD, and through data analyses from domestic trials/registry to contribute to practice guidelines.
Acknowledgments
It is partly supported by the Ministry of Science and Technology of Taiwan (108-2314-B-418-002-MY3).
AUTHOR CONTRIBUTIONS
The first author, Dr. Kai-Hung Cheng, participated in generating original ideas, in manuscript design and in drafting of the manuscript, in revising it critically for important intellectual content and in final approval of the manuscript submitted. Other authors participate in 1) conception and design and interpretation of collecting information: CMH, YWW and CJYH; 2) drafting of the manuscript or revising it for important intellectual content: YWW and CJYH; and 3) All authors provided final approval for publication submission and revised the manuscript for important intellectual content.
DISCLOSURE SUMMARY
The authors have nothing to disclose.
REFERENCES
- 1.Taiwan Ministry of Health and Welfare. Cause of Death Statistics. 2020 [Google Scholar]
- 2.McCune JS. Rapid advances in immunotherapy to treat cancer. Clin Pharmacol Ther. 2018;103:540–544. doi: 10.1002/cpt.985. [DOI] [PubMed] [Google Scholar]
- 3.Siegel RL, Miller KD, Jemal A. Cancer statistics, 2019. CA Cancer J Clin. 2019;69:7–34. doi: 10.3322/caac.21551. [DOI] [PubMed] [Google Scholar]
- 4.Zamorano JL, Lancellotti P, Rodriguez Munoz D, et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur Heart J. 2016;37:2768–2801. doi: 10.1093/eurheartj/ehw211. [DOI] [PubMed] [Google Scholar]
- 5.Armstrong GT, Oeffinger KC, Chen Y, et al. Modifiable risk factors and major cardiac events among adult survivors of childhood cancer. J Clin Oncol. 2013;31:3673–3680. doi: 10.1200/JCO.2013.49.3205. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Ewer MS, Lippman SM. Type II chemotherapy-related cardiac dysfunction: time to recognize a new entity. J Clin Oncol. 2005;23:2900–2902. doi: 10.1200/JCO.2005.05.827. [DOI] [PubMed] [Google Scholar]
- 7.Wang CC, Wu CK, Tsai ML, et al. 2019 focused update of the guidelines of the Taiwan Society of Cardiology for the diagnosis and treatment of heart failure. Acta Cardiol Sin. 2019;35:244–283. doi: 10.6515/ACS.201905_35(3).20190422A. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Plana JC, Galderisi M, Barac A, et al. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging. 2014;15:1063–1093. doi: 10.1093/ehjci/jeu192. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Pilleron S, Sarfati D, Janssen-Heijnen M, et al. Global cancer incidence in older adults, 2012 and 2035: a population-based study. Int J Cancer. 2019;144:49–58. doi: 10.1002/ijc.31664. [DOI] [PubMed] [Google Scholar]
- 10.Cardinale D, Colombo A, Lamantia G, et al. Anthracycline-induced cardiomyopathy: clinical relevance and response to pharmacologic therapy. J Am Coll Cardiol. 2010;55:213–220. doi: 10.1016/j.jacc.2009.03.095. [DOI] [PubMed] [Google Scholar]
- 11.Cardinale D, Colombo A, Bacchiani G, et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation. 2015;131:1981–1988. doi: 10.1161/CIRCULATIONAHA.114.013777. [DOI] [PubMed] [Google Scholar]
- 12.de Azambuja E, Procter MJ, van Veldhuisen DJ, et al. Trastuzumab-associated cardiac events at 8 years of median follow-up in the herceptin adjuvant trial (BIG 1-01). J Clin Oncol. 2014;32:2159–2165. doi: 10.1200/JCO.2013.53.9288. [DOI] [PubMed] [Google Scholar]
- 13.Mahmood SS, Fradley MG, Cohen JV, et al. Myocarditis in patients treated with immune checkpoint inhibitors. J Am Coll Cardiol. 2018;71:1755–1764. doi: 10.1016/j.jacc.2018.02.037. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Bates JE, Howell RM, Liu Q, et al. Therapy-related cardiac risk in childhood cancer survivors: an analysis of the childhood cancer survivor study. J Clin Oncol. 2019;37:1090–1101. doi: 10.1200/JCO.18.01764. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Henry ML, Niu J, Zhang N, et al. Cardiotoxicity and cardiac monitoring among chemotherapy-treated breast cancer patients. JACC Cardiovasc Imaging. 2018;11:1084–1093. doi: 10.1016/j.jcmg.2018.06.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Bijl JM, Roos MM, van Leeuwen-Segarceanu EM, et al. Assessment of valvular disorders in survivors of Hodgkin’s lymphoma treated by mediastinal radiotherapy +/- chemotherapy. Am J Cardiol. 2016;117:691–696. doi: 10.1016/j.amjcard.2015.11.027. [DOI] [PubMed] [Google Scholar]
- 17.Cheng KH, Handschumacher MD, Assuncao B, et al. Contraction timing patterns in patients treated for breast cancer before and after anthracyclines therapy. J Am Soc Echocardiogr. 2017;30:454–460. doi: 10.1016/j.echo.2016.12.013. [DOI] [PubMed] [Google Scholar]
- 18.Heidenreich PA, Hancock SL, Lee BK, et al. Asymptomatic cardiac disease following mediastinal irradiation. J Am Coll Cardiol. 2003;42:743–749. doi: 10.1016/s0735-1097(03)00759-9. [DOI] [PubMed] [Google Scholar]
- 19.Desai MY, Windecker S, Lancellotti P, et al. Prevention, diagnosis, and management of radiation-associated cardiac disease: JACC Scientific Expert Panel. J Am Coll Cardiol. 2019;74:905–927. doi: 10.1016/j.jacc.2019.07.006. [DOI] [PubMed] [Google Scholar]
- 20.Nielsen KM, Offersen BV, Nielsen HM, et al. Short and long term radiation induced cardiovascular disease in patients with cancer. Clin Cardiol. 2017;40:255–261. doi: 10.1002/clc.22634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Cheng YJ, Nie XY, Ji CC, et al. Long-term cardiovascular risk after radiotherapy in women with breast cancer. J Am Heart Assoc. 2017;6 doi: 10.1161/JAHA.117.005633. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Boekel NB, Jacobse JN, Schaapveld M, et al. Cardiovascular disease incidence after internal mammary chain irradiation and anthracycline-based chemotherapy for breast cancer. Br J Cancer. 2018;119:408–418. doi: 10.1038/s41416-018-0159-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Boekel NB, Duane FK, Jacobse JN, et al. Heart failure after treatment for breast cancer. Eur J Heart Fail. 2020;22:366–374. doi: 10.1002/ejhf.1620. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Taylor C, McGale P, Bronnum D, et al. Cardiac structure injury after radiotherapy for breast cancer: cross-sectional study with individual patient data. J Clin Oncol. 2018;36:2288–2296. doi: 10.1200/JCO.2017.77.6351. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.da Silva R. Effects of radiotherapy in coronary artery disease. Curr Atheroscler Rep. 2019;21:50. doi: 10.1007/s11883-019-0810-x. [DOI] [PubMed] [Google Scholar]
- 26.Mansouri I, Allodji RS, Hill C, et al. The role of irradiated heart and left ventricular volumes in heart failure occurrence after childhood cancer. Eur J Heart Fail. 2019;21:509–518. doi: 10.1002/ejhf.1376. [DOI] [PubMed] [Google Scholar]
- 27.Darby SC, Ewertz M, McGale P, et al. Risk of ischemic heart disease in women after radiotherapy for breast cancer. N Engl J Med. 2013;368:987–998. doi: 10.1056/NEJMoa1209825. [DOI] [PubMed] [Google Scholar]
- 28.Plummer C, Steingart RM, Jurczak W, et al. Treatment specific toxicities: hormones, antihormones, radiation therapy. Semin Oncol. 2019;46:414–420. doi: 10.1053/j.seminoncol.2019.01.006. [DOI] [PubMed] [Google Scholar]
- 29.Langendijk JA, Lambin P, De Ruysscher D, et al. Selection of patients for radiotherapy with protons aiming at reduction of side effects: the model-based approach. Radiother Oncol. 2013;107:267–273. doi: 10.1016/j.radonc.2013.05.007. [DOI] [PubMed] [Google Scholar]
- 30.Alvi RM, Frigault MJ, Fradley MG, et al. Cardiovascular events among adults treated with chimeric antigen receptor T-cells (CAR-T). J Am Coll Cardiol. 2019;74:3099–3108. doi: 10.1016/j.jacc.2019.10.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Salem JE, Manouchehri A, Moey M, et al. Cardiovascular toxicities associated with immune checkpoint inhibitors: an observational, retrospective, pharmacovigilance study. Lancet Oncol. 2018;19:1579–1589. doi: 10.1016/S1470-2045(18)30608-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Johnson DB, Balko JM, Compton ML, et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375:1749–1755. doi: 10.1056/NEJMoa1609214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Moslehi JJ, Salem JE, Sosman JA, et al. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet. 2018;391:933. doi: 10.1016/S0140-6736(18)30533-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Ganatra S, Neilan TG. Immune checkpoint inhibitor-associated myocarditis. Oncologist. 2018;23:879–886. doi: 10.1634/theoncologist.2018-0130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Johnson DB, Beckermann KE, Wang DY. Immune checkpoint inhibitor therapy in patients with autoimmune disease. Oncology (Williston Park) 2018;32:190–194. [PubMed] [Google Scholar]
- 36.Postow MA, Sidlow R, Hellmann MD. Immune-related adverse events associated with immune checkpoint blockade. N Engl J Med. 2018;378:158–168. doi: 10.1056/NEJMra1703481. [DOI] [PubMed] [Google Scholar]
- 37.Agrawal N, Khunger A, Vachhani P, et al. Cardiac toxicity associated with immune checkpoint inhibitors: case series and review of the literature. Case Rep Oncol. 2019;12:260–276. doi: 10.1159/000498985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Zhang L, Awadalla M, Mahmood SS, et al. Cardiovascular magnetic resonance in immune checkpoint inhibitor-associated myocarditis. Eur Heart J. 2020;41:1733–1743. doi: 10.1093/eurheartj/ehaa051. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Caforio AL, Pankuweit S, Arbustini E, et al. Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J. 2013;34:2636–2648, 2648a-2648d. doi: 10.1093/eurheartj/eht210. [DOI] [PubMed] [Google Scholar]
- 40.Lynce F, Barac A, Geng X, et al. Prospective evaluation of the cardiac safety of HER2-targeted therapies in patients with HER2-positive breast cancer and compromised heart function: the SAFE-HEaRt study. Breast Cancer Res Treat. 2019;175:595–603. doi: 10.1007/s10549-019-05191-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Hu JR, Florido R, Lipson EJ, et al. Cardiovascular toxicities associated with immune checkpoint inhibitors. Cardiovasc Res. 2019;115:854–868. doi: 10.1093/cvr/cvz026. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Moslehi JJ. Cardiovascular toxic effects of targeted cancer therapies. N Engl J Med. 2016;375:1457–1467. doi: 10.1056/NEJMra1100265. [DOI] [PubMed] [Google Scholar]
- 43.Ameri P, Canepa M, Anker MS, et al. Cancer diagnosis in patients with heart failure: epidemiology, clinical implications and gaps in knowledge. Eur J Heart Fail. 2018;20:879–887. doi: 10.1002/ejhf.1165. [DOI] [PubMed] [Google Scholar]
- 44.Calvillo-Arguelles O, Jaiswal S, Shlush LI, et al. Connections between clonal hematopoiesis, cardiovascular disease, and cancer: a review. JAMA Cardiol. 2019;4:380–387. doi: 10.1001/jamacardio.2019.0302. [DOI] [PubMed] [Google Scholar]
- 45.Armenian SH, Lacchetti C, Lenihan D. Prevention and monitoring of cardiac dysfunction in survivors of adult cancers: American Society of Clinical Oncology Clinical Practice Guideline Summary. J Oncol Pract. 2017;13:270–275. doi: 10.1200/JOP.2016.018770. [DOI] [PubMed] [Google Scholar]
- 46.Plana JC, Thavendiranathan P, Bucciarelli-Ducci C, Lancellotti P. Multi-modality imaging in the assessment of cardiovascular toxicity in the cancer patient. JACC Cardiovasc Imaging. 2018;11:1173–1186. doi: 10.1016/j.jcmg.2018.06.003. [DOI] [PubMed] [Google Scholar]
- 47.Curigliano G, Lenihan D, Fradley M, et al. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol. 2020;31:171–190. doi: 10.1016/j.annonc.2019.10.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Armenian SH, Lacchetti C, Barac A, et al. Prevention and monitoring of cardiac dysfunction in survivors of adult cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol. 2017;35:893–911. doi: 10.1200/JCO.2016.70.5400. [DOI] [PubMed] [Google Scholar]
- 49.Curigliano G, Cardinale D, Suter T, et al. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2012;23 Suppl 7:vii155–vii166. doi: 10.1093/annonc/mds293. [DOI] [PubMed] [Google Scholar]
- 50.Streiff MB, Abutalib SA, Farge D, et al. Update on guidelines for the management of cancer-associated thrombosis. Oncologist. 2021;26:e24–e40. doi: 10.1002/onco.13596. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Navi BB, Reiner AS, Kamel H, et al. Risk of arterial thromboembolism in patients with cancer. J Am Coll Cardiol. 2017;70:926–938. doi: 10.1016/j.jacc.2017.06.047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Lyman GH. Venous thromboembolism in the patient with cancer: focus on burden of disease and benefits of thromboprophylaxis. Cancer. 2011;117:1334–1349. doi: 10.1002/cncr.25714. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Lee AY, Levine MN, Baker RI, et al. Low-molecular-weight heparin versus a coumarin for the prevention of recurrent venous thromboembolism in patients with cancer. N Engl J Med. 2003;349:146–153. doi: 10.1056/NEJMoa025313. [DOI] [PubMed] [Google Scholar]
- 54.Kearon C, Akl EA, Ornelas J, et al. Antithrombotic therapy for VTE disease: CHEST Guideline and Expert Panel Report. Chest. 2016;149:315–352. doi: 10.1016/j.chest.2015.11.026. [DOI] [PubMed] [Google Scholar]
- 55.Ay C, Beyer-Westendorf J, Pabinger I. Treatment of cancer-associated venous thromboembolism in the age of direct oral anticoagulants. Ann Oncol. 2019;30:897–907. doi: 10.1093/annonc/mdz111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Lyman GH, Carrier M, Ay C, et al. American Society of Hematology 2021 guidelines for management of venous thromboembolism: prevention and treatment in patients with cancer. Blood Adv. 2021;5:927–974. doi: 10.1182/bloodadvances.2020003442. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Alvarez-Cardona JA, Ray J, Carver J, et al. Cardio-oncology education and training: JACC council perspectives. J Am Coll Cardiol. 2020;76:2267–2281. doi: 10.1016/j.jacc.2020.08.079. [DOI] [PMC free article] [PubMed] [Google Scholar]

