Open Access

Mechanism and prevention strategy of a bidirectional relationship between heart failure and cancer (Review)

  • Authors:
    • He Cai
    • Hongbo Men
    • Pengyu Cao
    • Yang Zheng
  • View Affiliations

  • Published online on: October 20, 2021     https://doi.org/10.3892/etm.2021.10898
  • Article Number: 1463
  • Copyright: © Cai et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The relationship between cancer and heart failure has been extensively studied in the last decade. These studies have focused on describing heart injury caused by certain cancer treatments, including radiotherapy, chemotherapy and targeted therapy. Previous studies have demonstrated a higher incidence of cancer in patients with heart failure. Heart failure enhances an over‑activation of the sympathetic nervous system and the renin‑angiotensin‑aldosterone system, and subsequently promotes cancer development. Other studies have found that heart failure and cancer both have a common pathological origin, flanked by chronic inflammation in certain organs. The present review aims to summarize and describe the recent discoveries, suggested mechanisms and relationships between heart failure and cancer. The current review provides more ideas on clinical prevention strategies according to the pathological mechanism involved.

1. Introduction

Heart failure and cancer are two major diseases that affect human health, and they represent most causes of death and disability in humans (1,2), The incidence of heart failure and cancer in oldindividualshas increased quickly in the world (3,4). In previous studies, epidemiologists have revealed that cancer treatment makes patients more likely to suffer from heart failure; this cardio-oncology research focused on the prevention and treatment of cardiac damage caused by cancer treatment. Also, the cardiac damage caused by cancer treatment and treatment of cancer patients with heart disease are discussed in the present review (Table I) (3,5-12). A number of studies have suggested that patients with heart failure are more likely to have cancer (Table II) (13-25); however, the mechanisms and relationships between heart failure and cancer remain unclear. Certain studies have even confirmed the presence of a precancerous lesion before carcinogenesis in heart failure (26,27), and suggested that both heart failure and cancer are chronic low-level inflammatory diseases (26). The pathogenesis of heart failure caused by cancer treatment and the mechanism of cancer occurrence in patients with heart failure is currently unclear. The present review outlines the relationship between heart failure and cancer, and provides clinical strategies towards prevention according to the pathological mechanism.

Table I

High risk of heart failure observed in different cancers and certain cancer treatments that can damage the heart.

Table I

High risk of heart failure observed in different cancers and certain cancer treatments that can damage the heart.

First author, yearCancerAnticancer therapy/drugsTargetHeart failure(Refs.)
Armenian et al, 2016Multiple myeloma, lung cancer, non-Hodgkin's lymphoma, breast cancerA variety of anticancer therapiesA variety of anticancer therapiesCompared with healthy individuals, the incidence of heart failure in patients with cancer is higher(3)
Swain et al, 2003Breast cancer and lung cancerDoxorubicinTopoisomerase 2Incidence rate: 26%(5)
Saiki et al, 2017Breast cancerRadiationCoronary artery endothelial cellsIncidence rate: 18%(6)
Slamon et al, 2001Breast cancerRecombinant monoclonal antibodyAnti-HER2Incidence rate: 27%(7)
Quarta et al, 2014Immunoglobulin amyloid light-chain diseaseDifferent therapiesCardiac amyloidosisVentricular wall thickening(8)
van Nimwegen et al, 2015Hodgkin's lymphomaAnthracyclinesDNAIncidence rate: 11%(9)
Laufer-Perl et al, 2020Breast cancerRecombinant monoclonal antibodyAnti-HER2Incidence rate: 5%(10)
Tian et al, 2020Soft-tissue sarcomaDoxorubicinTopoisomerase 2Incidence rate: 4.17%(11)
Degens et al, 2020Non-small cell lung cancerRadiationCoronary artery endothelial cellsIncidence rate: 7.6%(12)

Table II

Patients with heart failure have a high risk of developing cancer.

Table II

Patients with heart failure have a high risk of developing cancer.

First author, yearFollow-up, yearsNon-cardiovascular deaths attributable to cancer, %(Refs.)
Kjekshus et al, 20072.719.9(13)
Linde et al, 2008116.7(14)
Moss et al, 2009431.1(15)
Ruschitzka et al, 20131.621.7(16)
McMurray et al, 20142.327.8(17)
Doval et al, 19941.125.0(18)
Bigger, 19972.737.1(19)
Kadish et al, 20042.466.7(20)
Granger et al, 20032.847.8(21)
McMurray et al, 20033.438.6(22)
Roy et al, 20083.138.6(23)
Tavazzi et al, 20083.944.5(24)
Velazquez et al, 20114.731.5(25)

2. Mechanism of heart failure in patients with cancer

Cancer has a high risk of associated cardiac toxicity, and treatments such as radiation, chemotherapy or immunosuppressive therapy can also severely affect the heart. At present, the mechanisms behind cardiac toxicity and treatment-associated effects are described in Fig. 1 and the following text (28,29).

Cardiotoxicity of chemotherapy and radiotherapy

Anthracyclines are commonly used as chemotherapeutic drugs for solid and hematological cancer types. Anthracyclines produce a large number of reactive oxygen free radicals, which consequently cause myocardial injuries (30). Anthracycline effects induce acute or chronic cardiotoxicity depending on the dosage of the drug, ranging from 5% (cumulative dose of 400 mg/m2) to 26% (cumulative dose of 550 mg/m2) cardiotoxicity (5,9). However, a study has reported that patients with hematological diseases treated with low doses of anthracyclines still have cardiac malfunction (31). This class of drug has the advantage that after its injection, it is intercalated into the DNA and blocks the activity of topoisomerase 2, which subsequently inhibits the proliferation of cancerous cells (32). It has been demonstrated that cardiac topoisomerase is a key mediator of doxorubicin-induced cardiotoxicity, which may reduce the efficacy of treatment (33,34). In fact, doxorubicin induces apoptosis and DNA damage in a topoisomerase-dependent manner; it also ultimately affects oxidative phosphorylation and mitochondrial biogenesis (34). Dexrazoxane, a topoisomerase inhibitor, is currently used as an effective drug for preventing and treating heart injury caused by radiotherapy and chemotherapy (35,36). Other studies previously revealed that angiotensin-converting enzyme inhibitors (ACEIs) prevented heart injury, and that, phosphoinositide 3-kinase γ removed damaged mitochondria in a heart failure model induced by Adriamycin, suggesting possible treatments to prevent anthracycline-induced cardiotoxicity (37,38). Radiotherapy is the most common treatment for breast cancer. Usually, patients who have received total radiation exposure of >30 Gy, with daily radiation exposure of >2 Gy, have radiation exposure to the left or front of the chest; without radiation protection, the heart can easily manifest symptoms of cardiac damage, including left main coronary disease and pericarditis (39,40). One study revealed that radiation therapy can directly cause myocardial damage through reactive oxygen species (ROS)-induced activation of Ca2+/calmodulin-dependent protein kinase II (41). Radiation therapy can also cause vascular endothelial cell damage, which may contribute to coronary heart disease (6).

Targeted anticancer drug treatment causes heart failure

Trastuzumab, a monoclonal antibody against HER2, is an effective first-line drug for breast cancer (7). By binding to HER2, the trastuzumab molecule blocks the binding of human epidermal growth factor to HER2, thereby inhibiting the growth of cancerous cells (7). The cardiotoxicity of trastuzumab mostly results from symptomatic heart failure or subclinical left ventricular dysfunction (42). However, the effect of trastuzumab on the heart is reversible (43) through the activities of vascular endothelial growth factor (VEGF), which is an important regulator of angiogenesis (44). When cancer metastasizes, VEGF promotes neovascularization to provide nutrition to the cancer (44). The VEGF gene family consists of five members, which can activate downstream signaling pathways after binding to the corresponding VEGF receptor (VEGFR) (44,45); this phenomenon occurs by blocking the VEGF signaling pathway and includes the use of anti-VEGF/VEGFR monoclonal antibodies and VEGFR-tyrosine kinase inhibitor (TKIs). Drugs targeting VEGF signaling, including humanized anti-VEGF monoclonal antibody, humanized bevacizumab, TKIs and sorafenib, have certain cardiovascular side effects such as hypertension, thromboembolism and cardiomyopathy (44,46). It has been reported that the administration of bevacizumab combined with anthracyclines increases the incidence of heart failure from 4 to 14% (47). Meanwhile, VEGF can increase the release of nitric oxide, facilitate prostacyclin synthesis and decrease the expression of pro-inflammatory genes such as cyclooxygenase-2 and E-selectin. This suggests that anti-VEGF antibodies might cause hypertension and thromboembolic diseases (48,49). Certain studies have demonstrated that inhibitors of VEGF can damage endothelial cells and increase their microparticle production, while the microparticles can stimulate endothelial cells to generate certain reactions capable of causing further damage to the endothelial cells; among those reactions, massive production of endothelin-1, excessive oxidative stress and inflammatory activation are the most commonly observed (46,50-52). In order to improve the safety of TKI drugs, the need for further studies and an improved understanding of the mechanism of cardiac injury appears crucial. Finally, cardiotoxicity is also related to proteasome inhibitors, which are useful for the treatment of multiple myeloma and other hematological malignancies. According to a meta-analysis, the second-generation proteasome inhibitor carfilzomib was associated with higher cardiotoxicity, with an 18% incidence of cardiovascular adverse events (53,54). Furthermore, in pigs, inhibition of the ubiquitin-proteasome system of cardiomyocytes led to decreased cardiac function and the generation of possible cardiac damage (55).

The human ether-à-go-go-related (HERG) gene belongs to the voltage-activated outwardly-rectifying EAG family of K+ channels and is expressed in multiple tissue types, including cardiac, neural and smooth muscle tissues. HERG loss of function leads to long QT syndrome (56) and has been demonstrated to contribute to the occurrence of cancer. Furthermore, transfection of HERG can induce the malignant transformation of murine fibroblasts, while HERG blocker (dofetilide) can reverse this process (57). In previous, HERG channel antagonists have emerged as new target drugs for cancer treatment. However, the cardiotoxicity of HERG antagonists remains the major problem for this method of cancer treatment. HERG antagonists block the HERG channel, inhibit the proliferation and migration of cancerous cells, and inhibit the potassium channel of myocardial cells, resulting in severe arrhythmia (58,59). The most common arrhythmias are long QT syndrome and ventricular tachycardia; therefore, designing a drug that can be administered safely at a reasonable dosage is emerging as the main preventive strategy for HERG antagonist-related toxicity (60).

Cancer itself can cause heart failure

Cancer can cause cardiomyopathy, light chain amyloidosis and carcinoid heart disease. Amyloidosis is a disease that affects multiple organs, including the myocardium and heart valves in restrictive cardiomyopathy (61,62). Heart failure caused by light-chain amyloidosis is severe and might be related to the direct damage of light-chain amyloidosis in myocardial cells (8). Studies have also shown that oxidative stress may cause damage in myocardial cells (63,64). Currently, the treatment of events caused by cardiac light-chain amyloidosis is limited to the treatment of heart failure and related malignant cancer types, and there is no available treatment for light chain protein deposition (61). Carcinoid heart disease is caused by the release of vasoactive mediators, such as serotonin, bradykinin and histamine. Neuroendocrine tumors (NETs) are a rare type of cancer found in the gastrointestinal or respiratory tracts. Mediators released by NETs are inactivated in the liver and pulmonary blood vessels; therefore, carcinoid heart disease also occurs in the liver with cancer metastases in the stomach and intestines, which mainly damages the right ventricle, with bronchial carcinoids as an outcome (65,66). Carcinoid heart diseases are characterized by the formation of fibrotic plaques in the myocardium, which eventually leads to right-sided heart failure. In addition, fibrotic remodeling of the tricuspid valve results in regurgitation by the valve, leading to decompensation of the right ventricle. Medical treatment of carcinoid syndrome is limited to somatostatin analogs, but this treatment is not effective for the heart muscle itself or valvular disease (67). Clinically, although amyloidosis and carcinoid heart disease are the only forms of heart failure caused by cancer, it has been demonstrated in other studies that certain cancer types may affect cardiac function by releasing cardiac toxic cancer-related metabolites (68,69). In rats, this mutation stimulates the accumulation and release of D-2-hydroxyglutarate, which impairs Krebs cycle activity in the heart and inhibits contractile function (70).

3. Mechanism of cancer in patients with heart failure

In the last decade, epidemiology studies have reported a high incidence of cancer in patients with heart failure (71,72). However, these studies only indicate the relationship between cancer and heart failure; meanwhile, its related mechanism is still unclear. Possible mechanisms that interfere in such relationships (73) are shown in Fig. 1 and described in the following text.

Hypothesis from circulation factors

As an endocrine organ, during heart failure, the heart can secrete a number of circulating factors, including B-type natriuretic peptide, which can be used for the diagnosis/risk stratification and prognosis of heart failure (1,74). However, various cancer-generated circulating factors influence surrounding organs (75,76). In heart failure combined with cancer, increased secretion of important factors, including tumor necrosis factor, interleukin (IL)-6, IL-1 and VEGF, occurs. Numerous studies have shown that heart failure stimulates cancer growth. For example, Meijers et al (27) demonstrated that heart failure enhanced cancer growth in adenomatous polyposis coli mice. Compared with that in sham-operated mice, the number and size of the tumors in mice with heart failure increased by three-fold. The occurrence and development of cancer is related to cardiac remodeling markers such as left ventricular ejection fraction (LVEF) and myocardial fibrosis. In order to further verify these findings, Meijers et al (27) established a hemodynamic injury-free model and found that heart failure accelerated cancer growth. This suggested that heart failure stimulating cancer growth is not related to the myocardial infarction model, while some circulating factors secreted by the heart itself during heart failure may stimulate cancer growth (27). Certain proteomics studies discovered that several circulating protein factors were secreted into the blood during the occurrence of heart failure (77,78); those protein factors might have various effects on colon tissue in vitro. Among them, α-1-antichymotrypsin (SerpinA3) promotes cancer growth by phosphorylating Akt and ribosomal protein s6 in vitro (27). A community cohort study with a total of 8,592 subjects showed that over a follow-up time of 12 years, 1,132 subjects (13.1%) were diagnosed with cancer, and among these, 132 (11.7%) were diagnosed with colorectal cancer (27). The N-terminal pro-B-type natriuretic peptide is an independent risk factor for colorectal cancer in patients with heart failure, and the risk of cancer increases with increasing concentration of the peptide (27,79). Together, these studies indicate that the secretion of certain biomarkers produced by the heart is not only a signal of myocardial injury, but also affects the growth of distant cancer, possibly through cardiac exocrine effects. In addition, Bertero et al (26) conducted a study focusing on underlying mechanisms such as inflammation and neurohormones, which provided some preliminary evidence that heart failure could result from the adaptation of the body's environment to the onset or development of cancer.

Neurohormone activation

Activation of the renin-angiotensin-aldosterone system (RAAS) is one of the central compensatory homeostatic responses in patients with heart failure. RAAS activation functions to maintain blood pressure and cardiac output; however, chronic activation of RAAS can have deleterious effects on the heart, kidneys and blood vessels (80). In addition to systemic RAAS, most organ systems such as that of the heart, blood vessels and kidneys, and even cancerous cells, have local RAAS. The RAAS has different functions, hormones and receptors depending on its locality (81); for example, increased expression of angiotensin II receptor type 1 (AT1R) in cancerous cells suggests strong cancer aggressiveness and a poor prognosis (82). The regulation of RAAS may also affect the tumor size, although the results are inconsistent: Specifically, the angiotensin II (AngII)/AT1R axis is hypothesized to enhance tumor growth, while the AngII/AT2R signal serves the opposite role (83,84). RAAS inhibitors such as ACEIs or angiotensin II receptor blockers (ARBs) represent the cornerstone of heart failure treatment (85).

Oxidative stress

The ROS family is the key element for oxidative stress in eukaryotic cells. The heart inputs and outputs a consistent amount of energy and mainly relies on oxidative phosphorylation of mitochondria. ROS serve an important role in heart failure and cancer (86,87); however, oxidative phosphorylation of mitochondria also serves an important role in cancer development (88). Studies have found that dietary fiber supplementation has positive effects on heart oxidative stress responses (89,90). In addition, glycolysis increases the probability of heart failure and glucose oxidation leads to lactic acid production. Also, in response to rapid cancer growth, pyruvate dehydrogenase (PDH) and PDH kinase (PDK) play a major role in mitochondrial oxidative metabolism, which leads to increased glycolysis. PDH inhibits glucose oxidation and converts pyruvate to acetyl-CoA (91). PDK can phosphorylate and inhibit PDH. During heart failure, PDK is upregulated, but PDH is inhibited (91). Similar mechanisms for PDK upregulation and PDH inhibition are also present in cancerous cells (92). Dichloro-acetate, a PDK inhibitor, enhances PDH activity during heart failure, decreases ischemic damage and improves cardiac function; these changes consequently decrease the incidence/development of cancer (92).

Inflammation

Inflammation is closely related to heart failure. Heart failure increases inflammatory factor secretion, which supports the premise that inflammation leads to heart failure (93). Increased secretion of inflammatory factors during heart failure can cause bone marrow dysfunction. However, there is no direct evidence that proinflammatory cytokines released by cardiac cells affect cancer cells. Furthermore, Meijers et al (27) found that certain inflammatory factors, such as high-sensitivity C-reactive protein and central adrenomedullin precursors, are predictors or warning signs of cancers. The IL-1 inhibitor canakinumab decreased major cardiovascular events by 25% [hazard ratio (HR), 0.75; 95% confidence interval (CI), 0.66-0.85] in patients with myocardial infarction (94). Canakinumab also significantly decreased the incidence and mortality rate of lung cancer [highest dose (300 mg): HR, 0.33; 95% CI, 0.18-0.59; P<0.0001; and HR, 0.23; 95% CI, 0.10-0.54; P=0.0002, respectively] (95).

Immune system

Immune system dysfunction is closely related to the occurrence and development of cancer and heart failure (96,97). In the early stage of body damage, a large number of immune cells are beneficial; such cells are able to decrease and repair the area damaged by injury, but chronic immune activation will generate severe side effects in the body, such as severe or even fatal allergic reactions (98). A complete overview of immune system dysfunction and heart failure has recently been published by the Working Group on Myocardial Function of the European Society of Cardiology (99). It is important to note that the pathogenesis of heart failure is particular. As well as the differing pathophysiological mechanisms of heart disease, the immune activation methods of heart disease also vary. For example, during the first stage of myocardial infarction (a few hours), neutrophils invade the heart and start the inflammatory response immediately; furthermore, the infiltration of macrophages breaks down necrotic tissue and promotes scar formation. In the next stage of remodeling, the inflammatory response is weakened; the secreted cytokines will regulate the invasion of inflammatory cells after myocardial infarction (100-102). Heart failure with a normal ejection fraction is mostly due to obesity, hypertension, diabetes and metabolic syndrome (103). Recent studies have revealed that the immune system may also play a certain role in heart failure with a normal ejection system, in this case, cardiac hypertrophy and fibrosis often occur. In heart failure with ejection fraction retention, macrophages are involved in the process of cardiomyocyte apoptosis and cardiomyocyte fibrosis, but a decrease in macrophages can reduce myocardial hypertrophy. Immune system dysfunction is related to the development of cancer, as cancers can spread to different organs by weakening the immune system (104).

Cardiovascular drugs may cause cancer

To date, the impact of cardiovascular drugs on cancer is still unclear. A number ofmeta-analyses on all types of antihypertensive drugs showed that the use of ARB, ACEIs, β-receptor blockers, diuretics and calcium channel blockers has relatively increased the incidence of cancer and the risk associated with cancer death by 5.0-10.0% (105,106). However, some meta-analyses have confirmed that antihypertensive drugs are not associated with carcinogenesis and development (107,108). Studies on patients with type 2 diabetes showed a negative correlation between losartan and cancer risk; however, overall, candesartan and telmisartan resulted in an increased rate of cancer incidence (109). Another drug that affects cancer is aspirin. A study has found that the use of a low dose of aspirin results in the acceleration of the progression of cancer in older individuals (≥70 years old), potentially because aspirin inhibits antitumor inflammatory or immune responses, which regulate later stage growth and metastasis (110).

Gene-related hypothesis

Myocardial fibrosis leads to cardiac remodeling, promoting heart failure. It has been indicated that the delta like non-canonical notch ligand 1 (DLK1) gene is a key factor during the differentiation of fibroblasts into myoblasts (111). The knockdown of the DLK1 gene leads to the downregulation of microRNA-370 (miR-370), activates the TGF-β/Smad3 pathway and promotes myocardial cell fibrosis (112). Excessive deposition of extracellular matrix infiltrated by myofibroblasts can cause cardiac dysfunction (112). However, DLK1 is a type of imprinted gene that participates in the regulation of the differentiation of a variety of cells; its expression is increased in a number of cancer types, such as liver, pancreatic and colorectal cancer. Therefore, this gene plays an important role in carcinogenesis and cancer development (113). Sialyl-Lewis X (sLex) is the smallest recognition motif of the P-selectin ligand, which plays an important role in the adhesion and migration of cancerous cells. A study has found that miR-370 can specifically inhibit sLex expression and inhibit cell adhesion in colo-320 cells (114), justifying the fact that, inhibition of the DLK1 gene and downregulation of miR-370 lead to myocardial fibrosis/heart failure and cancer metastasis. Thus, some common targets might exist for heart failure and cancer. The metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) gene has previously been shown to be involved in the proliferation, metastasis and function of cancerous cells and the reproduction of endothelial and smooth muscle cells (115,116). Studies have found that MALAT1 is the key regulatory factor of mouse atherosclerosis, where its knockout can significantly increase coronary plaques and affected area (117). Also, a decrease in MALAT1 expression in patients with coronary plaques is associated with a poor prognosis (including heart failure, arrhythmia and sudden death) (118). At present, there is no study to determine which genes are directly related to heart failure and cancer, thus identification and characterization of genes involved in both cancer and heart failure is required to improve cancer therapeutic methods in the future.

4. Prevention strategies

Strategies for preventing heart failure in patients with cancer

From the perspective of pathogenesis, the main cause of heart failure in patients with cancer appears to be cardiac toxicity caused by cancer-related treatment. At present, the protective measures for such injury mainly include two schemes: The use of cardioprotective agents and standardized rehabilitation exercise (Fig. 2). The cardioprotective agents mainly include the use of traditional drugs, such as ACEI, to inhibit myocardial remodeling and topoisomerase inhibitors. In a recent meta-analysis, 15 patients within randomized controlled trials were selected to analyze the protective effect of myocardial remodeling drugs on preventing cardiac toxicity induced by cancer treatment. The study found that aldosterone antagonists, ACEIs, statins and β-blockers could substantially improve left ventricular systolic function, while ARBs displayed no cardioprotective effect and failed to improve the left ventricular systolic function (measured as LVEF) (119). However, another study proposed that ARBs are effective in the prevention of heart failure. The study found that patients administered acetyl-based chemotherapy had a moderate yet significant benefit in terms of LVEF following use of β-blockers or ACEIs/ARBs. The β-blocker analysis included 769 patients with cancer, and the ACEI/ARB analysis included a total of 581 patients with cancer. The mean LVEF difference between ACEIs and ARBs groups was 4.71% (120). Topoisomerase is a new target to prevent cancer treatment-related cardiotoxicity, and dexamethasone and other topoisomerase inhibitors inhibit topoisomerase II (121). It has been reported that dapagliflozin protects against doxorubicin-induced cardiotoxicity in patients with breast cancer and diabetes. Moreover, dapagliflozin inhibits doxorubicin-induced myocardial fibrosis and greatly improves cardiac function by inhibiting the apoptosis of cardiomyocytes and the generation of ROS (122). Therefore, topoisomerase inhibitors and dapagliflozin can protect the heart from the toxicity of chemotherapy drugs by inhibiting myocardial remodeling.

Exercise therapy is a new treatment for cancer-related heart failure. Cardiorespiratory fitness (CRF) is closely related to the prognosis of patients with heart failure. CRF decreases with age, and short-term (12- to 26-week) anticancer therapy can reduce CRF by 26% (123). The maximum oxygen consumption rate represents the extent of CRF, which can be improved by exercise therapy in patients with cancer-related heart failure. MacVicar et al (124) formulated an intermittent aerobic exercise prescription for 45 patients with breast cancer who received different chemotherapy regimens. This treatment recommended exercise three times a week at 60-80% of the normal maximum heart rate, for 10 weeks. The VO2 peak average of patients receiving this exercise prescription was increased by 40% compared with that of the non-exercise group. In another randomized controlled study, 20 patients with advanced breast cancer were randomly divided into two groups: The chemotherapy group and the chemotherapy + aerobic exercise group. After 12 weeks, the VO2 peak of the chemotherapy group decreased by 9%, while the VO2 peak of the chemotherapy + exercise group increased by 13% (125).

In addition to findings in breast cancer studies, another study found that exercise therapy was also effective for prostate cancer and Hodgkin's lymphoma, among others (126). Non-linear aerobic exercise could maintain the VO2 peak in patients with prostate cancer, while it could increase the VO2 peak in patients with Hodgkin's lymphoma from 5 to 17% (126). However, the impact of exercise therapy on the prognosis of cancer-related heart failure patients is controversial. In the follow-up period of 35 months, one study revealed that the all-cause mortality and readmission rate increased in the exercise group compared with that in the non-exercise group, and the VO2 peak showed no significant difference between the two groups (127). However, this result needs to be further confirmed due to the lack of a long-term exercise therapy group as a control. Other studies have previously shown that exercise therapy can improve the VO2 peak and the short-term prognosis in patients with cancer-related heart failure (128,129). Therefore, non-linear aerobic exercise is the recommended exercise for patients with cancer-related heart failure. It was advised that the patients keep non-linear aerobic exercise three times a week for 10-12 weeks and the exercise intensity was 60-85% of the normal maximum heart rate (128,129).

Strategies for the prevention of cancer in patients with heart failure

There are currently no drugs or treatments that can prevent cancer in patients with heart failure. As the mechanism by which patients with heart failure are more likely to develop cancer is known, the inhibition of excessive inflammation during myocardial remodeling can be a good asset (Fig. 2). Both heart failure and cancer are aging-related diseases. Rochette et al (130) found that the anti-aging protein humanin (HN), which is a 24-amino acid, endogenous, mitochondrial-derived peptide, can inhibit myocardial remodeling and inflammation. Studies have found that HN can protect cardiomyocytes through anti-oxidative stress (131,132); furthermore, Qin et al (133) demonstrated that the exogenous injection of HN analogs could inhibit age-related myocardial fibrosis, while HN was able to inhibit cancer metastasis. In fact, it has been revealed that HN was able to inhibit the lung metastasis of mouse melanoma cancer cells (134). However, whether HN can prevent cancer in patients with chronic heart failure is currently debated. Large-scale clinical randomized controlled trials and animal studies are needed to prove its effectiveness in the future.

5. Conclusion

With the increase in anticancer drugs discoveries and prescriptions, the incidence of cancer-related heart disease has recently increased. Heart failure affects the development of cancer through a variety of mechanisms. Therefore, cancer and heart failure are related and interact with each other, by sharing some usual risks, such as hypertension, diabetes mellitus and obesity, and even pathogenic genes. The pathophysiological mechanism of heart failure and cancer remains to be explored in depth. Currently, preventive strategies are limited to heart failure in patients with cancer. Further clinical trials are required to determine how to prevent patients with heart failure from suffering from cancer.

Acknowledgements

Not applicable.

Funding

No funding was received.

Availability of data and materials

Data sharing is not applicable to this article, as no datasets were generated or analyzed during the current study.

Authors' contributions

YZ conceived the study. HC and YZ wrote the manuscript. HM, PC, HC and YZ revised and edited the manuscript. Data authentication is not applicable. All authors have read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Christ M, Störk S, Dörr M, Heppner HJ, Müller C, Wachter R and Riemer U: Trend HF Germany Project. Heart failure epidemiology 2000-2013: Insights from the German federal health monitoring system. Eur J Heart Fail. 18:1009–1018. 2016.PubMed/NCBI View Article : Google Scholar

2 

DeSantis CE, Siegel RL, Sauer AG, Miller KD, Fedewa SA, Alcaraz KI and Jemal A: Cancer statistics for African Americans, 2016: Progress and opportunities in reducing racial disparities. CA Cancer J Clin. 66:290–308. 2016.PubMed/NCBI View Article : Google Scholar

3 

Armenian SH, Xu L, Ky B, Sun C, Farol LT, Pal SK, Douglas PS, Bhatia S and Chao C: Cardiovascular disease among survivors of adult-onset cancer: A community-based retrospective cohort study. J Clin Oncol. 34:1122–1130. 2016.PubMed/NCBI View Article : Google Scholar

4 

Choi HM, Park MS and Youn JC: Update on heart failure management and future directions. Korean J Intern Med. 34:11–43. 2019.PubMed/NCBI View Article : Google Scholar

5 

Swain SM, Whaley FS and Ewer MS: Congestive heart failure in patients treated with doxorubicin: A retrospective analysis of three trials. Cancer. 97:2869–2879. 2003.PubMed/NCBI View Article : Google Scholar

6 

Saiki H, Petersen IA, Scott CG, Bailey KR, Dunlay SM, Finley RR, Ruddy KJ, Yan E and Redfield MM: Risk of heart failure with preserved ejection fraction in older women after contemporary radiotherapy for breast cancer. Circulation. 135:1388–1396. 2017.PubMed/NCBI View Article : Google Scholar

7 

Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, Eiermann W, Wolter J, Pegram M, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 344:783–792. 2001.PubMed/NCBI View Article : Google Scholar

8 

Quarta CC, Solomon SD, Uraizee I, Kruger J, Longhi S, Ferlito M, Gagliardi C, Milandri A, Rapezzi C and Falk RH: Left ventricular structure and function in transthyretin-related versus light-chain cardiac amyloidosis. Circulation. 129:1840–1849. 2014.PubMed/NCBI View Article : Google Scholar

9 

van Nimwegen FA, Schaapveld M, Janus CP, Krol AD, Petersen EJ, Raemaekers JM, Kok WE, Aleman BM and van Leeuwen FE: Cardiovascular disease after Hodgkin lymphoma treatment: 40-year disease risk. JAMA Intern Med. 175:1007–1017. 2015.PubMed/NCBI View Article : Google Scholar

10 

Laufer-Perl M, Mor L, Milwidsky A, Derakhshesh M, Amrami N, Moshkovits Y, Arnold J, Topilsky Y, Arbel Y and Rozenbaum Z: Cancer therapeutics-related cardiac dysfunction among patients with active breast cancer: A cardio-oncology registry. Isr Med Assoc J. 22:564–568. 2020.PubMed/NCBI

11 

Tian Z, Yang Y, Yang Y, Zhang F, Li P, Wang J, Yang J, Zhang P, Yao W and Wang X: High cumulative doxorubicin dose for advanced soft tissue sarcoma. BMC Cancer. 20(1139)2020.PubMed/NCBI View Article : Google Scholar

12 

Degens J, De Ruysscher D, Houben R, Kietselaer B, Bootsma G, Hendriks L, Huijbers E, Schols A and Dingemans AC: Are patients with stage III non-small cell lung cancer treated with chemoradiotherapy at risk for cardiac events? Results from a retrospective cohort study. BMJ Open. 10(e036492)2020.PubMed/NCBI View Article : Google Scholar

13 

Kjekshus J, Apetrei E, Barrios V, Böhm M, Cleland JG, Cornel JH, Dunselman P, Fonseca C, Goudev A, Grande P, et al: Rosuvastatin in older patients with systolic heart failure. N Engl J Med. 357:2248–2261. 2007.PubMed/NCBI View Article : Google Scholar

14 

Linde C, Abraham WT, Gold MR, St John Sutton M, Ghio S and Daubert C: REVERSE (REsynchronization reVErses Remodeling in Systolic left vEntricular dysfunction) Study Group. Randomized trial of cardiac resynchronization in mildly symptomatic heart failure patients and in asymptomatic patients with left ventricular dysfunction and previous heart failure symptoms. J Am Coll Cardiol. 52:1834–1843. 2008.PubMed/NCBI View Article : Google Scholar

15 

Moss AJ, Hall WJ, Cannom DS, Klein H, Brown MW, Daubert JP, Estes NA III, Foster E, Greenberg H, Higgins SL, et al: Cardiac-resynchronization therapy for the prevention of heart-failure events. N Engl J Med. 361:1329–1338. 2009.PubMed/NCBI View Article : Google Scholar

16 

Ruschitzka F, Abraham WT, Singh JP, Bax JJ, Borer JS, Brugada J, Dickstein K, Ford I, Gorcsan J III, Gras D, et al: Cardiac-resynchronization therapy in heart failure with a narrow QRS complex. N Engl J Med. 369:1395–1405. 2013.PubMed/NCBI View Article : Google Scholar

17 

McMurray JJ, Packer M, Desai AS, Gong J, Lefkowitz MP, Rizkala AR, Rouleau JL, Shi VC, Solomon SD, Swedberg K, et al: Angiotensin-neprilysin inhibition versus enalapril in heart failure. N Engl J Med. 371:993–1004. 2014.PubMed/NCBI View Article : Google Scholar

18 

Doval HC, Nul DR, Grancelli HO, Perrone SV, Bortman GR and Curiel R: Randomised trial of low-dose amiodarone in severe congestive heart failure. Grupo de Estudio de la Sobrevida en la Insuficiencia Cardiaca en Argentina (GESICA). Lancet. 344:493–498. 1994.PubMed/NCBI View Article : Google Scholar

19 

Bigger JT Jr: Prophylactic use of implanted cardiac defibrillators in patients at high risk for ventricular arrhythmias after coronary-artery bypass graft surgery. Coronary artery bypass graft (CABG) patch trial investigators. N Engl J Med. 337:1569–1575. 1997.PubMed/NCBI View Article : Google Scholar

20 

Kadish A, Dyer A, Daubert JP, Quigg R, Estes NA, Anderson KP, Calkins H, Hoch D, Goldberger J, Shalaby A, et al: Prophylactic defibrillator implantation in patients with nonischemic dilated cardiomyopathy. N Engl J Med. 350:2151–2158. 2004.PubMed/NCBI View Article : Google Scholar

21 

Granger CB, McMurray JJ, Yusuf S, Held P, Michelson EL, Olofsson B, Ostergren J, Pfeffer MA and Swedberg K: CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function intolerant to angiotensin-converting-enzyme inhibitors: The CHARM-alternative trial. Lancet. 362:772–776. 2003.PubMed/NCBI View Article : Google Scholar

22 

McMurray JJ, Ostergren J, Swedberg K, Granger CB, Held P, Michelson EL, Olofsson B, Yusuf S and Pfeffer MA: CHARM Investigators and Committees. Effects of candesartan in patients with chronic heart failure and reduced left-ventricular systolic function taking angiotensin-converting-enzyme inhibitors: The CHARM-added trial. Lancet. 362:767–771. 2003.PubMed/NCBI View Article : Google Scholar

23 

Roy D, Talajic M, Nattel S, Wyse DG, Dorian P, Lee KL, Bourassa MG, Arnold JM, Buxton AE, Camm AJ, et al: Rhythm control versus rate control for atrial fibrillation and heart failure. N Engl J Med. 358:2667–2677. 2008.PubMed/NCBI View Article : Google Scholar

24 

Tavazzi L, Maggioni AP, Marchioli R, Barlera S, Franzosi MG, Latini R, Lucci D, Nicolosi GL, Porcu M and Tognoni G: Gissi-HF Investigators. Effect of n-3 polyunsaturated fatty acids in patients with chronic heart failure (the GISSI-HF trial): A randomised, double-blind, placebo-controlled trial. Lancet. 372:1223–1230. 2008.PubMed/NCBI View Article : Google Scholar

25 

Velazquez EJ, Lee KL, Deja MA, Jain A, Sopko G, Marchenko A, Ali IS, Pohost G, Gradinac S, Abraham WT, et al: Coronary-artery bypass surgery in patients with left ventricular dysfunction. N Engl J Med. 364:1607–1616. 2011.PubMed/NCBI View Article : Google Scholar

26 

Bertero E, Canepa M, Maack C and Ameri P: Linking heart failure to cancer: Background evidence and research perspectives. Circulation. 138:735–742. 2018.PubMed/NCBI View Article : Google Scholar

27 

Meijers WC, Maglione M, Bakker SJL, Oberhuber R, Kieneker LM, de Jong S, Haubner BJ, Nagengast WB, Lyon AR, van der Vegt B, et al: Heart failure stimulates tumor growth by circulating factors. Circulation. 138:678–691. 2018.PubMed/NCBI View Article : Google Scholar

28 

Bertero E, Ameri P and Maack C: Bidirectional relationship between cancer and heart failure: Old and new issues in cardio-oncology. Card Fail Rev. 5:106–111. 2019.PubMed/NCBI View Article : Google Scholar

29 

Ausoni S and Azzarello G: Development of cancer in patients with heart failure: How systemic inflammation can lay the groundwork. Front Cardiovasc Med. 7(598384)2020.PubMed/NCBI View Article : Google Scholar

30 

Tan C, Tasaka H, Yu KP, Murphy ML and Karnofsky DA: Daunomycin, an antitumor antibiotic, in the treatment of neoplastic disease. Clinical evaluation with special reference to childhood leukemia. Cancer. 20:333–353. 1967.PubMed/NCBI View Article : Google Scholar

31 

Vandecruys E, Mondelaers V, De Wolf D, Benoit Y and Suys B: Late cardiotoxicity after low dose of anthracycline therapy for acute lymphoblastic leukemia in childhood. J Cancer Surviv. 6:95–101. 2012.PubMed/NCBI View Article : Google Scholar

32 

Frederick CA, Williams LD, Ughetto G, van der Marel GA, van Boom JH, Rich A and Wang AH: Structural comparison of anticancer drug-DNA complexes: Adriamycin and daunomycin. Biochemistry. 29:2538–2549. 1990.PubMed/NCBI

33 

Martin E, Thougaard AV, Grauslund M, Jensen PB, Bjorkling F, Hasinoff BB, Tjørnelund J, Sehested M and Jensen LH: Evaluation of the topoisomerase II-inactive bisdioxopiperazine ICRF-161 as a protectant against doxorubicin-induced cardiomyopathy. Toxicology. 255:72–79. 2009.PubMed/NCBI View Article : Google Scholar

34 

Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF and Yeh ET: Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 18:1639–1642. 2012.PubMed/NCBI View Article : Google Scholar

35 

Jones RL: Utility of dexrazoxane for the reduction of anthracycline-induced cardiotoxicity. Expert Rev Cardiovasc Ther. 6:1311–1317. 2008.PubMed/NCBI View Article : Google Scholar

36 

Hasinoff BB, Patel D and Wu X: The role of topoisomerase IIβ in the mechanisms of action of the doxorubicin cardioprotective agent dexrazoxane. Cardiovasc Toxicol. 20:312–320. 2020.PubMed/NCBI View Article : Google Scholar

37 

Abd El-Aziz MA, Othman AI, Amer M and El-Missiry MA: Potential protective role of angiotensin-converting enzyme inhibitors captopril and enalapril against adriamycin-induced acute cardiac and hepatic toxicity in rats. J Appl Toxicol. 21:469–473. 2001.PubMed/NCBI View Article : Google Scholar

38 

Li M, Sala V, De Santis MC, Cimino J, Cappello P, Pianca N, Di Bona A, Margaria JP, Martini M, Lazzarini E, et al: Phosphoinositide 3-kinase gamma inhibition protects from anthracycline cardiotoxicity and reduces tumor growth. Circulation. 138:696–711. 2018.PubMed/NCBI View Article : Google Scholar

39 

Paris F, Fuks Z, Kang A, Capodieci P, Juan G, Ehleiter D, Haimovitz-Friedman A, Cordon-Cardo C and Kolesnick R: Endothelial apoptosis as the primary lesion initiating intestinal radiation damage in mice. Science. 293:293–297. 2001.PubMed/NCBI View Article : Google Scholar

40 

Orzan F, Brusca A, Conte MR, Presbitero P and Figliomeni MC: Severe coronary artery disease after radiation therapy of the chest and mediastinum: Clinical presentation and treatment. Br Heart J. 69:496–500. 1993.PubMed/NCBI View Article : Google Scholar

41 

Sag CM, Wolff HA, Neumann K, Opiela MK, Zhang J, Steuer F, Sowa T, Gupta S, Schirmer M, Hünlich M, et al: Ionizing radiation regulates cardiac Ca handling via increased ROS and activated CaMKII. Basic Res Cardiol. 108(385)2013.PubMed/NCBI View Article : Google Scholar

42 

De Keulenaer GW, Doggen K and Lemmens K: The vulnerability of the heart as a pluricellular paracrine organ: Lessons from unexpected triggers of heart failure in targeted ErbB2 anticancer therapy. Circ Res. 106:35–46. 2010.PubMed/NCBI View Article : Google Scholar

43 

Suter TM, Procter M, van Veldhuisen DJ, Muscholl M, Bergh J, Carlomagno C, Perren T, Passalacqua R, Bighin C, Klijn JG, et al: Trastuzumab-associated cardiac adverse effects in the herceptin adjuvant trial. J Clin Oncol. 25:3859–3865. 2007.PubMed/NCBI View Article : Google Scholar

44 

Bergers G and Benjamin LE: Tumorigenesis and the angiogenic switch. Nat Rev Cancer. 3:401–410. 2003.PubMed/NCBI View Article : Google Scholar

45 

Carlsson L, Ronquist G, Elisasson R, Dubois L, Ronquist KG and Larsson A: High concentrations of the angiogenic peptide VEGF-A in seminal fluid and its association to prostasomes. Clin Lab. 62:1515–1520. 2016.PubMed/NCBI View Article : Google Scholar

46 

Elice F, Jacoub J, Rickles FR, Falanga A and Rodeghiero F: Hemostatic complications of angiogenesis inhibitors in cancer patients. Am J Hematol. 83:862–270. 2008.PubMed/NCBI View Article : Google Scholar

47 

Cobleigh MA, Langmuir VK, Sledge GW, Miller KD, Haney L, Novotny WF, Reimann JD and Vassel A: A phase I/II dose-escalation trial of bevacizumab in previously treated metastatic breast cancer. Semin Oncol. 30 (5 Suppl 16):S117–S124. 2003.PubMed/NCBI View Article : Google Scholar

48 

Kappers MH, van Esch JH, Sluiter W, Sleijfer S, Danser AH and van den Meiracker AH: Hypertension induced by the tyrosine kinase inhibitor sunitinib is associated with increased circulating endothelin-1 levels. Hypertension. 56:675–681. 2010.PubMed/NCBI View Article : Google Scholar

49 

Li W, Croce K, Steensma DP, McDermott DF, Ben-Yehuda O and Moslehi J: Vascular and metabolic implications of novel targeted cancer therapies: Focus on kinase inhibitors. J Am Coll Cardiol. 66:1160–1178. 2015.PubMed/NCBI View Article : Google Scholar

50 

Neves KB, Rios FJ, Jones R, Evans T, Montezano AC and Touyz RM: Microparticles from vascular endothelial growth factor pathway inhibitor-treated cancer patients mediate endothelial cell injury. Cardiovasc Res. 115:978–988. 2019.PubMed/NCBI View Article : Google Scholar

51 

Maurea N, Coppola C, Piscopo G, Galletta F, Riccio G, Esposito E, De Lorenzo C, De Laurentiis M, Spallarossa P and Mercuro G: Pathophysiology of cardiotoxicity from target therapy and angiogenesis inhibitors. J Cardiovasc Med (Hagerstown). 17 (Suppl 1):S19–S26. 2016.PubMed/NCBI View Article : Google Scholar

52 

Schmidinger M, Zielinski CC, Vogl UM, Bojic A, Bojic M, Schukro C, Ruhsam M, Hejna M and Schmidinger H: Cardiac toxicity of sunitinib and sorafenib in patients with metastatic renal cell carcinoma. J Clin Oncol. 26:5204–5212. 2008.PubMed/NCBI View Article : Google Scholar

53 

Richardson PG, Sonneveld P, Schuster MW, Irwin D, Stadtmauer EA, Facon T, Harousseau JL, Ben-Yehuda D, Lonial S, Goldschmidt H, et al: Bortezomib or high-dose dexamethasone for relapsed multiple myeloma. N Engl J Med. 352:2487–2498. 2005.PubMed/NCBI View Article : Google Scholar

54 

Waxman AJ, Clasen S, Hwang WT, Garfall A, Vogl DT, Carver J, O'Quinn R, Cohen AD, Stadtmauer EA, Ky B and Weiss BM: Carfilzomib-associated cardiovascular adverse events: A systematic review and meta-analysis. JAMA Oncol. 4(e174519)2018.PubMed/NCBI View Article : Google Scholar

55 

Herrmann J, Wohlert C, Saguner AM, Flores A, Nesbitt LL, Chade A, Lerman LO and Lerman A: Primary proteasome inhibition results in cardiac dysfunction. Eur J Heart Fail. 15:614–623. 2013.PubMed/NCBI View Article : Google Scholar

56 

Vandenberg JI, Perry MD, Perrin MJ, Mann SA, Ke Y and Hill AP: hERG K(+) channels: Structure, function, and clinical significance. Physiol Rev. 92:1393–1478. 2012.PubMed/NCBI View Article : Google Scholar

57 

Pier DM, Shehatou GS, Giblett S, Pullar CE, Trezise DJ, Pritchard CA, Challiss RA and Mitcheson JS: Long-term channel block is required to inhibit cellular transformation by human ether-à-go-go-related gene (hERG1) potassium channels. Mol Pharmacol. 86:211–221. 2014.PubMed/NCBI View Article : Google Scholar

58 

Raschi E, Vasina V, Poluzzi E and De Ponti F: The hERG K+ channel: Target and antitarget strategies in drug development. Pharmacol Res. 57:181–195. 2008.PubMed/NCBI View Article : Google Scholar

59 

Stansfeld PJ, Sutcliffe MJ and Mitcheson JS: Molecular mechanisms for drug interactions with hERG that cause long QT syndrome. Expert Opin Drug Metab Toxicol. 2:81–94. 2006.PubMed/NCBI View Article : Google Scholar

60 

He S, Moutaoufik MT, Islam S, Persad A, Wu A, Aly KA, Fonge H, Babu M and Cayabyab FS: HERG channel and cancer: A mechanistic review of carcinogenic processes and therapeutic potential. Biochim Biophys Acta Rev Cancer. 1873(188355)2020.PubMed/NCBI View Article : Google Scholar

61 

Falk RH, Alexander KM, Liao R and Dorbala S: AL (Light-Chain) cardiac amyloidosis: A review of diagnosis and therapy. J Am Coll Cardiol. 68:1323–1341. 2016.PubMed/NCBI View Article : Google Scholar

62 

Gertz MA, Benson MD, Dyck PJ, Grogan M, Coelho T, Cruz M, Berk JL, Plante-Bordeneuve V, Schmidt HHJ and Merlini G: Diagnosis, prognosis, and therapy of transthyretin amyloidosis. J Am Coll Cardiol. 66:2451–2466. 2015.PubMed/NCBI View Article : Google Scholar

63 

Brenner DA, Jain M, Pimentel DR, Wang B, Connors LH, Skinner M, Apstein CS and Liao R: Human amyloidogenic light chains directly impair cardiomyocyte function through an increase in cellular oxidant stress. Circ Res. 94:1008–1010. 2004.PubMed/NCBI View Article : Google Scholar

64 

Sawyer DB, Siwik DA, Xiao L, Pimentel DR, Singh K and Colucci WS: Role of oxidative stress in myocardial hypertrophy and failure. J Mol Cell Cardiol. 34:379–388. 2002.PubMed/NCBI View Article : Google Scholar

65 

Lundin L, Norheim I, Landelius J, Oberg K and Theodorsson-Norheim E: Carcinoid heart disease: Relationship of circulating vasoactive substances to ultrasound-detectable cardiac abnormalities. Circulation. 77:264–269. 1988.PubMed/NCBI View Article : Google Scholar

66 

Modlin IM and Sandor A: An analysis of 8305 cases of carcinoid tumors. Cancer. 79:813–829. 1997.PubMed/NCBI View Article : Google Scholar

67 

Hassan SA, Banchs J, Iliescu C, Dasari A, Lopez-Mattei J and Yusuf SW: Carcinoid heart disease. Heart. 103:1488–1495. 2017.PubMed/NCBI View Article : Google Scholar

68 

Yeh ET, Tong AT, Lenihan DJ, Yusuf SW, Swafford J, Champion C, Durand JB, Gibbs H, Zafarmand AA and Ewer MS: Cardiovascular complications of cancer therapy: Diagnosis, pathogenesis, and management. Circulation. 109:3122–3131. 2004.PubMed/NCBI View Article : Google Scholar

69 

McKenney AS and Levine RL: Isocitrate dehydrogenase mutations in leukemia. J Clin Invest. 123:3672–3677. 2013.PubMed/NCBI View Article : Google Scholar

70 

Karlstaedt A, Zhang X, Vitrac H, Harmancey R, Vasquez H, Wang JH, Goodell MA and Taegtmeyer H: Oncometabolite d-2-hydroxyglutarate impairs α-ketoglutarate dehydrogenase and contractile function in rodent heart. Proc Natl Acad Sci USA. 113:10436–10441. 2016.PubMed/NCBI View Article : Google Scholar

71 

Rinde LB, Småbrekke B, Hald EM, Brodin EE, Njølstad I, Mathiesen EB, Løchen ML, Wilsgaard T, Brækkan SK, Vik A and Hansen JB: Myocardial infarction and future risk of cancer in the general population-the Tromsø study. Eur J Epidemiol. 32:193–201. 2017.PubMed/NCBI View Article : Google Scholar

72 

Berton G, Cordiano R, Cavuto F, Bagato F, Segafredo B and Pasquinucci M: Neoplastic disease after acute coronary syndrome: Incidence, duration, and features: The ABC-4* study on heart disease. J Cardiovasc Med (Hagerstown). 19:546–553. 2018.PubMed/NCBI View Article : Google Scholar

73 

de Boer RA, Meijers WC, van der Meer P and van Veldhuisen DJ: Cancer and heart disease: Associations and relations. Eur J Heart Fail. 21:1515–1525. 2019.PubMed/NCBI View Article : Google Scholar

74 

Arab S, Gramolini AO, Ping P, Kislinger T, Stanley B, van Eyk J, Ouzounian M, MacLennan DH, Emili A and Liu PP: Cardiovascular proteomics: Tools to develop novel biomarkers and potential applications. J Am Coll Cardiol. 48:1733–1741. 2006.PubMed/NCBI View Article : Google Scholar

75 

Isaac DL: Biomarkers in heart failure management. Curr Opin Cardiol. 23:127–133. 2008.PubMed/NCBI View Article : Google Scholar

76 

Dewey CM, Spitler KM, Ponce JM, Hall DD and Grueter CE: Cardiac-secreted factors as peripheral metabolic regulators and potential disease biomarkers. J Am Heart Assoc. 5(e003101)2016.PubMed/NCBI View Article : Google Scholar

77 

Pavo N, Raderer M, Hülsmann M, Scheithauer W, Adlbrecht C, Strunk G, Goliasch G, Gisslinger H, Steger GG, Hejna M, et al: Cardiovascular biomarkers in patients with cancer and their association with all-cause mortality. Heart. 101:1874–1880. 2015.PubMed/NCBI View Article : Google Scholar

78 

Tuñón J, Higueras J, Tarín N, Cristóbal C, Lorenzo Ó, Blanco-Colio L, Martín-Ventura JL, Huelmos A, Alonso J, Aceña Á, et al: N-terminal pro-brain natriuretic peptide is associated with a future diagnosis of cancer in patients with coronary artery disease. PLoS One. 10(e0126741)2015.PubMed/NCBI View Article : Google Scholar

79 

de Boer RA, Daniels LB, Maisel AS and Januzzi JL Jr: State of the Art: Newer biomarkers in heart failure. Eur J Heart Fail. 17:559–569. 2015.PubMed/NCBI View Article : Google Scholar

80 

Hartupee J and Mann DL: Neurohormonal activation in heart failure with reduced ejection fraction. Nat Rev Cardiol. 14:30–38. 2017.PubMed/NCBI View Article : Google Scholar

81 

George AJ, Thomas WG and Hannan RD: The renin-angiotensin system and cancer: Old dog, new tricks. Nat Rev Cancer. 10:745–759. 2010.PubMed/NCBI View Article : Google Scholar

82 

Ino K, Shibata K, Kajiyama H, Yamamoto E, Nagasaka T, Nawa A, Nomura S and Kikkawa F: Angiotensin II type 1 receptor expression in ovarian cancer and its correlation with tumour angiogenesis and patient survival. Br J Cancer. 94:552–560. 2006.PubMed/NCBI View Article : Google Scholar

83 

Catarata MJ, Ribeiro R, Oliveira MJ, Robalo Cordeiro C and Medeiros R: Renin-angiotensin system in lung tumor and microenvironment interactions. Cancers (Basel). 12(1457)2020.PubMed/NCBI View Article : Google Scholar

84 

Lever AF, Hole DJ, Gillis CR, McCallum IR, McInnes GT, MacKinnon PL, Meredith PA, Murray LS, Reid JL and Robertson JW: Do inhibitors of angiotensin-I-converting enzyme protect against risk of cancer? Lancet. 352:179–184. 1998.PubMed/NCBI View Article : Google Scholar

85 

Werner CM and Böhm M: The therapeutic role of RAS blockade in chronic heart failure. Ther Adv Cardiovasc Dis. 2:167–177. 2008.PubMed/NCBI View Article : Google Scholar

86 

Reuter S, Gupta SC, Chaturvedi MM and Aggarwal BB: Oxidative stress, inflammation, and cancer: How are they linked? Free Radic Biol Med. 49:1603–1616. 2010.PubMed/NCBI View Article : Google Scholar

87 

Takimoto E and Kass DA: Role of oxidative stress in cardiac hypertrophy and remodeling. Hypertension. 49:241–248. 2007.PubMed/NCBI View Article : Google Scholar

88 

Maiuri MC and Kroemer G: Essential role for oxidative phosphorylation in cancer progression. Cell Metab. 21:11–12. 2015.PubMed/NCBI View Article : Google Scholar

89 

Diniz YS, Cicogna AC, Padovani CR, Silva MD, Faine LA, Galhardi CM, Rodrigues HG and Novelli EL: Dietary restriction and fibre supplementation: Oxidative stress and metabolic shifting for cardiac health. Can J Physiol Pharmacol. 81:1042–1048. 2003.PubMed/NCBI View Article : Google Scholar

90 

Aune D, Chan DS, Greenwood DC, Vieira AR, Rosenblatt DA, Vieira R and Norat T: Dietary fiber and breast cancer risk: A systematic review and meta-analysis of prospective studies. Ann Oncol. 23:1394–1402. 2012.PubMed/NCBI View Article : Google Scholar

91 

Lopaschuk GD, Ussher JR, Folmes CD, Jaswal JS and Stanley WC: Myocardial fatty acid metabolism in health and disease. Physiol Rev. 90:207–258. 2010.PubMed/NCBI View Article : Google Scholar

92 

Bonnet S, Archer SL, Allalunis-Turner J, Haromy A, Beaulieu C, Thompson R, Lee CT, Lopaschuk GD, Puttagunta L, Bonnet S, et al: A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell. 11:37–51. 2007.PubMed/NCBI View Article : Google Scholar

93 

Mann DL: Innate immunity and the failing heart: The cytokine hypothesis revisited. Circ Res. 116:1254–1268. 2015.PubMed/NCBI View Article : Google Scholar

94 

Ridker PM, MacFadyen JG, Everett BM, Libby P, Thuren T and Glynn RJ: CANTOS Trial Group. Relationship of C-reactive protein reduction to cardiovascular event reduction following treatment with canakinumab: A secondary analysis from the CANTOS randomised controlled trial. Lancet. 391:319–328. 2018.PubMed/NCBI View Article : Google Scholar

95 

Ridker PM, MacFadyen JG, Thuren T, Everett BM, Libby P and Glynn RJ: CANTOS Trial Group. Effect of interleukin-1β inhibition with canakinumab on incident lung cancer in patients with atherosclerosis: Exploratory results from a randomised, double-blind, placebo-controlled trial. Lancet. 390:1833–1842. 2017.PubMed/NCBI View Article : Google Scholar

96 

Swann JB and Smyth MJ: Immune surveillance of tumors. J Clin Invest. 117:1137–1146. 2007.PubMed/NCBI View Article : Google Scholar

97 

Zhang Y, Bauersachs J and Langer HF: Immune mechanisms in heart failure. Eur J Heart Fail. 19:1379–1389. 2017.PubMed/NCBI View Article : Google Scholar

98 

Epelman S, Liu PP and Mann DL: Role of innate and adaptive immune mechanisms in cardiac injury and repair. Nat Rev Immunol. 15:117–129. 2015.PubMed/NCBI View Article : Google Scholar

99 

Frantz S, Falcao-Pires I, Balligand JL, Bauersachs J, Brutsaert D, Ciccarelli M, Dawson D, de Windt LJ, Giacca M, Hamdani N, et al: The innate immune system in chronic cardiomyopathy: A European society of cardiology (ESC) scientific statement from the working group on myocardial function of the ESC. Eur J Heart Fail. 20:445–459. 2018.PubMed/NCBI View Article : Google Scholar

100 

Stakos DA, Kambas K, Konstantinidis T, Mitroulis I, Apostolidou E, Arelaki S, Tsironidou V, Giatromanolaki A, Skendros P, Konstantinides S and Ritis K: Expression of functional tissue factor by neutrophil extracellular traps in culprit artery of acute myocardial infarction. Eur Heart J. 36:1405–1414. 2015.PubMed/NCBI View Article : Google Scholar

101 

Frangogiannis NG, Smith CW and Entman ML: The inflammatory response in myocardial infarction. Cardiovasc Res. 53:31–47. 2002.PubMed/NCBI View Article : Google Scholar

102 

Sun K, Li YY and Jin J: A double-edged sword of immuno-microenvironment in cardiac homeostasis and injury repair. Signal Transduct Target Ther. 6(79)2021.PubMed/NCBI View Article : Google Scholar

103 

Oktay AA, Rich JD and Shah SJ: The emerging epidemic of heart failure with preserved ejection fraction. Curr Heart Fail Rep. 10:401–410. 2013.PubMed/NCBI View Article : Google Scholar

104 

Pandya PH, Murray ME, Pollok KE and Renbarger JL: The immune system in cancer pathogenesis: Potential therapeutic approaches. J Immunol Res. 2016(4273943)2016.PubMed/NCBI View Article : Google Scholar

105 

Wei J, Galaviz KI, Kowalski AJ, Magee MJ, Haw JS, Narayan KMV and Ali MK: Comparison of cardiovascular events among users of different classes of antihypertension medications: A systematic review and network meta-analysis. JAMA Netw Open. 3(e1921618)2020.PubMed/NCBI View Article : Google Scholar

106 

Sipahi I, Debanne SM, Rowland DY, Simon DI and Fang JC: Angiotensin-receptor blockade and risk of cancer: Meta-analysis of randomised controlled trials. Lancet Oncol. 11:627–636. 2010.PubMed/NCBI View Article : Google Scholar

107 

Bangalore S, Kumar S, Kjeldsen SE, Makani H, Grossman E, Wetterslev J, Gupta AK, Sever PS, Gluud C and Messerli FH: Antihypertensive drugs and risk of cancer: Network meta-analyses and trial sequential analyses of 324,168 participants from randomised trials. Lancet Oncol. 12:65–82. 2011.PubMed/NCBI View Article : Google Scholar

108 

ARB Trialists Collaboration. Effects of telmisartan, irbesartan, valsartan, candesartan, and losartan on cancers in 15 trials enrolling 138,769 individuals. J Hypertens. 29:623–635. 2011.PubMed/NCBI View Article : Google Scholar

109 

Chang CH, Lin JW, Wu LC and Lai MS: Angiotensin receptor blockade and risk of cancer in type 2 diabetes mellitus: A nationwide case-control study. J Clin Oncol. 29:3001–3007. 2011.PubMed/NCBI View Article : Google Scholar

110 

McNeil JJ, Gibbs P, Orchard SG, Lockery JE, Bernstein WB, Cao Y, Ford L, Haydon A, Kirpach B, Macrae F, et al: Effect of aspirin on cancer incidence and mortality in older adults. J Natl Cancer Inst. 113:258–265. 2021.PubMed/NCBI View Article : Google Scholar

111 

Falix FA, Aronson DC, Lamers WH and Gaemers IC: Possible roles of DLK1 in the Notch pathway during development and disease. Biochim Biophys Acta. 1822:988–995. 2012.PubMed/NCBI View Article : Google Scholar

112 

Rodriguez P, Sassi Y, Troncone L, Benard L, Ishikawa K, Gordon RE, Lamas S, Laborda J, Hajjar RJ and Lebeche D: Deletion of delta-like 1 homologue accelerates fibroblast-myofibroblast differentiation and induces myocardial fibrosis. Eur Heart J. 40:967–978. 2019.PubMed/NCBI View Article : Google Scholar

113 

Akiyama J, Okamoto R, Iwasaki M, Zheng X, Yui S, Tsuchiya K, Nakamura T and Watanabe M: Delta-like 1 expression promotes goblet cell differentiation in Notch-inactivated human colonic epithelial cells. Biochem Biophys Res Commun. 393:662–667. 2010.PubMed/NCBI View Article : Google Scholar

114 

Wei Y, Shao J, Wang Y, Shen H, Yu S, Zhang J and Yin L: Hsa-miR-370 inhibited P-selectin-induced cell adhesion in human colon adenocarcinoma cells. Mol Cell Biochem. 450:159–166. 2019.PubMed/NCBI View Article : Google Scholar

115 

Wu XS, Wang XA, Wu WG, Hu YP, Li ML, Ding Q, Weng H, Shu YJ, Liu TY, Jiang L, et al: MALAT1 promotes the proliferation and metastasis of gallbladder cancer cells by activating the ERK/MAPK pathway. Cancer Biol Ther. 15:806–814. 2014.PubMed/NCBI View Article : Google Scholar

116 

Liu JY, Yao J, Li XM, Song YC, Wang XQ, Li YJ, Yan B and Jiang Q: Pathogenic role of lncRNA-MALAT1 in endothelial cell dysfunction in diabetes mellitus. Cell Death Dis. 5(e1506)2014.PubMed/NCBI View Article : Google Scholar

117 

Gast M, Rauch BH, Nakagawa S, Haghikia A, Jasina A, Haas J, Nath N, Jensen L, Stroux A, Böhm A, et al: Immune system-mediated atherosclerosis caused by deficiency of long non-coding RNA MALAT1 in ApoE-/-mice. Cardiovasc Res. 115:302–314. 2019.PubMed/NCBI View Article : Google Scholar

118 

Cremer S, Michalik KM, Fischer A, Pfisterer L, Jaé N, Winter C, Boon RA, Muhly-Reinholz M, John D, Uchida S, et al: Hematopoietic deficiency of the long noncoding RNA MALAT1 promotes atherosclerosis and plaque inflammation. Circulation. 139:1320–1334. 2019.PubMed/NCBI View Article : Google Scholar

119 

Li X, Li Y, Zhang T, Xiong X, Liu N, Pang B, Ruan Y, Gao Y, Shang H and Xing Y: Role of cardioprotective agents on chemotherapy-induced heart failure: A systematic review and network meta-analysis of randomized controlled trials. Pharmacol Res. 151(104577)2020.PubMed/NCBI View Article : Google Scholar

120 

Totzeck M, Mincu RI, Heusch G and Rassaf T: Heart failure from cancer therapy: Can we prevent it? ESC Heart Fail. 6:856–862. 2019.PubMed/NCBI View Article : Google Scholar

121 

Kollárová-Brázdová P, Jirkovská A, Karabanovich G, Pokorná Z, Bavlovič Piskáčková H, Jirkovský E, Kubeš J, Lenčová-Popelová O, Mazurová Y, Adamcová M, et al: Investigation of structure-activity relationships of dexrazoxane analogs reveals topoisomerase IIβ interaction as a prerequisite for effective protection against anthracycline cardiotoxicity. J Pharmacol Exp Ther. 373:402–415. 2020.PubMed/NCBI View Article : Google Scholar

122 

Chang WT, Lin YW, Ho CH, Chen ZC, Liu PY and Shih JY: Dapagliflozin suppresses ER stress and protects doxorubicin-induced cardiotoxicity in breast cancer patients. Arch Toxicol. 95:659–671. 2021.PubMed/NCBI View Article : Google Scholar

123 

Fitzgerald MD, Tanaka H, Tran ZV and Seals DR: Age-related declines in maximal aerobic capacity in regularly exercising vs sedentary women: A meta-analysis. J Appl Physiol (1985). 83:160–165. 1997.PubMed/NCBI View Article : Google Scholar

124 

MacVicar MG, Winningham ML and Nickel JL: Effects of aerobic interval training on cancer patients' functional capacity. Nurs Res. 38:348–351. 1989.PubMed/NCBI

125 

Jones LW, Fels DR, West M, Allen JD, Broadwater G, Barry WT, Wilke LG, Masko E, Douglas PS, Dash RC, et al: Modulation of circulating angiogenic factors and tumor biology by aerobic training in breast cancer patients receiving neoadjuvant chemotherapy. Cancer Prev Res (Phila). 6:925–937. 2013.PubMed/NCBI View Article : Google Scholar

126 

Courneya KS, Sellar CM, Stevinson C, McNeely ML, Peddle CJ, Friedenreich CM, Tankel K, Basi S, Chua N, Mazurek A and Reiman T: Randomized controlled trial of the effects of aerobic exercise on physical functioning and quality of life in lymphoma patients. J Clin Oncol. 27:4605–4612. 2009.PubMed/NCBI View Article : Google Scholar

127 

Jones LW, Douglas PS, Khouri MG, Mackey JR, Wojdyla D, Kraus WE, Whellan DJ and O'Connor CM: Safety and efficacy of aerobic training in patients with cancer who have heart failure: An analysis of the HF-ACTION randomized trial. J Clin Oncol. 32:2496–2502. 2014.PubMed/NCBI View Article : Google Scholar

128 

Hughes DC, Lenihan DJ, Harrison CA and Basen-Engquist KM: Exercise intervention for cancer survivors with heart failure: Two case reports. J Exerc Sci Fit. 9:65–73. 2011.PubMed/NCBI View Article : Google Scholar

129 

Maginador G, Lixandrão ME, Bortolozo HI, Vechin FC, Sarian LO, Derchain S, Telles GD, Zopf E, Ugrinowitsch C and Conceição MS: Aerobic exercise-induced changes in cardiorespiratory fitness in breast cancer patients receiving chemotherapy: A systematic review and meta-analysis. Cancers (Basel). 12(2240)2020.PubMed/NCBI View Article : Google Scholar

130 

Rochette L, Meloux A, Zeller M, Cottin Y and Vergely C: Role of humanin, a mitochondrial-derived peptide, in cardiovascular disorders. Arch Cardiovasc Dis. 113:564–571. 2020.PubMed/NCBI View Article : Google Scholar

131 

Cai H, Liu Y, Men H and Zheng Y: Protective mechanism of humanin against oxidative stress in aging-related cardiovascular diseases. Front Endocrinol (Lausanne). 12(683151)2021.PubMed/NCBI View Article : Google Scholar

132 

Thummasorn S, Shinlapawittayatorn K, Khamseekaew J, Jaiwongkam T, Chattipakorn SC and Chattipakorn N: Humanin directly protects cardiac mitochondria against dysfunction initiated by oxidative stress by decreasing complex I activity. Mitochondrion. 38:31–40. 2018.PubMed/NCBI View Article : Google Scholar

133 

Qin Q, Mehta H, Yen K, Navarrete G, Brandhorst S, Wan J, Delrio S, Zhang X, Lerman LO, Cohen P and Lerman A: Chronic treatment with the mitochondrial peptide humanin prevents age-related myocardial fibrosis in mice. Am J Physiol Heart Circ Physiol. 315:H1127–H1136. 2018.PubMed/NCBI View Article : Google Scholar

134 

Lue Y, Swerdloff R, Wan J, Xiao J, French S, Atienza V, Canela V, Bruhn KW, Stone B, Jia Y, et al: The potent humanin analogue (HNG) protects germ cells and leucocytes while enhancing chemotherapy-induced suppression of cancer metastases in male mice. Endocrinology. 156:4511–4521. 2015.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2021
Volume 22 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Cai H, Men H, Cao P and Zheng Y: Mechanism and prevention strategy of a bidirectional relationship between heart failure and cancer (Review). Exp Ther Med 22: 1463, 2021
APA
Cai, H., Men, H., Cao, P., & Zheng, Y. (2021). Mechanism and prevention strategy of a bidirectional relationship between heart failure and cancer (Review). Experimental and Therapeutic Medicine, 22, 1463. https://doi.org/10.3892/etm.2021.10898
MLA
Cai, H., Men, H., Cao, P., Zheng, Y."Mechanism and prevention strategy of a bidirectional relationship between heart failure and cancer (Review)". Experimental and Therapeutic Medicine 22.6 (2021): 1463.
Chicago
Cai, H., Men, H., Cao, P., Zheng, Y."Mechanism and prevention strategy of a bidirectional relationship between heart failure and cancer (Review)". Experimental and Therapeutic Medicine 22, no. 6 (2021): 1463. https://doi.org/10.3892/etm.2021.10898