ecancermedicalscience

Review

Cancer and cardiovascular disease: can understanding the mechanisms of cardiovascular injury guide us to optimise care in cancer survivors?

Lan-Linh Truong1, Laura Scott1, Raveen S Pal1, Mathew Jalink2, Sanjeeva Gunasekara3,4 and Don Thiwanka Wijeratne1,2,5

1Department of Medicine, Queen’s University, Kingston, ON K7L 3N6, Canada

2Department of Public Health, Queen’s University, Kingston, ON K7L 3N6, Canada

3National Cancer Institute, Maharagama 10280, Sri Lanka

4Sri Lanka Cancer Research Group, Colombo 10230, Sri Lanka

5Division of Cancer Care and Epidemiology, Queen’s University Cancer Research Institute, Kingston ON K7L 3N6, Canada


Abstract

Cancer and cardiovascular disease (CVD) are the leading causes of morbidity and mortality. Therefore, CVD deaths in cancer survivors remain a major challenge in improving cancer outcomes, especially in low and middle income countries (LMICs). Cancer and CVD share many common risk factors, both modifiable risk factors (obesity, diabetes and smoking) and non-modifiable factors such as inflammation. Additionally, some cancer therapies are associated with cardiac toxicity. These mechanisms drive increased CVD outcomes in cancer survivors, and understanding this relationship allows us to target therapies to combat such risks. Several commonly used pharmacotherapies for CVD demonstrate promise in cancer survivors for both primary and secondary prevention. Beta blockers and Angiotensin converting enzyme (ACE)-inhibitors have been shown in several studies to improve left ventricular ejection fraction (LVEF) in patients with already established LVEF decline following cancer therapy. Statin use during chemotherapy was associated with lower risk of heart failure and smaller declines in LVEF. Recent studies into the effects of anti-inflammatory medications on cardiovascular events in the non-cancer population have demonstrated promising results and may prove to be an area of further investigation and possible benefit in the cancer population [Canakinumab Anti-Inflammatory Thrombosis Outcomes Study (CANTOS) and Colchicine Cardiovascular Outcomes Trial (COLCOT)]. Additionally, several other medications including PCSK9 inhibitors, sodium-glucose cotransporter-2 inhibitors (SGLT2i) and glucagon-like peptide 1 (GLP-1) agonists have been shown to modify inflammation, and therefore may provide cardiovascular benefits. While common pharmacotherapies used in CVD show promise in cancer survivors, their exact mechanisms remain poorly understood. Few studies evaluate their clinical effectiveness specifically in cancer survivors, as this patient population is excluded from most studies. Further investigation is warranted with more representation of cancer survivors before cost-effective recommendations are made. This is especially true in LMICs where resources are sparse for primary and secondary prevention in order to optimise care in this unique, high-risk population for CVD.

Keywords: chemotherapy-induced cardiotoxicity, cardiovascular disease, cancer survivors, cancer survivorship, chemotherapy-related cardiac dysfunction, heart failure, coronary artery disease, primary prevention, secondary prevention, cardio-oncology

Correspondence to: Don Thiwanka Wijeratne
Email: dtdw@queensu.ca

Published: 14/07/2022
Received: 03/03/2022

Publication costs for this article were supported by ecancer (UK Charity number 1176307).

Copyright: © the authors; licensee ecancermedicalscience. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.


Introduction

Cardiovascular disease (CVD) and cancer account for nearly two-thirds of all non-communicable disease deaths globally [1]. CVD and cancer have long been shown to be leading causes of morbidity and mortality in high income countries and are increasingly becoming leading causes of morbidity and mortality in Low and Middle Income Countries (LMIC) due to alterations in demographics and socio-economic characteristics, as well as a lack of access to medical care in this population [2]. Despite high mortality rates, recent advancements in cancer therapeutics have led to significant improvements in cancer survival, with an estimated 14 million cancer survivors in the United States in 2014 and a projected 19 million by 2024 [3]. CVD is the leading cause of non-cancer mortality among cancer survivors, with the majority of CVD mortality being attributed to either ischaemic heart disease or congestive heart failure (CHF) [4, 5]. In recent years, there have been incredible advancements in cancer treatment; however, there remains an opportunity to further improve survival outcomes in cancer survivors through the prevention and treatment of CVD and its risk factors. The objective of this narrative review is to explore the pathophysiology behind and clinical consequences of CVD in cancer survivors, and the prevention and treatment of CVD in this population. We investigated the shared risk factors that exist between cancer and CVD, the role of surveillance of CVD in cancer survivors and the evidence of pharmacotherapies in management of cancer and cancer-therapy related CVD. We searched the Embase and Medline databases for relevant articles published in the last 10 years (January 2012 to December 2021) using the search terms related to Cardiovascular, Cancer and Pharmacotherapy.


Shared risk factors in cancer and CVD

The development of CVD amongst cancer survivors is likely caused by shared risk factors between the two conditions including: lifestyle factors, cancer-associated inflammation and the iatrogenic effects of cancer therapy. Cancer and CVD share many modifiable lifestyle risk factors including: tobacco use, obesity, sedentary lifestyle, poor diet and excessive alcohol intake [6]. Comorbidities are more common in cancer survivors, who when compared to healthy controls are more likely to have hypertension (59.5% versus 65.9%, p < 0.01), diabetes (21.5% versus 23.4%, p < 0.01), be overweight/obese (35.4% versus 43.4%, p < 0.01) and have a history of smoking (21.2% versus 32.7%, respectively, p < 0.01) [7]. The presence of these risk factors may translate to atherosclerosis and CVD [7].

Inflammation, both preceding and as a result of cancer, is a risk factor for CVD. Over 25% of cancers are linked with increased inflammatory states, whether it be infection, chronic inflammation or autoimmunity [8, 9]. For example, inflammatory bowel disease is known to increase risk of colorectal cancer (CRC) by 10-fold, and management of colitis with anti-inflammatory therapy reduces this risk [10, 11]. Mechanisms of inflammation seen in patients with both cancer and CVD include high levels of chronic inflammation, oxidative stress, altered telomere length and clonal haematopoiesis of indeterminate potential (CHIP) [12, 13]. From a cardiovascular perspective, CHIP is associated with accelerated atherosclerosis, as genes encoded by CHIP are also involved in de-activation of interleukin-6 (IL-6), ultimately leading to increased circulating levels of IL-6 and increased inflammation [14, 15]. Systemic inflammation caused by the malignancy itself and inflammation associated with cancer therapeutics have also been associated with increased CVD and accelerated development of coronary artery disease (CAD) [16, 17].

Cancer therapies can also be associated with the development of cardiac disease [18]. Traditional cytotoxic chemotherapy, such as anthracyclines, have been associated with a variety of cardiovascular side effects. Anthracyclines, for example doxorubicin, are associated with significant, irreversible left ventricular (LV) dysfunction due to decreased LV wall thickness, mass and LV fractional shortening [1922]. While higher cumulative doses of anthracyclines (>300 mg/m2) have been associated with increased risk of cardiotoxicity [23], subclinical echocardiographic abnormalities have been seen in patients treated with lower cumulative doses (<100 mg/m2) [24]. Up to half of survivors treated with anthracyclines experience some form of cardiac dysfunction within 20 years of treatment, causing them to be vulnerable to cardiac dysfunction at younger ages compared to the general population [25, 26]. In more recent years, there have been substantial developments in cancer related therapies, in particular the development of immunotherapy. While immunotherapy has proved to be effective in the treatment of cancer, some agents have also been noted to have cardiovascular side effects. Immune checkpoint inhibitors have been associated with the development of myocarditis, which has been found to be fatal in up to 50% of cases [27, 28]. Chemotherapy related cardiac dysfunction (CTRCD), defined as a >10% drop in left ventricular ejection fraction (LVEF) from baseline, or an absolute value of 53% or less, is a known complication of various cancer therapies (Trastuzumab, Pertuzumab) [29]. CTRCD occurs due to the medication’s interaction with DNA, inhibiting cell replication and subsequent myocyte death [19]. Other cardiotoxic effects include dilated cardiomyopathy, restrictive cardiomyopathy, myocardial infarction, conduction defects, valvular disease, pericardial disease and hypertension [23, 25, 30].

Mediastinal radiation has long been associated with an increased risk of cardiac morbidity and mortality [31]. Cancer survivors who underwent mediastinal irradiation during childhood have significant coronary vessel damage [32]. Almost half had mean coronary artery calcium (CAC) scores of patients 15 years older than their age, although they were often asymptomatic secondary to radiation-related nerve impairment or reduced exercise tolerance [32, 33]. The extent of CAD after mediastinal irradiation is dependent on multiple factors including radiation dose, patient age at the time of radiation, radiation field and time interval since radiation, in addition to the patient’s own personal risk factors [32]. Patients undergoing radiation therapy had a 2.3-fold higher risk for atherosclerotic disease [Odds ratio (OR): 2.3; 95% confidence interval (CI): 1.04–5.3]; and patients undergoing both radiation and chemotherapy had a 4.8-fold increased risk (95% CI: 1.6–14.4) compared to those undergoing surgery alone [34]. Other implications of radiation therapy on the heart include pericarditis, cardiomyopathies and valvular heart disease [35].

The shared risk factors between CVD and cancer are an important area for further investigation, as they may provide opportunities for targeted therapies to reduce the burden of CVD in the cancer population. Additionally, awareness of, and further investigation into the mechanisms of the cardiovascular effects of cancer therapies may serve to provide an opportunity for surveillance and earlier recognition of CVD in cancer patients.


Importance of disease surveillance for CVD in cancer survivors

Screening for cardiotoxicity in cancer survivors is primarily risk-based and is dependent on patient symptoms, cumulative doses of chemotherapy/radiation therapy and the patient’s overall health. Examples of cardiotoxicity screening among cancer patients include both cardiac troponin (cTnT) and N-terminal probrain natriuretic peptide (NT-proBNP), which have been validated as surrogate markers for late LV structural status in long-term survivors of childhood cancer [36, 37]. A study by Lipshultz et al [36] found that elevations in serum cTnT during the first 90 days of anthracycline treatment were associated with reduced LV thickness and increased pathologic LV remodelling 4 years later. Similar findings were true for elevated serum NT-proBNP [36]. An additional study by Cardinale et al [38] found that elevated TnI, another component of the troponin complex, is a risk marker for future development of reduced LVEF. A normal TnI identified patients at lower risk of developing reduced LVEF. This study also found that patients with a normal TnI had no cardiac damage in the first year following high-dose chemotherapy, thus helping separate patients in which close monitoring of cardiac function is required [38].

Studies looking at myocardial strain on imaging have shown a high prevalence of abnormal longitudinal strain in childhood survivors exposed to anthracyclines and/or chest radiotherapy, despite having preserved systolic function [39]. The SUCCOUR (Strain Surveillance of Chemotherapy for Improving Cardiovascular Outcomes) study compared the use of global longitudinal strain (GLS) compared to LVEF as a measure of LV dysfunction to initiate cardioprotective therapy in patients at risk of CTRCD. Researchers noted that at 1-year follow-up, the LVEF was less in the Ejection Fraction (EF)-guided arm compared to the GLS-guided arm (55% ± 7% compared to 57% ± 6%, respectively, p = 0.050) [40]. Routine surveillance with echocardiography is recommended in cancer survivors with high risk for cardiomyopathy, beginning within 2 years after exposure and repeated a minimum of every 5 years thereafter [41]. This recommendation was made after an international collaboration to harmonise existing cardiomyopathy surveillance recommendations in North America and across Europe. The Children’s Oncology Group, a National Cancer Institute supported clinical trials group, recommends surveillance echocardiograms every 1, 2 or 5 years for survivors of childhood cancer, depending on the presence and degree of three risk factors: age at treatment, cumulative anthracycline dose and whether patients received chest radiation [42]. Chest radiation is defined as any radiation in which the heart was in the field of treatment, including mediastinal, thoracic, spinal, left or whole upper abdominal or total body irradiation. Although there are no guidelines directed towards risk stratification of adult cancer survivors in North America, the American College of Cardiology (ACC), the American Society of Echocardiography and the European Association of Cardiovascular Imaging recommend screening echocardiogram 10 years after treatment, and then at 5-year intervals thereafter [43, 44]. For patients deemed high-risk for radiation-induced heart disease, they recommend screening at 5 years post-treatment and non-invasive stress testing every 5 years [44].


Supportive pharmacotherapy to reduce CVD in cancer survivors

Given our understanding of the above mechanisms, primary and secondary prevention in cancer survivors is crucial in reducing the burden of CVD. In this section, we explore the role of individual therapies.

Beta-blockers and renin angiotensin aldosterone system (RAAS) blockade

Beta-blockers (BB) work by blocking the action of endogenous catecholamines on beta-adrenergic receptors, thereby reducing blood pressure, cardiac remodelling and increase filling time with reduced oxygen demand on the heart [45]. The RAAS system regulates blood volume and systemic vascular resistance and its activation triggers sodium reabsorption, shifting fluid into the intravascular space and increasing arterial pressure. The RAAS system also stimulates the release of aldosterone causing further sodium resorption. Additionally, it stimulates thirst, the release of antidiuretic hormone and decreases the sensitivity of the baroreceptor reflex, leading to net increased sodium, total body water and vascular tone [46]. A study by Seicean et al [47] demonstrated that breast cancer patients with structurally normal hearts at baseline who were taking BB during treatment with anthracycline (mean total dose 268.3 ± 163.7 mg) or trastuzumab, with or without radiotherapy, had lower incidence of new heart failure (HF) events. The cumulative incidence of HF at 3 years in patients on continuous BB therapy was 2% (95% CI: 0.8–3.2) compared to 9% (95% CI: 8.3–10.3) in control patients [47]. Another study found that patients taking prophylactic BB therapy with carvedilol or nebivolol prior to anthracycline-based chemotherapy had less LV function decline at 6 months (63.8% ± 3.9% from a baseline of 65.5% ± 4.8%) compared to those who received placebo (LVEF 57.5% ± 5.6% from a baseline of 66.6% ± 5.5%) [48, 49]. The Carvedilol for Prevention of Chemotherapy-Related Cardiotoxicity (CECCY) trial found that carvedilol had no impact on the incidence of early onset or LVEF reduction with a 14.5% incidence of cardiotoxicity compared to 13.5% in the placebo group [50].

A randomised controlled trial by Cardinale et al [51] investigated cancer patients with elevated cardiac enzymes during treatment, a marker for cardiac injury. Participants were randomly assigned to either receive enalapril or no treatment. At baseline evaluation, LVEF was normal in all patients and comparable in the two groups. After 12 months, 25 control subjects (43%) showed a decrease in LVEF by >10% from baseline compared to no patients in the enalapril group (p < 0.001) [51]. In addition, the cumulative number of adverse cardiac events was lower in patients treated with enalapril compared to controls [51]. A separate study by Tallaj et al [52] investigated the use of BB combined with RAAS blockade. In patients with chemotherapy-induced cardiomyopathy, those treated with a combination of ACE-inhibitor (ACEi) and BB had significant improvement in LVEF (26% ± 10.20% versus 37% ± 17.6%, p = 0.028), which was not seen with Angiotensin converting enzyme inhibitor (ACEi) treatment alone [52]. The SAFE-HEaRt (Cardiac Safety of HER2 Targeted Therapy in Patients with HER2 Positive Breast Cancer and Reduced Left Ventricular Function) study was the first prospective study to demonstrate safety of Human Epidermal growth factor Receptor 2 (HER-2) targeted therapies in patients with reduced cardiac function (LVEF 40%–49%) [53]. 90% of patients (27 of 30) receiving cardioprotective therapy with BB and ACEi/ Angiotensin Receptor Blocker (ARBs) throughout the duration of their HER-2 treatment completed planned oncologic therapy without developing a cardiac event or asymptomatic decline in LVEF, thus proving that collaboration between cardiology and oncology can allow for this specific patient population to achieve optimal cancer therapy while minimising the risk of poor cardiac outcomes [53].

The OVERCOME (Prevention of Left Ventricular Dysfunction with Enalapril and Carbedilol in Patients Submitted to Intensive Chemotherapy for Treatment of Malignant Hemopathies) study demonstrated that concomitant treatment with enalapril and carvedilol can prevent LV systolic dysfunction, with a mild 3.1% absolute difference in the mean LVEF between the intervention and control groups [54]. A similar effect was seen in breast cancer patients with suspected trastuzumab-induced LV dysfunction. Once HF symptoms and LVEF were stable following treatment, 25 women who were given ACEi and BB were re-challenged with trastuzumab. Of them, 22 (88%) had stable LVEF without HF symptoms during follow-up, thus speculating that the combined use of ACEi and BB can lead to LV systolic recovery [55]. The MANTICORE (Multidisciplinary Approach to Novel Therapies in Cardio-Oncology Research) study however, found that while prophylactic ACEi and BB were associated with smaller declines in LVEF, it did not prevent concurrent LV remodelling, the primary outcome measure of this study [56].

In addition to LV systolic recovery, BB have been shown to reduce cancer-related death, reoccurrence and metastasis [57]. A retrospective study including a sample size of 466 consecutive patients with operable breast cancer and a follow-up period of >10 years demonstrated that patients taking a BB had a significantly reduced risk of developing metastasis and tumour recurrence and had longer disease-free survival [58]. Breast cancer patients treated with propranolol are significantly less likely to present with advanced disease and have lower cumulative probability of breast cancer-specific mortality compared with matched non-users [59]. A similar effect was seen in a study which included 24,238 patients with head and neck, lung, ovarian, gastric, colon and prostate cancers, where patients using propranolol had a 25% reduction in cancer recurrence compared to non-propranolol users [60]. This is attributed to the ability of Beta-adrenergic signalling to regulate immune responses to tumour cells, the inhibition of apoptosis and the induction of vascular endothelial growth factor [6163].

Sacubitril/Valsartan is recommended in current guidelines for patients with heart failure with reduced ejection fraction (HFrEF) to reduce mortality and hospitalisations, but there remains limited evidence in its effectiveness in patients’ concurrent cancer and HFrEF. One study found significantly improved LVEF, with 8 of the included 67 patients’ LVEF normalising. They also noted a significant reduction in NT-proBNP levels and improvement in exercise tolerance, as indicated by change in New York Heart Association (NYHA) functional class [64]. A separate study that included 635 patients noted similar findings, with baseline median NT-proBNP 997.5 pg/ml (InterQuartile Range (IQR): 663.8–2,380.8), which decreased to a median of 416.5 pg/ml (IQR: 192.0–798.2) with p < 0.001 [65]. They also found improvements in baseline NYHA functional class and increased LVEF from 26.7% ± 5.4% to 32.3% ± 5.5% (p < 0.001) [65].

Sacubitril/Valsartan is recommended in current guidelines for patients with heart failure with reduced ejection fraction (HFrEF) to reduce mortality and hospitalisations, but there remains limited evidence in its effectiveness in patients’ concurrent cancer and HFrEF. One study found significantly improved LVEF, with 8 of the included 67 patients’ LVEF normalising. They also noted a significant reduction in NT-proBNP levels and improvement in exercise tolerance, as indicated by change in NYHA functional class [64]. A separate study that included 635 patients noted similar findings, with baseline median NT-proBNP 997.5 pg/ml (IQR: 663.8–2,380.8), which decreased to a median of 416.5 pg/ml (IQR: 192.0–798.2) with p < 0.001 [65]. They also found improvements in baseline NYHA functional class and increased LVEF from 26.7% ± 5.4% to 32.3% ± 5.5% (p < 0.001) [65].

Aspirin

Aspirin is an antiplatelet agent that has long been the mainstay for both secondary prevention of Myocardial Infarction (MI) and stroke [66]. It acts by inhibiting cyclooxygenase (COX) activity, thus inhibits the synthesis of prostanoids, which are involved in modulating inflammatory responses, gastrointestinal cytoprotection and ulceration, atheroprotection and haemostasis among many other functions [67, 68]. Aspirin is administered at low doses to preferentially affect platelet COX-1 activity, but also has some COX-2 activity [69]. In terms of cancer progression, multiple studies have shown that aberrant COX-2 expression is a contributing factor in promoting CRC, with increased expression allowing for elevated prostanoid biosynthesis, and in turn, contribution to the initial steps of tumorigenesis [70]. Overexpression of COX-2 also increases cell migration and proliferation in intestinal epithelial cells [71].

Use of aspirin in primary prevention is currently not recommended in guidelines for conventional diabetic and hypertensive populations; however, cancer patients often have augmented risks of atherosclerosis. A meta-analysis found that aspirin was associated with a reduction in adverse cardiovascular events, but was also associated with an increased risk of major bleeding [72]. Its role in CRC specifically has been evaluated, where it was found to be effective in reducing the risk of CRC-related death in Randomized control trial (RCTs), thus suggesting that the antiplatelet action contributes to the prevention of both atherosclerosis and cancer [73, 74]. As such, the decision to start a patient on aspirin would require assessment of each patient’s risk–benefit profile [75]. The American College of Cardiology (ACC) recommends calculating 10-year CVD risk for patients aged 40–79 [66]. The Coronary artery calcium (CAC) score that is calculated from a cardiac Computed tomography (CT) may have added utility in predicting a nearly 10-fold increase in CVD events in patients with elevated calcium scores [76]. Using CAC score, and distribution of calcification amongst the coronaries, could help guide therapy to higher-risk patients especially those patients with premature atherosclerosis in cancer survivors [77, 78].

The medications discussed below have limited evidence on the utility in cancer survivors with very little reference to cancer populations. We discuss some of the potential benefits that may be applicable to these patients that warrant further study.

Lipid lowering medication

Statins

Statins have long been used in management of hypercholesterolaemia by inhibiting the rate-limiting enzyme involved in cholesterol synthesis [79]. The intermediate products involved in cholesterol synthesis activate various downstream signalling pathways, including regulation of inflammatory cytokines and chemokines [80]. Therefore, beyond cholesterol reduction, statins also decrease oxidative stress, inflammation and the number of inflammatory cells in atherosclerotic plaques [8082]. With these considerations, it is suggestive that statins can be considered another mechanism to protect against cardiotoxicity induced by cancer and cancer therapies.

Several small, prospective studies have evaluated the effect of atorvastatin on inflammatory markers in patients with LV systolic dysfunction with most noting a significant decrease in the concentration of inflammatory markers in patients with ischaemic and non-ischaemic cardiomyopathy [8385]. A cohort study demonstrated that women treated with concomitant statins during anthracycline-based chemotherapy had lower risk of HF [Hazard ratio (HR): 0.3; CI: 0.1–0.9; p = 0.03] compared to those in the non-statin treated comparison group [82]. Another noted that statin therapy resulted in smaller declines in mean LVEF compared to those not using a statin (−1.3 [3.8%] versus −7.9% [8.0%]) [86]. In terms of LV remodelling, one study noted a significant decrease in LV end-diastolic dimension (57.1 mm to 53.4 mm), compared to patients in the placebo group, who experienced an increase in LV end-diastolic dimension (56.1 mm to 60.3 mm) over a 12-month period [83]. The results of these studies are promising and suggest that statins may have a beneficial effect on CVD in individuals with cancer; however, further investigation is needed to determine the extent of the possible benefit of statins in this population.

PCSK-9 inhibitors

Proprotein convertase subtilisin/kexin type 9 serine protease (PCSK9) encodes a protein that prevents removal of low density lipoproteins (LDL) particles from the blood stream, thus PCSK9 inhibitors lead to decreased LDL concentrations and reduced risk of CVD [87, 88]. PCSK9 is also expressed in tissues including the brain, kidney and vascular wall [89]. In smooth muscle cells, it directly increases inflammation via the Nuclear Factor kappa B (NF-kB) pathway, thus promotes plaque monocyte infiltration and macrophage inflammation, and in turn contributing to the development of atherosclerosis [90, 91]. Interventions using monoclonal antibodies (MABs) have been the most commonly investigated and have consistently demonstrated significant efficacy in reducing LDL. A study by Shapiro et al [92] saw reductions in LDL by approximately 50% when MABs inhibiting PCSK9 were used as monotherapy, and approximately 70% when used in combination with statins and ezetimibe with excellent short-term safety and tolerability profile.

There was a strong correlation between intracellular lipid accumulation and expression of C-C chemokine Receptor type 2 (CCR2), a chemokine receptor involved in monocyte chemotaxis, implying a causal relation between lipid levels and pro-inflammatory changes [93]. As such, the use of PCSK9 MABs was associated with reversing the pro-inflammatory profile of monocytes in patients with familial hypercholesterolaemia, decreased tumour necrosis factor (TNF) release and increased secretion of the anti-inflammatory cytokine IL-10 [93]. This suggests that in addition to its lipid lowering properties, PCSK9 MABs also reduce CVD by reducing the inflammation that underlies atherosclerosis. The component involving inflammation is especially relevant in cancer survivors, given the increased inflammation occurring as a consequence of cancer diagnosis and therapeutics and the relationship between increased inflammation and CVD. Given this same hypothesis, PCSK9 MABs have also demonstrated reduction in cancer risks [94]. At this time, there are no long-term studies evaluating the effect of PCSK9 MABs in reducing CVD risk factors in cancer survivors, though is likely a new emerging area for research.

Role of antidiabetics

Sodium-glucose cotransporter-2 (SGLT2) inhibitors

SGLT2 inhibitors are used in the management of type 2 diabetes mellitus (T2DM) and work by inhibiting glucose reabsorption in kidneys [95]. SGLT2 cotransports sodium (Na) with glucose, such that the inhibition of SGLT2 leads to reduced reabsorption of both glucose and Na, leading to plasma volume contraction, decreased systolic and diastolic blood pressure with cardiovascular and renal benefits [96]. The plasma volume contraction haemodynamically unloads the LV, thereby decreasing myocardial oxygen demand, ventricular wall tension and filling pressures [97]. The Empagliflozin, Cardiovascular Outcomes, and Mortality in Type 2 Diabetes (EMPA-REG OUTCOME) trial found that in patients with diabetes, when compared to those receiving placebo, patients receiving empagliflozin had a 14% reduction in cardiovascular death, nonfatal MI, nonfatal stroke and >30% reduction in cardiovascular mortality, overall mortality and HF hospitalisations. This finding is especially notable, as there was only a marginal difference in HbA1c between the two groups [96]. A meta-analysis by Zhang et al [98] including 351,476 patients demonstrated similar results with statistically significant reduction in risk of major adverse cardiac events, all-cause mortality, cardiovascular mortality, nonfatal MI and hospitalisation for HF. SGLT2 inhibitors were also shown to have favourable cardiovascular outcomes in patients with pre-existing HF with both reduced and preserved ejection fraction [99, 100].

Decreased glucose metabolism associated with SGLT2 inhibitors could also help modulate inflammatory processes that contribute to CVD. For example, inflammatory macrophages preferentially use glucose through the glycolysis pathway [101]. SGLT2 inhibitors also improve the differentiation of epicardial adipose tissue, a known source of inflammatory mediators, and thus reduce the secretion of proinflammatory cytokines [102]. As a result, it is hypothesised that the use of SGLT2 inhibitors could reduce inflammatory processes in vascular endothelial cells and also contribute to weight loss thereby reducing the risk of CVD and cancer [101]. These results appear to be encouraging; however, further investigation in the cancer survivor population is warranted to determine the benefit of SGLT2 inhibitors in this population.

Glucagon-like peptide 1 (GLP1) agonists

GLP1 is a peptide secreted by enteroendocrine cells primarily involved in stimulating glucose-dependent insulin secretion and incretin signalling, though has also been linked with regulating inflammation and cardiovascular function [103]. Activation of the GLP1 receptor (GLP1-R) leads to inhibition of gastric and small bowel motility, leading to delayed nutrient absorption and reduced appetite. Hence, the use of GLP1 agonists in the management of diabetes has the added benefit of weight loss [104].

In addition, GLP1 is linked with regulating local and systemic inflammation and cardiovascular function [103]. GLP1 levels are shown to be elevated in chronic and acute inflammatory processes such as sepsis or chronic kidney disease, and correlated with the severity of illness and clinical outcomes [105]. The LEADER (Liraglutide and Cardiovascular Outcomes in Type 2 Diabetes) trial, which included 9,340 patients with T2DM, found that the occurrence of major adverse cardiac events was reduced in the liraglutide group [106]. In addition, fewer patients died from cardiovascular causes in the liraglutide group than in the control group [106]. A similar finding occurred in a separate study with 14,752 subjects using either once-a-week administration of exenatide, a long-acting GLP1 agonist, compared to placebo [107].

The added effect of weight loss with GLP1 agonists further reduces cardiovascular risk factors in cancer survivors. Long-term survivors of cancer are more likely to have poor adherence to dietary and physical activity guidelines, and cancer treatment regimens often include corticosteroids, which are known to increase percentage body fat and caloric intake in survivors [108, 109]. Vigorous exercise has been associated with a lower risk of CVD in a dose-dependent manner, independent of clinical and treatment-related risk factors [110]. As previously discussed, cancer therapeutics are known to cause LV dysfunction and in turn, can be attributed to impaired cardiovascular fitness and exercise tolerance [109]. Hence, the combined effect of reducing inflammation and weight loss makes GLP1 agonists a promising medication for the prevention of CVD in cancer survivors who are also diabetic, although they have not specifically been studied in these patients.

Other medications

Dexrazoxane

Dexrazoxane is a chelating agent that binds to intracellular iron before it enters cardiomyocytes, thus decreasing free radical formation and reducing cardiomyocyte apoptosis. It is currently the only Food and Drug Administration (FDA)-approved drug used for the prevention of anthracycline-related cardiotoxicity [111]. Several trials evaluating breast cancer patients undergoing chemotherapy with doxorubicin have shown a decrease in the risk of developing of HF when dexrazoxane was added to treatment [112]. A meta-analysis estimated an overall reduction in cardiac events by 65% while others have estimated a reduction of up to 82% [113]. It has also been shown to prevent cardiotoxicity in children and adolescents treated with anthracyclines [114]. This same study also highlighted that dexrazoxane exhibits its cardioprotective effects without decreasing the effectiveness of anthracyclines or affecting event-free survival [114]. It should be noted however, that the cardioprotective activity of dexrazoxane is not fool-proof as anthracyclines have several mechanisms causing cardiotoxicity, and dexrazoxane offers protection for some, but not all mechanisms of cardiotoxicity [115117]. This may benefit cancer survivors and is an area that warrants further study.

Biologics

TNF is a pro-inflammatory cytokine secreted primarily by immune cells and is involved in both inflammation, cell proliferation, apoptosis and lipid metabolism [118]. While anti-TNF agents have been long used in the treatment of autoimmune inflammatory conditions, its role in other inflammatory conditions such as CVD has not been extensively studied. TNF drives inflammation and plaque formation in atherosclerosis and thus its inhibition is a potential target for the prevention of CVD [119]. Multiple observational studies have shown that TNF inhibition reduces atherosclerosis and cardiovascular events when administered to patients with rheumatoid arthritis [120]. Interestingly, TNF levels post-MI are a strong predictor of recurrent events [121]. In addition, higher TNF levels are associated with higher odds of CAD (OR: 2.25, 95% CI: 1.50–3.37) and ischaemic stroke (OR: 0.54, 95% CI: 0.42–0.96) [118].

The CANTOS study, which sought to test the inflammatory hypothesis of atherosclerosis, included patients with a history of MI who were either assigned to receive placebo or a MAB canakinumab, which targets IL-1β, an inflammatory cytokine [122, 123]. Patients taking canakinumab had significantly reduced levels of C-reactive protein (CRP) and IL-6 compared to placebo. Patients receiving a 150 mg dose of canakinumab had 15% lower risk of primary end point (non-fatal MI, non-fatal stroke or cardiovascular death) compared to the placebo group (3.86 versus 4.50 events per 100 person years). Interestingly, cancer mortality was found to be significantly lower in patients receiving canakinumab than those receiving placebo, consistent with the notion that inflammation is a risk factor for cancer morbidity and mortality [8, 123]. It specifically showed a dose-dependent reduction in risk of lung cancer, up to 67% [124]. However, the use of Canakinumab as first-, second- or third-line treatment with chemotherapy in non-small cell lung cancer did not confirm the benefit [125]. Unfortunately, separate trials investigating the use TNFa inhibitors (etanercept and infliximab) in HF were stopped due to worsening CHF and worse prognosis compared to placebo [126, 127]. This mechanism is poorly understood, especially considering that elevated TNFa is consistently associated with CHF. Further investigation is needed to determine that if there is a roll for TNFa inhibitors in the cancer population.


Conclusion

Our narrative review outlines several strategies in reducing the burden of CVD in cancer survivors. The suggested strategies are based on the notion that CVD and cancer share many common risk factors, both modifiable risk factors and biochemical properties such as inflammation. Multiple cancer therapeutics are also associated with cardiac toxicity. We hypothesise that these three mechanisms are the driving forces behind poor CVD outcomes in cancer survivors, and that this understanding can facilitate therapies to reduce such risks.

Of the various drug classes investigated, several demonstrate promise and may warrant further investigation for potential integration into management of CVD in cancer survivors. The use of BB combined with ACEi was shown in several studies to prevent LVEF decline in patients receiving anti-cancer therapies and improve LVEF in patients who have already suffered LV dysfunction following cancer therapy [4749, 52, 128]. The use of statins during chemotherapy was associated with lower risk of HF and smaller declines in LVEF [82, 86]. PCSK9 MABs were associated with decreased TNF release and increased secretion of IL-10, an anti-inflammatory cytokine [93]. The use of SGLT2 inhibitors is known to improve cardiovascular outcomes and has been shown to reduce oxidative stress and inflammation [96, 98, 101]. GLP1 agonists, in addition to their ability to manage diabetes, have been associated with weight loss and increased GLP1 levels have been correlated with severity of inflammatory processes [105].

While common pharmacotherapies for CVD show promise in cancer survivors, their exact mechanism remains poorly understood. Only a few studies evaluated the clinical utility of these therapies in cancer survivors, often excluding this very population from most studies. Further investigation with more representation of cancer survivors assessing their clinical outcomes is warranted before recommendations are made for primary and secondary prevention in this unique high-risk population for CVD.

Due to shifting patterns of demographic and socio-economic characteristics in LMICs, CVD and cancer are fast becoming the leading causes of morbidity and mortality [129]. This problem is further exacerbated by limited access to expertise, investigations and medicines needed for treating these patients [2]. Therefore, CVD deaths in cancer survivors are a major challenge in improving cancer outcomes in LMICs. In these countries, adapting risk based cardiac screening protocols for cancer survivors, controlling CVD risk factors in cancer patients and choosing less cardiotoxic treatment regimens might help in compensating for some of these existing care gaps [12, 42, 130].


Conflicts of interest

The authors in this paper have no conflicts of interest to declare.


Funding statement

This article was not directly funded.


References

1. World Health Organization (2021) Noncommunicable diseases fact sheet 2021 [https://www.who.int/news-room/fact-sheets/detail/noncommunicable-diseases] Date accessed: 19/01/22

2. Agyemang C and van den Born BJ (2018) Limited access to CVD medicines in low-income and middle-income countries: poverty is at the heart of the matter Lancet Glob Health 6 e234–e235 https://doi.org/10.1016/S2214-109X(18)30048-2 PMID: 29433655

3. DeSantis CE, Lin CC, and Mariotto AB, et al (2014)Cancer treatment and survivorship statistics, 2014 CA Cancer J Clin 64 252–271 https://doi.org/10.3322/caac.21235 PMID: 24890451

4. Zaorsky NG, Churilla TM, and Egleston BL, et al (2017) Causes of death among cancer patients Ann Oncol 28 400–407 https://doi.org/10.1093/annonc/mdw604

5. Fidler MM, Reulen RC, and Henson K, et al (2017) Population-based long-term cardiac-specific mortality among 34 489 five-year survivors of childhood cancer in Great Britain Circulation 135 951–963 https://doi.org/10.1161/CIRCULATIONAHA.116.024811 PMID: 28082386 PMCID: 5338891

6. Johnson CB, Davis MK, and Law A, et al (2016) Shared risk factors for cardiovascular disease and cancer: implications for preventive health and clinical care in oncology patients Can J Cardiol 32 900–907 https://doi.org/10.1016/j.cjca.2016.04.008 PMID: 27343745

7. Armenian SH, Xu L, and Ky B, et al (2016) Cardiovascular disease among survivors of adult-onset cancer: a community-based retrospective cohort study J Clin Oncol 34 1122–1130 https://doi.org/10.1200/JCO.2015.64.0409 PMID: 26834065 PMCID: 7357493

8. Grivennikov SI, Greten FR, and Karin M (2010) Immunity, inflammation, and cancer Cell 140 883–899 https://doi.org/10.1016/j.cell.2010.01.025 PMID: 20303878 PMCID: 2866629

9. Mantovani A, Allavena P, and Sica A, et al (2008) Cancer-related inflammation Nature 454 436–444 https://doi.org/10.1038/nature07205 PMID: 18650914

10. Prior P, Gyde SN, and Macartney JC, et al (1982) Cancer morbidity in ulcerative colitis Gut 23 490–497 https://doi.org/10.1136/gut.23.6.490 PMID: 7076024 PMCID: 1419715

11. Eaden J, Abrams K, and Ekbom A, et al (2000) Colorectal cancer prevention in ulcerative colitis: a case-control study Aliment Pharmacol Ther 14 145–153 https://doi.org/10.1046/j.1365-2036.2000.00698.x PMID: 10651654

12. Koene RJ, Prizment AE, and Blaes A, et al (2016) Shared risk factors in cardiovascular disease and cancer Circulation 133 1104–1114 https://doi.org/10.1161/CIRCULATIONAHA.115.020406 PMID: 26976915 PMCID: 4800750

13. Masoudkabir F, Sarrafzadegan N, and Gotay C, et al (2017) Cardiovascular disease and cancer: Evidence for shared disease pathways and pharmacologic prevention Atherosclerosis 263 343–351 https://doi.org/10.1016/j.atherosclerosis.2017.06.001 PMID: 28624099 PMCID: 6207942

14. Zhang Q, Zhao K, and Shen Q, et al (2015) Tet2 is required to resolve inflammation by recruiting Hdac2 to specifically repress IL-6 Nature 525 389–393 https://doi.org/10.1038/nature15252 PMID: 26287468 PMCID: 4697747

15. Ferrone CK, Blydt-Hansen M, and Rauh MJ (2020) Age-associated TET2 mutations: common drivers of myeloid dysfunction, cancer and cardiovascular disease Int J Mol Sci 21 626 https://doi.org/10.3390/ijms21020626 PMCID: 7014315

16. Okwuosa TM, Anzevino S, and Rao R (2017) Cardiovascular disease in cancer survivors Postgrad Med J 93 82–90 https://doi.org/10.1136/postgradmedj-2016-134417 PMID: 28123076

17. Chang HM, Moudgil R, and Scarabelli T, et al (2017) Cardiovascular complications of cancer therapy: best practices in diagnosis, prevention, and management: part 1 J Am Coll Cardiol 70 2536–2551 https://doi.org/10.1016/j.jacc.2017.09.1096 PMID: 29145954 PMCID: 5825187

18. Whitlock MC, Yeboah J, and Burke GL, et al (2015) Cancer and its association with the development of coronary artery calcification: an assessment from the multi-ethnic study of atherosclerosis J Am Heart Assoc 4 e002533 https://doi.org/10.1161/JAHA.115.002533 PMID: 26553214 PMCID: 4845242

19. McGowan JV, Chung R, and Maulik A, et al (2017) Anthracycline chemotherapy and cardiotoxicity Cardiovasc Drugs Ther 31 63–75 https://doi.org/10.1007/s10557-016-6711-0 PMID: 28185035 PMCID: 5346598

20. Lipshultz SE, Colan SD, and Gelber RD, et al (1991) Late cardiac effects of doxorubicin therapy for acute lymphoblastic leukemia in childhood N Engl J Med 324 808–815 https://doi.org/10.1056/NEJM199103213241205 PMID: 1997853

21. Lipshultz SE (2006) Exposure to anthracyclines during childhood causes cardiac injury Semin Oncol 33 S8–14 https://doi.org/10.1053/j.seminoncol.2006.04.019 PMID: 16781284

22. Lipshultz SE, Lipsitz SR, and Sallan SE, et al (2005) Chronic progressive cardiac dysfunction years after doxorubicin therapy for childhood acute lymphoblastic leukemia J Clin Oncol 23 2629–2636 https://doi.org/10.1200/JCO.2005.12.121 PMID: 15837978

23. Lipshultz SE, Franco VI, and Miller TL, et al (2015) Cardiovascular disease in adult survivors of childhood cancer Annu Rev Med 66 161–176 https://doi.org/10.1146/annurev-med-070213-054849 PMID: 25587648 PMCID: 5057395

24. Leger K, Slone T, and Lemler M, et al (2015) Subclinical cardiotoxicity in childhood cancer survivors exposed to very low dose anthracycline therapy Pediatr Blood Cancer 62 123–127 https://doi.org/10.1002/pbc.25206

25. Mulrooney DA, Armstrong GT, and Huang S, et al (2016) Cardiac outcomes in adult survivors of childhood cancer exposed to cardiotoxic therapy: a cross-sectional study Ann Intern Med 164 93–101 https://doi.org/10.7326/M15-0424 PMID: 26747086 PMCID: 4809016

26. Lipshultz SE, Lipsitz SR, Mone SM, et al (1995) Female sex and higher drug dose as risk factors for late cardiotoxic effects of doxorubicin therapy for childhood cancer N Engl J Med 332 1738–1743 https://doi.org/10.1056/NEJM199506293322602 PMID: 7760889

27. Ganatra S and Neilan TG (2018) Immune checkpoint inhibitor-associated myocarditis Oncologist 23 879–886 https://doi.org/10.1634/theoncologist.2018-0130 PMID: 29802219 PMCID: 6156176

28. Palaskas N, Lopez-Mattei J, and Durand JB, et al (2020) Immune checkpoint inhibitor myocarditis: pathophysiological characteristics, diagnosis, and treatment J Am Heart Assoc 9 e013757 https://doi.org/10.1161/JAHA.119.013757 PMID: 31960755 PMCID: 7033840

29. Cardinale D, Colombo A, and Bacchiani G, et al (2015) Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy Circulation 131 1981–1988 https://doi.org/10.1161/CIRCULATIONAHA.114.013777 PMID: 25948538

30. Lipshultz SE, Cochran TR, and Franco VI, et al (2013) Treatment-related cardiotoxicity in survivors of childhood cancer Nat Rev Clin Oncol 10 697–710 https://doi.org/10.1038/nrclinonc.2013.195 PMID: 24165948

31. Hancock SL, Donaldson SS, and Hoppe RT (1993) Cardiac disease following treatment of Hodgkin’s disease in children and adolescents J Clin Oncol 11 1208–1215 https://doi.org/10.1200/JCO.1993.11.7.1208 PMID: 8315419

32. Rademaker J, Schoder H, and Ariaratnam NS, et al (2008) Coronary artery disease after radiation therapy for Hodgkin’s lymphoma: coronary CT angiography findings and calcium scores in nine asymptomatic patients AJR Am J Roentgenol 191 32–37 https://doi.org/10.2214/AJR.07.3112 PMID: 18562721

33. Yusuf SW, Daraban N, and Abbasi N, et al (2012) Treatment and outcomes of acute coronary syndrome in the cancer population Clin Cardiol 35 443–450 https://doi.org/10.1002/clc.22007 PMID: 22588943 PMCID: 6652695

34. Haugnes HS, Wethal T, and Aass N, et al (2010) Cardiovascular risk factors and morbidity in long-term survivors of testicular cancer: a 20-year follow-up study J Clin Oncol 28 4649–4657 https://doi.org/10.1200/JCO.2010.29.9362 PMID: 20855830

35. Wang H, Wei J, and Zheng Q, et al (2019) Radiation-induced heart disease: a review of classification, mechanism and prevention Int J Biol Sci 15 2128–2138 https://doi.org/10.7150/ijbs.35460 PMID: 31592122 PMCID: 6775290

36. Lipshultz SE, Miller TL, and Lipsitz SR, et al (2012) Continuous versus bolus infusion of doxorubicin in children with all: long-term cardiac outcomes Pediatrics 130 1003–1011 https://doi.org/10.1542/peds.2012-0727 PMID: 23166343 PMCID: 3507254

37. Lipshultz SE, Rifai N, and Dalton VM, et al (2004) The effect of dexrazoxane on myocardial injury in doxorubicin-treated children with acute lymphoblastic leukemia N Engl J Med 351 145–153 https://doi.org/10.1056/NEJMoa035153 PMID: 15247354

38. Cardinale D, Sandri MT, and Martinoni A, et al (2002) Myocardial injury revealed by plasma troponin I in breast cancer treated with high-dose chemotherapy Ann Oncol 13 710–715 https://doi.org/10.1093/annonc/mdf170 PMID: 12075738

39. Armstrong GT, Joshi VM, and Ness KK, et al (2015) Comprehensive echocardiographic detection of treatment-related cardiac dysfunction in adult survivors of childhood cancer: results from the St. Jude lifetime cohort study J Am Coll Cardiol 65 2511–2522 https://doi.org/10.1016/j.jacc.2015.04.013 PMID: 26065990 PMCID: 4539123

40. Thavendiranathan P, Negishi T, and Somerset E, et al (2021) Strain-guided management of potentially cardiotoxic cancer therapy J Am Coll Cardiol 77 392–401 https://doi.org/10.1016/j.jacc.2020.11.020

41. Armenian SH, Hudson MM, and Mulder RL, et al (2015) Recommendations for cardiomyopathy surveillance for survivors of childhood cancer: a report from the International Late Effects of Childhood Cancer Guideline Harmonization Group Lancet Oncol 16 e123–e136 https://doi.org/10.1016/S1470-2045(14)70409-7 PMID: 25752563 PMCID: 4485458

42. Wong FL, Bhatia S, and Landier W, et al (2014) Cost-effectiveness of the children’s oncology group long-term follow-up screening guidelines for childhood cancer survivors at risk for treatment-related heart failure Ann Intern Med 160 672–683 https://doi.org/10.7326/M13-2498 PMID: 24842414 PMCID: 4073480

43. Desai MY, Windecker S, and Lancellotti P, et al (2019) Prevention, diagnosis, and management of radiation-associated cardiac disease: JACC scientific expert panel J Am Coll Cardiol 74 905–927 https://doi.org/10.1016/j.jacc.2019.07.006 PMID: 31416535

44. Lancellotti P, Nkomo VT, and Badano LP, et al (2013) Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography Eur Heart J Cardiovasc Imaging 14 721–740 https://doi.org/10.1093/ehjci/jet123 PMID: 23847385

45. Collaborators GBDRF (2016) Global, regional, and national comparative risk assessment of 79 behavioural, environmental and occupational, and metabolic risks or clusters of risks, 1990-2015: a systematic analysis for the Global Burden of Disease Study 2015 Lancet 388 1659–1724 https://doi.org/10.1016/S0140-6736(16)31679-8

46. Fountain JH and Lappin SL (2020) Physiology, Renin Angiotensin System (FL: StatPearls)

47. Seicean S, Seicean A, and Alan N, et al (2013) Cardioprotective effect of beta-adrenoceptor blockade in patients with breast cancer undergoing chemotherapy: follow-up study of heart failure Circ Heart Fail 6 420–426 https://doi.org/10.1161/CIRCHEARTFAILURE.112.000055 PMID: 23425978

48. Kaya MG, Ozkan M, and Gunebakmaz O, et al (2013) Protective effects of nebivolol against anthracycline-induced cardiomyopathy: a randomized control study Int J Cardiol 167 2306–2310 https://doi.org/10.1016/j.ijcard.2012.06.023

49. Kalay N, Basar E, and Ozdogru I, et al (2006) Protective effects of carvedilol against anthracycline-induced cardiomyopathy J Am Coll Cardiol 48 2258–2262 https://doi.org/10.1016/j.jacc.2006.07.052 PMID: 17161256

50. Avila MS, Ayub-Ferreira SM, and de Barros Wanderley MR, Jr., et al (2018) Carvedilol for prevention of chemotherapy-related cardiotoxicity: the CECCY trial J Am Coll Cardiol 71 2281–2290 https://doi.org/10.1016/j.jacc.2018.02.049 PMID: 29540327

51. Cardinale D, Colombo A, and Sandri MT, et al (2006) Prevention of high-dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition Circulation 114 2474–2481 https://doi.org/10.1161/CIRCULATIONAHA.106.635144 PMID: 17101852

52. Tallaj JA, Franco V, and Rayburn BK, et al (2005) Response of doxorubicin-induced cardiomyopathy to the current management strategy of heart failure J Heart Lung Transplant 24 2196–2201 https://doi.org/10.1016/j.healun.2004.12.108 PMID: 16364871

53. Lynce F, Barac A, and Geng X, et al (2019) 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 175 595–603 https://doi.org/10.1007/s10549-019-05191-2 PMID: 30852761 PMCID: 6534513

54. Bosch X, Rovira M, and Sitges M, et al (2013) Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies) J Am Coll Cardiol 61 2355–2362 https://doi.org/10.1016/j.jacc.2013.02.072 PMID: 23583763

55. Ewer MS, Vooletich MT, and Durand JB, et al (2005) Reversibility of trastuzumab-related cardiotoxicity: new insights based on clinical course and response to medical treatment J Clin Oncol 23 7820–7826 https://doi.org/10.1200/JCO.2005.13.300 PMID: 16258084

56. Pituskin E, Mackey JR, and Koshman S, et al (2017) Multidisciplinary approach to novel therapies in cardio-oncology research (MANTICORE 101-Breast): a randomized trial for the prevention of trastuzumab-associated cardiotoxicity J Clin Oncol 35 870–877 https://doi.org/10.1200/JCO.2016.68.7830

57. Botteri E, Munzone E, and Rotmensz N, et al (2013) Therapeutic effect of beta-blockers in triple-negative breast cancer postmenopausal women Breast Cancer Res Treat 140 567–575 https://doi.org/10.1007/s10549-013-2654-3 PMID: 23912960

58. Powe DG, Voss MJ, and Zanker KS, et al (2010) Beta-blocker drug therapy reduces secondary cancer formation in breast cancer and improves cancer specific survival Oncotarget 1 628–638 https://doi.org/10.18632/oncotarget.197

59. Barron TI, Connolly RM, and Sharp L, et al (2011) Beta blockers and breast cancer mortality: a population- based study J Clin Oncol 29 2635–2644 https://doi.org/10.1200/JCO.2010.33.5422 PMID: 21632503

60. Chang PY, Huang WY, and Lin CL, et al (2015) Propranolol reduces cancer risk: a population-based cohort study Medicine (Baltimore) 94 e1097 https://doi.org/10.1097/MD.0000000000001097

61. Sloan EK, Priceman SJ, and Cox BF, et al (2010) The sympathetic nervous system induces a metastatic switch in primary breast cancer Cancer Res 70 7042–7052 https://doi.org/10.1158/0008-5472.CAN-10-0522 PMID: 20823155 PMCID: 2940980

62. Sood AK, Armaiz-Pena GN, and Halder J, et al (2010) Adrenergic modulation of focal adhesion kinase protects human ovarian cancer cells from anoikis J Clin Invest 120 1515–1523 https://doi.org/10.1172/JCI40802 PMID: 20389021 PMCID: 2860925

63. Lutgendorf SK, Cole S, and Costanzo E, et al (2003) Stress-related mediators stimulate vascular endothelial growth factor secretion by two ovarian cancer cell lines Clin Cancer Res 9 4514–4521 PMID: 14555525

64. Martin-Garcia A, Lopez-Fernandez T, and Mitroi C, et al (2020) Effectiveness of sacubitril-valsartan in cancer patients with heart failure ESC Heart Fail 7 763–767 https://doi.org/10.1002/ehf2.12627 PMID: 32022485 PMCID: 7160493

65. Gregorietti V, Fernandez TL, and Costa D, et al (2020) Use of Sacubitril/valsartan in patients with cardio toxicity and heart failure due to chemotherapy Cardiooncology 6 24 PMID: 33292750 PMCID: 7643279

66. Arnett DK, Blumenthal RS, and Albert MA, et al (2019) 2019 ACC/AHA guideline on the primary prevention of cardiovascular disease: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines Circulation 140 e596–e646 PMID: 30879355 PMCID: 7734661

67. Capone ML, Tacconelli S, and Di Francesco L, et al (2007) Pharmacodynamic of cyclooxygenase inhibitors in humans Prostaglandins Other Lipid Mediat 82 85–94 https://doi.org/10.1016/j.prostaglandins.2006.05.019

68. Smyth EM, Grosser T, and Wang M, et al (2009) Prostanoids in health and disease J Lipid Res 50 Suppl S423–S428 https://doi.org/10.1194/jlr.R800094-JLR200

69. Patrignani P and Patrono C (2015) Cyclooxygenase inhibitors: from pharmacology to clinical read-outs Biochim Biophys Acta 1851 422–432 https://doi.org/10.1016/j.bbalip.2014.09.016

70. Dixon DA, Blanco FF, and Bruno A, et al (2013) Mechanistic aspects of COX-2 expression in colorectal neoplasia Recent Results Cancer Res 191 7–37 https://doi.org/10.1007/978-3-642-30331-9_2

71. Wang D and Dubois RN (2010) The role of COX-2 in intestinal inflammation and colorectal cancer Oncogene 29 781–788 https://doi.org/10.1038/onc.2009.421

72. Zheng SL and Roddick AJ (2019) Association of aspirin use for primary prevention with cardiovascular events and bleeding events: a systematic review and meta-analysis J Am Med Assoc 321 277–287 https://doi.org/10.1001/jama.2018.20578

73. Rothwell PM, Wilson M, and Elwin CE, et al (2010) Long-term effect of aspirin on colorectal cancer incidence and mortality: 20-year follow-up of five randomised trials Lancet 376 1741–1750 https://doi.org/10.1016/S0140-6736(10)61543-7 PMID: 20970847

74. Rothwell PM, Price JF, and Fowkes FG, et al (2012) Short-term effects of daily aspirin on cancer incidence, mortality, and non-vascular death: analysis of the time course of risks and benefits in 51 randomised controlled trials Lancet 379 1602–1612 https://doi.org/10.1016/S0140-6736(11)61720-0 PMID: 22440946

75. Dasa O, Pepine CJ, and Pearson TA (2020) Aspirin in primary prevention: what changed? A critical appraisal of current evidence Am J Cardiol 141 38–48 https://doi.org/10.1016/j.amjcard.2020.11.014 PMID: 33221264

76. Detrano R, Guerci AD, and Carr JJ, et al (2008) Coronary calcium as a predictor of coronary events in four racial or ethnic groups N Engl J Med 358 1336–1345 https://doi.org/10.1056/NEJMoa072100 PMID: 18367736

77. Cooney MT, Dudina A, and Whincup P, et al (2009) Re-evaluating the Rose approach: comparative benefits of the population and high-risk preventive strategies Eur J Cardiovasc Prev Rehabil 16 541–549 https://doi.org/10.1097/HJR.0b013e32832b38a1 PMID: 19773660

78. Ferencik M, Pencina KM, and Liu T, et al (2017) Coronary artery calcium distribution is an independent predictor of incident major coronary heart disease events: results from the Framingham heart study Circ Cardiovasc Imaging 10 e006592 https://doi.org/10.1161/CIRCIMAGING.117.006592 PMID: 28956774 PMCID: 5659296

79. LaRosa JC, He J, and Vupputuri S (1999) Effect of statins on risk of coronary disease: a meta-analysis of randomized controlled trials J Am Med Assoc 282 2340–2346 https://doi.org/10.1001/jama.282.24.2340

80. Ramasubbu K, Estep J, and White DL, et al (2008) Experimental and clinical basis for the use of statins in patients with ischemic and nonischemic cardiomyopathy J Am Coll Cardiol 51 415–426 https://doi.org/10.1016/j.jacc.2007.10.009 PMID: 18222351

81. Zhou Q and Liao JK (2009) Statins and cardiovascular diseases: from cholesterol lowering to pleiotropy Curr Pharm Des 15 467–478 https://doi.org/10.2174/138161209787315684 PMID: 19199975 PMCID: 2896785

82. Seicean S, Seicean A, and Plana JC, et al (2012) Effect of statin therapy on the risk for incident heart failure in patients with breast cancer receiving anthracycline chemotherapy: an observational clinical cohort study J Am Coll Cardiol 60 2384–2390 https://doi.org/10.1016/j.jacc.2012.07.067 PMID: 23141499

83. Sola S, Mir MQ, and Lerakis S, et al (2006) Atorvastatin improves left ventricular systolic function and serum markers of inflammation in nonischemic heart failure J Am Coll Cardiol 47 332–337 https://doi.org/10.1016/j.jacc.2005.06.088 PMID: 16412856

84. Mozaffarian D, Minami E, and Letterer RA, et al (2005) The effects of atorvastatin (10 mg) on systemic inflammation in heart failure Am J Cardiol 96 1699–1704 https://doi.org/10.1016/j.amjcard.2005.07.092 PMID: 16360360

85. Tousoulis D, Antoniades C, and Vassiliadou C, et al (2005) Effects of combined administration of low dose atorvastatin and vitamin E on inflammatory markers and endothelial function in patients with heart failure Eur J Heart Fail 7 1126–1132 https://doi.org/10.1016/j.ejheart.2005.03.007 PMID: 16051518

86. Acar Z, Kale A, and Turgut M, et al (2011) Efficiency of atorvastatin in the protection of anthracycline-induced cardiomyopathy J Am Coll Cardiol 58 988–989 https://doi.org/10.1016/j.jacc.2011.05.025 PMID: 21851890

87. Zhang DW, Garuti R, and Tang WJ, et al (2008) Structural requirements for PCSK9-mediated degradation of the low-density lipoprotein receptor Proc Natl Acad Sci U S A 105 13045–13050 https://doi.org/10.1073/pnas.0806312105 PMID: 18753623 PMCID: 2526098

88. Cohen JC, Boerwinkle E, and Mosley TH Jr, et al (2006) Sequence variations in PCSK9, low LDL, and protection against coronary heart disease N Engl J Med 354 1264–1272 https://doi.org/10.1056/NEJMoa054013 PMID: 16554528

89. Seidah NG, Abifadel M, and Prost S, et al (2017) The proprotein convertases in hypercholesterolemia and cardiovascular diseases: emphasis on proprotein convertase subtilisin/Kexin 9 Pharmacol Rev 69 33–52 https://doi.org/10.1124/pr.116.012989

90. Giunzioni I, Tavori H, and Covarrubias R, et al (2016) Local effects of human PCSK9 on the atherosclerotic lesion J Pathol 238 52–62 https://doi.org/10.1002/path.4630

91. Bai XQ, Peng J, and Wang MM, et al (2018) PCSK9: a potential regulator of apoE/apoER2 against inflammation in atherosclerosis? Clin Chim Acta 483 192–196 https://doi.org/10.1016/j.cca.2018.04.040 PMID: 29727700

92. Shapiro MD, Fazio S, and Tavori H (2015) Targeting PCSK9 for therapeutic gains Curr Atheroscler Rep 17 499 https://doi.org/10.1007/s11883-015-0499-4 PMID: 25712137 PMCID: 5560054

93. Bernelot Moens SJ, Neele AE, and Kroon J, et al (2017) PCSK9 monoclonal antibodies reverse the pro-inflammatory profile of monocytes in familial hypercholesterolaemia Eur Heart J 38 1584–1593 https://doi.org/10.1093/eurheartj/ehx002 PMID: 28329114

94. Nowak C and Arnlov J (2018) A Mendelian randomization study of the effects of blood lipids on breast cancer risk Nat Commun 9 3957 https://doi.org/10.1038/s41467-018-06467-9 PMID: 30262900 PMCID: 6160471

95. Marx N and McGuire DK (2016) Sodium-glucose cotransporter-2 inhibition for the reduction of cardiovascular events in high-risk patients with diabetes mellitus Eur Heart J 37 3192–3200 https://doi.org/10.1093/eurheartj/ehw110 PMID: 27153861

96. Heerspink HJ, Perkins BA, and Fitchett DH, et al (2016) Sodium glucose cotransporter 2 inhibitors in the treatment of diabetes mellitus: cardiovascular and kidney effects, potential mechanisms, and clinical applications Circulation 134 752–772 https://doi.org/10.1161/CIRCULATIONAHA.116.021887 PMID: 27470878

97. Cherney DZ, Perkins BA, and Soleymanlou N, et al (2014) The effect of empagliflozin on arterial stiffness and heart rate variability in subjects with uncomplicated type 1 diabetes mellitus Cardiovasc Diabetol 13 28 https://doi.org/10.1186/1475-2840-13-28 PMID: 24475922 PMCID: 3915232

98. Zhang XL, Zhu QQ, and Chen YH, et al (2018) Cardiovascular safety, long-term noncardiovascular safety, and efficacy of sodium-glucose cotransporter 2 inhibitors in patients with type 2 diabetes mellitus: a systemic review and meta-analysis with trial sequential analysis J Am Heart Assoc 7 e007165 https://doi.org/10.1161/JAHA.117.007165 PMID: 29353233 PMCID: 5850151

99. Anker SD, Butler J, and Filippatos GS, et al (2019) Evaluation of the effects of sodium–glucose co‐transporter 2 inhibition with empagliflozin on morbidity and mortality in patients with chronic heart failure and a preserved ejection fraction: rationale for and design of the EMPEROR‐Preserved Trial Eur J Heart Fail 21 1279–1287 https://doi.org/10.1002/ejhf.1596 PMID: 31523904

100. Packer M, Butler J, and Filippatos GS, et al (2019) Evaluation of the effect of sodium–glucose co‐transporter 2 inhibition with empagliflozin on morbidity and mortality of patients with chronic heart failure and a reduced ejection fraction: rationale for and design of the EMPEROR‐Reduced trial Euro J Heart Fail 21 1270–1278 https://doi.org/10.1002/ejhf.1536

101. Staels B (2017) Cardiovascular protection by sodium glucose cotransporter 2 inhibitors: potential mechanisms Am J Med 130 S30–S39 https://doi.org/10.1016/j.amjmed.2017.04.009 PMID: 28526184

102. Mazurek T, Zhang L, and Zalewski A, et al (2003) Human epicardial adipose tissue is a source of inflammatory mediators Circulation 108 2460–2466 https://doi.org/10.1161/01.CIR.0000099542.57313.C5 PMID: 14581396

103. Drucker DJ (2016) The cardiovascular biology of glucagon-like peptide-1 Cell Metab 24 15–30. https://doi.org/10.1016/j.cmet.2016.06.009 PMID: 27345422

104. Meier JJ (2012) GLP-1 receptor agonists for individualized treatment of type 2 diabetes mellitus Nat Rev Endocrinol 8 728–742 https://doi.org/10.1038/nrendo.2012.140 PMID: 22945360

105. Lebherz C, Schlieper G, and Mollmann J, et al (2017) GLP-1 levels predict mortality in patients with critical illness as well as end-stage renal disease Am J Med 130 833–841e3 https://doi.org/10.1016/j.amjmed.2017.03.010 PMID: 28366423

106. Marso SP, Daniels GH, and Brown-Frandsen K, et al (2016) Liraglutide and cardiovascular outcomes in type 2 diabetes N Engl J Med 375 311–322 https://doi.org/10.1056/NEJMoa1603827 PMID: 27295427 PMCID: 4985288

107. Holman RR, Bethel MA, and Mentz RJ, et al (2017) Effects of once-weekly exenatide on cardiovascular outcomes in type 2 diabetes N Engl J Med 377 1228–1239 https://doi.org/10.1056/NEJMoa1612917 PMID: 28910237

108. Zhang FF, Kelly MJ, and Must A (2017) Early nutrition and physical activity interventions in childhood cancer survivors Curr Obes Rep 6 168–177 https://doi.org/10.1007/s13679-017-0260-0 PMID: 28455678 PMCID: 5501167

109. Chow EJ, Pihoker C, and Hunt K, et al (2007) Obesity and hypertension among children after treatment for acute lymphoblastic leukemia Cancer 110 2313–2320 https://doi.org/10.1002/cncr.23050 PMID: 17896787

110. Jones LW, Liu Q, and Armstrong GT, et al (2014) Exercise and risk of major cardiovascular events in adult survivors of childhood hodgkin lymphoma: a report from the childhood cancer survivor study J Clin Oncol 32 3643–3650 https://doi.org/10.1200/JCO.2014.56.7511 PMID: 25311213 PMCID: 4220043

111. Timm KN and Tyler DJ (2020) The role of AMPK activation for cardioprotection in doxorubicin-induced cardiotoxicity Cardiovasc Drugs Ther 34 255–269 https://doi.org/10.1007/s10557-020-06941-x PMID: 32034646 PMCID: 7125062

112. Bhave M, Shah AN, and Akhter N, Ret al (2014) An update on the risk prediction and prevention of anticancer therapy-induced cardiotoxicity Curr Opin Oncol 26 590–599 https://doi.org/10.1097/CCO.0000000000000132 PMID: 25233068

113. Kalam K and Marwick TH (2013) Role of cardioprotective therapy for prevention of cardiotoxicity with chemotherapy: a systematic review and meta-analysis Eur J Cancer 49 2900–2909 https://doi.org/10.1016/j.ejca.2013.04.030 PMID: 23706982

114. Lipshultz SE, Adams MJ, and Colan SD, et al (2013) Long-term cardiovascular toxicity in children, adolescents, and young adults who receive cancer therapy: pathophysiology, course, monitoring, management, prevention, and research directions: a scientific statement from the American Heart Association Circulation 128 1927–1995 https://doi.org/10.1161/CIR.0b013e3182a88099 PMID: 24081971

115. Limat S, Demesmay K, and Fagnoni P, et al (2005) Cost effectiveness of cardioprotective strategies in patients with aggressive non-Hodgkin’s lymphoma Clin Drug Investig 25 719–729 https://doi.org/10.2165/00044011-200525110-00005

116. Lipshultz SE, Lipsitz SR, and Sallan SE, et al (2002) Long-term enalapril therapy for left ventricular dysfunction in doxorubicin-treated survivors of childhood cancer J Clin Oncol 20 4517–4522 https://doi.org/10.1200/JCO.2002.12.102 PMID: 12454107

117. Silber JH, Cnaan A, and Clark BJ, et al (2004) Enalapril to prevent cardiac function decline in long-term survivors of pediatric cancer exposed to anthracyclines J Clin Oncol 22 820–828 https://doi.org/10.1200/JCO.2004.06.022 PMID: 14990637

118. Yuan S, Carter P, and Bruzelius M, et al (2020) Effects of tumour necrosis factor on cardiovascular disease and cancer: a two-sample Mendelian randomization study EBioMedicine 59 102956

119. Branen L, Hovgaard L, and Nitulescu M, et al (2004) Inhibition of tumor necrosis factor-alpha reduces atherosclerosis in apolipoprotein E knockout mice Arterioscler Thromb Vasc Biol 24 2137–2142 https://doi.org/10.1161/01.ATV.0000143933.20616.1b PMID: 15345516

120. Greenberg JD, Furer V, and Farkouh ME (2011) Cardiovascular safety of biologic therapies for the treatment of RA Nat Rev Rheumatol 8 13–21 https://doi.org/10.1038/nrrheum.2011.168 PMID: 22083220

121. Ridker PM, Rifai N, and Pfeffer M, et al (2000) Elevation of tumor necrosis factor-alpha and increased risk of recurrent coronary events after myocardial infarction Circulation 101 2149–2153 https://doi.org/10.1161/01.CIR.101.18.2149 PMID: 10801754

122. Ruperto N, Brunner HI, and Quartier P, et al (2012) Two randomized trials of canakinumab in systemic juvenile idiopathic arthritis N Engl J Med 367 2396–2406 https://doi.org/10.1056/NEJMoa1205099 PMID: 23252526

123. Tardif JC, Kouz S, and Waters DD, et al (2019) Efficacy and safety of low-dose colchicine after myocardial infarction N Engl J Med 381(26) 2497-505

124. Ridker PM, Howard CP, and Walter V, et al (2012) Effects of interleukin-1beta inhibition with canakinumab on hemoglobin A1c, lipids, C-reactive protein, interleukin-6, and fibrinogen: a phase IIb randomized, placebo-controlled trial Circulation 126 2739–2748 https://doi.org/10.1161/CIRCULATIONAHA.112.122556 PMID: 23129601

125. Novartis (2021) Novartis provides update on Phase III study evaluating canakinumab (ACZ885) as second or third-line treatment in combination with chemotherapy in non-small cell lung cancer [https://www.novartis.com/news/media-releases/novartis-provides-update-phase-iii-study-evaluating-canakinumab-acz885-second-or-third-line-treatment-combination-chemotherapy-non-small-cell-lung-cancer: Novartis]

126. Mann DL, McMurray JJ, and Packer M, et al (2004) Targeted anticytokine therapy in patients with chronic heart failure: results of the Randomized Etanercept Worldwide Evaluation (RENEWAL) Circulation 109 1594–1602 https://doi.org/10.1161/01.CIR.0000124490.27666.B2 PMID: 15023878

127. Chung ES, Packer M, and Lo KH, et al (2003) Randomized, double-blind, placebo-controlled, pilot trial of infliximab, a chimeric monoclonal antibody to tumor necrosis factor-alpha, in patients with moderate-to-severe heart failure: results of the anti-TNF Therapy Against Congestive Heart Failure (ATTACH) trial Circulation 107 3133–3140 https://doi.org/10.1161/01.CIR.0000077913.60364.D2 PMID: 12796126

128. Noori A, Lindenfeld J, and Wolfel E, et al (2000) Beta-blockade in adriamycin-induced cardiomyopathy J Card Fail 6 115–119 PMID: 10908085

129. Ezzati M, Pearson-Stuttard J, and Bennett JE, et al (2018) Acting on non-communicable diseases in low-and middle-income tropical countries Nature 559 507–516 https://doi.org/10.1038/s41586-018-0306-9 PMID: 30046068

130. Schindera C, Zürcher SJ, and Jung R, et al (2021) Physical fitness and modifiable cardiovascular disease risk factors in survivors of childhood cancer: a report from the SURfit study Cancer 127 1690–1698 https://doi.org/10.1002/cncr.33351 PMID: 33405260

Related Articles

Fernanda J Martin, Isabel M Saffie, Mabel A Hurtado, Diana Avila-Jaque, Rodrigo A Lagos, Carolina A Selman, Jonathan Z Huserman, Valentina A Castillo, Badir J Chahuán
Salah Eddine El Herrag, Soraya Moulessehoul, Douniazad El Mehadji, Djamila Yekrou, Méghit Boumediène Khaled
Nancy S Bolous, Peter Mercredi, Miguel Bonilla, Paola Friedrich, Nickhill Bhakta, Monika L Metzger, Pascale Y Gassant
Table of Contents
Table of Contents