Next Article in Journal
Validation of the Standardized Needs Evaluation Questionnaire in Polish Cancer Patients
Previous Article in Journal
Chromatin Profiles Are Prognostic of Clinical Response to Bortezomib-Containing Chemotherapy in Pediatric Acute Myeloid Leukemia: Results from the COG AAML1031 Trial
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cancer and Cardiovascular Disease: The Conjoined Twins

by
Mohammad Zmaili
1,
Jafar Alzubi
2,
Motasem Alkhayyat
3,
Almaza Albakri
4,
Feras Alkhalaileh
5,
Joshua Longinow
5 and
Rohit Moudgil
1,5,*
1
Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart, Vascular and Thoracic Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
2
Department of Medicine, Division of Cardiology, Einstein Medical Center, Philadelphia, PA 19141, USA
3
Department of Gastroenterology, Hepatology and Nutrition, Digestive Disease and Surgery Institute, Cleveland Clinic Foundation, Cleveland, OH 44195, USA
4
Jordanian Royal Medical Services, Department of Internal Medicine, King Abdullah II Ben Al-Hussein Street, Amman 11855, Jordan
5
Department of Internal Medicine, Cleveland Clinic, Cleveland, OH 44195, USA
*
Author to whom correspondence should be addressed.
Cancers 2024, 16(8), 1450; https://doi.org/10.3390/cancers16081450
Submission received: 15 January 2024 / Revised: 21 March 2024 / Accepted: 26 March 2024 / Published: 9 April 2024
(This article belongs to the Section Cancer Epidemiology and Prevention)

Abstract

:

Simple Summary

Cardiovascular disease and cancer are two major causes of morbidity and mortality worldwide. The significant overlap between cardiovascular medicine and oncology led to the emergence of the cardio-oncology field. Understanding the pathophysiological basis of the interconnected relationship between cardiovascular disease and cancer is fundamental to improving patient care and clinical outcomes. Several cardiovascular therapies have proven beneficial in the oncologic field, and thus, may need to be incorporated into the therapeutic armamentarium against cancer.

Abstract

Cancer and cardiovascular disease are the two most common causes of death worldwide. As the fields of cardiovascular medicine and oncology continue to expand, the area of overlap is becoming more prominent demanding dedicated attention and individualized patient care. We have come to realize that both fields are inextricably intertwined in several aspects, so much so that the mere presence of one, with its resultant downstream implications, has an impact on the other. Nonetheless, cardiovascular disease and cancer are generally approached independently. The focus that is granted to the predominant pathological entity (either cardiovascular disease or cancer), does not allow for optimal medical care for the other. As a result, ample opportunities for improvement in overall health care are being overlooked. Herein, we hope to shed light on the interconnected relationship between cardiovascular disease and cancer and uncover some of the unintentionally neglected intricacies of common cardiovascular therapeutics from an oncologic standpoint.

1. Introduction

1.1. Burden and Epidemiology of Cardiovascular Disease and Cancer

In spite of the remarkable improvement in cardiovascular outcomes over the last several years, cardiovascular disease (CVD) continues to be the number one cause of morbidity and mortality worldwide [1,2]. Additionally, cancer is also a leading cause of death globally, with a significant economic impact [3]. The most recent report from the Centers for Disease Control and Prevention (CDC) showed that in 2019, CVD and cancer were the leading causes of death in the United States, with 659,041 and 599,601 deaths, respectively [4]. The World Health Organization (WHO) estimates that 17.9 million people die every year from CVD and nearly 9–10 million die from cancer [5,6]. While the disease processes are an independent cause of increased mortality, recent evidence suggests that they are intertwined and therefore, mitigation of risk factors from one (such as CVD) can protect patients from the other (such as cancer) [7]. In this review, we highlight the significant points of intersection between CVD and cancer and discuss the major clinical implications in regard to prevention and management.

1.2. Significance of Co-Occurrence of Cardiovascular Disease and Cancer

CVDs including coronary artery disease, carotid artery disease, peripheral vascular disease, cerebrovascular disease, and heart failure co-exist in more than 40% of patients with lung cancer, 30% with hematologic malignancies, 35% with renal cancer, 25% with head and neck cancers, 25% with colon cancer, 15% with breast cancer [8]. As cancer survivorship continues to improve with the advent of new therapies, an increasing number of cancer survivors are being followed by general practitioners [9]. As will be discussed in this review, these patients have a higher risk of cardiovascular complications and diseases, apart from the cumulative risk and increased prevalence of cardiovascular disease with aging, which in turn affects their long-term prognosis, and may even encumber their candidacy for cancer therapies. The realization of this association calls for a multidisciplinary approach with the incorporation of cardiovascular healthcare into the management and follow-up of cancer patients [10]. The aim of the cardio-oncology field is to ensure comprehensive medical care for the cancer patient, with an emphasis on risk stratification, complication prevention, and cardiovascular health optimization, which may collectively dictate eligibility for and response to various cancer therapies, and thus, overall outcome and survival [11].
The occurrence of CVD in cancer patients, not only affects long-term survival, but may also increase the susceptibility to cardiotoxic effects of certain therapies necessitating suboptimal dosage administration, or even worse, premature treatment cessation [12,13]. This has important ramifications as an analysis published by Copeland-Halperin et al. in 2020 showed that holding trastuzumab for 6 weeks or longer -owing to cardiotoxicity- increases the risk of invasive breast cancer recurrence or death, with an adjusted hazard ratio (HR) of 1.56% (95% CI, 1.10–2.21), in patients with early-stage human epidermal growth factor receptor 2 (ERBB2)-positive breast cancer [14]. It is clear that CVDs and their risk factors are pivotal predictors of cardiotoxicity associated with cancer therapy. More recent studies have shown that CAD, hypertension, and diabetes are considered strong predictors of left ventricular dysfunction among patients receiving anthracycline chemotherapy, whereas CAD, obesity, and hypertension, increase the risk of left ventricular dysfunction in breast cancer patients receiving trastuzumab [15,16]. Similarly, pre-existing hypertension was shown to be the strongest predictor of resistant hypertension requiring interruption of cancer therapy in patients receiving anti-angiogenic targeted agents [17]. This transcends into newer therapies also as more evidence emerge that therapies such as immune checkpoint inhibitors can cause myocarditis, valvulitis, and increases incidence of atherosclerotic disease [18,19,20].

1.3. Shared Risk Factors and Pathophysiological Processes

Even though CVD and cancer may appear to be two distinct entities, there are multiple areas of intersection. The two conditions tend to overlap, not only in regard to risk factors, but also in some underlying pathophysiological processes (Figure 1). In a recent large retrospective cohort study evaluating the effect of presence of CVD on cancer incidence, it was found that CVD, particularly atherosclerotic disease, was associated with an increased occurrence of specific cancer subtypes compared with those without CVD [21]. Interestingly, the risk of malignancy in patients with CVD was noted to be cancer-specific, with higher risk of lung, bladder, liver, colon, and other hematologic cancers. However, those patients were also found to have a lower risk of other cancer subtypes like breast, ovarian, and uterine cancers. There is a growing level of evidence that suggests shared etiologic mechanisms, of which inflammation stands out as a pivotal contributor to the manifestation of both conditions [22]. Nonetheless, other notable factors may contribute to the relationship between both disease processes.
The role of inflammation in CVD pathogenesis is well-established, and many of the known CVD risk factors such as hypertension, smoking, dyslipidemia, obesity, and insulin resistance, can trigger atherosclerosis and lead to CVD events by inducing inflammation via various pathways [23]. This includes a heightened expression of pro-inflammatory cytokines, which leads to intensified oxidative stress due to increased production of reactive oxygen species and lipid peroxidation [23,24].
On the other hand, our understanding of the interaction between the immune system, inflammation, and cancer development is expanding, as many tumors have been shown to arise from sites of infection, chronic irritation, and inflammation [23,25,26]. Whether promoted by viral infections, smoking, or carcinogenic chemicals, inflammation can lead to cancer formation via complex pathways that include, whether directly or indirectly, enhanced cellular proliferation, dysregulated inflammatory response, amplified release of various cytokines, recruitment of inflammatory cells, or increased production of reactive oxygen species, which eventually can culminate in oxidative DNA damage, and disruption of DNA repair [23,27].
Other mechanisms that may explain the shared risk factors of cancer and CVD include hormonal-mediated disturbances seen obesity, diabetes mellitus, and physical inactivity [28]. In obesity, there is an increased expression of proinflammatory cytokines and hormones that are produced within adipose tissue, such as interleukin-6 (IL-6), tumor necrosis factor-alpha (TNFa), leptin, angiotensinogen, resistin, and C-reactive protein [29,30]. These molecules are believed to promote a steady state of low-grade inflammation and oxidative stress through formation of reaction oxygen species, which in turn may lead to DNA disruption, and eventually a higher risk of cancer development [31,32,33]. Additionally, leptin, which is secreted by adipose tissue, plays a major role in obesity-r,elated CVD [34]. IL-6 and TNFa can also induce hypertension and atherosclerosis, both of which play pivotal role in heart failure with preserved ejection fraction pathogenesis [35,36]. In this context, the CANTOS trial, which was randomized double-blinded study, investigated the effects of canakinumab, monoclonal antibody to proinflammatory cytokine IL-1β, in patients with prior myocardial infarction and elevated serum level of high high-sensitivity C-reactive protein (≥2 mg/dL) [37]. It was found that patients treated with canakinumab had a significantly lower rate of recurrent cardiovascular events compared to placebo group. But more interestingly, canakinumab was associated with a lower incidence of lung cancer, which further support the central role of inflammation in cancer and CVD pathogenesis [38].
Numerous studies have linked diabetes to cancer risk and its progression [39]. In diabetes, insulin resistance triggers atherosclerosis through oxidative stress, glycosylation, and high triglyceride levels, leading to endothelial damage in the vascular beds and thus atheroma formation [40,41]. Similarly, diabetes promotes a pro-inflammatory state that can mediate and increase the risk for cancer development and progression [29,42,43]. Additionally, sex hormone disturbances, which may result from hyperinsulinemia in the setting of disturbed glycemic homeostasis, are implicated in the carcinogenesis of some tumors such as breast and endometrial cancers [42,44,45,46]. Moreover, insulin-like growth factors, which are multifunctional peptides that regulate cell proliferation, differentiation, and apoptosis, are increased in the setting of impaired glycemic regulation and insulin resistance. This in turn can, not only enhance tumorigenesis and promote the development of some cancers, but also mediate smooth muscle proliferation in blood vessels with resultant atherosclerosis [39,47,48,49,50]. Correlation exists at the molecular level also as recent studies showed that potency of immune checkpoint inhibitors are greatly enhanced by PCSK9 inhibitors [51].
Furthermore, several studies have showed that metabolic reprogramming play a significant role in both heart failure and cancer molecular pathogenesis [52]. This metabolic shift at a cellular level occurs in tumors cells and failing cardiomyocytes in response to various stressors. Specifically, healthy myocardium mainly relies on beta oxidation of fatty acids for ATP synthesis [53]; however, metabolic source of energy in cardiomyocytes switches away from fatty acid utilization to glucose, ketone bodies, and amino acids (such as glutamine) under stress like pressure overload conditions [54,55]. This metabolic switch in failing cardiomyocytes leads to production of metabolites that are essential for biosynthesis as well as ventricular hypertrophy and contributes to calcium mishandling and ultimately cardiac dysfunction [42,56]. A similar shift in metabolic dependency is seen in cancerous cells to meet the catabolic and anabolic needs of growing tumors where the source of energy of proliferating cells shifts to glucose consumption “Warburg effect” along with preferential production of lactate, even in the presence of adequate tissue supply of oxygen, unlike healthy tissues that depends mainly on fatty acid oxidation for its energy expenditure [57,58]. This metabolic alteration in cancer cells is thought to support de novo synthesis of nucleotides, lipids, and proteins that are needed for tumor growth. [57] It is important to understand that the effects of these metabolic alteration in cancers and failing hearts extend beyond just altering source of energy but also have pleiotropic effects mediated by the byproducts of the metabolic changes and are also directly linked to organ dysfunction [59] In addition, certain oncometabolites produced by cancers, such D-2-hydroxyglutarate and succinate, have been directly implicated in development of cardiomyopathy in animal studies [60,61].

2. Cardiovascular Health in Cancer Patients

2.1. Role of Cardiovascular Disease Screening

The importance of primary prevention to reduce the overall burden of cardiovascular disease cannot be overemphasized. The first message of the 2019 American College of Cardiology/American Heart Association (ACC/AHA) Task Force on Clinical Practice Guidelines Report states that promotion of healthy lifestyle is the most important measure to reduce and prevent cardiovascular disease [62]. Additionally, the report lays out detailed recommendations to determine an individual’s risk for cardiovascular disease and to facilitate decisions in regards to preventive strategies and therapies. Several measures have been adopted to estimate such a risk, utilizing clinical tools and data such as the 10-year atherosclerotic cardiovascular disease (ASCVD) risk, the SCORE (Systematic COronary Risk Evaluation) project, among others [63,64,65,66,67,68]. The ultimate goal is to identify patients who are at a high risk of cardiovascular disease to a point where the benefit of a preventive intervention such as statin prescription surpasses the risks of possible adverse effects. As discussed, cancer patients represent a vulnerable population with an elevated risk of cardiovascular disease given the inherent properties of the risk factors that are shared among both conditions [28,69]. In various types of cancer, cardiovascular disease and mortality were shown to be greater in cancer survivors compared to age-matched controls [70,71,72,73]. Therefore, additional emphasis should be placed on risk assessment and reduction in this patient population. This may not only improve the overall cardiovascular health and outcome, but may also prove beneficial from an oncologic perspective. The development of cardiotoxicity has been reported to adversely affect outcomes of patients with cancer, and its prevention may be beneficial to overall patient survival [74,75,76]. Thus, it is fundamental to involve a cardiologist in the care of cancer patients, not only to detect early cardiovascular side effects of cancer therapy, but also to optimize the overall cardiovascular care of cancer patients from the initial cancer diagnosis to survivorship.

2.2. Cardiovascular Disease Screening in Cancer Patients. Where We Stand, and What Are the Implications?

While it has become more evident that cardiovascular disease and cancer share several risk factors, and therefore tend to commonly coexist, preventive measures have not converged as the two conditions are generally approached independently. The American Society of Clinical Oncology (ASCO) advocates for early implementation of routine cardiovascular surveillance in high-risk cancer patients, along with cardiovascular risk factor screening and modification [77]. Similarly, the American Heart Association (AHA) delineated a significant interrelation between cardiovascular disease and breast cancer, and provided a comprehensive review of the substantial areas of overlap [78]. The diagnosis of cancer often leads to mental stress, anxiety, and significantly impacts patients’ lifestyle. The influence of cancer detection affects medical providers as well. The focus granted to cancer management and prognosis often shifts the attention away from cardiovascular health promotion, and therefore, the opportunity for risk factor screening and modification is inadvertently missed [79]. The untoward sequelae of this cognitive distraction are expected to become more substantial as advancements in cancer detection and therapies have improved cancer survival rates [80]. This in turn is expected to increase the burden of cardiovascular disease, either as a complication from cancer therapy or from the cumulative effect of risk factors with increased longevity [78]. Previous studies have shown that cancer survivors may have poor overall control of their traditional cardiovascular risk factors [81]. There are also significant numbers of cancer patients with established CVD diagnosis who do not get referred to cardiologists, and are thus suboptimally managed [8]. Even if prompt action is undertaken to estimate the cardiovascular disease risk, the commonly used clinical assessment tools tend to underestimate the risk in patients with cancer, whether active or in remission [82]. As an example, a study of 561 breast cancer patients assessed the coronary artery calcium score derived from computed tomography (CT) scans that were performed for radiotherapy planning, and found that one third of patients who demonstrated a high coronary artery calcium score lacked other cardiovascular risk factors [83]. As such, these patients would not have been classified in the appropriate risk category group by clinical risk assessment tools. The implication of a heightened cardiovascular disease risk translates into higher rates of cardiovascular mortality in cancer patients compared to the general population [84]. An observational population-based study scrutinized the rates of cardiovascular mortality among 28 cancer types in cancer survivors from 1973 to 2012. Results showed that 11.3% of deaths were attributable to cardiovascular disease. Interestingly, the risk of cardiovascular mortality exceeded that of cancer mortality in eight types of cancer in at least one calendar year [84].

3. Cardiovascular Interventions in the Armamentarium of Cancer Therapy

Given the similarities and significant overlap between cardiovascular disease and cancer as discussed above, it is not surprising that cardiovascular interventions may provide a benefit and be useful in the oncologic field. Significant observations were noted in the past, in regards to a beneficial effect on the natural history of cancer, from cardiovascular medications. This favorable role encompasses a range of effects, from providing a protective effect against cancer development and progression, to augmenting the antitumor actions of cancer therapeutics. Below is a brief overview of commonly prescribed cardiovascular medications from an oncologic perspective, which is also summarized in Table 1.

3.1. Aspirin

Aspirin is an irreversible inhibitor of cyclooxygenase-1 (COX-1) enzyme that produces precursors for prostaglandins and thromboxanes [85]. This results in dose- and time-dependent inhibition of thromboxane A2 (TXA2) formation, an important mediator in platelet recruitment and aggregation [86].
Aspirin is generally considered the mainstay antiplatelet therapy for the treatment of acute coronary syndromes as well as the secondary prevention of atherothrombotic events in patients with various atherosclerotic diseases [87]. A metanalysis of sixteen secondary prevention trials showed that treatment with low-dose aspirin is effective in preventing approximately one-fifth of atherothrombotic vascular complications in patients with previous myocardial infarction, stroke, or transient cerebral ischemia [88]. Nonetheless, aspirin use for primary prevention of cardiovascular diseases is still controversial with uncertain balance between cardiovascular benefits and bleeding risk [89].
In the last few decades, compelling data have emerged suggesting an association between the regular use of COX inhibitors, including aspirin and other nonsteroidal anti-inflammatory drugs, and reduced risk of colorectal cancers [90]. The protective effects of aspirin against cancer are believed to be related to the prevention of early neoplastic transformation in addition to an anti-metastatic action [91]. It is evident that platelets play a pivotal role in neoplastic transformation via enhanced biosynthesis of prostaglandin E2 (PGE2), among several lipid mediators that are synthesized and released by activated platelets. PGE2 influences the adhesive, migratory, and invasive behavior of cells and create an environment that facilitates tumor formation and progression [90].
Hence, multiple randomized controlled trials (RCTs) were designed to study the chemo-preventive effects of COX inhibitors, given the data that have suggested an important role of COX enzymes, particularly COX-2, in gastrointestinal carcinogenesis [92,93]. Nonetheless, some of these trials that were designed to evaluate these chemo-preventive effects of selective COX-2 inhibitors, such as celecoxib and rofecoxib, showed an increased risk of major adverse cardiovascular events and were halted [94,95]. On contrast to selective COX-2 inhibitors, RCTs, observational case-control and meta-analysis studies have demonstrated a chemo-preventive effect of aspirin against colorectal cancers [96,97,98,99,100,101]. Other studies have also demonstrated that aspirin reduced the risk of death from several non-colonic solid cancers including esophageal, pancreatic, brain, lung, stomach, and prostate cancer in patients with Lynch syndrome [101,102]. However, it is unclear whether there is a chemoprotective benefits against malignancies other than colorectal cancer in patients without Lynch syndrome. Nonetheless, the evidence of use of aspirin and its mitigating effect on colorectal cancer morbidity and mortality has been identified [103,104,105].

3.2. Angiotensin Converting Enzyme Inhibitors (ACEi) and Angiotensin Receptor Blockers (ARB)

Angiotensin converting enzyme inhibitors (ACEi) and angiotensin receptor blockers (ARB) are widely used in the management of hypertension and heart failure [106,107]. In addition, they have renal protective effects in patients with diabetes [108]. It is evident that the renin–angiotensin–aldosterone system (RAAS), including angiotensin II and angiotensin II type 2 (AT2) receptor subtypes, regulates blood pressure homeostasis and electrolyte balance [109,110]. The functions of RAAS extend not only to the cardiovascular system but also involve multiple organ systems such as the kidney, brain, pituitary, adrenal, gonad and adipose [111,112]. Previous research and human studies have demonstrated RAAS signaling within various organs and tissues, indicating its essential role in several biological processes with involvement in different pathophysiological mechanisms, including inflammation [113,114,115]. Additionally, other studies have shown that angiotensin II functions as a paracrine and/or autocrine signal in some cancers and mediates recruitment of inflammatory cells. This leads to an enhanced secretion of cytokines that accelerate cell proliferation and tumor angiogenesis, such as up-regulating vascular endothelial growth factor (VEGF) expression [113,116,117]. Furthermore, the RAAS may modulate cancer growth and progression at different levels, including sustained angiogenesis, evasion of apoptosis, self-sufficiency in growth signals, insensitivity to anti-growth signals, tissue invasion and metastases, and limitless replicative potential [117]. In preclinical models, RAAS inhibitors have shown efficacy in reducing metastases, whereas AT1 R expression frequently correlates with the degree of cancer invasiveness. In two separate lung metastases models, the treatment of mice with candesartan (ARB) significantly reduced lung metastatic burden [118,119]. whereas captopril (ACEi) significantly reduced cancer size and was associated with a decreased lymph node metastases in a non-small-cell lung cancer xenograft model [120]. Wherein human retrospective studies, they have provided some evidence that long-term use of RAAS inhibitors might modulate cancer growth and progression [121,122,123]. One of the first studies that assessed the risk of cancer in hypertensive patients who received ACE inhibitors over a 15-year period, showed that the relative risk of incident and fatal cancer among 1559 patients receiving ACE inhibitors was significantly reduced in comparison with control subjects, most markedly for female-specific cancers [124]. In three other retrospective case–controlled studies, ACE inhibitor use was associated with reduction in the incidence of esophageal, pancreatic, and colon cancer [117,125,126]. Other studies showed that the use of RAAS antagonism has also been associated with a reduced risk of melanomas and a lower risk of developing prostate cancer [121,122]. These retrospective studies suggested that dysregulation of RAAS components plays a role in a broad range of human malignancies and may correlate with disease outcome. More importantly, these studies present a large body of evidence that RAAS inhibitors play an important prognostic indicator and novel molecular target for a wide range of cancers. Other retrospective studies have reported that losartan (ARB) use prior to chemotherapy potentiates the anticancer response by improving chemotherapy delivery to cancer cells [127,128,129]. A more recent large meta-analysis of nine studies (n = 1362) that evaluated the effects of beta blockers, ACEi, and ARBs on cardiotoxicity of trastuzumab and anthracycline reported the those therapies were associated with the preservation of LVEF [130]. Although the link between RAAS, cancer angiogenesis and invasion arguably represent a therapeutic opportunity for clinical intervention, few clinical trials have been initiated to investigate the efficacy of RAAS modulators in cancer. This may be attributed mainly to the complex nature of RAAS signaling, making the response to RAAS inhibitors, either individually or in combination with other drugs, difficult to predict [117]. On the other hand, some observational studies have shown that the use of some RAAS inhibitors was associated with an increased risk of lung cancer [131].

3.3. Beta Blockers (βB)

Beta (β)-adrenoceptors are broadly distributed in various tissues and regulate wide range of important physiological functions and disease states [132]. β-adrenergic receptor blockers have been widely used for the treatment of hypertension, ischemic heart disease, and congestive heart failure [133,134]. Several studies have shown that catecholamines can significantly enhance the ability of tumor cells to invade the surrounding extracellular matrix via beta adrenergic system, thereby enhancing tumor growth through activation of invasive ability and stimulating VEGF secretion [135,136,137]. It has been evident that the β-adrenergic system plays pivotal roles in cancer development and progression and is involved in almost every step of cancer development, including stimulation of continuous proliferation along with evasion of growth suppressors, resistance to apoptosis, enhancement of invasion and metastasis, and induction of angiogenesis [137]. A translational study showed that norepinephrine and the β-adrenergic agonist isoproterenol can enhance the production of a proangiogenic cytokine, vascular endothelial growth factor, by ovarian cancer cells [138]. Interestingly, these effects were completely blocked by propranolol (βB), suggesting that β-adrenergic receptors mediate production of proangiogenic factors and thereby facilitation tumor metastasis [138]. Hence, the potential benefits of βB to alleviate the deleterious progression of cancers influenced by β-adrenergic system have been further investigated. A number of studies have evaluated the effect of βB use on cancer, but they have had conflicting or inconsistent findings. First, a large retrospective case-control study of prostate cancer patients, which investigated the effect of different classes of antihypertensives on cancer (including βB, ACEi, calcium channel blockers, alpha-blockers), showed that only βB use was associated with a significant reduction in prostate cancer risk [139]. In another observational study that looked at outcomes of patients with cardiovascular disease on a 10-year-follow up, it was noted that βB use was associated with a significant reduction in cancer incidence [140]. Other studies did not show or support protective benefits for βB against cancers [141,142]. Several prospective clinical trials that are assessing propranolol use in patients with ovarian (NCT01504126, NCT01308944), colorectal cancer (NCT00888797), breast (NCT01847001, NCT00502684, NCT02596867), and melanoma (NCT01988831) are under investigation. The result of these trials would illuminate the potential chemoprotective effects of βB against some cancers.

3.4. Calcium Channel Blockers (CCB)

Calcium channel blockers (CCBs) are one of the first-line treatments for hypertension that function by blocking T-type calcium channels [143]. These channels regulate calcium homeostasis that controls various cellular processes, including those relevant to tumorigenesis, such as proliferation, apoptosis, gene transcription and angiogenesis [144]. Thus, it is evident that increased T-type calcium channel expression and function have an important role in the abnormal proliferation of cells in many types of cancers [145]. Interestingly, it has been suggested that the expression of T-type calcium channels in cancer cells may vary depending on the rate of cellular proliferation. [145,146]. Calcium has the ability to function both as a promoter of cellular proliferation, and as an inducer of cell death, depending on the amplitude of the increase in the intracellular calcium concentration, and the duration of this change. Consequently, both activators and inhibitors of calcium channels may have potential anti-cancerous effects [146]. The role of T-type calcium channels in cellular proliferation has been described in breast, brain, colorectal, gastric, ovarian, and prostate tumors as well as leukemia [146,147,148,149,150].

3.5. Statins (β-Hydroxy β-Methylglutaryl-CoA (HMG-CoA) Reductase Inhibitors)

Statins are considered a first-line treatment for hypercholesterolemia and a cornerstone therapy for atherosclerotic diseases [151]. Statins suppress cholesterol synthesis via inhibition of 3-hydroxy-3-methyl-glutarylcoenzyme A reductase, a pivotal enzyme in cholesterol synthesis [152]. Some cholesterol precursor molecules, which are also inhibited by statin therapy, are essential components of other critical cellular functions including cell membrane integrity [153]. Therefore, suppression of these molecules may interfere with cellular growth and development of cancers. In addition to their effects on cholesterol, multiple in-vivo and in-vitro studies have shown antiproliferative effects on various types of cancers [154,155,156,157,158,159]. Most data on the potential effects of statins on cancers are derived from observational studies. Multiple studies and metanalysis have described a decreased risk of prostate cancer in association with statins use [160,161,162,163]. Nevertheless, there were conflicting data on effect of statins on prostate cancer progression with some studies showing no effect of statins on progression-free survival after radiotherapy for prostate cancer and radical prostatectomy [164]. While other retrospective cohort studies showed a reduced biochemical recurrence in prostate cancer patients treated with radical prostatectomy [165,166]. Similarly, chemoprotective effects of statins on gastrointestinal cancers were also extensively studied. For example, a large meta-analysis has reported a protective association between statin use and gastric cancer risk among both Asian and Western population, in a dose-dependent manner [167]. Additionally, several studies that examined the association between statin use and colorectal cancer risk and survival have reported conflicting results. In one case-control study, statin use was associated with lower risk of colorectal cancer but no significant association with colorectal cancer specific mortality was found [168]. In another study, statin use was not associated with reduced risk of colon cancer, but was associated with reduced risk of rectal cancer [169]. Moreover, studies that have evaluated the association between statin use and risk of breast cancer reported conflicting results. it appears that statin with lipophilic structures had more chemoprotective effect against breast cancers compared to hydrophilic ones [170,171,172]. Some data have also suggested that statins have a favorable effect, particularly in patients with triple negative breast cancer [173,174,175,176,177]. Furthermore, the effect of statins on response to systemic anticancer therapy in patients with solid cancers was evaluated in observational studies. In one meta-analysis, statin did not appear to improve response to cytotoxic therapy in patients with solid tumors [178]. In a recent RCT involving patients with lymphoma who were scheduled to receive anthracycline-based chemotherapy, the odds of cardiotoxicity after anthracycline treatment were approximately three times greater for patients randomized to the placebo compared with those randomized to atorvastatin [179]. Although statin therapy shows some benefits in several types of cancer, these chemoprotective effects need to be further evaluated with RCTs.

3.6. Proprotein Convertase Subtilisin Kexin Type 9 Inhibitors (PCSK9-i)

Proprotein convertase subtilisin kexin type 9 (PCSK9) is a proteolytic enzyme that plays an important role in hepatic cholesterol homeostasis [180,181,182]. Among its various functions, it is known for its role in the reduction of low-density lipoprotein (LDL) receptor expression on hepatocytes’ cell surface [183,184,185]. Downregulation of these receptors, which is mediated by intracellular signaling and targeting for degradation, leads to higher LDL plasma levels with resultant hypercholesterolemia and heightened cardiovascular disease risk [186,187]. As the understanding of PCSK9 role in the pathogenesis of hypercholesterolemia evolved, efforts devoted to this unique pathway led to the development of monoclonal antibodies against PCSK9 [188]. Two FDA-approved injectable human monoclonal antibodies (Alirocumab and Evolucumab) are now available for use, either as monotherapy or as an add-on to other anti-lipid therapies, in patients with familial hypercholesterolemia or in patients who have intolerance to traditional first-line therapies, i.e., statin therapy [189,190,191,192,193]. Both have shown remarkable reductions in LDL levels compared to placebo, with a range of approximately 30–70% reduction, and up to 60% reduction in patients who are already on statin therapy [194,195,196,197,198,199,200,201,202,203].
Similar to the effects on LDL receptors, PCSK9 has been increasingly recognized as a regulator for other cell surface receptors, some of which are key modulators in immune signaling and response [204]. Moreover, cholesterol was found to have inhibitory effects on the antitumor responses of CD8+ T cells, and it is also involved in major histocompatibility protein class I (MHC-I) recycling on cell membranes [205,206]. These pathophysiological observations provoked the hypothesis of PCSK9 being a regulator of anti-tumor responses and thus, a potential target to enhance immunity against tumors [51]. Eliminating the effect of PCSK9, whether via direct inhibition or gene knock-out, was shown to attenuate tumor growth in mice cancer cells by decreasing the barriers or checkpoints to T call signaling, which then boosts the anti-tumor immune response to cancer cells [51]. PCSK9 deficiency also increased the expression of MHC-I on the surface of tumor cells, exposing them to the immune defenses and allowing greater intra-tumoral cytotoxic T cell infiltration [51]. These anti-tumoral effects were most notable in the presence of immune checkpoint therapy with anti-programmed cell death ligand 1 (anti-PD1) agents, where a synergistic action has been demonstrated [51]. Furthermore, the introduction of small interfering RNA (siRNA) against PCSK9 into human lung adenocarcinoma cells downregulated anti-apoptotic molecules and induced mitochondrial dysfunction, which then hampered tumor activity by promoting cell apoptosis [207]. A recent study in Japan showed that higher levels of PCSK9 antibodies in the serum was associated with favorable postoperative prognosis in esophageal cancer patients compared to low antibody levels [208]. In another Italian pilot study of 44 elderly patients with advanced and pretreated non-small cell lung cancer, serum levels of PCSK9 below a certain cutoff (95 ng/mL) at the second nivolumab dose was associated with better overall survival in comparison to higher levels [209]. In an experimental mouse models of breast and colon cancer, anti-PCSK9 vaccine, which led to lower plasma level and activity of PCSK9, was associated with moderate but not significant tumor growth reduction and prolongation of lifespan [210,211]. In a prior observational Mendelian randomization study, genetic variants that simulate PCSK9 inhibition were associated with lower breast cancer risk [212]. This emerging data makes PCSK9 inhibitors a plausible consideration for future trials to evaluate their clinical effect as an adjunct therapies for various cancers, especially those in which immune therapy have proven efficacious.

3.7. Sodium-Glucose Co-Transporter-2 Inhibitors (SGLT-2 Inhibitors)

SGLT-2 inhibitors were initially developed for diabetes treatment. They work through inhibition of sodium-glucose co-transporter-2 that is responsible for active cellular uptake of glucose and sodium, thereby decreasing glucose reabsorption in the proximal convoluted tubules of the nephrons and causing glycosuria, which leads ultimately to lowering blood glucose level [213]. However, it became evident over the past decade that the benefits of SGLT-2 inhibitors extend beyond blood glucose control in diabetic patient as multiple studies have shown significant beneficial cardiovascular effects in patients treated with SGLT-2 inhibitors [214,215]. Several randomized control trials have shown cardiovascular benefits in heart failure patients. The most recent data from DAPA-HF (Dapagliflozin and Prevention of Adverse Outcomes in Heart Failure) [216] and EMPEROR-Reduced (Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Reduced Ejection Fraction) [217] showed reduction in heart failure hospitalizations or cardiovascular death when compared with placebo. More recently, EMPEROR-Preserved (Empagliflozin Outcome Trial in Patients With Chronic Heart Failure With Preserved Ejection Fraction) trial have reported that treatment with Empagliflozin in patients with heart failure and a preserved ejection fraction is associated with a reduced combined risk of cardiovascular death or hospitalization for heart failure in patients, regardless of the presence or absence of diabetes [218]. Furthermore, CANVAS study (Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes) that combined data from two trials including more than ten thousands patients with type 2 diabetes and high cardiovascular risk, has showed that canagliflozin was associated with a reduction in major adverse cardiovascular events compared with placebo [219]. Similar results of cardiovascular benefits were replicated in other studies [220,221]. Interestingly, the cardiovascular benefits derived from SGLT2 inhibitors were independent of diabetes status of patients, which strongly suggest alternative mechanisms for the reported cardioprotective effects [221]. Several mechanism were proposed to explain the those cardioprotective effects such as decreased production of leptin and reduced pericardial adipose tissue deposition as well as inflammation [222,223]. In addition, SGLT2 inhibition was found to shift metabolism toward more lipid oxidation and ketone production along with reduced glucose oxidation [224,225]. other potential mechanisms for cardiovascular benefits of SGLT2 inhibitors include enhanced natriuresis, reduction in plasma volume and blood pressure as well as improvement of systemic endothelial function and arterial stiffness [226,227]. In cancer context, several meta-analysis studies have examined cancer risk in patients treated with various SGLT2 inhibitors, but they did not show significant chemoprotective effects [228,229,230]. Several translational studies have reported antiproliferative and chemoprotective effects with SGLT2 inhibition in certain types of cancers [231,232,233]. As described above, cancer cells exhibit metabolic reprogramming “Warburg effect” that promotes the survival and progression of cancers. Data from in-vitro studies showed amelioration of metabolic reprogramming seen in some cancers. For example, it was shown that SGLT2 inhibitor suppresses hepatocellular cancer (HCC) in-vitro growth through blockage of glucose influx-induced β-catenin action, which is a pro-oncogenic protein [234]. In another study, in-vitro treatment with SGLT2 inhibitors demonstrated disruption of adhesion capacity of certain cancer cells and suppression of oxidative phosphorylation via inhibition of mitochondrial electron transport chain in HCC, breast, prostate, and lung cancer cells [228,235,236].

3.8. Exercise

Before we conclude our discussion on the oncologic effects of cardiovascular interventions, it would be worth mentioning that exercise, which is thought of as a cardiovascular lifestyle intervention, can also boost cancer immunity. Current epidemiological evidence suggests that regular physical activity and exercise influence cardiovascular health in several ways. This include antiatherogenic effects in blood vessels, including improvements in vascular endothelial function and structural vascular adaptations, and a healthy autonomic balance (regular exercise increases vagal tone to the heart and prevents malignant arrhythmias) [237,238]. Additionally, regular exercise can also prevent fatal arrhythmias by inducing cardiac preconditioning, which provides a cardio-protective effect against ischemia-reperfusion injury [239,240,241,242]. Nonetheless, the cardiac preconditioning effects of exercise is still widely under-recognized and mostly evident in preclinical studies [243].
The benefits of exercise training for cancer patients are also becoming increasingly evident. It has been shown that aerobic exercise reduces cancer incidence and inhibits tumor growth [244,245]. Epidemiological studies have shown that physical activity reduces the risk of at least 13 different cancer types with an exercise-dependent reduction in the risk of disease recurrence for certain cancers [246]. Likewise, numerous preclinical exercise studies showed similar exercise-dependent protection against cancer [247]. Across the vast majority of preclinical studies investigating the effect of exercise on cancer outcomes, exercise has been shown to reduce the rate of tumor growth [244,245,247].
Additionally, exercise training not only can reduce tumor growth, but may also have the potential to augment the potency and efficacy of traditional cancer therapies [248]. As the efficacy of both chemotherapy and immunotherapy relies on adequate blood perfusion to the tumor, exercise training strongly affects blood circulation and oxygen delivery to peripheral tissues and thereby enhance delivery of the cytotoxic drugs to the interior of tumors [245,249,250].

4. Future Perspectives… Will We Be Ready to Change Our Protocols in the Near Future?

As elucidated, the interrelation between cancer and CVDs manifests through shared risk factors and molecular mechanisms. For many years, the field of cardio-oncology has predominantly focused on the development of CVD during or following cancer treatment. However, contemporary investigations have unveiled the potential for cancer to instigate or exacerbate CVDs, resulting in exacerbated prognostic outcomes.
Importantly, advancements in cancer therapeutics and increased longevity among cancer patients have brought to the fore a concomitant rise in the prevalence of CVDs within this demographic. Nonetheless, it is evident that even cancer patients with established diagnoses of cardiovascular diseases are less likely to receive standard treatments compared with those without cancer. Furthermore, many standard chemotherapy regimens are associated with direct cardiotoxic effects; many of which are unpredictable and associated with increased worse outcomes that account in part for increased mortality and morbidity in this cohort. Additional cancer- or cancer therapy-related factors that may complicate or undermine treatment of cardiovascular diseases including elevated risk of bleeding, thrombosis, and hypercoagulability in certain patients. For example, coronary artery disease in cancer patients may not be adequately treated, and they are less likely to undergo percutaneous coronary revascularization [251,252,253]. This reluctance may stem from concerns about or perceptions of an increased risk of stent thrombosis, or bleeding with optimal antiplatelet therapy. Therefore, it is crucial to study effects and safety of routinely used cardiovascular therapies in cancer patients, which may ultimately assist in development of cancer-specific risk scoring system that guide clinicians for treatment of cardiovascular diseases in cancer patients.
On management front, cardiovascular medications may be used as potential adjuvant therapies to standard anticancer treatments that could improve anti-tumor response and effectiveness. Although further studies are needed, repurposing cardiovascular therapies for non-cardiovascular indications and extending these interventions to cancer patients could be a promising therapeutic opportunity for patients with different stages of cancer who may not respond to standard cancer therapy or even lack an effective therapeutic treatment. Although preclinical studies have shown that these medications can demonstrate chemoprotective effects, or improve the effectiveness of standard chemotherapy regimens by modulating various pathways and molecular targets that are integral parts of tumorigenesis, the exact mechanisms by which some of the cardiovascular therapies provide beneficial effects for certain types of cancers are neither well-understood nor quite predictable.
Therefore, randomized clinical trials involving human subjects are imperative to ascertaining the efficacy of repurposed therapies for specific cancers, along with determining the optimal dosages necessary to achieve these significant therapeutic effects. Additionally, future research endeavors should prioritize elucidating the intricate interplay between CVD and cancer, thereby fostering the development of preventive strategies, and facilitating the co-management of cancer and CVD. This comprehensive approach is pivotal for ensuring successful cancer survivorship and optimizing patient outcomes in cardio-oncology.

5. Conclusions

In conclusion, the intricate nexus between CVD and cancer is increasingly acknowledged as a pivotal determinant of patient prognosis and therapeutic paradigms. Emerging evidence highlights a mutual, bidirectional relationship in which cancer and CVD distinctly influence one another’s outcomes. It is important to note that CVD does not increase the risk of cancer per se; rather, shared risk factors in patients with CVD may also promote cancer development. This convergence of disciplines presents formidable challenges alongside promising avenues for augmenting patient care. Recognizing the interconnected risk factors and molecular cascades between cancer and CVD, as well as the potential cardiotoxic ramifications of cancer therapies, is of paramount importance. The exploration of cardiovascular pharmacotherapeutics repurposed as adjunctive modalities in cancer management holds substantial potential, albeit remaining speculative and necessitating rigorous validation through randomized clinical trials. Moreover, adopting a holistic approach to the concurrent management of cancer and CVD is imperative for fostering resilient cancer survivorship and optimizing patient outcomes within the burgeoning domain of cardio-oncology. Hence, the advancement of our understanding pertaining to this complex interplay and the formulation of pre-emptive strategies stand as pivotal strides towards elevating the standard of healthcare delivery for individuals confronting the dual burden of cancer and cardiovascular disease.

Author Contributions

Conceptualization, M.Z., J.A. and R.M.; validation, M.Z. and J.A.; resources, M.A., A.A., J.L. and F.A.; writing—original draft preparation, M.Z. and J.A.; writing—review and editing, R.M.; supervision, R.M. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Acknowledgments

We extend our sincere appreciation to Razan Alrawashdeh and Ranya Baddourah for their invaluable contributions to this manuscript. Their expertise and support have greatly enhanced the quality of our work, and we are deeply grateful for their dedication throughout this endeavor.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Centers for Disease Control and Prevention. WHO Guidelines. Underlying Cause Death, 1999–2018; CDC WONDER Online Database: Atlanta, GA, USA, 2018. [Google Scholar]
  2. Weir, H.K. Heart Disease and Cancer Deaths—Trends and Projections in the United States, 1969–2020. Prev. Chronic Dis. 2019, 13, E157. [Google Scholar] [CrossRef] [PubMed]
  3. Bray, F.; Ferlay, J.; Soerjomataram, I.; Siegel, R.L.; Torre, L.A.; Jemal, A. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA. Cancer J. Clin. 2018, 68, 394–424. [Google Scholar] [CrossRef] [PubMed]
  4. Xu, J.; Murphy, S.L.; Kochanek, K.D.; Arias, E. Deaths: Final data for 2019. Natl. Vital Stat. Rep. 2021, 70, 8. [Google Scholar] [CrossRef]
  5. The World Health Organization (WHO). Cardiovascular Diseases (CVDs); WHO: Geneva, Switzerland, 2021. [Google Scholar]
  6. The World Health Organization (WHO). Cancer; WHO: Geneva, Switzerland, 2022. [Google Scholar]
  7. de Boer, R.A.; Meijers, W.C.; van der Meer, P.; van Veldhuisen, D.J. Cancer and heart disease: Associations and relations. Eur. J. Heart Fail. 2019, 21, 1515–1525. [Google Scholar] [CrossRef] [PubMed]
  8. Al-Kindi, S.G.; Oliveira, G.H. Prevalence of Preexisting Cardiovascular Disease in Patients with Different Types of Cancer the Unmet Need for Onco-Cardiology. Mayo Clin. Proc. 2016, 91, 81–83. [Google Scholar] [CrossRef] [PubMed]
  9. Herrmann, J.; Lerman, A.; Sandhu, N.P.; Villarraga, H.R.; Mulvagh, S.L.; Kohli, M. Evaluation and Management of Patients With Heart Disease and Cancer: Cardio-Oncology. Mayo Clin. Proc. 2014, 89, 1287. [Google Scholar] [CrossRef] [PubMed]
  10. Cardinale, D.; Colombo, A.; Lamantia, G.; Colombo, N.; Civelli, M.; De Giacomi, G.; Pandini, C.; Sandri, M.; Cipolla, C. Cardio-oncology: A new medical issue. Ecancermedicalscience 2008, 2, 126. [Google Scholar] [CrossRef] [PubMed]
  11. Koutsoukis, A.; Ntalianis, A.; Repasos, E.; Kastritis, E.; Dimopoulos, M.A.; Paraskevaidis, I. Cardio-oncology: A Focus on Cardiotoxicity. Eur. Cardiol. Rev. 2018, 13, 64. [Google Scholar] [CrossRef] [PubMed]
  12. Hershman, D.L.; McBride, R.B.; Eisenberger, A.; Wei, Y.T.; Grann, V.R.; Jacobson, J.S. Doxorubicin, cardiac risk factors, and cardiac toxicity in elderly patients with diffuse B-cell non-Hodgkin’s lymphoma. J. Clin. Oncol. 2008, 26, 3159–3165. [Google Scholar] [CrossRef]
  13. Wang, S.Y.; Long, J.B.; Hurria, A.; Owusu, C.; Steingart, R.M.; Gross, C.P.; Chen, J. Cardiovascular events, early discontinuation of trastuzumab, and their impact on survival. Breast Cancer Res. Treat. 2014, 146, 411–419. [Google Scholar] [CrossRef]
  14. Copeland-Halperin, R.S.; Al-Sadawi, M.; Patil, S.; Liu, J.E.; Steingart, R.M.; Dang, C.T.; Yu, A.F. Early Trastuzumab Interruption and Recurrence-Free Survival in ERBB2-Positive Breast Cancer. JAMA Oncol. 2020, 6, 1971. [Google Scholar] [CrossRef] [PubMed]
  15. Lotrionte, M.; Biondi-zoccai, G.; Abbate, A. Review and Meta-Analysis of Incidence and Clinical Predictors of Anthracycline Cardiotoxicity. Am. J. Cardiol. 2013, 112, 1980–1984. [Google Scholar] [CrossRef] [PubMed]
  16. Gunaldi, M.; Duman, B.B.; Afsar, C.U.; Paydas, S.; Erkisi, M.; Kara, I.O.; Sahin, B. Risk factors for developing cardiotoxicity of trastuzumab in breast cancer patients: An observational single-centre study. J. Oncol. Pharm. Pract. 2016, 22, 242–247. [Google Scholar] [CrossRef] [PubMed]
  17. Hamnvik, O.R.; Choueiri, T.K.; Turchin, A.; Mckay, R.R. Clinical Risk Factors for the Development of Hypertension in Patients Treated with Inhibitors of the VEGF Signaling Pathway. Cancer 2015, 121, 311–319. [Google Scholar] [CrossRef] [PubMed]
  18. Drobni, Z.D.; Alvi, R.M.; Taron, J.; Zafar, A.; Murphy, S.P.; Rambarat, P.K.; Mosarla, R.C.; Lee, C.; Zlotoff, D.A.; Raghu, V.K.; et al. Association between Immune Checkpoint Inhibitors with Cardiovascular Events and Atherosclerotic Plaque. Circulation 2020, 142, 2299–2311. [Google Scholar] [CrossRef]
  19. Wassif, H.; Hussain, M.; Collier, P.H.; Moudgil, R. Immunotherapy-mediated valvulitis: A new cardiovascular immunotherapy-related adverse event. Eur. Hear. J.—Cardiovasc. Imaging 2020, 21, 1102. [Google Scholar] [CrossRef]
  20. Moslehi, J.; Lichtman, A.H.; Sharpe, A.H.; Galluzzi, L.; Kitsis, R.N. Immune checkpoint inhibitor-associated myocarditis: Manifestations and mechanisms. J. Clin. Investig. 2021, 131, e145186. [Google Scholar] [CrossRef] [PubMed]
  21. Bell, C.F.; Lei, X.; Haas, A.; Baylis, R.A.; Gao, H.; Luo, L.; Giordano, S.H.; Wehner, M.R.; Nead, K.T.; Leeper, N.J. Risk of Cancer After Diagnosis of Cardiovascular Disease. JACC. CardioOncology 2023, 5, 431–440. [Google Scholar] [CrossRef]
  22. Koelwyn, G.J.; Aboumsallem, J.P.; Moore, K.J.; de Boer, R.A. Reverse cardio-oncology: Exploring the effects of cardiovascular disease on cancer pathogenesis. J. Mol. Cell. Cardiol. 2022, 163, 1–8. [Google Scholar] [CrossRef]
  23. Coussens, L.M.; Werb, Z. Inflammation and cancer. Nature 2002, 420, 860–867. [Google Scholar] [CrossRef]
  24. Libby, P. Inflammation and cardiovascular disease mechanisms. Am. J. Clin. Nutr. 2006, 83, 456S–460S. [Google Scholar] [CrossRef] [PubMed]
  25. Mantovani, A.; Allavena, P.; Sica, A.; Balkwill, F. Cancer-related inflammation. Nature 2008, 454, 436–444. [Google Scholar] [CrossRef] [PubMed]
  26. Elinav, E.; Nowarski, R.; Thaiss, C.A.; Hu, B.; Jin, C.; Flavell, R.A. Inflammation-induced cancer: Crosstalk between tumours, immune cells and microorganisms. Nat. Rev. Cancer 2013, 13, 759–771. [Google Scholar] [CrossRef] [PubMed]
  27. Diakos, C.I.; Charles, K.A.; McMillan, D.C.; Clarke, S.J. Cancer-related inflammation and treatment effectiveness. Lancet. Oncol. 2014, 15, e493–e503. [Google Scholar] [CrossRef] [PubMed]
  28. Koene, R.J.; Prizment, A.E.; Blaes, A.; Konety, S.H. Shared Risk Factors in Cardiovascular Disease and Cancer. Circulation 2016, 133, 1104–1114. [Google Scholar] [CrossRef] [PubMed]
  29. Steppan, C.M.; Bailey, S.T.; Bhat, S.; Brown, E.J.; Banerjee, R.R.; Wright, C.M.; Patel, H.R.; Ahima, R.S.; Lazar, M.A. The hormone resistin links obesity to diabetes. Nature 2001, 409, 307–312. [Google Scholar] [CrossRef] [PubMed]
  30. Makki, K.; Froguel, P.; Wolowczuk, I. Adipose Tissue in Obesity-Related Inflammation and Insulin Resistance: Cells, Cytokines, and Chemokines. ISRN Inflamm. 2013, 2013, 139239. [Google Scholar] [CrossRef] [PubMed]
  31. Iyengar, N.M.; Gucalp, A.; Dannenberg, A.J.; Hudis, C.A. Obesity and Cancer Mechanisms: Tumor Microenvironment and Inflammation. J. Clin. Oncol. Off. J. Am. Soc. Clin. Oncol. 2016, 34, 4270–4276. [Google Scholar] [CrossRef] [PubMed]
  32. Lauby-Secretan, B.; Scoccianti, C.; Loomis, D.; Grosse, Y.; Bianchini, F.; Straif, K. Body Fatness and Cancer—Viewpoint of the IARC Working Group. N. Engl. J. Med. 2016, 375, 794–798. [Google Scholar] [CrossRef]
  33. Gregor, M.F.; Hotamisligil, G.S. Inflammatory mechanisms in obesity. Annu. Rev. Immunol. 2011, 29, 415–445. [Google Scholar] [CrossRef]
  34. Katsiki, N.; Mikhailidis, D.P.; Banach, M. Leptin, cardiovascular diseases and type 2 diabetes mellitus. Acta Pharmacol. Sin. 2018, 39, 1176–1188. [Google Scholar] [CrossRef] [PubMed]
  35. Vlasova, M.; Purhonen, A.K.; Jarvelin, M.R.; Rodilla, E.; Pascual, J.; Herzig, K.H. Role of adipokines in obesity-associated hypertension. Acta Physiol. 2010, 200, 107–127. [Google Scholar] [CrossRef] [PubMed]
  36. de Wit, S.; Glen, C.; de Boer, R.A.; Lang, N.N. Mechanisms shared between cancer, heart failure, and targeted anti-cancer therapies. Cardiovasc. Res. 2023, 118, 3451–3466. [Google Scholar] [CrossRef] [PubMed]
  37. Ridker, P.M.; Everett, B.M.; Thuren, T.; MacFadyen, J.G.; Chang, W.H.; Ballantyne, C.; Fonseca, F.; Nicolau, J.; Koenig, W.; Anker, S.D.; et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N. Engl. J. Med. 2017, 377, 1119–1131. [Google Scholar] [CrossRef] [PubMed]
  38. Ridker, P.M.; MacFadyen, J.G.; Thuren, T.; Everett, B.M.; Libby, P.; Glynn, R.J. 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 2017, 390, 1833–1842. [Google Scholar] [CrossRef] [PubMed]
  39. Renehan, A.G.; Zwahlen, M.; Minder, C.; Dwyer, S.T.O.; Shalet, S.M.; Egger, M. Insulin-like growth factor (IGF)-I, IGF binding protein-3, and cancer risk: Systematic review and meta-regression analysis. Lancet 2004, 363, 1346–1353. [Google Scholar] [CrossRef] [PubMed]
  40. Hadi, H.A.R.; Al Suwaidi, J.A. Endothelial dysfunction in diabetes mellitus. Vasc. Health Risk Manag. 2007, 3, 853. [Google Scholar] [PubMed]
  41. Tabit, C.E.; Chung, W.B.; Hamburg, N.M.; Vita, J.A. Endothelial dysfunction in diabetes mellitus: Molecular mechanisms and clinical implications. Rev. Endocr. Metab. Disord. 2010, 11, 61. [Google Scholar] [CrossRef] [PubMed]
  42. Garg, S.K.; Maurer, H.; Reed, K.; Selagamsetty, R. Diabetes and cancer: Two diseases with obesity as a common risk factor. Diabetes, Obes. Metab. 2014, 16, 97–110. [Google Scholar] [CrossRef]
  43. Giri, B.; Dey, S.; Das, T.; Sarkar, M.; Banerjee, J.; Dash, S.K. Chronic hyperglycemia mediated physiological alteration and metabolic distortion leads to organ dysfunction, infection, cancer progression and other pathophysiological consequences: An update on glucose toxicity. Biomed. Pharmacother. 2018, 107, 306–328. [Google Scholar] [CrossRef]
  44. Byrne, F.L.; Martin, A.R.; Kosasih, M.; Caruana, B.T.; Farrell, R. The Role of Hyperglycemia in Endometrial Cancer Pathogenesis. Cancers 2020, 12, 1191. [Google Scholar] [CrossRef] [PubMed]
  45. Kaaks, R.; Lukanova, A.; Kurzer, M.S. Obesity, endogenous hormones, and endometrial cancer risk: A synthetic review. Cancer Epidemiol. Biomark. Prev. 2002, 11, 1531–1543. [Google Scholar]
  46. Wairagu, P.M.; Phan, A.N.H.; Kim, M.K.; Han, J.; Kim, H.W.; Choi, J.W.; Kim, K.W.; Cha, S.K.; Park, K.H.; Jeong, Y. Insulin priming effect on estradiol-induced breast cancer metabolism and growth. Cancer Biol. Ther. 2015, 16, 484. [Google Scholar] [CrossRef]
  47. Bayes-Genis, A.; Conover, C.A.; Schwartz, R.S. The Insulin-Like Growth Factor Axis. Circ. Res. 2000, 86, 125–130. [Google Scholar] [CrossRef] [PubMed]
  48. Giovannucci, E. Insulin, Insulin-Like Growth Factors and Colon Cancer: A Review of the Evidence. J. Nutr. 2001, 131, 3109S–3120S. [Google Scholar] [CrossRef] [PubMed]
  49. Cohen, D.H.; LeRoith, D. Obesity, type 2 diabetes, and cancer: The insulin and IGF connection. Endocr. Relat. Cancer 2012, 19, F27–F45. [Google Scholar] [CrossRef] [PubMed]
  50. Gallagher, E.J.; LeRoith, D. The proliferating role of insulin and insulin-like growth factors in cancer. Trends Endocrinol. Metab. 2010, 21, 610–618. [Google Scholar] [CrossRef] [PubMed]
  51. Liu, X.; Bao, X.; Hu, M.; Chang, H.; Jiao, M.; Cheng, J.; Xie, L.; Huang, Q.; Li, F.; Li, C.-Y. Inhibition of PCSK9 potentiates immune checkpoint therapy for cancer. Nature 2020, 588, 693–698. [Google Scholar] [CrossRef] [PubMed]
  52. de Boer, R.A.; Hulot, J.S.; Tocchetti, C.G.; Aboumsallem, J.P.; Ameri, P.; Anker, S.D.; Bauersachs, J.; Bertero, E.; Coats, A.J.S.; Čelutkienė, J.; et al. Common mechanistic pathways in cancer and heart failure. A scientific roadmap on behalf of the Translational Research Committee of the Heart Failure Association (HFA) of the European Society of Cardiology (ESC). Eur. J. Heart Fail. 2020, 22, 2272–2289. [Google Scholar] [CrossRef]
  53. Allard, M.F.; Schonekess, B.O.; Henning, S.L.; English, D.R.; Lopaschuk, G.D. Contribution of oxidative metabolism and glycolysis to ATP production in hypertrophied hearts. Am. J. Physiol.—Hear. Circ. Physiol. 1994, 267, H742–H750. [Google Scholar] [CrossRef]
  54. Goodwin, G.W.; Taylor, C.S.; Taegtmeyer, H. Regulation of energy metabolism of the heart during acute increase in heart work. J. Biol. Chem. 1998, 273, 29530–29539. [Google Scholar] [CrossRef]
  55. De Jong, K.A.; Lopaschuk, G.D. Complex Energy Metabolic Changes in Heart Failure With Preserved Ejection Fraction and Heart Failure With Reduced Ejection Fraction. Can. J. Cardiol. 2017, 33, 860–871. [Google Scholar] [CrossRef] [PubMed]
  56. Ritterhoff, J.; Young, S.; Villet, O.; Shao, D.; Carnevale Neto, F.; Bettcher, L.F.; Hsu, Y.W.A.; Kolwicz, S.C.; Raftery, D.; Tian, R. Metabolic remodeling promotes cardiac hypertrophy by directing glucose to aspartate biosynthesis. Circ. Res. 2020, 126, 182–196. [Google Scholar] [CrossRef] [PubMed]
  57. Liberti, M.V.; Locasale, J.W. The Warburg Effect: How Does it Benefit Cancer Cells? Trends Biochem. Sci. 2016, 41, 211. [Google Scholar] [CrossRef] [PubMed]
  58. DeBerardinis, R.J.; Mancuso, A.; Daikhin, E.; Nissim, I.; Yudkoff, M.; Wehrli, S.; Thompson, C.B. Beyond aerobic glycolysis: Transformed cells can engage in glutamine metabolism that exceeds the requirement for protein and nucleotide synthesis. Proc. Natl. Acad. Sci. USA 2007, 104, 19345–19350. [Google Scholar] [CrossRef] [PubMed]
  59. Karlstaedt, A.; Moslehi, J.; de Boer, R.A. Cardio-onco-metabolism: Metabolic remodelling in cardiovascular disease and cancer. Nat. Rev. Cardiol. 2022, 19, 414–425. [Google Scholar] [CrossRef] [PubMed]
  60. Akbay, E.A.; Moslehi, J.; Christensen, C.L.; Saha, S.; Tchaicha, J.H.; Ramkissoon, S.H.; Stewart, K.M.; Carretero, J.; Kikuchi, E.; Zhang, H.; et al. D-2-hydroxyglutarate produced by mutant IDH2 causes cardiomyopathy and neurodegeneration in mice. Genes Dev. 2014, 28, 479–490. [Google Scholar] [CrossRef] [PubMed]
  61. Karlstaedt, A.; Zhang, X.; Vitrac, H.; Harmancey, R.; Vasquez, H.; Wang, J.H.; Goodell, M.A.; Taegtmeyer, H. Oncometabolite d-2-hydroxyglutarate impairs α-ketoglutarate dehydrogenase and contractile function in rodent heart. Proc. Natl. Acad. Sci. USA 2016, 113, 10436–10441. [Google Scholar] [CrossRef] [PubMed]
  62. Arnett, D.K.; Blumenthal, R.S.; Albert, M.A.; Buroker, A.B.; Goldberger, Z.D.; Hahn, E.J.; Himmelfarb, C.D.; Khera, A.; Lloyd-Jones, D.; McEvoy, J.W.; et al. 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 2019, 140, e596–e646. [Google Scholar] [CrossRef]
  63. Lloyd-Jones, D.M.; Bennett, G.; Coady, S.; D’Agostino, R.B.; Gibbons, R.; Greenland, P.; Lackland, D.T.; Levy, D.; O’Donnell, C.J.; Robinson, J.G.; et al. 2013 ACC/AHA guideline on the assessment of cardiovascular risk: A report of the American College of Cardiology/American Heart Association Task Force on Practice Guidelines. Circulation 2014, 129, S49–S73. [Google Scholar] [CrossRef]
  64. Blum, C.B.; Eckel, R.H.; Goldberg, A.C.; Gordon, D.; Levy, D.; Lloyd-Jones, D.M.; McBride, P.; Sanford Schwartz, J.; Shero, S.T.; Smith, S.C.; et al. 2013 ACC/AHA Guideline on the Treatment of Blood Cholesterol to Reduce Atherosclerotic Cardiovascular Risk in Adults. Am. Hear. Assoc. Task Force Pract. Guidel. Circ. 2014, 129, 1–45. [Google Scholar] [CrossRef]
  65. Chia, Y.C.; Lim, H.M.; Ching, S.M. Validation of the pooled cohort risk score in an Asian population—A retrospective cohort study. BMC Cardiovasc. Disord. 2014, 14, 163. [Google Scholar] [CrossRef]
  66. Hanee Henderson, K.; Kaufman, B.G.; Stearns, S.; Couper, D.; Sueta, C.; Kucharska-Newton, A.M.; Foraker, R.E.; Asafu-Adjei, J.; Chang, P. Prevention Validation of the Atherosclerotic Cardiovascular Disease (Ascvd) Pooled Cohort Risk Equations by Education Level: The Atherosclerosis Risk in Communities (Aric) Study. J. Am. Coll. Cardiol. 2016, 5, 1842. [Google Scholar] [CrossRef]
  67. Rana, J.S.; Tabada, G.H.; Solomon, M.D.; Lo, J.C.; Jaffe, M.G.; Sung, S.H.; Ballantyne, C.M.; Go, A.S. Accuracy of the Atherosclerotic Cardiovascular Risk Equation in a Large Contemporary, Multiethnic Population. J. Am. Coll. Cardiol. 2016, 67, 2118–2130. [Google Scholar] [CrossRef]
  68. Conroy, R.M.; Pyörälä, K.; Fitzgerald, A.P.; Sans, S.; Menotti, A.; De Backer, G.; De Bacquer, D.; Ducimetière, P.; Jousilahti, P.; Keil, U.; et al. Estimation of ten-year risk of fatal cardiovascular disease in Europe: The SCORE project on behalf of the SCORE project group. Eur. Heart J. 2003, 24, 987–1003. [Google Scholar] [CrossRef] [PubMed]
  69. Meijers, W.C.; de Boer, R.A. Common risk factors for heart failure and cancer. Cardiovasc. Res. 2019, 115, 844–853. [Google Scholar] [CrossRef] [PubMed]
  70. Hooning, M.J.; Botma, A.; Aleman, B.M.P.; Baaijens, M.H.A.; Bartelink, H.; Klijn, J.G.M.; Taylor, C.W.; van Leeuwen, F.E. Long-term risk of cardiovascular disease in 10-year survivors of breast cancer. J. Natl. Cancer Inst. 2007, 99, 365–375. [Google Scholar] [CrossRef] [PubMed]
  71. van Nimwegen, F.A.; Schaapveld, M.; Janus, C.P.M.; Krol, A.D.G.; Petersen, E.J.; Raemaekers, J.M.M.; Kok, W.E.M.; Aleman, B.M.P.; Leeuwen, F.E. van Cardiovascular Disease After Hodgkin Lymphoma Treatment: 40-Year Disease Risk. JAMA Intern. Med. 2015, 175, 1007–1017. [Google Scholar] [CrossRef] [PubMed]
  72. Boyne, D.J.; Mickle, A.T.; Brenner, D.R.; Friedenreich, C.M.; Cheung, W.Y.; Tang, K.L.; Wilson, T.A.; Lorenzetti, D.L.; James, M.T.; Ronksley, P.E.; et al. Long-term risk of cardiovascular mortality in lymphoma survivors: A systematic review and meta-analysis. Cancer Med. 2018, 7, 4801. [Google Scholar] [CrossRef]
  73. Vaughn, D.J.; Palmer, S.C.; Carver, J.R.; Jacobs, L.A.; Mohler, E.R. Cardiovascular risk in long-term survivors of testicular cancer. Cancer 2008, 112, 1949–1953. [Google Scholar] [CrossRef]
  74. Dixon, J.M.; Farrell, C.; Jones, A.; Leonard, R.; Murray, N.; Palmieri, C.; Plummer, C.J.; Stanley, A.; Verrill, M.W. Expert opinion on the use of anthracyclines in patients with advanced breast cancer at cardiac risk. Ann. Oncol. 2009, 20, 816–827. [Google Scholar] [CrossRef]
  75. Rubio-Infante, N.; Ramírez-Flores, Y.A.; Castillo, E.C.; Lozano, O.; García-Rivas, G.; Torre-Amione, G. Cardiotoxicity associated with immune checkpoint inhibitor therapy: A meta-analysis. Eur. J. Heart Fail. 2021, 23, 1739–1747. [Google Scholar] [CrossRef] [PubMed]
  76. Vater, L.B.; Lefebvre, B.; Turk, A.; Clasen, S.C. Fluoropyrimidine Cardiotoxicity: Incidence, Outcomes, and Safety of Rechallenge. Curr. Oncol. Rep. 2022, 24, 943–950. [Google Scholar] [CrossRef] [PubMed]
  77. Armenian, S.H.; Lacchetti, C.; Barac, A.; Carver, J.; Constine, L.S.; Denduluri, N.; Dent, S.; Douglas, P.S.; Durand, J.-B.; Ewer, M.; et al. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2016, 35, 893–911. [Google Scholar] [CrossRef] [PubMed]
  78. Mehta, L.S.; Watson, K.E.; Barac, A.; Beckie, T.M.; Bittner, V.; Cruz-Flores, S.; Dent, S.; Kondapalli, L.; Ky, B.; Okwuosa, T.; et al. Cardiovascular Disease and Breast Cancer: Where These Entities Intersect: A Scientific Statement From the American Heart Association. Circulation 2018, 137, e30–e66. [Google Scholar] [CrossRef] [PubMed]
  79. Handy, C.E.; Quispe, R.; Pinto, X.; Blaha, M.J.; Blumenthal, R.S.; Michos, E.D.; Lima, J.A.C.; Guallar, E.; Ryu, S.; Cho, J.; et al. Synergistic Opportunities in the Interplay Between Cancer Screening and Cardiovascular Disease Risk Assessment. Circulation 2018, 138, 727–734. [Google Scholar] [CrossRef]
  80. Richards, M.A.; Stockton, D.; Babb, P.; Coleman, M.P. How many deaths have been avoided through improvements in cancer survival? BMJ 2000, 320, 895–898. [Google Scholar] [CrossRef] [PubMed]
  81. Enright, K.A.; Krzyzanowska, M.K. Control of cardiovascular risk factors among adult cancer survivors: A population-based survey. Cancer Causes Control 2010, 21, 1867–1874. [Google Scholar] [CrossRef] [PubMed]
  82. Gernaat, S.A.M.; Boer, J.M.A.; van den Bongard, D.H.J.; Maas, A.H.E.M.; van der Pol, C.C.; Bijlsma, R.M.; Grobbee, D.E.; Verkooijen, H.M.; Peeters, P.H. The risk of cardiovascular disease following breast cancer by Framingham risk score. Breast Cancer Res. Treat. 2018, 170, 119–127. [Google Scholar] [CrossRef]
  83. Gernaat, S.A.M.; Išgum, I.; de Vos, B.D.; Takx, R.A.P.; Young-Afat, D.A.; Rijnberg, N.; Grobbee, D.E.; van der Graaf, Y.; de Jong, P.A.; Leiner, T.; et al. Automatic Coronary Artery Calcium Scoring on Radiotherapy Planning CT Scans of Breast Cancer Patients: Reproducibility and Association with Traditional Cardiovascular Risk Factors. PLoS ONE 2016, 11, e0167925. [Google Scholar] [CrossRef]
  84. Sturgeon, K.M.; Deng, L.; Bluethmann, S.M.; Zhou, S.; Trifiletti, D.M.; Jiang, C.; Kelly, S.P.; Zaorsky, N.G. A population-based study of cardiovascular disease mortality risk in US cancer patients. Eur. Heart J. 2019, 40, 3889–3897. [Google Scholar] [CrossRef] [PubMed]
  85. Rauzi, F.; Kirkby, N.S.; Edin, M.L.; Whiteford, J.; Zeldin, D.C.; Mitchell, J.A.; Warner, T.D. Aspirin inhibits the production of proangiogenic 15(S)-HETE by platelet cyclooxygenase-1. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2016, 30, 4256–4266. [Google Scholar] [CrossRef]
  86. Davì, G.; Patrono, C. Platelet activation and atherothrombosis. N. Engl. J. Med. 2007, 357, 2482–2494. [Google Scholar] [CrossRef] [PubMed]
  87. Levine, G.N.; Bates, E.R.; Bittl, J.A.; Brindis, R.G.; Fihn, S.D.; Fleisher, L.A.; Granger, C.B.; Lange, R.A.; Mack, M.J.; Mauri, L.; et al. 2016 ACC/AHA Guideline Focused Update on Duration of Dual Antiplatelet Therapy in Patients With Coronary Artery Disease: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines: An Update of th. Circulation 2016, 134, e123–e155. [Google Scholar] [CrossRef] [PubMed]
  88. Collins, R.; Peto, R.; Hennekens, C.; Doll, R.; Bubes, V.; Buring, J.; Dushkesas, R.; Gaziano, M.; Brennan, P.; Meade, T.; et al. Aspirin in the primary and secondary prevention of vascular disease: Collaborative meta-analysis of individual participant data from randomised trials. Lancet 2009, 373, 1849–1860. [Google Scholar]
  89. Patrono, C. Low-dose aspirin in primary prevention: Cardioprotection, chemoprevention, both, or neither? Eur. Heart J. 2013, 34, 3403–3411. [Google Scholar] [CrossRef] [PubMed]
  90. Thun, M.J.; Henley, S.J.; Patrono, C. Nonsteroidal anti-inflammatory drugs as anticancer agents: Mechanistic, pharmacologic, and clinical issues. J. Natl. Cancer Inst. 2002, 94, 252–266. [Google Scholar] [CrossRef] [PubMed]
  91. Patrignani, P.; Patrono, C. Aspirin and Cancer. J. Am. Coll. Cardiol. 2016, 68, 967–976. [Google Scholar] [CrossRef] [PubMed]
  92. Giardiello, F.M.; Yang, V.W.; Hylind, L.M.; Krush, A.J.; Petersen, G.M.; Trimbath, J.D.; Piantadosi, S.; Garrett, E.; Geiman, D.E.; Hubbard, W.; et al. Primary chemoprevention of familial adenomatous polyposis with sulindac. N. Engl. J. Med. 2002, 346, 1054–1059. [Google Scholar] [CrossRef]
  93. Nugent, K.P.; Farmer, K.C.; Spigelman, A.D.; Williams, C.B.; Phillips, R.K. Randomized controlled trial of the effect of sulindac on duodenal and rectal polyposis and cell proliferation in patients with familial adenomatous polyposis. Br. J. Surg. 1993, 80, 1618–1619. [Google Scholar] [CrossRef]
  94. Bresalier, R.S.; Sandler, R.S.; Quan, H.; Bolognese, J.A.; Oxenius, B.; Horgan, K.; Lines, C.; Riddell, R.; Morton, D.; Lanas, A.; et al. Cardiovascular events associated with rofecoxib in a colorectal adenoma chemoprevention trial. N. Engl. J. Med. 2005, 352, 1092–1102. [Google Scholar] [CrossRef] [PubMed]
  95. Solomon, S.D.; McMurray, J.J.V.; Pfeffer, M.A.; Wittes, J.; Fowler, R.; Finn, P.; Anderson, W.F.; Zauber, A.; Hawk, E.; Bertagnolli, M. Cardiovascular risk associated with celecoxib in a clinical trial for colorectal adenoma prevention. N. Engl. J. Med. 2005, 352, 1071–1080. [Google Scholar] [CrossRef] [PubMed]
  96. Lin, J.-L.; Lin, J.-X.; Zheng, C.-H.; Li, P.; Xie, J.-W.; Wang, J.; Lu, J.; Chen, Q.-Y.; Cao, L.; Lin, M.; et al. Relationship between aspirin use of esophageal, gastric and colorectal cancer patient survival: A meta-analysis. BMC Cancer 2020, 20, 638. [Google Scholar] [CrossRef]
  97. Miller, E.J.; Patell, R.; Uhlmann, E.J.; Ren, S.; Southard, H.; Elavalakanar, P.; Weber, G.M.; Neuberg, D.; Zwicker, J.I. Antiplatelet medications and risk of intracranial hemorrhage in patients with metastatic brain tumors. Blood Adv. 2022, 6, 1559–1565. [Google Scholar] [CrossRef] [PubMed]
  98. Streicher, S.A.; Yu, H.; Lu, L.; Kidd, M.S.; Risch, H.A. Case-control study of aspirin use and risk of pancreatic cancer. CancerEpidemiol. Biomark. Prev. 2014, 23, 1254–1263. [Google Scholar] [CrossRef]
  99. Wang, L.; Zhang, R.; Yu, L.; Xiao, J.; Zhou, X.; Li, X.; Song, P.; Li, X. Aspirin Use and Common Cancer Risk: A Meta-Analysis of Cohort Studies and Randomized Controlled Trials. Front. Oncol. 2021, 11, 690219. [Google Scholar] [CrossRef] [PubMed]
  100. Algra, A.M.; Rothwell, P.M. Effects of regular aspirin on long-term cancer incidence and metastasis: A systematic comparison of evidence from observational studies versus randomised trials. Lancet Oncol. 2012, 13, 518–527. [Google Scholar] [CrossRef]
  101. Burn, J.; Gerdes, A.; Macrae, F.; Mecklin, J.; Moeslein, G.; Olschwang, S.; Eccles, D.; Evans, D.G.; Morrison, P.J.; Murday, V.; et al. Long-term eff ect of aspirin on cancer risk in carriers of hereditary colorectal cancer: An analysis from the CAPP2 randomised controlled trial. Lancet 2011, 378, 2081–2087. [Google Scholar] [CrossRef]
  102. Rothwell, P.M.; Fowkes, F.G.R.; Belch, J.F.F.; Ogawa, H.; Warlow, C.P.; Meade, T.W. Eff ect of daily aspirin on long-term risk of death due to cancer: Analysis of individual patient data from randomised trials. Lancet 2011, 377, 31–41. [Google Scholar] [CrossRef]
  103. Flossmann, E.; Rothwell, P.M. Effect of aspirin on long-term risk of colorectal cancer: Consistent evidence from randomised and observational studies. Lancet 2007, 369, 1603–1613. [Google Scholar] [CrossRef]
  104. Jacobs, E.J.; Thun, M.J.; Bain, E.B.; Rodriguez, C.; Henley, S.J.; Calle, E.E. A Large Cohort Study of Long-Term Daily Use of Adult-Strength Aspirin and Cancer Incidence. JNCI J. Natl. Cancer Inst. 2007, 99, 608–615. [Google Scholar] [CrossRef]
  105. Thun, M.J.; Namboodiri, M.M.; Heath, C.W.J. Aspirin use and reduced risk of fatal colon cancer. N. Engl. J. Med. 1991, 325, 1593–1596. [Google Scholar] [CrossRef]
  106. Gallo, G.; Volpe, M.; Rubattu, S. Angiotensin Receptor Blockers in the Management of Hypertension: A Real-World Perspective and Current Recommendations. Vasc. Health Risk Manag. 2022, 18, 507–515. [Google Scholar] [CrossRef]
  107. Wong, J.; Patel, R.A.; Kowey, P.R. The clinical use of angiotensin-converting enzyme inhibitors. Prog. Cardiovasc. Dis. 2004, 47, 116–130. [Google Scholar] [CrossRef]
  108. Messerli, F.H.; Bangalore, S.; Bavishi, C.; Rimoldi, S.F. Angiotensin-Converting Enzyme Inhibitors in Hypertension: To Use or Not to Use? J. Am. Coll. Cardiol. 2018, 71, 1474–1482. [Google Scholar] [CrossRef]
  109. Laghlam, D.; Jozwiak, M.; Nguyen, L.S. Renin-Angiotensin-Aldosterone System and Immunomodulation: A State-of-the-Art Review. Cells 2021, 10, 1767. [Google Scholar] [CrossRef]
  110. Paz Ocaranza, M.; Riquelme, J.A.; García, L.; Jalil, J.E.; Chiong, M.; Santos, R.A.S.; Lavandero, S. Counter-regulatory renin-angiotensin system in cardiovascular disease. Nat. Rev. Cardiol. 2020, 17, 116–129. [Google Scholar] [CrossRef]
  111. Campbell, D.J. Circulating and tissue angiotensin systems. J. Clin. Investig. 1987, 79, 1–6. [Google Scholar] [CrossRef]
  112. Vargas Vargas, R.A.; Varela Millán, J.M.; Fajardo Bonilla, E. Renin-angiotensin system: Basic and clinical aspects-A general perspective. Endocrinol. Diabetes Nutr. 2022, 69, 52–62. [Google Scholar] [CrossRef] [PubMed]
  113. Leung, P.S.; Chappell, M.C. A local pancreatic renin-angiotensin system: Endocrine and exocrine roles. Int. J. Biochem. Cell Biol. 2003, 35, 838–846. [Google Scholar] [CrossRef] [PubMed]
  114. Capettini, L.S.A.; Montecucco, F.; Mach, F.; Stergiopulos, N.; Santos, R.A.S.; da Silva, R.F. Role of renin-angiotensin system in inflammation, immunity and aging. Curr. Pharm. Des. 2012, 18, 963–970. [Google Scholar] [CrossRef] [PubMed]
  115. Wright, M.D.; Binger, K.J. Macrophage heterogeneity and renin-angiotensin system disorders. Pflugers Arch. 2017, 469, 445–454. [Google Scholar] [CrossRef] [PubMed]
  116. Rasha, F.; Ramalingam, L.; Gollahon, L.; Rahman, R.L.; Rahman, S.M.; Menikdiwela, K.; Moustaid-Moussa, N. Mechanisms linking the renin-angiotensin system, obesity, and breast cancer. Endocr. Relat. Cancer 2019, 26, R653–R672. [Google Scholar] [CrossRef] [PubMed]
  117. George, A.J.; Thomas, W.G.; Hannan, R.D. The renin-angiotensin system and cancer: Old dog, new tricks. Nat. Rev. Cancer 2010, 10, 745–759. [Google Scholar] [CrossRef] [PubMed]
  118. Fujita, M.; Hayashi, I.; Yamashina, S.; Itoman, M.; Majima, M. Blockade of angiotensin AT1a receptor signaling reduces tumor growth, angiogenesis, and metastasis. Biochem. Biophys. Res. Commun. 2002, 294, 441–447. [Google Scholar] [CrossRef] [PubMed]
  119. Miyajima, A.; Kosaka, T.; Asano, T.; Asano, T.; Seta, K.; Kawai, T.; Hayakawa, M. Angiotensin II type I antagonist prevents pulmonary metastasis of murine renal cancer by inhibiting tumor angiogenesis. Cancer Res. 2002, 62, 4176–4179. [Google Scholar] [PubMed]
  120. Attoub, S.; Gaben, A.M.; Al-Salam, S.; Al Sultan, M.A.H.; John, A.; Nicholls, M.G.; Mester, J.; Petroianu, G. Captopril as a potential inhibitor of lung tumor growth and metastasis. Ann. N. Y. Acad. Sci. 2008, 1138, 65–72. [Google Scholar] [CrossRef]
  121. Christian, J.B.; Lapane, K.L.; Hume, A.L.; Eaton, C.B.; Weinstock, M.A. Association of ACE inhibitors and angiotensin receptor blockers with keratinocyte cancer prevention in the randomized VATTC trial. J. Natl. Cancer Inst. 2008, 100, 1223–1232. [Google Scholar] [CrossRef] [PubMed]
  122. Ronquist, G.; García Rodríguez, L.A.; Ruigómez, A.; Johansson, S.; Wallander, M.A.; Frithz, G.; Svärdsudd, K. Association between Captopril, Other Antihypertensive Drugs and Risk of Prostate Cancer. Prostate 2004, 58, 50–56. [Google Scholar] [CrossRef]
  123. Wilop, S.; Von Hobe, S.; Crysandt, M.; Esser, A.; Osieka, R.; Jost, E. Impact of angiotensin I converting enzyme inhibitors and angiotensin II type 1 receptor blockers on survival in patients with advanced non-small-cell lung cancer undergoing first-line platinum-based chemotherapy. J. Cancer Res. Clin. Oncol. 2009, 135, 1429–1435. [Google Scholar] [CrossRef]
  124. Lever, A.F.; Hole, D.J.; Gillis, C.R.; McCallum, I.R.; McInnes, G.T.; MacKinnon, P.L.; Meredith, P.A.; Murray, L.S.; Reid, J.L.; Robertson, J.W.K. Do inhibitors of angiotensin-I-converting enzyme protect against risk of cancer? Lancet 1998, 352, 179–184. [Google Scholar] [CrossRef]
  125. Lanas, A.; Alcedo González, J. [Chemoprevention in adenocarcinoma of the esophagus]. Acta Gastroenterol. Latinoam. 2007, 37, 37–48. [Google Scholar] [PubMed]
  126. Cheung, K.S.; Chan, E.W.; Seto, W.K.; Wong, I.C.K.; Leung, W.K. ACE (Angiotensin-Converting Enzyme) Inhibitors/Angiotensin Receptor Blockers Are Associated with Lower Colorectal Cancer Risk: A Territory-Wide Study with Propensity Score Analysis. Hypertension 2020, 76, 968–975. [Google Scholar] [CrossRef]
  127. Liu, H.; Naxerova, K.; Pinter, M.; Incio, J.; Lee, H.; Shigeta, K.; Ho, W.W.; Crain, J.A.; Jacobson, A.; Michelakos, T.; et al. Use of Angiotensin System Inhibitors Is Associated with Immune Activation and Longer Survival in Nonmetastatic Pancreatic Ductal Adenocarcinoma. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2017, 23, 5959–5969. [Google Scholar] [CrossRef] [PubMed]
  128. Koay, E.J.; Truty, M.J.; Cristini, V.; Thomas, R.M.; Chen, R.; Chatterjee, D.; Kang, Y.; Bhosale, P.R.; Tamm, E.P.; Crane, C.H.; et al. Transport properties of pancreatic cancer describe gemcitabine delivery and response. J. Clin. Investig. 2014, 124, 1525–1536. [Google Scholar] [CrossRef] [PubMed]
  129. Zhao, Y.; Cao, J.; Melamed, A.; Worley, M.; Gockley, A.; Jones, D.; Nia, H.T.; Zhang, Y.; Stylianopoulos, T.; Kumar, A.S.; et al. Losartan treatment enhances chemotherapy efficacy and reduces ascites in ovarian cancer models by normalizing the tumor stroma. Proc. Natl. Acad. Sci. USA 2019, 116, 2210–2219. [Google Scholar] [CrossRef] [PubMed]
  130. Lewinter, C.; Nielsen, T.H.; Edfors, L.R.; Linde, C.; Bland, J.M.; LeWinter, M.; Cleland, J.G.F.; Køber, L.; Braunschweig, F.; Mansson-Broberg, A. A systematic review and meta-analysis of beta-blockers and renin–angiotensin system inhibitors for preventing left ventricular dysfunction due to anthracyclines or trastuzumab in patients with breast cancer. Eur. Heart J. 2021, 43, 2562–2569. [Google Scholar] [CrossRef] [PubMed]
  131. Kristensen, K.B.; Hicks, B.; Azoulay, L.; Pottegård, A. Use of ACE (Angiotensin-Converting Enzyme) Inhibitors and Risk of Lung Cancer: A Nationwide Nested Case-Control Study. Circ. Cardiovasc. Qual. Outcomes 2021, 14, 17–27. [Google Scholar] [CrossRef] [PubMed]
  132. Sano, M.; Yoshimasa, T.; Yagura, T.; Yamamoto, I. Non-homogeneous distribution of beta 1- and beta 2-adrenoceptors in various human tissues. Life Sci. 1993, 52, 1063–1070. [Google Scholar] [CrossRef]
  133. Bangalore, S.; Messerli, F.H.; Kostis, J.B.; Pepine, C.J. Cardiovascular protection using beta-blockers: A critical review of the evidence. J. Am. Coll. Cardiol. 2007, 50, 563–572. [Google Scholar] [CrossRef]
  134. do Vale, G.T.; Ceron, C.S.; Gonzaga, N.A.; Simplicio, J.A.; Padovan, J.C. Three Generations of β-blockers: History, Class Differences and Clinical Applicability. Curr. Hypertens. Rev. 2019, 15, 22–31. [Google Scholar] [CrossRef]
  135. Thaker, H.; Li, Y.; Gershenson, D.M.; Lutgendorf, S.; Cole, S.W. Stress Hormone—Mediated Invasion of Ovarian Cancer Cells. Clin. Cancer Res. 2011, 12, 369–375. [Google Scholar] [CrossRef]
  136. Chakroborty, D.; Sarkar, C.; Basu, B.; Dasgupta, P.S.; Basu, S. Catecholamines regulate tumor angiogenesis. Cancer Res. 2009, 69, 3727–3730. [Google Scholar] [CrossRef] [PubMed]
  137. Krizanova, O.; Babula, P.; Pacak, K. Stress, catecholaminergic system and cancer. Stress 2016, 19, 419–428. [Google Scholar] [CrossRef]
  138. Lutgendorf, S.K.; Cole, S.; Costanzo, E.; Bradley, S.; Coffin, J.; Jabbari, S.; Rainwater, K.; Ritchie, J.M.; Yang, M.; Sood, A.K. Stress-related mediators stimulate vascular endothelial growth factor secretion by two ovarian cancer cell lines. Clin. Cancer Res. 2003, 9, 4514–4521. [Google Scholar]
  139. Perron, L.; Bairati, I.; Harel, F.; Meyer, F. Antihypertensive drug use and the risk of prostate cancer (Canada). Cancer Causes Control 2004, 15, 535–541. [Google Scholar] [CrossRef]
  140. Algazi, M.; Plu-Bureau, G.; Flahault, A.; Dondon, M.G.; Lê, M.G. [Could treatments with beta-blockers be associated with a reduction in cancer risk?]. Rev. Epidemiol. Sante Publique 2004, 52, 53–65. [Google Scholar] [CrossRef]
  141. Bangalore, S.; Kumar, S.; Kjeldsen, S.E.; Makani, H.; Grossman, E.; Wetterslev, J.; Gupta, A.K.; Sever, P.S.; Gluud, C.; Messerli, F.H. Antihypertensive drugs and risk of cancer: Network meta-analyses and trial sequential analyses of 324,168 participants from randomised trials. Lancet. Oncol. 2011, 12, 65–82. [Google Scholar] [CrossRef]
  142. Rodriguez, C.; Jacobs, E.J.; Deka, A.; Patel, A.V.; Bain, E.B.; Thun, M.J.; Calle, E.E. Use of blood-pressure-lowering medication and risk of prostate cancer in the Cancer Prevention Study II Nutrition Cohort. Cancer Causes Control 2009, 20, 671–679. [Google Scholar] [CrossRef]
  143. Elliott, W.J.; Ram, C.V.S. Calcium channel blockers. J. Clin. Hypertens. 2011, 13, 687–689. [Google Scholar] [CrossRef] [PubMed]
  144. Mason, R.P. Calcium channel blockers, apoptosis and cancer: Is there a biologic relationship? J. Am. Coll. Cardiol. 1999, 34, 1857–1866. [Google Scholar] [CrossRef]
  145. Taylor, J.T.; Zeng, X.-B.; Pottle, J.E.; Lee, K.; Wang, A.R.; Yi, S.G.; Scruggs, J.A.S.; Sikka, S.S.; Li, M. Calcium signaling and T-type calcium channels in cancer cell cycling. World J. Gastroenterol. 2008, 14, 4984–4991. [Google Scholar] [CrossRef] [PubMed]
  146. Panner, A.; Wurster, R.D. T-type calcium channels and tumor proliferation. Cell Calcium 2006, 40, 253–259. [Google Scholar] [CrossRef] [PubMed]
  147. Li, W.; Zhang, S.-L.; Wang, N.; Zhang, B.-B.; Li, M. Blockade of T-type Ca(2+) channels inhibits human ovarian cancer cell proliferation. Cancer Investig. 2011, 29, 339–346. [Google Scholar] [CrossRef] [PubMed]
  148. Buchanan, P.J.; McCloskey, K.D. Ca(V) channels and cancer: Canonical functions indicate benefits of repurposed drugs as cancer therapeutics. Eur. Biophys. J. 2016, 45, 621–633. [Google Scholar] [CrossRef] [PubMed]
  149. Chen, R.; Zeng, X.; Zhang, R.; Huang, J.; Kuang, X.; Yang, J.; Liu, J.; Tawfik, O.; Thrasher, J.B.; Li, B. Cav1.3 channel α1D protein is overexpressed and modulates androgen receptor transactivation in prostate cancers. Urol. Oncol. 2014, 32, 524–536. [Google Scholar] [CrossRef]
  150. Wang, C.-Y.; Lai, M.-D.; Phan, N.N.; Sun, Z.; Lin, Y.-C. Meta-Analysis of Public Microarray Datasets Reveals Voltage-Gated Calcium Gene Signatures in Clinical Cancer Patients. PLoS ONE 2015, 10, e0125766. [Google Scholar] [CrossRef] [PubMed]
  151. Grundy, S.M.; Stone, N.J.; Bailey, A.L.; Beam, C.; Birtcher, K.K.; Blumenthal, R.S.; Braun, L.T.; de Ferranti, S.; Faiella-Tommasino, J.; Forman, D.E.; et al. 2018 AHA/ACC/AACVPR/AAPA/ABC/ACPM/ADA/AGS/APhA/ASPC/NLA/PCNA Guideline on the Management of Blood Cholesterol: A Report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines. Circulation 2019, 139, e1082–e1143. [Google Scholar] [CrossRef] [PubMed]
  152. Stancu, C.; Sima, A. Statins: Mechanism of action and effects. J. Cell. Mol. Med. 2001, 5, 378–387. [Google Scholar] [CrossRef]
  153. Chimento, A.; Casaburi, I.; Avena, P.; Trotta, F.; De Luca, A.; Rago, V.; Pezzi, V.; Sirianni, R. Cholesterol and Its Metabolites in Tumor Growth: Therapeutic Potential of Statins in Cancer Treatment. Front. Endocrinol. 2018, 9, 807. [Google Scholar] [CrossRef]
  154. Boudreau, D.M.; Yu, O.; Johnson, J. Statin Use and Cancer Risk: A Comprehensive Review. Expert Opin. Drug Saf. 2010, 9, 603. [Google Scholar] [CrossRef] [PubMed]
  155. Vanova, K.; Boukalova, S.; Gbelcova, H.; Muchova, L.; Neuzil, J.; Gurlich, R.; Ruml, T.; Vitek, L. Heme oxygenase is not involved in the anti-proliferative effects of statins on pancreatic cancer cells. BMC Cancer 2016, 16, 1–11. [Google Scholar] [CrossRef] [PubMed]
  156. Chen, Y.; Li, L.B.; Zhang, J.; Tang, D.P.; Wei, J.J.; Zhuang, Z.H. Simvastatin, but not pravastatin, inhibits the proliferation of esophageal adenocarcinoma and squamous cell carcinoma cells: A cell-molecular study. Lipids Health Dis. 2018, 17, 1–7. [Google Scholar] [CrossRef] [PubMed]
  157. Jones, H.M.; Fang, Z.; Sun, W.; Clark, L.H.; Stine, J.E.; Tran, A.Q.; Sullivan, S.A.; Gilliam, T.P.; Zhou, C.; Bae-Jump, V.L. Atorvastatin exhibits anti-tumorigenic and anti-metastatic effects in ovarian cancer in vitro. Am. J. Cancer Res. 2017, 7, 2478. [Google Scholar] [PubMed]
  158. Wang, S.T.; Ho, H.J.; Lin, J.T.; Shieh, J.J.; Wu, C.Y. Simvastatin-induced cell cycle arrest through inhibition of STAT3/SKP2 axis and activation of AMPK to promote p27 and p21 accumulation in hepatocellular carcinoma cells. Cell Death Dis. 2017, 8, e2626. [Google Scholar] [CrossRef] [PubMed]
  159. Schointuch, M.N.; Gilliam, T.P.; Stine, J.E.; Han, X.; Zhou, C.; Gehrig, P.A.; Kim, K.; Bae-Jump, V.L. Simvastatin, an HMG-CoA reductase inhibitor, exhibits anti-metastatic and anti-tumorigenic effects in endometrial cancer. Gynecol. Oncol. 2014, 134, 346. [Google Scholar] [CrossRef] [PubMed]
  160. Yu, O.; Eberg, M.; Benayoun, S.; Aprikian, A.; Batist, G.; Suissa, S.; Azoulay, L. Use of statins and the risk of death in patients with prostate cancer. J. Clin. Oncol. 2014, 32, 5–11. [Google Scholar] [CrossRef] [PubMed]
  161. Tan, N.; Klein, E.A.; Li, J.; Moussa, A.S.; Jones, J.S. Statin use and risk of prostate cancer in a population of men who underwent biopsy. J. Urol. 2011, 186, 86–90. [Google Scholar] [CrossRef]
  162. Murtola, T.J.; Tammela, T.L.J.; Lahtela, J.; Auvinen, A. Cholesterol-Lowering Drugs and Prostate Cancer Risk: A Population-based Case-Control Study. Cancer Epidemiol. Prev. Biomark. 2007, 16, 2226–2232. [Google Scholar] [CrossRef]
  163. Bansal, D.; Undela, K.; D’Cruz, S.; Schifano, F. Statin use and risk of prostate cancer: A meta-analysis of observational studies. PLoS ONE 2012, 7, e46691. [Google Scholar] [CrossRef]
  164. Prabhu, N.; Kapur, N.; Catalona, W.; Leikin, R.; Helenowski, I.; Jovanovich, B.; Gurley, M.; Okwuosa, T.M.; Kuzel, T.M. Statin use and risk of prostate cancer biochemical recurrence after radical prostatectomy. Urol. Oncol. Semin. Orig. Investig. 2021, 39, 130.e9–130.e15. [Google Scholar] [CrossRef] [PubMed]
  165. Scosyrev, E.; Tobis, S.; Donsky, H.; Wu, G.; Joseph, J.; Rashid, H.; Messing, E. Statin use and the risk of biochemical recurrence of prostate cancer after definitive local therapy: A meta-analysis of eight cohort studies. BJU Int. 2013, 111, E71–E77. [Google Scholar] [CrossRef]
  166. Allott, E.H.; Howard, L.E.; Cooperberg, M.R.; Kane, C.J.; Aronson, W.J.; Terris, M.K.; Amling, C.L.; Freedland, S.J. Postoperative statin use and risk of biochemical recurrence following radical prostatectomy: Results from the Shared Equal Access Regional Cancer Hospital (SEARCH) database. BJU Int. 2014, 114, 661–666. [Google Scholar] [CrossRef] [PubMed]
  167. Singh, P.P.; Singh, S. Statins are associated with reduced risk of gastric cancer: A systematic review and meta-analysis. Ann. Oncol. 2013, 24, 1721–1730. [Google Scholar] [CrossRef] [PubMed]
  168. Lakha, F.; Theodoratou, E.; Farrington, S.M.; Tenesa, A.; Cetnarskyj, R.; Din, F.V.N.; Porteous, M.E.; Dunlop, M.G.; Campbell, H. Statin use and association with colorectal cancer survival and risk: Case control study with prescription data linkage. BMC Cancer 2012, 12, 1–10. [Google Scholar] [CrossRef] [PubMed]
  169. Lee, J.E.; Baba, Y.; Ng, K.; Giovannucci, E.; Fuchs, C.S.; Ogino, S.; Chan, A.T. Statin Use and Colorectal Cancer Risk According to Molecular Subtypes in Two Large Prospective Cohort Studies. Cancer Prev. Res. 2011, 4, 1808–1815. [Google Scholar] [CrossRef]
  170. Ahern, T.P.; Pedersen, L.; Tarp, M.; Cronin-Fenton, D.P.; Garne, J.P.; Silliman, R.A.; Sørensen, H.T.; Lash, T.L. Statin Prescriptions and Breast Cancer Recurrence Risk: A Danish Nationwide Prospective Cohort Study. JNCI J. Natl. Cancer Inst. 2011, 103, 1461–1468. [Google Scholar] [CrossRef]
  171. Brewer, T.M.; Masuda, H.; Liu, D.D.; Shen, Y.; Liu, P.; Iwamoto, T.; Kai, K.; Barnett, C.M.; Woodward, W.A.; Reuben, J.M.; et al. Statin use in primary inflammatory breast cancer: A cohort study. Br. J. Cancer 2013, 109, 318–324. [Google Scholar] [CrossRef]
  172. Nickels, S.; Vrieling, A.; Seibold, P.; Heinz, J.; Obi, N.; Flesch-Janys, D.; Chang-Claude, J. Mortality and Recurrence Risk in Relation to the Use of Lipid-Lowering Drugs in a Prospective Breast Cancer Patient Cohort. PLoS ONE 2013, 8, 75088. [Google Scholar] [CrossRef]
  173. Nowakowska, M.K.; Lei, X.; Thompson, M.T.; Shaitelman, S.F.; Wehner, M.R.; Woodward, W.A.; Giordano, S.H.; Nead, K.T. Association of statin use with clinical outcomes in patients with triple-negative breast cancer. Cancer 2021, 127, 4142–4150. [Google Scholar] [CrossRef]
  174. Van Wyhe, R.D.; Rahal, O.M.; Woodward, W.A. Effect of statins on breast cancer recurrence and mortality: A review. Breast Cancer 2017, 9, 559. [Google Scholar] [CrossRef] [PubMed]
  175. Jung, H.H.; Lee, S.H.; Kim, J.Y.; Ahn, J.S.; Park, Y.H.; Im, Y.H. Statins affect ETS1-overexpressing triple-negative breast cancer cells by restoring DUSP4 deficiency. Sci. Rep. 2016, 6, 33035. [Google Scholar] [CrossRef] [PubMed]
  176. Za, Y.C.C.E.; Wu, S.; Huang, L.; Buquet, C.; Shen, R.; Nzalez, B.S.G.; Latorre, E.A.G.; Boyer, O.; Varin, R.; Jiménez-Zamudio, L.A.; et al. Synergistic promoting effects of pentoxifylline and simvastatin on the apoptosis of triple-negative MDA-MB-231 breast cancer cells. Int. J. Oncol. 2018, 52, 1246–1254. [Google Scholar]
  177. Lin, Z.; Zhang, Z.; Jiang, X.; Kou, X.; Bao, Y.; Liu, H.; Sun, F.; Ling, S.; Qin, N.; Jiang, L.; et al. Mevastatin blockade of autolysosome maturation stimulates LBH589-induced cell death in triple-negative breast cancer cells. Oncotarget 2017, 8, 17833. [Google Scholar] [CrossRef] [PubMed]
  178. Jang, H.J.; Kim, H.S.; Kim, J.H.; Lee, J. The Effect of Statin Added to Systemic Anticancer Therapy: A Meta-Analysis of Randomized, Controlled Trials. J. Clin. Med. 2018, 7, 325. [Google Scholar] [CrossRef] [PubMed]
  179. Neilan, T.G.; Quinaglia, T.; Onoue, T.; Mahmood, S.S.; Drobni, Z.D.; Gilman, H.K.; Smith, A.; Heemelaar, J.C.; Brahmbhatt, P.; Ho, J.S.; et al. Atorvastatin for Anthracycline-Associated Cardiac Dysfunction: The STOP-CA Randomized Clinical Trial. JAMA 2023, 330, 528–536. [Google Scholar] [CrossRef] [PubMed]
  180. Maxwell, K.N.; Soccio, R.E.; Duncan, E.M.; Sehayek, E.; Breslow, J.L. Novel putative SREBP and LXR target genes identified by microarray analysis in liver of cholesterol-fed mice. J. Lipid Res. 2003, 44, 2109–2119. [Google Scholar] [CrossRef] [PubMed]
  181. Seidah, N.G.; Benjannet, S.; Wickham, L.; Marcinkiewicz, J.; Jasmin, S.B.; Stifani, S.; Basak, A.; Prat, A.; Chrétien, M. The secretory proprotein convertase neural apoptosis-regulated convertase 1 (NARC-1): Liver regeneration and neuronal differentiation. Proc. Natl. Acad. Sci. USA 2003, 100, 928–933. [Google Scholar] [CrossRef] [PubMed]
  182. Abifadel, M.; Varret, M.; Rabès, J.-P.; Allard, D.; Ouguerram, K.; Devillers, M.; Cruaud, C.; Benjannet, S.; Wickham, L.; Erlich, D.; et al. Mutations in PCSK9 cause autosomal dominant hypercholesterolemia. Nat. Genet. 2003, 34, 154–156. [Google Scholar] [CrossRef]
  183. Ferri, N.; Tibolla, G.; Pirillo, A.; Cipollone, F.; Mezzetti, A.; Pacia, S.; Corsini, A.; Catapano, A.L. Proprotein convertase subtilisin kexin type 9 (PCSK9) secreted by cultured smooth muscle cells reduces macrophages LDLR levels. Atherosclerosis 2012, 220, 381–386. [Google Scholar] [CrossRef]
  184. Maxwell, K.N.; Breslow, J.L. Adenoviral-mediated expression of Pcsk9 in mice results in a low-density lipoprotein receptor knockout phenotype. Proc. Natl. Acad. Sci. USA 2004, 101, 7100–7105. [Google Scholar] [CrossRef] [PubMed]
  185. McNutt, M.C.; Kwon, H.J.; Chen, C.; Chen, J.R.; Horton, J.D.; Lagace, T.A. Antagonism of secreted PCSK9 increases low density lipoprotein receptor expression in HepG2 cells. J. Biol. Chem. 2009, 284, 10561–10570. [Google Scholar] [CrossRef] [PubMed]
  186. Surdo, P.L.; Bottomley, M.J.; Calzetta, A.; Settembre, E.C.; Cirillo, A.; Pandit, S.; Ni, Y.G.; Hubbard, B.; Sitlani, A.; Carfí, A. Mechanistic implications for LDL receptor degradation from the PCSK9/LDLR structure at neutral pH. EMBO Rep. 2011, 12, 1300. [Google Scholar] [CrossRef]
  187. Horton, J.D.; Cohen, J.C.; Hobbs, H.H. PCSK9: A convertase that coordinates LDL catabolism. J. Lipid Res. 2009, 50, S172–S177. [Google Scholar] [CrossRef] [PubMed]
  188. Sabatine, M.S. PCSK9 inhibitors: Clinical evidence and implementation. Nat. Rev. Cardiol. 2018, 16, 155–165. [Google Scholar] [CrossRef] [PubMed]
  189. Mcgovern, T.J. Tertiary Pharmacology/Toxicology Review; US Food and Drug Administration: Silver Spring, MD, USA, 2015. [Google Scholar]
  190. Rosenson, R.S.; Hegele, R.A.; Fazio, S.; Cannon, C.P. The Evolving Future of PCSK9 Inhibitors. J. Am. Coll. Cardiol. 2018, 72, 314–329. [Google Scholar] [CrossRef] [PubMed]
  191. Stein, E.A.; Mellis, S.; Yancopoulos, G.D.; Stahl, N.; Logan, D.; Smith, W.B.; Lisbon, E.; Gutierrez, M.; Webb, C.; Wu, R.; et al. Effect of a monoclonal antibody to PCSK9 on LDL cholesterol. N. Engl. J. Med. 2012, 366, 1108–1118. [Google Scholar] [CrossRef] [PubMed]
  192. Manniello, M.; Pisano, M. Alirocumab (Praluent): First in the New Class of PCSK9 Inhibitors. Pharm. Ther. 2016, 41, 28. [Google Scholar]
  193. Vega, A. Center for Drug Evaluation and Research Application Number: 125559orig1s000 Summary Review; US Food and Drug Administration: Silver Spring, MD, USA, 2014. [Google Scholar]
  194. Roth, E.M.; McKenney, J.M.; Hanotin, C.; Asset, G.; Stein, E.A. Atorvastatin with or without an antibody to PCSK9 in primary hypercholesterolemia. N. Engl. J. Med. 2012, 367, 1891–1900. [Google Scholar] [CrossRef] [PubMed]
  195. Sullivan, D.; Olsson, A.G.; Scott, R.; Kim, J.B.; Xue, A.; Gebski, V.; Wasserman, S.M.; Stein, E.A. Effect of a monoclonal antibody to PCSK9 on low-density lipoprotein cholesterol levels in statin-intolerant patients: The GAUSS randomized trial. JAMA 2012, 308, 2497–2506. [Google Scholar] [CrossRef]
  196. Giugliano, R.P.; Desai, N.R.; Kohli, P.; Rogers, W.J.; Somaratne, R.; Huang, F.; Liu, T.; Mohanavelu, S.; Hoffman, E.B.; McDonald, S.T.; et al. Efficacy, safety, and tolerability of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 in combination with a statin in patients with hypercholesterolaemia (LAPLACE-TIMI 57): A randomised, placebo-controlled, dose-ranging, phase 2 study. Lancet 2012, 380, 2007–2017. [Google Scholar] [CrossRef] [PubMed]
  197. Koren, M.J.; Scott, R.; Kim, J.B.; Knusel, B.; Liu, T.; Lei, L.; Bolognese, M.; Wasserman, S.M. Efficacy, safety, and tolerability of a monoclonal antibody to proprotein convertase subtilisin/kexin type 9 as monotherapy in patients with hypercholesterolaemia (MENDEL): A randomised, double-blind, placebo-controlled, phase 2 study. Lancet 2012, 380, 1995–2006. [Google Scholar] [CrossRef] [PubMed]
  198. Raal, F.J.; Stein, E.; Dufour, R.; Turner, T.; Civeira, F.; Burgess, L.; Langslet, G.; Scott, R.; Olsson, A.G.; Sullivan, D.; et al. PCSK9 inhibition with evolocumab (AMG 145) in heterozygous familial hypercholesterolaemia (RUTHERFORD-2): A randomised, double-blind, placebo-controlled trial. Lancet 2015, 385, 331–340. [Google Scholar] [CrossRef] [PubMed]
  199. Koren, M.J.; Lundqvist, P.; Bolognese, M.; Neutel, J.M.; Monsalvo, M.L.; Yang, J.; Kim, J.B.; Scott, R.; Wasserman, S.M.; Bays, H. Anti-PCSK9 monotherapy for hypercholesterolemia: The MENDEL-2 randomized, controlled phase III clinical trial of evolocumab. J. Am. Coll. Cardiol. 2014, 63, 2531–2540. [Google Scholar] [CrossRef] [PubMed]
  200. Stroes, E.; Colquhoun, D.; Sullivan, D.; Civeira, F.; Rosenson, R.S.; Watts, G.F.; Bruckert, E.; Cho, L.; Dent, R.; Knusel, B.; et al. Anti-PCSK9 antibody effectively lowers cholesterol in patients with statin intolerance: The GAUSS-2 randomized, placebo-controlled phase 3 clinical trial of evolocumab. J. Am. Coll. Cardiol. 2014, 63, 2541–2548. [Google Scholar] [CrossRef] [PubMed]
  201. Everett, B.M.; Smith, R.J.; Hiatt, W.R. Reducing LDL with PCSK9 Inhibitors—The Clinical Benefit of Lipid Drugs. N. Engl. J. Med. 2015, 373, 1588–1591. [Google Scholar] [CrossRef] [PubMed]
  202. Robinson, J.G.; Farnier, M.; Krempf, M.; Bergeron, J.; Luc, G.; Averna, M.; Stroes, E.S.; Langslet, G.; Raal, F.J.; El Shahawy, M.; et al. Efficacy and Safety of Alirocumab in Reducing Lipids and Cardiovascular Events. N. Engl. J. Med. 2015, 372, 1489–1499. [Google Scholar] [CrossRef] [PubMed]
  203. Blom, D.J.; Hala, T.; Bolognese, M.; Lillestol, M.J.; Toth, P.D.; Burgess, L.; Ceska, R.; Roth, E.; Koren, M.J.; Ballantyne, C.M.; et al. A 52-Week Placebo-Controlled Trial of Evolocumab in Hyperlipidemia. N. Engl. J. Med. 2014, 370, 1809–1819. [Google Scholar] [CrossRef]
  204. Yang, W.; Bai, Y.; Xiong, Y.; Zhang, J.; Chen, S.; Zheng, X.; Meng, X.; Li, L.; Wang, J.; Xu, C.; et al. Potentiating the antitumour response of CD8(+) T cells by modulating cholesterol metabolism. Nature 2016, 531, 651–655. [Google Scholar] [CrossRef]
  205. Ma, X.; Bi, E.; Huang, C.; Lu, Y.; Xue, G.; Guo, X.; Wang, A.; Yang, M.; Qian, J.; Dong, C.; et al. Cholesterol negatively regulates IL-9-producing CD8+ T cell differentiation and antitumor activity. J. Exp. Med. 2018, 215, 1555–1569. [Google Scholar] [CrossRef]
  206. Naslavsky, N.; Weigert, R.; Donaldson, J.G. Characterization of a Nonclathrin Endocytic Pathway: Membrane Cargo and Lipid Requirements. Mol. Biol. Cell 2004, 15, 3542. [Google Scholar] [CrossRef] [PubMed]
  207. Xu, X.; Cui, Y.; Cao, L.; Zhang, Y.; Yin, Y.; Hu, X. PCSK9 regulates apoptosis in human lung adenocarcinoma A549 cells via endoplasmic reticulum stress and mitochondrial signaling pathways. Exp. Ther. Med. 2017, 13, 1993–1999. [Google Scholar] [CrossRef] [PubMed]
  208. Ito, M.; Hiwasa, T.; Oshima, Y.; Yajima, S.; Suzuki, T.; Nanami, T.; Sumazaki, M.; Shiratori, F.; Funahashi, K.; Li, S.-Y.; et al. Association of Serum Anti-PCSK9 Antibody Levels with Favorable Postoperative Prognosis in Esophageal Cancer. Front. Oncol. 2021, 11, 708039. [Google Scholar] [CrossRef] [PubMed]
  209. Bonaventura, A.; Grossi, F.; Carbone, F.; Vecchié, A.; Minetti, S.; Bardi, N.; Elia, E.; Ansaldo, A.M.; Ferrara, D.; Rijavec, E.; et al. Serum PCSK9 levels at the second nivolumab cycle predict overall survival in elderly patients with NSCLC: A pilot study. Cancer Immunol. Immunother. 2019, 68, 1351–1358. [Google Scholar] [CrossRef] [PubMed]
  210. Momtazi-Borojeni, A.A.; Nik, M.E.; Jaafari, M.R.; Banach, M.; Sahebkar, A. Effects of immunization against PCSK9 in an experimental model of breast cancer. Arch. Med. Sci. 2019, 15, 570. [Google Scholar] [CrossRef] [PubMed]
  211. Momtazi-Borojeni, A.A.; Nik, M.E.; Jaafari, M.R.; Banach, M.; Sahebkar, A. Potential anti-tumor effect of a nanoliposomal antiPCSK9 vaccine in mice bearing colorectal cancer. Arch. Med. Sci. 2019, 15, 559–569. [Google Scholar] [CrossRef] [PubMed]
  212. Nowak, C.; Ärnlöv, J. A Mendelian randomization study of the effects of blood lipids on breast cancer risk. Nat. Commun. 2018, 9, 3957. [Google Scholar] [CrossRef] [PubMed]
  213. Tamargo, J. Heart Failure and Arrhythmias Sodium—Glucose Cotransporter 2 Inhibitors in Heart Failure: Potential Mechanisms of Action, Adverse Effects and Future Developments Heart Failure and Arrhythmias. Eur Cardiol. 2019, 14, 23–32. [Google Scholar] [CrossRef] [PubMed]
  214. Fathi, A.; Vickneson, K.; Singh, J.S. SGLT2-inhibitors; more than just glycosuria and diuresis. Heart Fail. Rev. 2021, 26, 623–642. [Google Scholar] [CrossRef]
  215. Cowie, M.R. SGLT2 inhibitors: Mechanisms of cardiovascular benefit beyond glycaemic control. Nat. Rev. Cardiol. 2020, 17, 761–772. [Google Scholar] [CrossRef]
  216. McMurray, J.J.V.; Solomon, S.D.; Inzucchi, S.E.; Køber, L.; Kosiborod, M.N.; Martinez, F.A.; Ponikowski, P.; Sabatine, M.S.; Anand, I.S.; Bělohlávek, J.; et al. Dapagliflozin in Patients with Heart Failure and Reduced Ejection Fraction. N. Engl. J. Med. 2019, 381, 1995–2008. [Google Scholar] [CrossRef] [PubMed]
  217. Packer, M.; Anker, S.D.; Butler, J.; Filippatos, G.; Pocock, S.J.; Carson, P.; Januzzi, J.; Verma, S.; Tsutsui, H.; Brueckmann, M.; et al. Cardiovascular and Renal Outcomes with Empagliflozin in Heart Failure. N. Engl. J. Med. 2020, 383, 1413–1424. [Google Scholar] [CrossRef] [PubMed]
  218. Anker, S.D.; Butler, J.; Filippatos, G.; Ferreira, J.P.; Bocchi, E.; Böhm, M.; Brunner-La Rocca, H.-P.; Choi, D.-J.; Chopra, V.; Chuquiure-Valenzuela, E.; et al. Empagliflozin in Heart Failure with a Preserved Ejection Fraction. N. Engl. J. Med. 2021, 385, 1451–1461. [Google Scholar] [CrossRef] [PubMed]
  219. Neal, B.; Perkovic, V.; Mahaffey, K.W.; de Zeeuw, D.; Fulcher, G.; Erondu, N.; Shaw, W.; Law, G.; Desai, M.; Matthews, D.R. Canagliflozin and Cardiovascular and Renal Events in Type 2 Diabetes. N. Engl. J. Med. 2017, 377, 644–657. [Google Scholar] [CrossRef]
  220. Kondo, H.; Takahashi, N. Reduced hospitalization for heart failure using anti-diabetic drug dapagliflozin: Implications of DECLARE-TIMI 58 for the basic science community. Cardiovasc. Res. 2019, 115, e54–e57. [Google Scholar] [CrossRef] [PubMed]
  221. Wanner, C.; Lachin, J.M.; Inzucchi, S.E.; Fitchett, D.; Mattheus, M.; George, J.; Woerle, H.J.; Broedl, U.C.; von Eynatten, M.; Zinman, B. Empagliflozin and Clinical Outcomes in Patients With Type 2 Diabetes Mellitus, Established Cardiovascular Disease, and Chronic Kidney Disease. Circulation 2018, 137, 119–129. [Google Scholar] [CrossRef] [PubMed]
  222. Naznin, F.; Sakoda, H.; Okada, T.; Tsubouchi, H.; Waise, T.M.Z. Canagli fl ozin, a sodium glucose cotransporter 2 inhibitor, attenuates obesity-induced in fl ammation in the nodose ganglion, hypothalamus, and skeletal muscle of mice. Eur. J. Pharmacol. 2017, 794, 37–44. [Google Scholar] [CrossRef] [PubMed]
  223. Yagi, S.; Hirata, Y.; Ise, T.; Yamada, H.; Fukuda, D.; Matsuura, T.; Yamaguchi, K.; Tobiume, T.; Soeki, T.; Wakatsuki, T.; et al. Canagliflozin reduces epicardial fat in patients with type 2 diabetes mellitus. Diabetol. Metab. Syndr. 2017, 9, 78. [Google Scholar] [CrossRef]
  224. Ferrannini, E.; Mark, M.; Mayoux, E. CV Protection in the EMPA-REG OUTCOME Trial: A “Thrifty Substrate” Hypothesis. Diabetes Care 2016, 39, 1108–1114. [Google Scholar] [CrossRef]
  225. Rajeev, S.P.; Cuthbertson, D.J.; Wilding, J.P.H. Energy balance and metabolic changes with sodium-glucose co-transporter 2 inhibition. Diabetes. Obes. Metab. 2016, 18, 125–134. [Google Scholar] [CrossRef]
  226. Solini, A.; Giannini, L.; Seghieri, M.; Vitolo, E.; Taddei, S.; Ghiadoni, L.; Bruno, R.M. Dapagliflozin acutely improves endothelial dysfunction, reduces aortic stiffness and renal resistive index in type 2 diabetic patients: A pilot study. Cardiovasc. Diabetol. 2017, 16, 138. [Google Scholar] [CrossRef]
  227. Lambers Heerspink, H.J.; de Zeeuw, D.; Wie, L.; Leslie, B.; List, J. Dapagliflozin a glucose-regulating drug with diuretic properties in subjects with type 2 diabetes. Diabetes. Obes. Metab. 2013, 15, 853–862. [Google Scholar] [CrossRef] [PubMed]
  228. Dutka, M.; Bobiński, R.; Francuz, T.; Garczorz, W.; Zimmer, K.; Ilczak, T.; Ćwiertnia, M.; Hajduga, M.B. SGLT-2 Inhibitors in Cancer Treatment-Mechanisms of Action and Emerging New Perspectives. Cancers 2022, 14, 5811. [Google Scholar] [CrossRef] [PubMed]
  229. Dicembrini, I.; Nreu, B.; Mannucci, E.; Monami, M. Sodium-glucose co-transporter-2 (SGLT-2) inhibitors and cancer: A meta-analysis of randomized controlled trials. Diabetes. Obes. Metab. 2019, 21, 1871–1877. [Google Scholar] [CrossRef] [PubMed]
  230. Ptaszynska, A.; Cohen, S.M.; Messing, E.M.; Reilly, T.P.; Johnsson, E.; Johnsson, K. Assessing Bladder Cancer Risk in Type 2 Diabetes Clinical Trials: The Dapagliflozin Drug Development Program as a “Case Study”. Diabetes Ther. Res. Treat. Educ. Diabetes Relat. Disord. 2015, 6, 357–375. [Google Scholar] [CrossRef] [PubMed]
  231. Basak, D.; Gamez, D.; Deb, S. SGLT2 Inhibitors as Potential Anticancer Agents. Biomedicines 2023, 11, 1867. [Google Scholar] [CrossRef] [PubMed]
  232. Wu, W.; Zhang, Z.; Jing, D.; Huang, X.; Ren, D.; Shao, Z.; Zhang, Z. SGLT2 inhibitor activates the STING/IRF3/IFN-β pathway and induces immune infiltration in osteosarcoma. Cell Death Dis. 2022, 13, 523. [Google Scholar] [CrossRef] [PubMed]
  233. Zhou, J.; Zhu, J.; Yu, S.-J.; Ma, H.-L.; Chen, J.; Ding, X.-F.; Chen, G.; Liang, Y.; Zhang, Q. Sodium-glucose co-transporter-2 (SGLT-2) inhibition reduces glucose uptake to induce breast cancer cell growth arrest through AMPK/mTOR pathway. Biomed. Pharmacother. 2020, 132, 110821. [Google Scholar] [CrossRef] [PubMed]
  234. Hung, M.H.; Chen, Y.L.; Chen, L.J.; Chu, P.Y.; Hsieh, F.S.; Tsai, M.H.; Shih, C.T.; Chao, T.I.; Huang, C.Y.; Chen, K.F. Canagliflozin inhibits growth of hepatocellular carcinoma via blocking glucose-influx-induced β-catenin activation. Cell Death Dis. 2019, 10, 420. [Google Scholar] [CrossRef]
  235. Okada, J.; Yamada, E.; Saito, T.; Yokoo, H.; Osaki, A.; Shimoda, Y.; Ozawa, A.; Nakajima, Y.; Pessin, J.E.; Okada, S.; et al. Dapagliflozin Inhibits Cell Adhesion to Collagen I and IV and Increases Ectodomain Proteolytic Cleavage of DDR1 by Increasing ADAM10 Activity. Molecules 2020, 25, 495. [Google Scholar] [CrossRef]
  236. Osataphan, S.; Macchi, C.; Singhal, G.; Chimene-Weiss, J.; Sales, V.; Kozuka, C.; Dreyfuss, J.M.; Pan, H.; Tangcharoenpaisan, Y.; Morningstar, J.; et al. SGLT2 inhibition reprograms systemic metabolism via FGF21-dependent and -independent mechanisms. JCI Insight 2019, 4, e123130. [Google Scholar] [CrossRef] [PubMed]
  237. Pearson, M.J.; Smart, N.A. Exercise therapy and autonomic function in heart failure patients: A systematic review and meta-analysis. Hear. Fail. Rev. 2018, 23, 91–108. [Google Scholar] [CrossRef] [PubMed]
  238. Ashor, A.W.; Lara, J.; Siervo, M.; Celis-Morales, C.; Oggioni, C.; Jakovljevic, D.G.; Mathers, J.C. Exercise Modalities and Endothelial Function: A Systematic Review and Dose–Response Meta-Analysis of Randomized Controlled Trials. Sport. Med. 2014, 45, 279–296. [Google Scholar] [CrossRef] [PubMed]
  239. Asimakis, G.K.; Inners-McBride, K.; Medellin, G.; Conti, V.R. Ischemic preconditioning attenuates acidosis and postischemic dysfunction in isolated rat heart. Am. J. Physiol. 1992, 263, H887–H894. [Google Scholar] [CrossRef] [PubMed]
  240. Cohen, M.V.; Yang, X.M.; Downey, J.M. Smaller infarct after preconditioning does not predict extent of early functional improvement of reperfused heart. Am. J. Physiol. 1999, 277, H1754–H1761. [Google Scholar] [CrossRef] [PubMed]
  241. Parratt, J.; Vegh, A. Pronounced antiarrhythmic effects of ischemic preconditioning. Cardioscience 1994, 5, 9–18. [Google Scholar] [PubMed]
  242. Shiki, K.; Hearse, D.J. Preconditioning of ischemic myocardium: Reperfusion-induced arrhythmias. Am. J. Physiol. 1987, 253, H1470–H1476. [Google Scholar] [CrossRef]
  243. Fiuza-Luces, C.; Santos-Lozano, A.; Joyner, M.; Carrera-Bastos, P.; Picazo, O.; Zugaza, J.L.; Izquierdo, M.; Ruilope, L.M.; Lucia, A. Exercise benefits in cardiovascular disease: Beyond attenuation of traditional risk factors. Nat. Rev. Cardiol. 2018, 15, 731–743. [Google Scholar] [CrossRef]
  244. Cormie, P.; Zopf, E.M.; Zhang, X.; Schmitz, K.H. The Impact of Exercise on Cancer Mortality, Recurrence, and Treatment-Related Adverse Effects. Epidemiol. Rev. 2017, 39, 71–92. [Google Scholar] [CrossRef]
  245. Hojman, P.; Gehl, J.; Christensen, J.F.; Pedersen, B.K. Molecular Mechanisms Linking Exercise to Cancer Prevention and Treatment. Cell Metab. 2018, 27, 10–21. [Google Scholar] [CrossRef]
  246. Moore, S.C.; Lee, I.M.; Weiderpass, E.; Campbell, P.T.; Sampson, J.N.; Kitahara, C.M.; Keadle, S.K.; Arem, H.; De Gonzalez, A.B.; Hartge, P.; et al. Association of Leisure-Time Physical Activity with Risk of 26 Types of Cancer in 1.44 Million Adults. JAMA Intern. Med. 2016, 176, 816–825. [Google Scholar] [CrossRef]
  247. Ashcraft, K.A.; Peace, R.M.; Betof, A.S.; Dewhirst, M.W.; Jones, L.W. Efficacy and Mechanisms of Aerobic Exercise on Cancer Initiation, Progression, and Metastasis: A Critical Systematic Review of In Vivo Preclinical Data. Cancer Res. 2016, 76, 4032–4050. [Google Scholar] [CrossRef]
  248. Aveseh, M.; Nikooie, R.; Aminaie, M. Exercise-induced changes in tumour LDH-B and MCT1 expression are modulated by oestrogen-related receptor alpha in breast cancer-bearing BALB/c mice. J. Physiol. 2015, 593, 2635–2648. [Google Scholar] [CrossRef]
  249. Loughney, L.; West, M.A.; Kemp, G.J.; Grocott, M.P.W.; Jack, S. Exercise intervention in people with cancer undergoing neoadjuvant cancer treatment and surgery: A systematic review. Eur. J. Surg. Oncol. J. Eur. Soc. Surg. Oncol. Br. Assoc. Surg. Oncol. 2016, 42, 28–38. [Google Scholar] [CrossRef]
  250. Schmitz, K.H.; Courneya, K.S.; Matthews, C.; Demark-Wahnefried, W.; Galvão, D.A.; Pinto, B.M.; Irwin, M.L.; Wolin, K.Y.; Segal, R.J.; Lucia, A.; et al. American College of Sports Medicine roundtable on exercise guidelines for cancer survivors. Med. Sci. Sports Exerc. 2010, 42, 1409–1426. [Google Scholar] [CrossRef]
  251. Iannaccone, M.; D’Ascenzo, F.; Vadalà, P.; Wilton, S.B.; Noussan, P.; Colombo, F.; Raposeiras Roubín, S.; Abu Assi, E.; González-Juanatey, J.R.; Simao Henriques, J.P.; et al. Prevalence and outcome of patients with cancer and acute coronary syndrome undergoing percutaneous coronary intervention: A BleeMACS substudy. Eur. Heart J. Acute Cardiovasc. Care 2018, 7, 631–638. [Google Scholar] [CrossRef]
  252. Bharadwaj, A.; Potts, J.; Mohamed, M.O.; Parwani, P.; Swamy, P.; Lopez-Mattei, J.C.; Rashid, M.; Kwok, C.S.; Fischman, D.L.; Vassiliou, V.S.; et al. Acute myocardial infarction treatments and outcomes in 6.5 million patients with a current or historical diagnosis of cancer in the USA. Eur. Heart J. 2020, 41, 2183–2193. [Google Scholar] [CrossRef] [PubMed]
  253. Leiva, O.; AbdelHameid, D.; Connors, J.M.; Cannon, C.P.; Bhatt, D.L. Common Pathophysiology in Cancer, Atrial Fibrillation, Atherosclerosis, and Thrombosis: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncology 2021, 3, 619–634. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Illustration of shared pathophysiological pathways in cancer and cardiovascular disease (CVD) Cancer-related therapeutics include chemotherapy, radiotherapy, and immune checkpoint inhibitors.
Figure 1. Illustration of shared pathophysiological pathways in cancer and cardiovascular disease (CVD) Cancer-related therapeutics include chemotherapy, radiotherapy, and immune checkpoint inhibitors.
Cancers 16 01450 g001
Table 1. Potential desirable oncologic effects of commonly prescribed cardiovascular medications.
Table 1. Potential desirable oncologic effects of commonly prescribed cardiovascular medications.
MedicationPotential Cancer TargetsCancer Outcomes
AspirinCRC, esophageal, pancreatic, brain, lung, stomach, and prostate cancers
Prevention of early neoplastic transformation and possible anti-metastatic effect
Mitigation of morbidity and mortality of some cancers
Angiotensin converting enzyme inhibitors (ACEi)
Angiotensin receptor blockers (ARB)
Non-small-cell lung cancer Esophageal, pancreatic, and colon, prostate cancers and melanoma
Reduction of incidence of some cancers and mitigation of metastatic burden
Enhance anticancer response prior to chemotherapy
Some observational data showed increased risk of lung cancer
Beta blockers (βB)Breast, prostate, and ovarian cancers
Prevention of certain types of cancers
Tumor growth reduction
Calcium channel blockers (CCB)Breast, brain, colorectal, gastric, ovarian, and prostate tumors as well as leukemia
Possible antiproliferative effects in preclinical studies
StatinCRC, gastric, prostate and breast cancers
Prevention of certain cancers.
Improvement of biochemical recurrence in prostate cancer
Proprotein convertase subtilisin kexin type 9 inhibitors (PCSK9-i)Non-small-cell lung cancer, breast and colon cancers
Improvement of anti-tumor immune response to cancer cells
Prevention of certain type of cancers
Tumor growth reduction and improved survival in certain cancers
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zmaili, M.; Alzubi, J.; Alkhayyat, M.; Albakri, A.; Alkhalaileh, F.; Longinow, J.; Moudgil, R. Cancer and Cardiovascular Disease: The Conjoined Twins. Cancers 2024, 16, 1450. https://doi.org/10.3390/cancers16081450

AMA Style

Zmaili M, Alzubi J, Alkhayyat M, Albakri A, Alkhalaileh F, Longinow J, Moudgil R. Cancer and Cardiovascular Disease: The Conjoined Twins. Cancers. 2024; 16(8):1450. https://doi.org/10.3390/cancers16081450

Chicago/Turabian Style

Zmaili, Mohammad, Jafar Alzubi, Motasem Alkhayyat, Almaza Albakri, Feras Alkhalaileh, Joshua Longinow, and Rohit Moudgil. 2024. "Cancer and Cardiovascular Disease: The Conjoined Twins" Cancers 16, no. 8: 1450. https://doi.org/10.3390/cancers16081450

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop