Next Article in Journal
Terpenoid-Mediated Targeting of STAT3 Signaling in Cancer: An Overview of Preclinical Studies
Next Article in Special Issue
Beyond Natriuretic Peptides: Unveiling the Power of Emerging Biomarkers in Heart Failure
Previous Article in Journal
Factors and Mechanisms of Thyroid Hormone Activity in the Brain: Possible Role in Recovery and Protection
Previous Article in Special Issue
The Dark Side of Sphingolipids: Searching for Potential Cardiovascular Biomarkers
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiovascular Biomarkers in Cardio-Oncology: Antineoplastic Drug Cardiotoxicity and Beyond

by
Umberto Attanasio
1,†,
Elena Di Sarro
1,†,
Lucia Tricarico
2,
Daniela Di Lisi
3,4,
Giuseppe Armentaro
5,
Sofia Miceli
5,
Francesco Fioretti
6,
Martino Deidda
7,
Michele Correale
2,
Giuseppina Novo
3,4,
Angela Sciacqua
5,
Savina Nodari
6,
Christian Cadeddu
7,
Carlo Gabriele Tocchetti
1,8,9,10,
Alberto Palazzuoli
11 and
Valentina Mercurio
1,8,9,*
1
Department of Translational Medical Sciences, Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
2
Cardiology Unit, Department of Medical and Surgical Sciences, University of Foggia, Viale Pinto 1, 71122 Foggia, Italy
3
Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Piazza delle Cliniche 2, 90127 Palermo, Italy
4
Division of Cardiology, University Hospital Paolo Giaccone, Via del Vespro 129, 90127 Palermo, Italy
5
Department of Medical and Surgical Sciences, University Magna Græcia of Catanzaro, Campus Universitario di Germaneto, V.le Europa, 88100 Catanzaro, Italy
6
Cardiology Section, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, Spedali Civili Hospital and University of Brescia, Piazzale Spedali Civili 1, 25123 Brescia, Italy
7
Department of Medical Sciences and Public Health, University of Cagliari, 09042 Monserrato, Italy
8
Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
9
Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
10
Center for Basic and Clinical Immunology Research (CISI), Federico II University, Via Sergio Pansini 5, 80131 Naples, Italy
11
Cardiovascular Diseases Unit, Cardio-thoracic and Vascular Department Le Scotte Hospital, University of Siena, Strada delle Scotte 14, 53100 Siena, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomolecules 2024, 14(2), 199; https://doi.org/10.3390/biom14020199
Submission received: 30 December 2023 / Revised: 29 January 2024 / Accepted: 3 February 2024 / Published: 7 February 2024
(This article belongs to the Special Issue Molecular Biomarkers In Cardiology 2022–2023)

Abstract

:
Serum biomarkers represent a reproducible, sensitive, minimally invasive and inexpensive method to explore possible adverse cardiovascular effects of antineoplastic treatments. They are useful tools in risk stratification, the early detection of cardiotoxicity and the follow-up and prognostic assessment of cancer patients. In this literature review, we aim at describing the current state of knowledge on the meaning and the usefulness of cardiovascular biomarkers in patients with cancer; analyzing the intricate relationship between cancer and cardiovascular disease (especially HF) and how this affects cardiovascular and tumor biomarkers; exploring the role of cardiovascular biomarkers in the risk stratification and in the identification of chemotherapy-induced cardiotoxicity; and providing a summary of the novel potential biomarkers in this clinical setting.

1. Introduction

Cardio-oncology is a discipline that studies the relationship between cancer and cardiovascular diseases (CVDs), and it is mostly focused on the prevention and management of cardiovascular damage resulting from anticancer therapies [1]. The possible cardiotoxic events induced by anticancer treatments include myocardial dysfunction, heart failure (HF), coronary artery disease, valvular disease, arrhythmias, pericardial disease, hypertension and thrombolytic events. In addition to the cardiotoxic effects of oncological therapies, it is believed that cancer and HF are linked by a bidirectional relationship, where one disease favors the other [2,3]. Serum biomarkers represent a reproducible, sensitive, minimally invasive and inexpensive method to explore these effects. They are useful tools in risk stratification, the early detection of cardiotoxicity, follow-up and prognostic assessment [2,4]. They may be good tools to identify patients at high risk of adverse cardiovascular effects before the initiation of therapy and to detect subclinical diseases during active therapy in combination with imaging, identifying patients who should receive cardioprotective therapies [5,6]. Troponins and natriuretic peptides have garnered the broadest evidence base for cardiotoxicity risk prediction, but other markers are being investigated. However, further studies are needed to assess the diagnostic and prognostic roles of other potential biomarkers, including inflammatory and other novel markers. This review article aims at describing the current state of knowledge on the meaning and the usefulness of cardiovascular biomarkers in patients with cancer; analyzing the intricate relationship between cancer and cardiovascular disease (especially HF) and how this affects cardiovascular and tumor biomarkers; exploring the role of cardiovascular biomarkers in the risk stratification and in the identification of chemotherapy-induced cardiotoxicity; and providing a summary of the novel potential biomarkers in this clinical setting.

2. Cardiovascular Biomarkers: Troponins and Natriuretic Peptides

A biomarker, a biological molecule detected in blood, body fluids or tissues, is linked to vital parameters and imaging tests. These markers play a crucial role in revealing disease characteristics, serving as indicators of the risk and disease state and predicting outcomes, including the rate of progression [7]. Cardiac biomarkers, such as troponin and natriuretic peptides (NPs), play pivotal roles in assessing heart health, offering valuable insights into myocardial damage and HF.
NPs, notably brain natriuretic peptide (BNP) and atrial natriuretic peptide (ANP), serve as widely utilized biomarkers in HF. Synthesized as prohormones, they undergo cleavage into active hormones (BNP and ANP) and inactive forms (NT-proBNP and MR-proANP). While BNP and ANP have short circulating half-lives, NT-proBNP and MR-proANP persist for longer, with renal clearance [8,9]. Considered the gold-standard biomarkers in HF, BNP and NT-proBNP receive Class IA recommendation in the guidelines of major societies, including the American Heart Association (AHA) and European Society of Cardiology (ESC) [10,11]. However, clinical interpretation is influenced by factors such as obesity, which lowers serum levels, and various conditions like age, heart diseases, valve disorders, atrial fibrillation and renal failure, which increase plasmatic concentrations [12,13]. BNP and NT-proBNP have similar clinical value for the evaluation of cardiac function, although NT-proBNP is more stable and does not appear to be affected by changes in anticoagulants, collection containers, body position or circadian rhythm [14]. They offer a non-invasive means to estimate intracardiac filling pressures and end-diastolic wall stress, enhancing the diagnostic accuracy when combined with clinical assessment, ECG, chest X-ray and echocardiography [11,15]. Several randomized clinical trials support the additional use of BNP or NT-proBNP concentrations, leading to improved medical and economic outcomes [16,17]. BNP and N-terminal pro-B-type natriuretic peptides (NT-proBNPs) are biomarkers of long-term cardiovascular dysfunction in asymptomatic patients [18,19,20,21] and represent an important screening tool for patients presenting with dyspnea during anticancer treatment. High diagnostic accuracy is observed in discriminating HF from other causes of dyspnea, with NP concentrations correlating with the likelihood of HF. The optimal cut-off concentrations for acute and chronic HF differ, with higher thresholds in cases of acute dyspnea [11]. NPs exhibit high prognostic accuracy in various conditions, including HF hospitalization, myocardial infarction, valvular heart disease, atrial fibrillation and pulmonary embolism. They also track variations in myocardial stress and dysfunction, making them valuable in conditions like Takotsubo syndrome and during cancer treatment. Myocardial ischemia triggers NP expression independently of mechanical stress, highlighting their role in HF worsening [11,22].
NPs are valuable markers for cardiotoxicity assessment. They can identify acute cardiotoxicity, particularly within 24 h of exposure to anthracycline chemotherapy [23]. While NPs are useful for HF screening in cancer patients with dyspnea, a cut-off value of 100 ng/L for NT-proBNP has high sensitivity [24,25]. Several studies indicate that elevated baseline NT-proBNP in cancer patients is a significant predictor of mortality risk [26,27,28,29].
Troponins are biomarkers that have always been used to diagnose acute coronary syndromes, but have proven to be useful in identifying cardiotoxicity. There are three types of troponins: troponin I, troponin T and troponin C. Cardiac troponin T (cTnT) and cardiac troponin I (cTnI), released during cardiac muscle cell injury, are heart-specific markers, but not disease-specific. Increased levels can be found in various conditions, both physiological (i.e., physical or psycho-emotional stress) and pathological, including chronic HF, diabetes, arterial hypertension, inflammatory heart disease, pulmonary embolism, chronic renal failure and sepsis. The methods of determining cTnT and cTnI have been continuously improved, increasing their analytical sensitivity and specificity [30]. Highly sensitive (hs) immunoassays are now available to determine hs-cTnT and hs-cTnI concentrations, detecting very low but diagnostically significant concentrations of circulating cardiac troponins, especially to identify subclinical cardiac damage [31,32,33]. Small changes in plasma high-sensitivity cTnI (hs-cTnI) below the 99th centile have prognostic value in various heart-related conditions. Temporal changes in high-sensitivity cardiac troponin concentrations help to differentiate acute from chronic cardiomyocyte injury, with a near-linear association between cTnT/cTnI concentrations and the risk of developing clinical HF, hospitalization, atrial fibrillation and death. Managing cardiomyocyte injury requires individualization based on dominant mechanisms, though this can be challenging [34].
In 2020, a meta-analysis of 61 trials involving 5691 adult cancer patients revealed that anticancer therapy often leads to an increase in troponin levels (OR 14.3, 95% CI 6.0–34.1; n = 3049). Elevated troponins were associated with a higher risk of left ventricular dysfunction (LVD) (OR 11.9, 95% CI 4.4–32.1; n  =  2163) [35]. This underscores the potential of troponin assessment in identifying patients at risk for cardiotoxicity during cancer treatment. Moreover, beta-blockers, but not candesartan or low-dose enalapril, were found effective in preventing troponin rises when used as a primary prevention strategy in two independent prospective trials, emphasizing the role of troponin monitoring in evaluating the response to cardioprotective treatment [36,37,38]. The combination of these biomarkers provides a comprehensive evaluation of cardiac function, aiding clinicians in making informed decisions about diagnosis, risk stratification and treatment planning. Their distinct specificities contribute to a more nuanced understanding of cardiac conditions, enhancing the overall effectiveness of cardiovascular care.

3. Cardiovascular Biomarkers in Cancer Patients vs. Tumor Biomarkers in Heart Failure Patients

The recent investigations on the possible interplay between cardiovascular biomarkers and cancer have unveiled a complex relationship, challenging the traditional perspectives on the exclusive association of cardiac biomarkers, such as troponin and natriuretic peptides, with some cardiovascular diseases or with the development of cancer therapy-related cardiovascular toxicity (CTRCD). In fact, even though the most recent recommendations on the management of patients at risk of developing CTRCD suggest the broad use of troponin and natriuretic peptides to stratify cardiovascular risk (at baseline) and to identify the early development of toxicity (during follow-up) [39], some evidence suggests that the circulating levels of NPs and troponins can be elevated in patients with neoplasms, even prior to the introduction of cancer therapy, with no evidence of an abnormal cardiac status [26]. Indeed, it is known that malignant cells are able to produce vasoactive peptides, such as vasopressin [40] or endothelin-1 [41], as well as cardiac neurohormones, like atrial natriuretic peptide [42] and B-type natriuretic peptide (BNP) [43,44]. Importantly, BNP elevation cannot be always explained by the presence of an underlying cardiac or hemodynamic condition that would typically affect BNP levels [45]. Moreover, the levels of troponin T (TnT) and troponin I (TnI) have been recently demonstrated to be elevated in cancer patients [46,47], being the former also associated with a worse prognosis [46]. The evidence that such cardiovascular biomarkers tend to increase proportionally with the advancing tumor stage, even without other signs of cardiac damage or dysfunction [26], may reinforce the idea that, in some cases, the elevation of these biomarkers could be hypothesized to be driven by the neoplastic disease, rather than the presence of an underlying cardiopathy or the occurrence of CTRCD. Certainly, this should not discourage the clinical use of these biomarkers according to the most recent guidelines [39], especially taking into account that they seem to be useful in the prediction of prognosis [26,46]. Of note, even though elevated levels of cardiovascular biomarkers were found in subjects without evidence of manifest cardiac involvement, the association between late-stage cancer, cancer cachexia and cardiac wasting has been extensively described and may represent a possible explanation for this phenomenon [48,49,50].
Conversely, a reciprocal relationship may be identified within cardiovascular conditions, specifically HF, presenting elevated levels of tumor biomarkers [51]. In fact, it has emerged that patients affected by HF present with high serum levels in several biomarkers presumed to be tumor-related, such as CA19-9, CA125 and human epididymis protein 4 (HE4) [51,52,53,54,55,56]. The possible interconnections between HF and cancer extend far beyond HF being a manifestation of CTRCD, and they have been deeply investigated in the last few decades [57,58,59,60,61,62,63]. Interestingly, an analysis of 2079 patients affected by HF in the BIOSTAT-CHF cohort [64] proved that the blood levels of five out of the six tumor biomarkers that they investigated (CA125, CA15-3, CA19-9, CEA and CYFRA 21-1) were significantly correlated with all-cause mortality, while CA125 also showed a strong correlation with HF hospitalization risk, and CYFRA 21-1 had equivalent predictive utility for all-cause mortality when compared with NTproBNP levels [51]. In particular, CA125 is probably the most investigated tumor biomarker in this setting, having shown prognostic value in different cardiac settings, being elevated in response to both congestion and inflammatory stress conditions [65,66,67,68,69]. This tumor biomarker is also associated with hospitalization, a worse prognosis and elevated NTproBNP serum levels [65,66,67,68,69]. Likewise, in recent studies, HE4, which is a serum biomarker currently used to monitor the recurrence of epithelial ovarian cancer, has been demonstrated to be strongly associated with HF and it seems to represent an independent predictor of HF outcomes [70,71,72].
Thus, the associations between established tumor biomarkers and indices of HF severity, along with their independent prognostic value for the HF outcomes of these biomarkers, and between classic cardiovascular biomarkers and cancer patient prognosis, may suggest the presence of dysregulated pathophysiologic pathways common to both cancer and cardiovascular diseases [61]. As already mentioned, the existent correlation between these two conditions is well recognized in the scientific literature and, remarkably, it has been highlighted that cancer and cardiovascular disease share many risk factors, as well as a common underlying inflammatory condition [57,58,59,61,62,73]. Of note, this chronic phlogistic state has been also observed in clinical practice and reported in the literature, with the help of serum inflammatory markers such as C-reactive protein (CRP) and proinflammatory cytokines such as interleukin 6 (IL-6) in both cancer and HF [19,74,75,76,77,78]. As highlighted for the other biomarkers, IL-6 seems to play a role in predicting prognosis in HF patients [79], and it was also found, together with CRP, to be proportionally elevated to the cardiovascular peptides (such as NTproBNP) in a population of 555 individuals with different types and stages of cancer that had not yet undergone chemotherapy; in addition, similarly to the trend of cardiac biomarkers, IL-6 serum levels were found to be higher in patients with more advanced stages of cancer [26].
To summarize, the intricate interplay between cardiovascular biomarkers and cancer extends beyond their traditional roles, revealing a complex relationship influenced by systemic inflammation and shared risk factors and pathophysiological pathways. These biomarkers, while often utilized to detect potential cardiac toxicity from cancer therapies, can also rise independently, even before chemotherapy, being sometimes able to stratify risk and predict prognosis. Such a bidirectional association, where cancer patients exhibit elevated cardiovascular biomarkers and cardiac patients present increased levels of tumor biomarkers, underscores the need for a deeper understanding of these markers in the context of the more precise and targeted use of these tools in both clinical scenarios.

4. Role of Cardiovascular Biomarkers in the Risk Stratification and in the Identification of Chemotherapy-Induced Cardiotoxicity in Cancer Patients

In recent years, important advances have been made in the field of cardio-oncology [39]. The ESC Guidelines on Cardio-Oncology recommend cardiovascular (CV) toxicity risk stratification before starting potentially cardiotoxic cancer therapy [39,80]. Baseline CV risk stratification in cancer patients is important in order to prescribe cardioprotective treatment before starting antineoplastic treatment when needed, to schedule a cardiology referral before treatment and to plan the most appropriate surveillance program during and after treatment [39]. Cardiotoxicity risk is a dynamic variable related not only to traditional cardiovascular risk factors but also to treatment-related factors; therefore, it is advisable to conduct its assessment by using a dedicated cancer patient tool such as the HFA-ICOS risk score [39,80]. Few studies have so far validated this score in patients with solid or hematological tumors [81,82]. The HFA-ICOS risk score takes into consideration lifestyle risk factors, demographic and CV risk factors, previous CV diseases, previous and concomitant cardiotoxic cancer treatment and baseline cardiac biomarkers (elevated baseline troponin—Tn, elevated baseline brain natriuretic peptide—BNP or NT-proBNP) in patients receiving six of the most frequently used anticancer treatments [80]. Monitoring biomarkers, and particularly Tn and BNP/NT-proBNP, during cancer treatment aids in the early diagnosis of cardiotoxicity in cancer patients [34].
Symptomatic CTRCD is defined by the presence of HF symptoms; the diagnosis of asymptomatic CTRCD is based on the left ventricular ejection fraction (LVEF) reduction and/or relative decline in global longitudinal strain (GLS) and/or a new rise in cardiac biomarkers (cTnI/cTnT gammaGT; 99th percentile, BNP ≥ 35 pg/mL, NT-proBNP ≥ 125 pg/mL, or a new significant rise from baseline beyond the biological and analytical variations of the assay used) [39]. Several studies have shown the prognostic role of monitoring cardiac troponin. In particular, Cardinale et al. demonstrated, in patients treated with high-dose anthracyclines, that persistently negative troponin is able to identify low-risk patients who do not need close echocardiographic monitoring, while a persistent troponin elevation preceded an LVEF drop and identified high-risk patients [83,84,85]. In patients treated with trastuzumab, a troponin increase identified patients at risk of non-reversible left ventricular dysfunction [86]. In a recent meta-analysis, Michel et al. confirmed the predictive role of a troponin increase after anthracycline-based chemotherapy or human epidermal receptor 2 (HER2) inhibitor therapy for the development of left ventricular dysfunction.
Petricciulo et al. showed that, in patients treated with immune checkpoint inhibitors (ICI), baseline hs-TnT predicted a composite cardiovascular endpoint (cardiovascular death, stroke or transient ischemic attack, pulmonary embolism and new-onset HF) and the progression of cardiac involvement at 3 months, with 14 ng/L as the best cut-off [87]. In patients treated with ICI, guidelines recommend measuring baseline cardiac troponin to stratify cardiotoxicity risk; moreover, it should be monitored in order to detect ICI-induced myocarditis [39]. Less evidence exists regarding the increase in BNP/NT-proBNP as a marker for cancer therapy-related cardiotoxicity [20]. In fact, a BNP/NT-proBNP increase after chemotherapy may be associated with fluid overload, limiting its diagnostic and prognostic role. Elevated BNP levels have been found at baseline in patients with malignancies and without HF and/or sepsis; it is likely that tumor-related mechanisms and oxidative stress contribute to increasing this biomarker [88].
In patients with multiple myeloma receiving proteasome inhibitors, baseline high levels of BNP (>100 pg/mL) and NT-proBNP (>125 pg/mL) predicted cardiovascular adverse events associated with worse overall outcomes [89]. It is recommended to monitor NP at baseline, before starting proteosome inhibitors, and during treatment, especially in the setting of cardiac amyloidosis [39]. Regarding a biomarker-guided cardioprotective strategy, the recently published multicenter prospective Cardiac Care trial failed to demonstrate a cardioprotective effect of a troponin-measurement-guided strategy in patients receiving anthracycline-based chemotherapy [90]. Certainly, the use of cardiac biomarkers shows advantages compared to imaging in the management of cancer patients and, in particular, they are easier to obtain by oncologists and more reproducible than imaging; they are also less time-consuming for patients. However, many problems still limit their implementation in clinical practice, such as the lack of standardization in trial methodologies evaluating biomarkers, the wide heterogeneity in terms of malignancy types, cancer treatment schedules and the definition of cardiotoxicity across the trials.

5. Cancer-Therapy-Related Cardiac Dysfunction: Clinical Utility of Biomarkers and the Role of Genetic Polymorphisms

Anthracyclines are the oldest chemotherapeutic drugs known to cause cardiovascular toxicity. Anthracycline-induced cardiotoxicity is cumulative, dose-dependent, irreversible and can present with symptomatic or asymptomatic CTRCD. The baseline measurement of NP and cTn is recommended before receiving anthracycline chemotherapy, especially in high- and very high-risk patients, and before each cycle (in high- and very high-risk patients) or every other cycle during anthracycline chemotherapy and either 3 or 12 months after the completion of cancer therapy [39].
Concerning HER2-targeted therapies, trastuzumab-induced cardiotoxicity has been widely studied and it is largely reversible and dose-independent, suggesting that genetic factors may play an important role in its occurrence [91,92]. An increase in cTn levels identifies patients at higher risk of trastuzumab-induced CTRCD, and the serial measurement of NP was more sensitive in predicting subsequent declines in LVEF during trastuzumab treatment [39]. Fluoropyrimidines such as 5-fluorouracil and its oral prodrug capecitabine represent the second cause of cardiotoxicity from chemotherapy, following that from anthracyclines, although their cardiotoxicity is generally underestimated and underdiagnosed [93]. Vascular endothelial growth factor (VEGF) inhibitors, including monoclonal antibodies and tyrosine kinase inhibitors (TKIs), may cause relevant cardiovascular effects, leading to an impairment in the balance between vasodilation and vasoconstriction, undermining endothelial cell integrity and interacting with off-target pathways [94]. The measurement of NP may be considered in all patients, at baseline and then every 4 months during the first year in moderate-risk patients, or at baseline and 4 weeks after starting treatment, and then every 3 months during the first year, in high- and very high-risk patients [39].
Multitargeted kinase inhibitors targeting BCR-ABL (including imatinib, bosutinib, dasatinib, nilotinib and ponatinib) are associated with unique toxicity, because of the “off-target” effects of each drug [39].
Radiotherapy (RT) can also negatively affect cardiac function by promoting myocardial fibrosis and microvascular damage. The risk of developing heart disease in subjects irradiated to the chest is greater than in the general population. In a prospective study of a cohort of patients with Hodgkin’s or non-Hodgkin’s lymphoma treated with anthracyclines, it was demonstrated that RT may have an additional unfavorable impact on the myocardial longitudinal systolic deformation [95].
The most common forms of CTRCD are summarized in Table 1. Examples of increases in serum biomarkers correlated to cardiotoxicity are listed in Table 2.
Knowledge of the pathophysiology of chemotherapy-induced cardiotoxicity is essential in order to develop strategies aimed at reducing such toxicity. Furthermore, it is important to highlight that the type of damage and the pathways involved can guide the search for genetic polymorphisms associated with a reduction or an increase in the risk of cardiotoxicity. A genetic polymorphism is a variation in the DNA sequence present in at least 1% of the population. It has been shown that genetic polymorphisms can influence traits and susceptibility to diseases or the effectiveness of drug therapies (i.e., the effectiveness of chemotherapy or the possible onset of adverse events in oncological treatments).
In the most recent literature, there are many examples of the application of genetic polymorphisms to identify genetic susceptibility in the onset of cardiac damage related to oncological therapy. A study that included 176 women with breast cancer without concurrent cardiovascular diseases, who were scheduled for polychemotherapy with anthracyclines, recommended the evaluation of genetic polymorphisms in the p53 protein (rs1042522) and NOS3 (rs1799983), NADPH-oxidase (rs4673), GPX1 (rs1050450), ADRB1 (Arg389Gly, rs1801253) and MMP-3 (rs3025058) genes prior to starting chemotherapy. This study shows that the maximum risk of cardiotoxicity is associated with the presence of the p53 protein gene Arg/Arg genotype and NOS3 gene T/T genotype [103]. A genome-wide association study (GWAS) and pathway analysis of changes in LVEF after exposure to anthracyclines in in 385 subjects revealed that the presence of SNP rs10443221 on chromosome 1 (1p32.1) near PRDM2 conferred protection from cardiotoxicity (4.11-point increase in LVEF for each alternate allele) [104]. Numerous pharmacogenomics studies have identified the synonymous genomic variant rs7853758 and the intronic variant rs885004 in SLC28A3 as statistically associated with a lower incidence of anthracycline-induced cardiotoxicity. A study conducted on six well-phenotyped, doxorubicin-treated pediatric patients showed that patient-derived cardiomyocytes recapitulate the cardioprotective effect of the SLC28A3 locus and that SLC28A3 expression influences the severity of doxorubicin-induced cardiotoxicity (DIC). This study provides two potential therapeutic options to attenuate DIC, repurposing FDA-approved desipramine or SLC28A3-AS1 long noncoding RNA therapy, and proposes a simple clinical test to detect the presence of rs11140490 to predict that a patient will be less likely to experience DIC [105]. A case–control association study of 2258 genetic variants between nine cases of trastuzumab-induced cardiotoxicity and controls from the Japanese general population, registered in the Human Genetic Variation Database, identified a novel variant in the EYS gene associated with trastuzumab-induced cardiotoxicity. This finding provides new insights into personalized trastuzumab therapy for patients with HER2-positive cancer [106]. The VEGF-634 CC polymorphism is associated with higher risks of both bevacizumab-induced hypertension and thromboembolism [107]. It has been reported that polymorphisms of the KDR gene decrease VEGF-A’s binding ability to the KDR protein, leading to an increased risk of coronary artery disease [108]. Further studies in this field are needed to aim at personalizing therapeutic schemes also according to the genetic profile of the patient, targeting the treatment and avoiding adverse events.

6. Multiparametric Approach Integrating Biomarkers, Imaging and Clinic: The Example of Multiple Myeloma

Biomarkers and imaging play a key role in the early detection and subsequent monitoring of CTRCD in cancer patients, allowing the early detection of cardiac dysfunction before clinical manifestations [84,109,110,111]. In fact, several studies have shown how natriuretic peptide (NP) determination at baseline or NP changes during follow-up are able to predict future CTRCD [17,112,113].
In particular, in patients with multiple myeloma (MM), who are at a high risk of developing cardiovascular (CV) complications, the measurement of NPs before cancer treatment predicts subsequent CV adverse events with good accuracy. In line with this, in a study of 109 patients with relapsed MM, circulating levels of BNP > 100 pg/mL or NT-proBNP > 125 pg/mL before the initiation of carfilzomib were associated with an odds ratio of 10.8 for subsequent CV adverse events [89].
In addition, a finding of particular interest comes from a study of 555 patients with different types of cancer, in which elevated values at baseline of cardiac biomarkers including NT-proBNP and troponin (cTn) were strongly correlated with all-cause mortality. This observation further supports the hypothesis that the presence of subclinical myocardial damage could be directly related to disease progression [26].
However, in the CARDIOTOX (CARDIOvascular TOXicity induced by cancer-related therapies) registry, which enrolled 855 patients treated with different types of therapy, including radiation therapy (RT), both NT-proBNP and cTn elevations at baseline were not associated with the development of severe CTRCD identified as LVEF < 40% or clinical HF [114].
Therefore, based on these results, the ESC cardio-oncology guidelines recommend NT-pro-BNP and cTn dosages in high- and very-high-risk patients; moreover, their measurement should be considered in low- and moderate-risk patients before treatment with proteasome inhibitors. Due to the emerging role of biomarkers in CRTCD identification, there is great interest in the search for new biomarkers that can further support risk stratification in this patient setting. Candidates include myeloperoxidase, galectin-3, micro-ribonucleic acids and immunoglobulin E. However, at present, there is insufficient evidence to support the use of these biomarkers in clinical practice and further studies are needed [39].
In the absence of biomarkers that are applicable to the entire cancer population, the integration of biomarkers and cardiovascular imaging is the best method to detect chemotherapy cardiotoxicity at an early stage. Indeed, the onset of chemotherapy may be followed by myocardial cell damage with the release of troponins and natriuretic peptides, which occurs about 3 months before the clinical manifestation of cardiomyopathy [34,85]. The latter is followed by a reduction in GLS, which precedes a reduction in LVEF and then the appearance of HF clinical symptoms [39].
Indeed, the determination of GLS is recommended before chemotherapy in patients with previous CV disease or at risk of CTRCD. In particular, GLS is a more sensitive and reproducible measure of LV systolic function than LVEF and may identify subclinical cardiac dysfunction before the detection of abnormal LVEF [115,116,117,118]. Indeed, a 15% reduction from baseline in GLS and/or a reduction in circumferential strain during follow-up is indicative of CRTCD or future left ventricular dysfunction [39]. Thus, both the absolute GLS value and relative reduction may be indicators of subclinical CRTCD [119].
This is even more evident in the case of MM; in fact, in addition to the changes in GLS, the Global Myocardial Work Efficiency (GWE), another echocardiographic parameter, can undergo significant changes in the case of CTRCD. Indeed, in MM patients on bortezomib therapy, reduced GWE values can detect early CTRCD but also cardiac adverse events after six months of bortezomib therapy [120].
In the event of poor-quality echocardiographic images or when a specific condition is diagnosed (e.g., hypertrophic cardiomyopathy), CMR is recommended for better risk assessment.
In symptomatic patients (stable angina, limiting dyspnea) with clinical suspicion of coronary artery disease (CAD), functional imaging testing is recommended, especially before the use of anticancer therapies associated with vascular toxicity (e.g., fluoropyrimidines, VEGFi, breakpoint cluster region–Abelson oncogene locus (BCR-ABL), tyrosine kinase inhibitors (TKIs)). Alternatively, in patients with a low to intermediate pretest probability of CAD, coronary computed tomography angiography (CCTA) is a robust alternative modality with high sensitivity to rule out obstructive CAD [39].
In conclusion, the integrated approach of clinical presentation, biomarkers and imaging methods is the best strategy for the recognition and monitoring of CRTCD, allowing early identification and thus timely therapeutic optimization, reducing CRTCD-related adverse events and improving patients’ quality of life.

7. Novel Potential Biomarkers

Reliable biomarkers that could predict cardiotoxicity and/or the early onset of CTRCD are not currently available in the clinical setting. However, troponins and natriuretic peptides have been indicated in the last ESC Guidelines on Cardio-Oncology as risk factors to be evaluated as part of the baseline cardiotoxicity risk assessment because of their usefulness as indicators of cardiomyocyte injury, but their predictive capacity for the onset of cardiotoxicity still lacks reliability. Predictive biomarkers for the early and late onset of CTRCD are urgently needed to mitigate the risks associated with cardiac complications [121]
Novel emerging biomarkers were recently evaluated in a CTRCD setting.
-
MicroRNAs (miRNAs) [122]: small endogenous single-stranded non-coding RNAs, which act as modifiers of gene expression post-transcriptionally through the binding to protein-coding messenger RNA [123]. The dysregulation of miRNAs has been associated with various diseases, so they are of great interest as biomarkers, particularly due to their properties of being potentially disease-specific, stable, quantifiable and easily extracted from a range of clinical samples. Several studies show that circulating miRNAs are correlated with CTRCD, especially in breast cancer and in patients treated with anthracyclines and trastuzumab [124]. However, further studies are needed to accurately evaluate the potential use of miRNAs in this clinical setting.
-
Myeloperoxidase (MPO): myeloid-lineage-restricted enzyme with bactericidal properties, found in the azurophilic granules of neutrophils, involved in the neutrophil extracellular traps (NETs) that are implicated in myocardial infarction and in serious cardiovascular events [121]. Elevated circulating levels of MPO were found in breast cancer patients that experienced cardiotoxicity, so MPO is now considered a promising biomarker for the early detection of anthracycline-related and anthracycline–trastuzumab cardiac dysfunction, based on the results of several studies [125,126].
-
Galectin-3 (Gal-3): a β-galactoside-binding protein and a member of the lectin family, implicated in various pathophysiological processes including fibrosis, inflammation and oxidative stress and known to induce cardiac fibroblast proliferation and collagen production and deposition [127]. Recent studies have investigated Gal-3 as a potential diagnostic biomarker for cancer-therapy-induced cardiac dysfunction in breast cancer patients [37,128,129]. In particular, some studies show changes in the circulating levels of Gal-3 in response to treatment with cardiotoxic breast cancer therapies [37], without clear evidence of an association between elevated levels of Gal-3 and the incidence of cardiotoxicity [129]. Its prognostic impact has not yet been fully understood.
-
C-Reactive Protein (CRP): an inflammatory marker assessed as a biomarker for the detection of CTRCD. In the past, various studies have shown a change in CRP levels in response to chemotherapy in breast cancer patients, with no clear association with the onset of CTRCD [37,130,131]. Other studies have found no association between the levels of CRP and subsequent cardiotoxicity [132].
-
Growth Differentiation Factor 15 (GDF-15): a hormonal peptide of the transforming growth factor-β superfamily. Increased concentrations of GDF-15 were noted in cardiomyocytes during ischemia–reperfusion injury and myocardial infarction [133], in response to treatment with anthracyclines and trastuzumab in breast cancer patients [132,134]. A study by Tromp et al. [135] showed a strong and significant association between GDF-15 and changes in left ventricular ejection fraction in late breast cancer survivors after correction for potential confounders. Further studies with a larger number of patients are required to evaluate the predictive role of GDF-15 in CTRCD and overcome inconsistencies.
-
Other Biomarkers under Investigation: other potentially promising biomarkers are currently under investigation for their utility in the detection and/or prediction of CTRCD. These include endothelin-1, neuregulin-1, plasma bioactive adrenomedullin (ADM), D-dimer, soluble fms-like tyrosine kinase receptor (sFlt)-1, soluble ST2 (sST2), CK-MB, topoisomerase II α gene (TOP2A), cardiac myosin light chain 1 (cMLC-1), vascular endothelial growth factor (VEGF), placental growth factor (PIGF), procollagen-derived type-I C-terminal-propeptide (PICP), epicardial adipose tissue (EAT) volume, circulating bilirubin, hemopexin, glycated hemoglobin (HbA(1)c), advanced oxidation protein products (AOPP), human resistin and vascular adhesion molecule 1 (VCAM-1) [121].
Recently, Varkoly et al. [136] demonstrated for the first time that circulating RNA virus gene sequences (as orthomyxovirus) could be detected in the blood samples of patients with hematological malignancies or breast cancer, and that there is an association between them and myocarditis and lower LVEF in response to chemotherapy.
In summary, traditional biomarkers (such as troponins and natriuretic peptides) are the most frequently used CTRCD biomarkers in clinical trials and in clinical practice, albeit not universally, and their strengths and limitations are well documented. In particular, they could be indicators of cardiomyocyte injury, with a predictive capacity for the onset of cardiotoxicity of poor reliability. Several promising biomarkers are reported in the literature, with the aim to develop a more advanced and accurate risk stratification model for the prediction and early detection of CTRCD in the cancer population. To date, there is a need for larger additional and prospective clinical studies with sufficient statistical power for the validation of novel biomarkers of CTRCD. In any case, a combination of both serum and imaging biomarkers is encouraged for the best and most precise risk stratification of patients at risk of developing cardiotoxicity in response to cancer therapy.

8. Non-Conventional Potential Biomarkers: Omics

To date, conventional cardiotoxicity parameters and biomarkers have typically shown significant changes only after evident heart damage. In recent decades, researchers have shifted their focus to unraveling the mechanisms underlying cardiotoxicity to identify potential biomarkers for early cardiac damage. Omics sciences, including genomics, transcriptomics, proteomics and metabolomics, have been extensively employed in this pursuit. The sensitivity of metabolomics, with its ability to capture the complexity of entire metabolic networks, makes it one of the most promising approaches in the quest for early cardiovascular damage biomarkers [137].
Doxorubicin (DOX) is well known for exhibiting cardiotoxicity at cumulative doses, leading to increased intracellular reactive oxygen species in the heart. Palmer et al. conducted a two-phase study on human-induced pluripotent stem-cell-derived cardiomyocytes (hiPSC-CM) to predict cardiotoxicity development. They identified four metabolites (lactic acid, arachidonic acid, thymidine and 2′-deoxycytidine) with essential roles in regulating mitochondrial function, oxidative stress and replication, associated with cardiotoxicity [138].
Similarly, Dionisio et al. explored cardiotoxicity induced by cyclophosphamide, an anticancer prodrug known to cause cardiotoxicity and other severe adverse effects. Their in vitro metabolomic approach revealed a connection between cyclophosphamide’s active metabolite, 4-hydroxycyclophosphamide, and acrolein. Results indicated that these metabolites led to mitochondrial and lysosomal impairment, increased intracellular sugar levels, affected Krebs cycle metabolites and altered amino acid levels [139].
Li et al. identified 39 biomarkers for the detection of doxorubicin, isoproterenol and 5-fluorouracil cardiotoxicity earlier than biochemical and histopathological analyses in a mouse model. They established a predictive model characterized by L-carnitine, 19-hydroxydeoxycorticosterone, lysophosphatidylcholine (LPC) (14:0) and LPC (20:2) [140].
Schnackenberg’s group discovered 18 metabolites significantly altered in the plasma and another 22 metabolites increased in cardiac tissue after a cumulative doxorubicin dose of 6 mg/kg. The detection of myocardial injury and cardiac pathology, however, was not possible until cumulative doses of 18 and 24 mg/kg, respectively. In both serum and tissue specimens, the levels of many amino acids (including arginine and citrulline), biogenic amines, acylcarnitines (carnitine) and tricarboxylic acid cycle (TCA)-related metabolites (e.g., lactate, succinate) were found to be altered [141].
Tan et al. identified DOX-induced cardiotoxicity constituted by 24 metabolites involved in glycolysis, the citrate cycle and the metabolism of some amino acids and lipids [142].
Jensen et al. demonstrated significant decreases in docosahexaenoic acid, arachidonic acid/eicosatetraenoic acid, o-phosphocolamine and 6-hydroxynicotinic acid in the hearts of mice treated with sunitinib [143], along with alterations in taurine/hypotaurine metabolism, after sorafenib treatment [144].
Unger’s group identified, for the first time, a six-metabolite panel constituted by SM (d18:1/16:0), SM (d18:1/18:0), PC (16:0/14:0), PE (16:0/20:4), 1-(1,2-dihexanoylphosphatidyl) inositol-4,5-bisphosphate and Gly-Arg-Gly-Asp-Asn-Pro. All these metabolites were found to be upregulated in the plasma of patients showing cardiotoxicity, using samples from both rat models exposed to radiation and patients receiving radiation therapy for esophageal cancer [145].
In the context of breast cancer patients—the most studied population—Asnani’s group emphasized the importance of intermediary metabolism in anthracyclines and trastuzumab-treated patients. The authors recognized variations in aconitic and citric acid capable of discriminating patients who developed cardiotoxicity from those who did not. Moreover, citric acid levels correlated with the modification in LVEF at three months and with absolute LVEF values at nine months. Patients with cardiotoxicity also exhibited increased purine metabolites inosine, hypoxanthine and uric acid, while pyrimidine metabolites pseudouridine and orotic acid characterized those patients who did not show cardiotoxicity [146].
Preliminary data from Cocco et al. seem to demonstrate, for the first time, that early anthracycline-induced cardiotoxicity (detected by asymptomatic GLS reduction) is correlated with a characteristic metabolic profile: higher concentrations of Krebs cycle intermediates (fumarate and succinate) and fatty acids (such as linoleic acid) in patients with cardiotoxicity and increased levels of cardioprotective metabolites, such as tryptophan, in patients without cardiotoxicity. Interestingly, the damage induced by cardiotoxicity showed the upregulation of metabolites similar to those identified in clinical HF and mice models of cardiotoxicity [147].
Regarding carfilzomib-related cardiotoxicity, the group of Tantawy demonstrated lower plasma levels of pyruvate and higher values of lactate [148].
More recently, Yang et al. identified mitochondrial-regulated glycerolipid metabolism as a key player in metabolic changes due to immunotherapy-related cardiotoxicity [149].
The discussed studies underscore how metabolomics could represent a sensitive and promising tool in cardio-oncology research. Its translational utilization could lead to a more efficient follow-up in all cardiotoxicity-prone patients.
The Central Figure (Figure 1) summarizes the complex bidirectional relationship between cancer and cardiovascular disease and the major role of widely utilized and novel biomarkers.

9. Central Figure

Figure 1. Beyond cancer therapy-related cardiac dysfunction (CTRCD): bidirectional relationship between cancer and heart disease. (a) Increased levels of natriuretic peptides and troponins have been found in cancer patients, even before the introduction of anticancer therapy, without evidence of abnormal cardiac function. Malignant cells are capable of producing vasoactive peptides, such as vasopressin or endothelin-1, as well as cardiac neurohormones, such as NPs. (b) Cancer and cardiovascular disease share many risk factors, as well as a common underlying inflammatory condition. Some serum inflammatory markers such as C-reactive protein (CRP) and proinflammatory cytokines like interleukin 6 (IL-6) are expressed in both cancer and heart failure. (c) Patients affected by HF present with high serum levels in several biomarkers presumed to be tumor-related, such as CA19-9, CA125 and human epididymis protein 4 (HE4). In particular, CA125 has shown prognostic value in different cardiac settings and HE4 is demonstrated to be strongly associated with HF. (d) Metabolomics could represent a sensitive and promising tool in cardio-oncology research, including more efficient follow-up in all patients at risk of cardiotoxicity. CTX-induced damage showed upregulation of metabolites similar to those known in clinical heart failure. (e) Novel emerging biomarkers in CTRCD setting are needed to mitigate the risks associated with cardiac complications.
Figure 1. Beyond cancer therapy-related cardiac dysfunction (CTRCD): bidirectional relationship between cancer and heart disease. (a) Increased levels of natriuretic peptides and troponins have been found in cancer patients, even before the introduction of anticancer therapy, without evidence of abnormal cardiac function. Malignant cells are capable of producing vasoactive peptides, such as vasopressin or endothelin-1, as well as cardiac neurohormones, such as NPs. (b) Cancer and cardiovascular disease share many risk factors, as well as a common underlying inflammatory condition. Some serum inflammatory markers such as C-reactive protein (CRP) and proinflammatory cytokines like interleukin 6 (IL-6) are expressed in both cancer and heart failure. (c) Patients affected by HF present with high serum levels in several biomarkers presumed to be tumor-related, such as CA19-9, CA125 and human epididymis protein 4 (HE4). In particular, CA125 has shown prognostic value in different cardiac settings and HE4 is demonstrated to be strongly associated with HF. (d) Metabolomics could represent a sensitive and promising tool in cardio-oncology research, including more efficient follow-up in all patients at risk of cardiotoxicity. CTX-induced damage showed upregulation of metabolites similar to those known in clinical heart failure. (e) Novel emerging biomarkers in CTRCD setting are needed to mitigate the risks associated with cardiac complications.
Biomolecules 14 00199 g001

10. Conclusions

In this literature review, we described the current state of knowledge on the meaning and the usefulness of cardiovascular biomarkers in patients with cancer, taking into account the intricate relationship between cancer and cardiovascular disease and how this affects cardiovascular and tumor biomarkers. Furthermore, we elucidated the role of cardiovascular biomarkers in the risk stratification and in the identification of chemotherapy-induced cardiotoxicity, according to the latest 2022 ESC Guidelines on Cardio-Oncology, providing a summary of the novel potential biomarkers in this clinical setting.
An increase in cardiac biomarkers reflects the hemodynamic overload (natriuretic peptides) and cardiomyocyte damage (high-sensitivity troponins). Cardiac biomarkers are sensitive parameters of cardiac damage. The current ESC cardio-oncology guidelines recommend the evaluation of biomarkers at the baseline evaluation and during follow-up visits, with different recommendations concerning the timing according to the anticancer treatment and baseline risk class. Cardiac damage in cancer patients may be secondary to antineoplastic treatment but can also be the consequence of cardiac damage secondary to pathophysiological mechanisms related to the cancer per se or secondary to cardiovascular comorbidities. Furthermore, cardiac biomarkers can increase in the case of worsening renal function. NT-proBNP is characterized by significant intrinsic variability and only variations greater than 20% should be considered as clinically relevant. A multiparametric approach evaluating cardiac biomarkers (and their changes over time) with other clinical/instrumental parameters (including imaging) can help in the identification of cardiac damage. Cardiac biomarkers allow the ruling out of cardiac involvement in specific onco-hematological settings (i.e., cardiac involvement in multiple myeloma). Novel potential inflammatory and non-inflammatory biomarkers are under investigation in this clinical setting. New omics sciences can identify early signs of cardiac damage in the setting of antineoplastic-drug-induced cardiotoxicity.

Author Contributions

Conceptualization, U.A., E.D.S. and V.M.; formal analysis, V.M.; writing—original draft preparation, U.A., E.D.S., L.T., D.D.L., G.A., S.M., F.F., M.D., M.C., G.N., A.S., S.N., C.C., C.G.T., A.P. and V.M.; writing—review and editing, V.M.; supervision, A.P., C.G.T. and V.M. All authors have read and agreed to the published version of the manuscript.

Funding

C.G.T.: Italian Ministry of Health (PNRR-MAD-2022-12376632).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brown, S.-A. Preventive Cardio-Oncology: The Time Has Come. Front. Cardiovasc. Med. 2020, 6, 187. [Google Scholar] [CrossRef]
  2. Chianca, M.; Panichella, G.; Fabiani, I.; Giannoni, A.; L’Abbate, S.; Aimo, A.; Del Franco, A.; Vergaro, G.; Grigoratos, C.; Castiglione, V.; et al. Bidirectional Relationship Between Cancer and Heart Failure: Insights on Circulating Biomarkers. Front. Cardiovasc. Med. 2022, 9, 936654. [Google Scholar] [CrossRef]
  3. Strongman, H.; Gadd, S.; Matthews, A.; Mansfield, K.E.; Stanway, S.; Lyon, A.R.; Dos-Santos-Silva, I.; Smeeth, L.; Bhaskaran, K. Medium and long-term risks of specific cardiovascular diseases in survivors of 20 adult cancers: A population-based cohort study using multiple linked UK electronic health records databases. Lancet 2019, 394, 1041–1054. [Google Scholar] [CrossRef]
  4. Tan, L.-L.; Lyon, A.R. Role of Biomarkers in Prediction of Cardiotoxicity During Cancer Treatment. Curr. Treat. Options Cardiovasc. Med. 2018, 20, 55. [Google Scholar] [CrossRef]
  5. Fabiani, I.; Panichella, G.; Aimo, A.; Grigoratos, C.; Vergaro, G.; Pugliese, N.R.; Taddei, S.; Cardinale, D.M.; Passino, C.; Emdin, M.; et al. Subclinical cardiac damage in cancer patients before chemotherapy. Heart Fail. Rev. 2022, 27, 1091–1104. [Google Scholar] [CrossRef]
  6. Sorodoc, V.; Sirbu, O.; Lionte, C.; Haliga, R.E.; Stoica, A.; Ceasovschih, A.; Petris, O.R.; Constantin, M.; Costache, I.I.; Petris, A.O.; et al. The Value of Troponin as a Biomarker of Chemotherapy-Induced Cardiotoxicity. Life 2022, 12, 1183. [Google Scholar] [CrossRef]
  7. Vasan, R.S. Biomarkers of Cardiovascular Disease. Circulation 2006, 113, 2335–2362. [Google Scholar] [CrossRef]
  8. Cui, K.; Huang, W.; Fan, J.; Lei, H. Midregional pro-atrial natriuretic peptide is a superior biomarker to N-terminal pro-B-type natriuretic peptide in the diagnosis of heart failure patients with preserved ejection fraction. Medicine 2018, 97, e12277. [Google Scholar] [CrossRef] [PubMed]
  9. Cowie, M. Clinical applications of B-type natriuretic peptide (BNP) testing. Eur. Heart J. 2003, 24, 1710–1718. [Google Scholar] [CrossRef] [PubMed]
  10. Heidenreich, P.A.; Bozkurt, B.; Aguilar, D.; Allen, L.A.; Byun, J.J.; Colvin, M.M.; Deswal, A.; Drazner, M.H.; Dunlay, S.M.; Evers, L.R.; et al. 2022 AHA/ACC/HFSA Guideline for the Management of Heart Failure. J. Am. Coll. Cardiol. 2022, 79, e263–e421. [Google Scholar] [CrossRef] [PubMed]
  11. McDonagh, T.A.; Metra, M.; Adamo, M.; Gardner, R.S.; Baumbach, A.; Böhm, M.; Burri, H.; Butler, J.; Čelutkienė, J.; Chioncel, O.; et al. 2021 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2021, 42, 3599–3726. [Google Scholar] [CrossRef] [PubMed]
  12. Gaggin, H.K.; Januzzi, J.L. Biomarkers and diagnostics in heart failure. Biochim. Biophys. Acta-Mol. Basis Dis. 2013, 1832, 2442–2450. [Google Scholar] [CrossRef] [PubMed]
  13. Madamanchi, C.; Alhosaini, H.; Sumida, A.; Runge, M.S. Obesity and natriuretic peptides, BNP and NT-proBNP: Mechanisms and diagnostic implications for heart failure. Int. J. Cardiol. 2014, 176, 611–617. [Google Scholar] [CrossRef]
  14. Masson, S. Direct Comparison of B-Type Natriuretic Peptide (BNP) and Amino-Terminal proBNP in a Large Population of Patients with Chronic and Symptomatic Heart Failure: The Valsartan Heart Failure (Val-HeFT) Data. Clin. Chem. 2006, 52, 1528–1538. [Google Scholar] [CrossRef] [PubMed]
  15. Januzzi, J.L.; Chen-Tournoux, A.A.; Christenson, R.H.; Doros, G.; Hollander, J.E.; Levy, P.D.; Nagurney, J.T.; Nowak, R.M.; Pang, P.S.; Patel, D.; et al. N-Terminal Pro–B-Type Natriuretic Peptide in the Emergency Department. J. Am. Coll. Cardiol. 2018, 71, 1191–1200. [Google Scholar] [CrossRef] [PubMed]
  16. Mueller, C.; Scholer, A.; Laule-Kilian, K.; Martina, B.; Schindler, C.; Buser, P.; Pfisterer, M.; Perruchoud, A.P. Use of B-Type Natriuretic Peptide in the Evaluation and Management of Acute Dyspnea. N. Engl. J. Med. 2004, 350, 647–654. [Google Scholar] [CrossRef] [PubMed]
  17. Mueller, C.; McDonald, K.; de Boer, R.A.; Maisel, A.; Cleland, J.G.F.; Kozhuharov, N.; Coats, A.J.S.; Metra, M.; Mebazaa, A.; Ruschitzka, F.; et al. Heart Failure Association of the European Society of Cardiology practical guidance on the use of natriuretic peptide concentrations. Eur. J. Heart Fail. 2019, 21, 715–731. [Google Scholar] [CrossRef] [PubMed]
  18. Mayer, O.; Šimon, J.; Plášková, M.; Cífková, R.; Trefil, L. N-terminal pro B-type natriuretic peptide as prognostic marker for mortality in coronary patients without clinically manifest heart failure. Eur. J. Epidemiol. 2009, 24, 363–368. [Google Scholar] [CrossRef]
  19. Ananthan, K.; Lyon, A.R. The Role of Biomarkers in Cardio-Oncology. J. Cardiovasc. Transl. Res. 2020, 13, 431–450. [Google Scholar] [CrossRef]
  20. Hinrichs, L.; Mrotzek, S.M.; Mincu, R.-I.; Pohl, J.; Röll, A.; Michel, L.; Mahabadi, A.A.; Al-Rashid, F.; Totzeck, M.; Rassaf, T. Troponins and Natriuretic Peptides in Cardio-Oncology Patients—Data From the ECoR Registry. Front. Pharmacol. 2020, 11, 740. [Google Scholar] [CrossRef]
  21. Cartas-Espinel, I.; Telechea-Fernández, M.; Manterola Delgado, C.; Ávila Barrera, A.; Saavedra Cuevas, N.; Riffo-Campos, A.L. Novel molecular biomarkers of cancer therapy-induced cardiotoxicity in adult population: A scoping review. ESC Heart Fail. 2022, 9, 1651–1665. [Google Scholar] [CrossRef] [PubMed]
  22. McCullough, P.A.; Nowak, R.M.; McCord, J.; Hollander, J.E.; Herrmann, H.C.; Steg, P.G.; Duc, P.; Westheim, A.; Omland, T.; Knudsen, C.W.; et al. B-Type Natriuretic Peptide and Clinical Judgment in Emergency Diagnosis of Heart Failure. Circulation 2002, 106, 416–422. [Google Scholar] [CrossRef] [PubMed]
  23. Lenihan, D.J.; Stevens, P.L.; Massey, M.; Plana, J.C.; Araujo, D.M.; Fanale, M.A.; Fayad, L.E.; Fisch, M.J.; Yeh, E.T.H. The Utility of Point-of-Care Biomarkers to Detect Cardiotoxicity During Anthracycline Chemotherapy: A Feasibility Study. J. Card. Fail. 2016, 22, 433–438. [Google Scholar] [CrossRef] [PubMed]
  24. Wieshammer, S.; Dreyhaupt, J.; Müller, D.; Momm, F.; Jakob, A. Limitations of N-Terminal Pro-B-Type Natriuretic Peptide in the Diagnosis of Heart Disease among Cancer Patients Who Present with Cardiac or Pulmonary Symptoms. Oncology 2016, 90, 143–150. [Google Scholar] [CrossRef] [PubMed]
  25. Hayakawa, H.; Komada, Y.; Hirayama, M.; Hori, H.; Ito, M.; Sakurai, M. Plasma levels of natriuretic peptides in relation to doxorubicin-induced cardiotoxicity and cardiac function in children with cancer. Med. Pediatr. Oncol. 2001, 37, 4–9. [Google Scholar] [CrossRef] [PubMed]
  26. Pavo, N.; Raderer, M.; Hülsmann, M.; Neuhold, S.; Adlbrecht, C.; Strunk, G.; Goliasch, G.; Gisslinger, H.; Steger, G.G.; Hejna, M.; et al. Cardiovascular biomarkers in patients with cancer and their association with all-cause mortality. Heart 2015, 101, 1874–1880. [Google Scholar] [CrossRef]
  27. Daugaard, G.; Lassen, U.; Bie, P.; Pedersen, E.B.; Jensen, K.T.; Abildgaard, U.; Hesse, B.; Kjaer, A. Natriuretic peptides in the monitoring of anthracycline induced reduction in left ventricular ejection fraction. Eur. J. Heart Fail. 2005, 7, 87–93. [Google Scholar] [CrossRef]
  28. De Iuliis, F.; Salerno, G.; Taglieri, L.; De Biase, L.; Lanza, R.; Cardelli, P.; Scarpa, S. Serum biomarkers evaluation to predict chemotherapy-induced cardiotoxicity in breast cancer patients. Tumor Biol. 2016, 37, 3379–3387. [Google Scholar] [CrossRef]
  29. Zardavas, D.; Suter, T.M.; Van Veldhuisen, D.J.; Steinseifer, J.; Noe, J.; Lauer, S.; Al-Sakaff, N.; Piccart-Gebhart, M.J.; de Azambuja, E. Role of Troponins I and T and N -Terminal Prohormone of Brain Natriuretic Peptide in Monitoring Cardiac Safety of Patients With Early-Stage Human Epidermal Growth Factor Receptor 2–Positive Breast Cancer Receiving Trastuzumab: A Herceptin Adjuvant Study C. J. Clin. Oncol. 2017, 35, 878–884. [Google Scholar] [CrossRef]
  30. Chauin, A. The Main Causes and Mechanisms of Increase in Cardiac Troponin Concentrations Other Than Acute Myocardial Infarction (Part 1): Physical Exertion, Inflammatory Heart Disease, Pulmonary Embolism, Renal Failure, Sepsis. Vasc. Health Risk Manag. 2021, 17, 601–617. [Google Scholar] [CrossRef]
  31. Specchia, G.; Buquicchio, C.; Pansini, N.; Di Serio, F.; Liso, V.; Pastore, D.; Greco, G.; Ciuffreda, L.; Mestice, A.; Liso, A. Monitoring of cardiac function on the basis of serum troponin I levels in patients with acute leukemia treated with anthracyclines. J. Lab. Clin. Med. 2005, 145, 212–220. [Google Scholar] [CrossRef] [PubMed]
  32. Jones, M.; O’Gorman, P.; Kelly, C.; Mahon, N.; Fitzgibbon, M.C. High-sensitive cardiac troponin-I facilitates timely detection of subclinical anthracycline-mediated cardiac injury. Ann. Clin. Biochem. Int. J. Lab. Med. 2017, 54, 149–157. [Google Scholar] [CrossRef]
  33. Nisticò, C.; Bria, E.; Cuppone, F.; Carpino, A.; Ferretti, G.; Vitelli, G.; Sperduti, I.; Calabretta, F.; Toglia, G.; Tomao, S.; et al. Troponin-T and myoglobin plus echocardiographic evaluation for monitoring early cardiotoxicity of weekly epirubicin–paclitaxel in metastatic breast cancer patients. Anticancer Drugs 2007, 18, 227–232. [Google Scholar] [CrossRef] [PubMed]
  34. Pudil, R.; Mueller, C.; Čelutkienė, J.; Henriksen, P.A.; Lenihan, D.; Dent, S.; Barac, A.; Stanway, S.; Moslehi, J.; Suter, T.M.; et al. Role of serum biomarkers in cancer patients receiving cardiotoxic cancer therapies: A position statement from the Cardio-Oncology Study Group of the Heart Failure Association and the Cardio-Oncology Council of the European Socie. Eur. J. Heart Fail. 2020, 22, 1966–1983. [Google Scholar] [CrossRef] [PubMed]
  35. Michel, L.; Mincu, R.I.; Mahabadi, A.A.; Settelmeier, S.; Al-Rashid, F.; Rassaf, T.; Totzeck, M. Troponins and brain natriuretic peptides for the prediction of cardiotoxicity in cancer patients: A meta-analysis. Eur. J. Heart Fail. 2020, 22, 350–361. [Google Scholar] [CrossRef] [PubMed]
  36. Avila, M.S.; Ayub-Ferreira, S.M.; de Barros Wanderley, M.R.; das Dores Cruz, F.; Gonçalves Brandão, S.M.; Rigaud, V.O.C.; Higuchi-dos-Santos, M.H.; Hajjar, L.A.; Kalil Filho, R.; Hoff, P.M.; et al. Carvedilol for Prevention of Chemotherapy-Related Cardiotoxicity: The CECCY Trial. J. Am. Coll. Cardiol. 2018, 71, 2281–2290. [Google Scholar] [CrossRef] [PubMed]
  37. Gulati, G.; Heck, S.L.; Røsjø, H.; Ree, A.H.; Hoffmann, P.; Hagve, T.A.; Norseth, J.; Gravdehaug, B.; Steine, K.; Geisler, J.; et al. Neurohormonal Blockade and Circulating Cardiovascular Biomarkers During Anthracycline Therapy in Breast Cancer Patients: Results From the PRADA (Prevention of Cardiac Dysfunction During Adjuvant Breast Cancer Therapy) Study. J. Am. Heart Assoc. 2017, 6, e006513. [Google Scholar] [CrossRef]
  38. Gulati, G.; Heck, S.L.; Ree, A.H.; Hoffmann, P.; Schulz-Menger, J.; Fagerland, M.W.; Gravdehaug, B.; von Knobelsdorff-Brenkenhoff, F.; Bratland, Å.; Storås, T.H.; et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): A 2 × 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol. Eur. Heart J. 2016, 37, 1671–1680. [Google Scholar] [CrossRef]
  39. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Hear. J.-Cardiovasc. Imaging 2022, 23, e333–e465. [Google Scholar] [CrossRef]
  40. Morawiec, B.; Kawecki, D. Copeptin. J. Cardiovasc. Med. 2013, 14, 19–25. [Google Scholar] [CrossRef]
  41. Nelson, J.; Bagnato, A.; Battistini, B.; Nisen, P. The endothelin axis: Emerging role in cancer. Nat. Rev. Cancer 2003, 3, 110–116. [Google Scholar] [CrossRef]
  42. Wigle, D.A.; Campling, B.G.; Sarda, I.R.; Shin, S.H.; Watson, J.D.; Frater, Y.; Flynn, T.G.; Pang, S.C. ANP secretion from small cell lung cancer cell lines: A potential model of ANP release. Am. J. Physiol. Circ. Physiol. 1995, 268, H1869–H1874. [Google Scholar] [CrossRef]
  43. Ohsaki, Y.; Gross, A.J.; Le, P.T.; Oie, H.; Johnson, B. Human Small Cell Lung Cancer Cells Produce Brain Natriuretic Peptide. Oncology 1999, 56, 155–159. [Google Scholar] [CrossRef]
  44. Popat, J.; Rivero, A.; Pratap, P.; Guglin, M. What Is Causing Extremely Elevated Amino Terminal Brain Natriuretic Peptide in Cancer Patients? Congest. Heart Fail. 2013, 19, 143–148. [Google Scholar] [CrossRef] [PubMed]
  45. Burjonroppa, S.C.; Tong, A.T.; Xiao, L.-C.; Johnson, M.M.; Yusuf, S.W.; Lenihan, D.J. Cancer Patients With Markedly Elevated B-Type Natriuretic Peptide May Not Have Volume Overload. Am. J. Clin. Oncol. 2007, 30, 287–293. [Google Scholar] [CrossRef] [PubMed]
  46. Lim, E.; Li Choy, L.; Flaks, L.; Mussa, S.; Van Tornout, F.; Van Leuven, M.; Parry, G.W. Detected troponin elevation is associated with high early mortality after lung resection for cancer. J. Cardiothorac. Surg. 2006, 1, 37. [Google Scholar] [CrossRef]
  47. Danese, E.; Montagnana, M.; Giudici, S.; Aloe, R.; Franchi, M.; Guidi, G.C.; Lippi, G. Highly-sensitive troponin I is increased in patients with gynecological cancers. Clin. Biochem. 2013, 46, 1135–1138. [Google Scholar] [CrossRef] [PubMed]
  48. Vonhaehling, S.; Doezhner, W.; Anker, S. Nutrition, metabolism, and the complex pathophysiology of cachexia in chronic heart failure. Cardiovasc. Res. 2007, 73, 298–309. [Google Scholar] [CrossRef]
  49. von Haehling, S.; Lainscak, M.; Springer, J.; Anker, S.D. Cardiac cachexia: A systematic overview. Pharmacol. Ther. 2009, 121, 227–252. [Google Scholar] [CrossRef]
  50. Springer, J.; Tschirner, A.; Haghikia, A.; von Haehling, S.; Lal, H.; Grzesiak, A.; Kaschina, E.; Palus, S.; Pötsch, M.; von Websky, K.; et al. Prevention of liver cancer cachexia-induced cardiac wasting and heart failure. Eur. Heart J. 2014, 35, 932–941. [Google Scholar] [CrossRef]
  51. Shi, C.; van der Wal, H.H.; Silljé, H.H.W.; Dokter, M.M.; van den Berg, F.; Huizinga, L.; Vriesema, M.; Post, J.; Anker, S.D.; Cleland, J.G.; et al. Tumour biomarkers: Association with heart failure outcomes. J. Intern. Med. 2020, 288, 207–218. [Google Scholar] [CrossRef] [PubMed]
  52. Varol, E.; Ozaydin, M.; Dogan, A.; Kosar, F. Tumour marker levels in patients with chronic heart failure. Eur. J. Heart Fail. 2005, 7, 840–843. [Google Scholar] [CrossRef] [PubMed]
  53. Yilmaz, M.B.; Nikolaou, M.; Cohen Solal, A. Tumour biomarkers in heart failure: Is there a role for CA-125? Eur. J. Heart Fail. 2011, 13, 579–583. [Google Scholar] [CrossRef] [PubMed]
  54. Faggiano, P.; DʼAloia, A.; Antonini-Canterin, F.; Vizzardi, E.; Nicolosi, G.L.; Cas, L.D. Tumour markers in chronic heart failure. Review of the literature and clinical implications. J. Cardiovasc. Med. 2006, 7, 573–579. [Google Scholar] [CrossRef] [PubMed]
  55. Swords, D.; Firpo, M.; Scaife, C.; Mulvihill, S. Biomarkers in pancreatic adenocarcinoma: Current perspectives. OncoTargets Ther. 2016, 9, 7459–7467. [Google Scholar] [CrossRef] [PubMed]
  56. Kosar, F.; Aksoy, Y.; Ozguntekin, G.; Ozerol, I.; Varol, E. Relationship between cytokines and tumour markers in patients with chronic heart failure. Eur. J. Heart Fail. 2006, 8, 270–274. [Google Scholar] [CrossRef]
  57. Ameri, P.; Bertero, E.; Meijers, W.C. Cancer is a comorbidity of heart failure. Eur. Heart J. 2023, 44, 1133–1135. [Google Scholar] [CrossRef]
  58. Meijers, W.C.; Maglione, M.; Bakker, S.J.L.; Oberhuber, R.; Kieneker, L.M.; De Jong, S.; Haubner, B.J.; Nagengast, W.B.; Lyon, A.R.; Van Der Vegt, B.; et al. Heart failure stimulates tumor growth by circulating factors. Circulation 2018, 138, 678–691. [Google Scholar] [CrossRef]
  59. Bertero, E.; Canepa, M.; Maack, C.; Ameri, P. Linking Heart Failure to Cancer. Circulation 2018, 138, 735–742. [Google Scholar] [CrossRef]
  60. Cuomo, A.; Rodolico, A.; Galdieri, A.; Russo, M.; Campi, G.; Franco, R.; Bruno, D.; Aran, L.; Carannante, A.; Attanasio, U.; et al. Heart Failure and Cancer: Mechanisms of Old and New Cardiotoxic Drugs in Cancer Patients. Card. Fail. Rev. 2019, 5, 112–118. [Google Scholar] [CrossRef]
  61. Cuomo, A.; Pirozzi, F.; Attanasio, U.; Franco, R.; Elia, F.; De Rosa, E.; Russo, M.; Ghigo, A.; Ameri, P.; Tocchetti, C.G.; et al. Cancer Risk in the Heart Failure Population: Epidemiology, Mechanisms, and Clinical Implications. Curr. Oncol. Rep. 2021, 23, 7. [Google Scholar] [CrossRef] [PubMed]
  62. Cuomo, A.; Paudice, F.; D’Angelo, G.; Perrotta, G.; Carannante, A.; Attanasio, U.; Iengo, M.; Fiore, F.; Tocchetti, C.G.; Mercurio, V.; et al. New-Onset Cancer in the HF Population: Epidemiology, Pathophysiology, and Clinical Management. Curr. Heart Fail. Rep. 2021, 18, 191–199. [Google Scholar] [CrossRef] [PubMed]
  63. Banke, A.; Schou, M.; Videbæk, L.; Møller, J.E.; Torp-Pedersen, C.; Gustafsson, F.; Dahl, J.S.; Køber, L.; Hildebrandt, P.R.; Gislason, G.H. Incidence of cancer in patients with chronic heart failure: A long-term follow-up study. Eur. J. Heart Fail. 2016, 18, 260–266. [Google Scholar] [CrossRef] [PubMed]
  64. Voors, A.A.; Anker, S.D.; Cleland, J.G.; Dickstein, K.; Filippatos, G.; van der Harst, P.; Hillege, H.L.; Lang, C.C.; ter Maaten, J.M.; Ng, L.; et al. A systems BIOlogy Study to Tailored Treatment in Chronic Heart Failure: Rationale, design, and baseline characteristics of BIOSTAT-CHF. Eur. J. Heart Fail. 2016, 18, 716–726. [Google Scholar] [CrossRef]
  65. Stanciu, A.E.; Stanciu, M.M.; Vatasescu, R.G. NT-proBNP and CA 125 levels are associated with increased pro-inflammatory cytokines in coronary sinus serum of patients with chronic heart failure. Cytokine 2018, 111, 13–19. [Google Scholar] [CrossRef] [PubMed]
  66. Núñez, J.; Miñana, G.; Núñez, E.; Chorro, F.J.; Bodí, V.; Sanchis, J. Clinical utility of antigen carbohydrate 125 in heart failure. Heart Fail. Rev. 2014, 19, 575–584. [Google Scholar] [CrossRef] [PubMed]
  67. Li, K.H.C.; Gong, M.; Li, G.; Baranchuk, A.; Liu, T.; Wong, M.C.S.; Jesuthasan, A.; Lai, R.W.C.; Lai, J.C.L.; Lee, A.P.W.; et al. Cancer antigen-125 and outcomes in acute heart failure: A systematic review and meta-analysis. Heart Asia 2018, 10, e011044. [Google Scholar] [CrossRef]
  68. Huang, F.; Zhang, K.; Chen, J.; Cai, Q.; Liu, X.; Wang, T.; Lv, Z.; Wang, J.; Huang, H. Elevation of carbohydrate antigen 125 in chronic heart failure may be caused by mechanical extension of mesothelial cells from serous cavity effusion. Clin. Biochem. 2013, 46, 1694–1700. [Google Scholar] [CrossRef]
  69. Yoon, J.Y.; Yang, D.H.; Cho, H.J.; Kim, N.K.; Kim, C.-Y.; Son, J.; Roh, J.-H.; Jang, S.Y.; Bae, M.H.; Lee, J.H.; et al. Serum levels of carbohydrate antigen 125 in combination with N-terminal pro-brain natriuretic peptide in patients with acute decompensated heart failure. Korean J. Intern. Med. 2019, 34, 811–818. [Google Scholar] [CrossRef]
  70. Piek, A.; Meijers, W.C.; Schroten, N.F.; Gansevoort, R.T.; de Boer, R.A.; Silljé, H.H.W. HE4 Serum Levels Are Associated with Heart Failure Severity in Patients With Chronic Heart Failure. J. Card. Fail. 2017, 23, 12–19. [Google Scholar] [CrossRef]
  71. de Boer, R.A.; Cao, Q.; Postmus, D.; Damman, K.; Voors, A.A.; Jaarsma, T.; van Veldhuisen, D.J.; Arnold, W.D.; Hillege, H.L.; Silljé, H.H.W. The WAP Four-Disulfide Core Domain Protein HE4: A Novel Biomarker for Heart Failure. JACC Heart Fail. 2013, 1, 164–169. [Google Scholar] [CrossRef]
  72. Piek, A.; Du, W.; de Boer, R.A.; Silljé, H.H.W. Novel heart failure biomarkers: Why do we fail to exploit their potential? Crit. Rev. Clin. Lab. Sci. 2018, 55, 246–263. [Google Scholar] [CrossRef]
  73. Mercurio, V.; Cuomo, A.; Dessalvi, C.C.; Deidda, M.; Di Lisi, D.; Novo, G.; Manganaro, R.; Zito, C.; Santoro, C.; Ameri, P.; et al. Redox imbalances in ageing and metabolic alterations: Implications in cancer and cardiac diseases. An overview from the working group of cardiotoxicity and cardioprotection of the Italian society of cardiology (SIC). Antioxidants 2020, 9, 641. [Google Scholar] [CrossRef]
  74. Prabhu, S.D. Cytokine-Induced Modulation of Cardiac Function. Circ. Res. 2004, 95, 1140–1153. [Google Scholar] [CrossRef]
  75. Wrigley, B.J.; Lip, G.Y.H.; Shantsila, E. The role of monocytes and inflammation in the pathophysiology of heart failure. Eur. J. Heart Fail. 2011, 13, 1161–1171. [Google Scholar] [CrossRef]
  76. Lippitz, B.E. Cytokine patterns in patients with cancer: A systematic review. Lancet Oncol. 2013, 14, e218–e228. [Google Scholar] [CrossRef]
  77. Pine, S.R.; Mechanic, L.E.; Enewold, L.; Chaturvedi, A.K.; Katki, H.A.; Zheng, Y.-L.; Bowman, E.D.; Engels, E.A.; Caporaso, N.E.; Harris, C.C. Increased Levels of Circulating Interleukin 6, Interleukin 8, C-Reactive Protein, and Risk of Lung Cancer. JNCI J. Natl. Cancer Inst. 2011, 103, 1112–1122. [Google Scholar] [CrossRef]
  78. Yan, G.; Liu, T.; Yin, L.; Kang, Z.; Wang, L. Levels of peripheral Th17 cells and serum Th17-related cytokines in patients with colorectal cancer: A meta-analysis. Cell. Mol. Biol. 2018, 64, 94–102. [Google Scholar] [CrossRef]
  79. Gwechenberger, M.; Hülsmann, M.; Berger, R.; Graf, S.; Springer, C.; Stanek, B.; Pacher, R. Interleukin-6 and B-type natriuretic peptide are independent predictors for worsening of heart failure in patients with progressive congestive heart failure. J. Heart Lung Transplant. 2004, 23, 839–844. [Google Scholar] [CrossRef]
  80. Lyon, A.R.; Dent, S.; Stanway, S.; Earl, H.; Brezden-Masley, C.; Cohen-Solal, A.; Tocchetti, C.G.; Moslehi, J.J.; Groarke, J.D.; Bergler-Klein, J.; et al. Baseline cardiovascular risk assessment in cancer patients scheduled to receive cardiotoxic cancer therapies: A position statement and new risk assessment tools from the Cardio-Oncology Study Group of the Heart Failure Association of the European Society of Cardiology in collaboration with the International Cardio-Oncology Society. Eur. J. Heart Fail. 2020, 22, 1945–1960. [Google Scholar] [CrossRef]
  81. Tini, G.; Cuomo, A.; Battistoni, A.; Sarocchi, M.; Mercurio, V.; Ameri, P.; Volpe, M.; Porto, I.; Tocchetti, C.G.; Spallarossa, P. Baseline cardio-oncologic risk assessment in breast cancer women and occurrence of cardiovascular events: The HFA/ICOS risk tool in real-world practice. Int. J. Cardiol. 2022, 349, 134–137. [Google Scholar] [CrossRef]
  82. Di Lisi, D.; Madaudo, C.; Alagna, G.; Santoro, M.; Rossetto, L.; Siragusa, S.; Novo, G. The new HFA/ICOS risk assessment tool to identify patients with chronic myeloid leukaemia at high risk of cardiotoxicity. ESC Heart Fail. 2022, 9, 1914–1919. [Google Scholar] [CrossRef] [PubMed]
  83. Cardinale, D.; Sandri, M.T.; Martinoni, A.; Tricca LabTech, A.; Civelli, M.; Lamantia, G.; Cinieri, S.; Martinelli, G.; Cipolla, C.M.; Fiorentini, C. Left ventricular dysfunction predicted by early troponin I release after high-dose chemotherapy. J. Am. Coll. Cardiol. 2000, 36, 517–522. [Google Scholar] [CrossRef]
  84. Cardinale, D.; Sandri, M.T.; Colombo, A.; Colombo, N.; Boeri, M.; Lamantia, G.; Civelli, M.; Peccatori, F.; Martinelli, G.; Fiorentini, C.; et al. Prognostic Value of Troponin I in Cardiac Risk Stratification of Cancer Patients Undergoing High-Dose Chemotherapy. Circulation 2004, 109, 2749–2754. [Google Scholar] [CrossRef]
  85. Cardinale, D.; Biasillo, G.; Salvatici, M.; Sandri, M.T.; Cipolla, C.M. Using biomarkers to predict and to prevent cardiotoxicity of cancer therapy. Expert Rev. Mol. Diagn. 2017, 17, 245–256. [Google Scholar] [CrossRef]
  86. Cardinale, D.; Colombo, A.; Torrisi, R.; Sandri, M.T.; Civelli, M.; Salvatici, M.; Lamantia, G.; Colombo, N.; Cortinovis, S.; Dessanai, M.A.; et al. Trastuzumab-Induced Cardiotoxicity: Clinical and Prognostic Implications of Troponin I Evaluation. J. Clin. Oncol. 2010, 28, 3910–3916. [Google Scholar] [CrossRef]
  87. Petricciuolo, S.; Delle Donne, M.G.; Aimo, A.; Chella, A.; De Caterina, R. Pre-treatment high-sensitivity troponin T for the short-term prediction of cardiac outcomes in patients on immune checkpoint inhibitors. Eur. J. Clin. Investig. 2021, 51, e13400. [Google Scholar] [CrossRef]
  88. Chen, L.L.; Dulu, A.O.; Pastores, S.M. Elevated brain natriuretic peptide in a patient with metastatic cancer without heart failure: A case study. J. Am. Assoc. Nurse Pract. 2023, 36, 73–76. [Google Scholar] [CrossRef]
  89. Cornell, R.F.; Ky, B.; Weiss, B.M.; Dahm, C.N.; Gupta, D.K.; Du, L.; Carver, J.R.; Cohen, A.D.; Engelhardt, B.G.; Garfall, A.L.; et al. Prospective Study of Cardiac Events During Proteasome Inhibitor Therapy for Relapsed Multiple Myeloma. J. Clin. Oncol. 2019, 37, 1946–1955. [Google Scholar] [CrossRef]
  90. Henriksen, P.A.; Hall, P.; MacPherson, I.R.; Joshi, S.S.; Singh, T.; Maclean, M.; Lewis, S.; Rodriguez, A.; Fletcher, A.; Everett, R.J.; et al. Multicenter, Prospective, Randomized Controlled Trial of High-Sensitivity Cardiac Troponin I–Guided Combination Angiotensin Receptor Blockade and Beta-Blocker Therapy to Prevent Anthracycline Cardiotoxicity: The Cardiac CARE Trial. Circulation 2023, 148, 1680–1690. [Google Scholar] [CrossRef]
  91. Tanaka, S.; Ikari, A.; Nitta, T.; Horiuchi, T. Long-term irreversible trastuzumab-induced cardiotoxicity for metastatic breast cancer in a patient without cardiac risk factors. Oxford Med. Case Rep. 2017, 2017, omx038. [Google Scholar] [CrossRef]
  92. Lunardi, M.; Al-Habbaa, A.; Abdelshafy, M.; Davey, M.G.; Elkoumy, A.; Ganly, S.; Elzomor, H.; Cawley, C.; Sharif, F.; Crowley, J.; et al. Genetic and RNA-related molecular markers of trastuzumab-chemotherapy-associated cardiotoxicity in HER2 positive breast cancer: A systematic review. BMC Cancer 2022, 22, 396. [Google Scholar] [CrossRef]
  93. Lestuzzi, C.; Stolfo, D.; De Paoli, A.; Banzato, A.; Buonadonna, A.; Bidoli, E.; Tartuferi, L.; Viel, E.; De Angelis, G.; Lonardi, S.; et al. Cardiotoxicity from Capecitabine Chemotherapy: Prospective Study of Incidence at Rest and During Physical Exercise. Oncologist 2022, 27, e158–e167. [Google Scholar] [CrossRef]
  94. Chen, H.X.; Cleck, J.N. Adverse effects of anticancer agents that target the VEGF pathway. Nat. Rev. Clin. Oncol. 2009, 6, 465–477. [Google Scholar] [CrossRef]
  95. Mercurio, V.; Cuomo, A.; Della Pepa, R.; Ciervo, D.; Cella, L.; Pirozzi, F.; Parrella, P.; Campi, G.; Franco, R.; Varricchi, G.; et al. What Is the Cardiac Impact of Chemotherapy and Subsequent Radiotherapy in Lymphoma Patients? Antioxid. Redox Signal. 2019, 31, 1166–1174. [Google Scholar] [CrossRef]
  96. EMA. Medicines European Medicines Agency. Eur Med Agency Sci Med Heal. Available online: https://www.ema.europa.eu/en/homepage (accessed on 29 December 2023).
  97. FDA. Drugs@FDA FDA-Approved Drugs; FDA US Food Drug Administration: Beltsville, MD, USA. Available online: https://www.fda.gov/drugs (accessed on 29 December 2023).
  98. Skála, M.; Hanousková, B.; Skálová, L.; Matoušková, P. MicroRNAs in the diagnosis and prevention of drug-induced cardiotoxicity. Arch. Toxicol. 2019, 93, 1–9. [Google Scholar] [CrossRef]
  99. Horie, T.; Ono, K.; Nishi, H.; Nagao, K.; Kinoshita, M.; Watanabe, S.; Kuwabara, Y.; Nakashima, Y.; Takanabe-Mori, R.; Nishi, E.; et al. Acute doxorubicin cardiotoxicity is associated with miR-146a-induced inhibition of the neuregulin-ErbB pathway. Cardiovasc. Res. 2010, 87, 656–664. [Google Scholar] [CrossRef]
  100. Mahmood, S.S.; Fradley, M.G.; Cohen, J.V.; Nohria, A.; Reynolds, K.L.; Heinzerling, L.M.; Sullivan, R.J.; Damrongwatanasuk, R.; Chen, C.L.; Gupta, D.; et al. Myocarditis in Patients Treated With Immune Checkpoint Inhibitors. J. Am. Coll. Cardiol. 2018, 71, 1755–1764. [Google Scholar] [CrossRef]
  101. Lee, D.W.; Gardner, R.; Porter, D.L.; Louis, C.U.; Ahmed, N.; Jensen, M.; Grupp, S.A.; Mackall, C.L. Current concepts in the diagnosis and management of cytokine release syndrome. Blood 2014, 124, 188–195. [Google Scholar] [CrossRef]
  102. Onitilo, A.A.; Engel, J.M.; Stankowski, R.V.; Liang, H.; Berg, R.L.; Doi, S.A.R. High-sensitivity C-reactive protein (hs-CRP) as a biomarker for trastuzumab-induced cardiotoxicity in HER2-positive early-stage breast cancer: A pilot study. Breast Cancer Res. Treat. 2012, 134, 291–298. [Google Scholar] [CrossRef]
  103. Kopeva, K.V.; Grakova, E.V.; Shilov, S.N.; Popova, A.A.; Berezikova, E.N.; Neupokoeva, M.N.; Ratushnyak, E.T.; Teplyakov, A.T. Anthra cycline-Induced Cardiotoxicity: The Role of Genetic Predictors. Kardiologiia 2023, 63, 22–28. [Google Scholar] [CrossRef]
  104. Wells, Q.S.; Veatch, O.J.; Fessel, J.P.; Joon, A.Y.; Levinson, R.T.; Mosley, J.D.; Held, E.P.; Lindsay, C.S.; Shaffer, C.M.; Weeke, P.E.; et al. Genome-wide association and pathway analysis of left ventricular function after anthracycline exposure in adults. Pharmacogenet. Genom. 2017, 27, 247–254. [Google Scholar] [CrossRef]
  105. Magdy, T.; Jouni, M.; Kuo, H.-H.; Weddle, C.J.; Lyra-Leite, D.; Fonoudi, H.; Romero-Tejeda, M.; Gharib, M.; Javed, H.; Fajardo, G.; et al. Identification of Drug Transporter Genomic Variants and Inhibitors That Protect Against Doxorubicin-Induced Cardiotoxicity. Circulation 2022, 145, 279–294. [Google Scholar] [CrossRef] [PubMed]
  106. Udagawa, C.; Nakamura, H.; Ohnishi, H.; Tamura, K.; Shimoi, T.; Yoshida, M.; Yoshida, T.; Totoki, Y.; Shibata, T.; Zembutsu, H. Whole exome sequencing to identify genetic markers for trastuzumab-induced cardiotoxicity. Cancer Sci. 2018, 109, 446–452. [Google Scholar] [CrossRef]
  107. Etienne-Grimaldi, M.; Formento, P.; Degeorges, A.; Pierga, J.; Delva, R.; Pivot, X.; Dalenc, F.; Espié, M.; Veyret, C.; Formento, J.; et al. Prospective analysis of the impact of VEGF-A gene polymorphisms on the pharmacodynamics of bevacizumab-based therapy in metastatic breast cancer patients. Br. J. Clin. Pharmacol. 2011, 71, 921–928. [Google Scholar] [CrossRef]
  108. Wang, Y.; Zheng, Y.; Zhang, W.; Yu, H.; Lou, K.; Zhang, Y.; Qin, Q.; Zhao, B.; Yang, Y.; Hui, R. Polymorphisms of KDRGene Are Associated With Coronary Heart Disease. J. Am. Coll. Cardiol. 2007, 50, 760–767. [Google Scholar] [CrossRef]
  109. Plana, J.C.; Galderisi, M.; Barac, A.; Ewer, M.S.; Ky, B.; Scherrer-Crosbie, M.; Ganame, J.; Sebag, I.A.; Agler, D.A.; Badano, L.P.; et al. Expert Consensus for Multimodality Imaging Evaluation of Adult Patients during and after Cancer Therapy: A Report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J. Am. Soc. Echocardiogr. 2014, 27, 911–939. [Google Scholar] [CrossRef]
  110. Armenian, S.H.; Hudson, M.M.; Mulder, R.L.; Chen, M.H.; Constine, L.S.; Dwyer, M.; Nathan, P.C.; Tissing, W.J.E.; Shankar, S.; Sieswerda, E.; et al. Recommendations for cardiomyopathy surveillance for survivors of childhood cancer: A report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet Oncol. 2015, 16, e123–e136. [Google Scholar] [CrossRef] [PubMed]
  111. Zamorano, J.L.; Lancellotti, P.; Rodriguez Muñoz, D.; Aboyans, V.; Asteggiano, R.; Galderisi, M.; Habib, G.; Lenihan, D.J.; Lip, G.Y.H.; Lyon, A.R.; et al. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines. Eur. Heart J. 2016, 37, 2768–2801. [Google Scholar] [CrossRef] [PubMed]
  112. Feola, M.; Garrone, O.; Occelli, M.; Francini, A.; Biggi, A.; Visconti, G.; Albrile, F.; Bobbio, M.; Merlano, M. Cardiotoxicity after anthracycline chemotherapy in breast carcinoma: Effects on left ventricular ejection fraction, troponin I and brain natriuretic peptide. Int. J. Cardiol. 2011, 148, 194–198. [Google Scholar] [CrossRef]
  113. Demissei, B.G.; Hubbard, R.A.; Zhang, L.; Smith, A.M.; Sheline, K.; McDonald, C.; Narayan, V.; Domchek, S.M.; DeMichele, A.; Shah, P.; et al. Changes in Cardiovascular Biomarkers With Breast Cancer Therapy and Associations With Cardiac Dysfunction. J. Am. Heart Assoc. 2020, 9, e014708. [Google Scholar] [CrossRef] [PubMed]
  114. López-Sendón, J.; Álvarez-Ortega, C.; Zamora Auñon, P.; Buño Soto, A.; Lyon, A.R.; Farmakis, D.; Cardinale, D.; Canales Albendea, M.; Feliu Batlle, J.; Rodríguez Rodríguez, I.; et al. Classification, prevalence, and outcomes of anticancer therapy-induced cardiotoxicity: The CARDIOTOX registry. Eur. Heart J. 2020, 41, 1720–1729. [Google Scholar] [CrossRef] [PubMed]
  115. Liu, J.E.; Barac, A.; Thavendiranathan, P.; Scherrer-Crosbie, M. Strain Imaging in Cardio-Oncology. JACC Cardio Oncol. 2020, 2, 677–689. [Google Scholar] [CrossRef] [PubMed]
  116. Brown, J.; Jenkins, C.; Marwick, T.H. Use of myocardial strain to assess global left ventricular function: A comparison with cardiac magnetic resonance and 3-dimensional echocardiography. Am. Heart J. 2009, 157, 102.e1–102.e5. [Google Scholar] [CrossRef]
  117. Tadic, M.; Genger, M.; Baudisch, A.; Kelle, S.; Cuspidi, C.; Belyavskiy, E.; Burkhardt, F.; Venneri, L.; Attanasio, P.; Pieske, B. Left Ventricular Strain in Chemotherapy-Naive and Radiotherapy-Naive Patients With Cancer. Can. J. Cardiol. 2018, 34, 281–287. [Google Scholar] [CrossRef] [PubMed]
  118. Sawaya, H.; Sebag, I.A.; Plana, J.C.; Januzzi, J.L.; Ky, B.; Tan, T.C.; Cohen, V.; Banchs, J.; Carver, J.R.; Wiegers, S.E.; et al. Assessment of Echocardiography and Biomarkers for the Extended Prediction of Cardiotoxicity in Patients Treated With Anthracyclines, Taxanes, and Trastuzumab. Circ. Cardiovasc. Imaging 2012, 5, 596–603. [Google Scholar] [CrossRef]
  119. Liang, S.; Liu, Z.; Li, Q.; He, W.; Huang, H. Advance of echocardiography in cardiac amyloidosis. Heart Fail. Rev. 2023, 28, 1345–1356. [Google Scholar] [CrossRef]
  120. Liu, Z.; Zhang, L.; Liu, M.; Wang, F.; Xiong, Y.; Tang, Z.; Li, Q.; Lu, Q.; Liang, S.; Niu, T.; et al. Myocardial Injury in Multiple Myeloma Patients With Preserved Left Ventricular Ejection Fraction: Noninvasive Left Ventricular Pressure-Strain Myocardial Work. Front. Cardiovasc. Med. 2022, 8, 782580. [Google Scholar] [CrossRef]
  121. Alexandraki, A.; Papageorgiou, E.; Zacharia, M.; Keramida, K.; Papakonstantinou, A.; Cipolla, C.M.; Tsekoura, D.; Naka, K.; Mazzocco, K.; Mauri, D.; et al. New Insights in the Era of Clinical Biomarkers as Potential Predictors of Systemic Therapy-Induced Cardiotoxicity in Women with Breast Cancer: A Systematic Review. Cancers 2023, 15, 3290. [Google Scholar] [CrossRef]
  122. Tocchetti, C.G.; Ameri, P.; de Boer, R.A.; D’Alessandra, Y.; Russo, M.; Sorriento, D.; Ciccarelli, M.; Kiss, B.; Bertrand, L.; Dawson, D.; et al. Cardiac dysfunction in cancer patients: Beyond direct cardiomyocyte damage of anticancer drugs: Novel cardio-oncology insights from the joint 2019 meeting of the ESC working groups of myocardial function and cellular biology of the heart. Cardiovasc. Res. 2020, 116, 1820–1834. [Google Scholar] [CrossRef]
  123. Friedman, R.C.; Farh, K.K.H.; Burge, C.B.; Bartel, D.P. Most mammalian mRNAs are conserved targets of microRNAs. Genome Res. 2009, 19, 92–105. [Google Scholar] [CrossRef]
  124. Brown, C.; Mantzaris, M.; Nicolaou, E.; Karanasiou, G.; Papageorgiou, E.; Curigliano, G.; Cardinale, D.; Filippatos, G.; Memos, N.; Naka, K.K.; et al. A systematic review of miRNAs as biomarkers for chemotherapy-induced cardiotoxicity in breast cancer patients reveals potentially clinically informative panels as well as key challenges in miRNA research. Cardio-Oncology 2022, 81, 1589–1600. [Google Scholar] [CrossRef]
  125. Lakhani, H.V.; Pillai, S.S.; Zehra, M.; Dao, B.; Tirona, M.T.; Thompson, E.; Sodhi, K. Detecting early onset of anthracyclines-induced cardiotoxicity using a novel panel of biomarkers in West-Virginian population with breast cancer. Sci. Rep. 2021, 11, 7954. [Google Scholar] [CrossRef] [PubMed]
  126. Putt, M.; Hahn, V.S.; Januzzi, J.L.; Sawaya, H.; Sebag, I.A.; Plana, J.C.; Picard, M.H.; Carver, J.R.; Halpern, E.F.; Kuter, I.; et al. Longitudinal Changes in Multiple Biomarkers Are Associated with Cardiotoxicity in Breast Cancer Patients Treated with Doxorubicin, Taxanes, and Trastuzumab. Clin. Chem. 2015, 61, 1164–1172. [Google Scholar] [CrossRef] [PubMed]
  127. Sharma, U.C.; Pokharel, S.; Brakel, T.J.; Berlo, J.H.; Cleutjens, J.P.; Schroen, B.; André, S.; Crijns, H.J.; Gabius, H.J.; Maessen, J.; et al. Galectin-3 marks activated macrophages in failure-prone hypertrophied hearts and contributes to cardiac dysfunction. Circulation 2004, 110, 3121–3128. [Google Scholar] [CrossRef] [PubMed]
  128. Van Boxtel, W.; Bulten, B.F.; Mavinkurve-Groothuis, A.M.C.; Bellersen, L.; Mandigers, C.M.P.W.; Joosten, L.A.B.; Kapusta, L.; De Geus-Oei, L.F.; Van Laarhoven, H.W.M. New biomarkers for early detection of cardiotoxicity after treatment with docetaxel, doxorubicin and cyclophosphamide. Biomarkers 2015, 20, 143–148. [Google Scholar] [CrossRef] [PubMed]
  129. Seidman, A.; Hudis, C.; Kathryn Pierri, M.; Shak, S.; Paton, V.; Ashby, M.; Murphy, M.; Stewart, S.J.; Keefe, D. Cardiac dysfunction in the trastuzumab clinical trials experience. J. Clin. Oncol. 2002, 69, 706–708. [Google Scholar] [CrossRef] [PubMed]
  130. Hasan, D.; Ismail, Y.; Al Tibi, A.; AL-Zeidaneen, S.A.; Ode, M.; Burghel, G.J.; Natsheh, I.; Abdelnour, A. Serum Biomarkers for Chemotherapy Cardiotoxicity Risk Detection of Breast Cancer Patients. Asian Pacific. J. Cancer Prev. 2021, 22, 3355–3363. [Google Scholar] [CrossRef] [PubMed]
  131. Micheletti, P.L.; Carla-da-Silva, J.; Scandolara, T.B.; Kern, R.; Alves, V.D.; Malanowski, J.; Victorino, V.J.; Herrera, A.C.S.A.; Rech, D.; Souza, J.A.O.; et al. Proinflammatory circulating markers: New players for evaluating asymptomatic acute cardiovascular toxicity in breast cancer treatment. J. Chemother. 2021, 33, 106–115. [Google Scholar] [CrossRef] [PubMed]
  132. Ky, B.; Putt, M.; Sawaya, H.; French, B.; Januzzi, J.L.; Sebag, I.A.; Plana, J.C.; Cohen, V.; Banchs, J.; Carver, J.R.; et al. Early Increases in Multiple Biomarkers Predict Subsequent Cardiotoxicity in Patients With Breast Cancer Treated With Doxorubicin, Taxanes, and Trastuzumab. J. Am. Coll. Cardiol. 2014, 63, 809–816. [Google Scholar] [CrossRef]
  133. Assadi, A.; Zahabi, A.; Hart, R.A. GDF15, an update of the physiological and pathological roles it plays: A review. Pflügers Arch.-Eur. J. Physiol. 2020, 472, 1535–1546. [Google Scholar] [CrossRef]
  134. Kirkham, A.A.; Pituskin, E.; Thompson, R.B.; MacKey, J.R.; Koshman, S.L.; Jassal, D.; Pitz, M.; Haykowsky, M.J.; Pagano, J.J.; Chow, K.; et al. Cardiac and cardiometabolic phenotyping of trastuzumab-mediated cardiotoxicity: A secondary analysis of the MANTICORE trial. Eur. Heart J.-Cardiovasc. Pharmacother. 2022, 8, 130–139. [Google Scholar] [CrossRef]
  135. Tromp, J.; Boerman, L.M.; Sama, I.E.; Maass, S.W.M.C.; Maduro, J.H.; Hummel, Y.M.; Berger, M.Y.; de Bock, G.H.; Gietema, J.A.; Berendsen, A.J.; et al. Long-term survivors of early breast cancer treated with chemotherapy are characterized by a pro-inflammatory biomarker profile compared to matched controls. Eur. J. Heart Fail. 2020, 22, 1239–1246. [Google Scholar] [CrossRef]
  136. Varkoly, K.; Tan, S.; Beladi, R.; Fonseca, D.; Zanetti, I.R.; Kraberger, S.; Shah, C.; Yaron, J.R.; Zhang, L.; Juby, M.; et al. RNA Virus Gene Signatures Detected in Patients With Cardiomyopathy After Chemotherapy; A Pilot Study. Front. Cardiovasc. Med. 2022, 9, 821162. [Google Scholar] [CrossRef] [PubMed]
  137. Deidda, M.; Mercurio, V.; Cuomo, A.; Noto, A.; Mercuro, G.; Cadeddu Dessalvi, C. Metabolomic Perspectives in Antiblastic Cardiotoxicity and Cardioprotection. Int. J. Mol. Sci. 2019, 20, 4928. [Google Scholar] [CrossRef] [PubMed]
  138. Palmer, J.A.; Smith, A.M.; Gryshkova, V.; Donley, E.L.R.; Valentin, J.-P.; Burrier, R.E. A Targeted Metabolomics-Based Assay Using Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Identifies Structural and Functional Cardiotoxicity Potential. Toxicol. Sci. 2020, 174, 218–240. [Google Scholar] [CrossRef] [PubMed]
  139. Dionísio, F.; Araújo, A.M.; Duarte-Araújo, M.; Bastos, M.d.L.; Guedes de Pinho, P.; Carvalho, F.; Costa, V.M. Cardiotoxicity of cyclophosphamide’s metabolites: An in vitro metabolomics approach in AC16 human cardiomyocytes. Arch. Toxicol. 2022, 96, 653–671. [Google Scholar] [CrossRef] [PubMed]
  140. Li, Y.; Ju, L.; Hou, Z.; Deng, H.; Zhang, Z.; Wang, L.; Yang, Z.; Yin, J.; Zhang, Y. Screening, Verification, and Optimization of Biomarkers for Early Prediction of Cardiotoxicity Based on Metabolomics. J. Proteome Res. 2015, 14, 2437–2445. [Google Scholar] [CrossRef]
  141. Schnackenberg, L.K.; Pence, L.; Vijay, V.; Moland, C.L.; George, N.; Cao, Z.; Yu, L.-R.; Fuscoe, J.C.; Beger, R.D.; Desai, V.G. Early metabolomics changes in heart and plasma during chronic doxorubicin treatment in B6C3F 1 mice. J. Appl. Toxicol. 2016, 36, 1486–1495. [Google Scholar] [CrossRef]
  142. Tan, G.; Lou, Z.; Liao, W.; Zhu, Z.; Dong, X.; Zhang, W.; Li, W.; Chai, Y. Potential Biomarkers in Mouse Myocardium of Doxorubicin-Induced Cardiomyopathy: A Metabonomic Method and Its Application. PLoS ONE 2011, 6, e27683. [Google Scholar] [CrossRef]
  143. Jensen, B.; Parry, T.; Huang, W.; Ilaiwy, A.; Bain, J.; Muehlbauer, M.; O’Neal, S.; Patterson, C.; Johnson, G.; Willis, M. Non-Targeted Metabolomics Analysis of the Effects of Tyrosine Kinase Inhibitors Sunitinib and Erlotinib on Heart, Muscle, Liver and Serum Metabolism In Vivo. Metabolites 2017, 7, 31. [Google Scholar] [CrossRef]
  144. Jensen, B.C.; Parry, T.L.; Huang, W.; Beak, J.Y.; Ilaiwy, A.; Bain, J.R.; Newgard, C.B.; Muehlbauer, M.J.; Patterson, C.; Johnson, G.L.; et al. Effects of the kinase inhibitor sorafenib on heart, muscle, liver and plasma metabolism in vivo using non-targeted metabolomics analysis. Br. J. Pharmacol. 2017, 174, 4797–4811. [Google Scholar] [CrossRef]
  145. Unger, K.; Li, Y.; Yeh, C.; Barac, A.; Srichai, M.B.; Ballew, E.A.; Girgis, M.; Jayatilake, M.; Sridharan, V.; Boerma, M.; et al. Plasma metabolite biomarkers predictive of radiation induced cardiotoxicity. Radiother. Oncol. 2020, 152, 133–145. [Google Scholar] [CrossRef]
  146. Asnani, A.; Shi, X.; Farrell, L.; Lall, R.; Sebag, I.A.; Plana, J.C.; Gerszten, R.E.; Scherrer-Crosbie, M. Changes in Citric Acid Cycle and Nucleoside Metabolism Are Associated with Anthracycline Cardiotoxicity in Patients with Breast Cancer. J. Cardiovasc. Transl. Res. 2020, 13, 349–356. [Google Scholar] [CrossRef] [PubMed]
  147. Cocco, D.; Ferro, E.; Ricci, S.; Deidda, M.; Noto, A.; Madeddu, C.; Atzori, F.; Scartozzi, M.; Mercuro, G.; Cadeddu Dessalvi, C. Defining the metabolomic profile associated with early cardiotoxicity in patients with breast cancer treated with anthracyclines. Eur. Heart J. 2020, 41, ehaa946.3289. [Google Scholar] [CrossRef]
  148. Tantawy, M.; Chekka, L.M.; Huang, Y.; Garrett, T.J.; Singh, S.; Shah, C.P.; Cornell, R.F.; Baz, R.C.; Fradley, M.G.; Waheed, N.; et al. Lactate Dehydrogenase B and Pyruvate Oxidation Pathway Associated With Carfilzomib-Related Cardiotoxicity in Multiple Myeloma Patients: Result of a Multi-Omics Integrative Analysis. Front. Cardiovasc. Med. 2021, 8, 645122. [Google Scholar] [CrossRef] [PubMed]
  149. Yang, X.; Duan, X.; Xia, Z.; Huang, R.; He, K.; Xiang, G. The Regulation Network of Glycerolipid Metabolism as Coregulators of Immunotherapy-Related Myocarditis. Cardiovasc. Ther. 2023, 2023, 8774971. [Google Scholar] [CrossRef] [PubMed]
Table 1. List of the most used chemotherapy classes of agents and their possible cardiovascular toxicity. The boxes are marked if at least one of the compounds, belonging to the respective class, is shown to cause the reported effect with at least a common frequency (≥1/100) according to EMA and FDA prescribing information and to the most recent European guidelines on cardio-oncology [39,96,97].
Table 1. List of the most used chemotherapy classes of agents and their possible cardiovascular toxicity. The boxes are marked if at least one of the compounds, belonging to the respective class, is shown to cause the reported effect with at least a common frequency (≥1/100) according to EMA and FDA prescribing information and to the most recent European guidelines on cardio-oncology [39,96,97].
ArrhythmiaHFVascular ToxicityVTE/PESystemic HTNOther
Anthracyclines
Alkylating agents
Antimetabolites
Immunomodulatory
drugs
DM
Taxanes
Platinum-based agents
Androgen deprivation therapy
Proteasome inhibitors PH
HER2 inhibitors
VEGF inhibitors
BCR-ABL1 inhibitors Pleuro-pericardial Effusion
PH
ALK inhibitors Dyslipidaemia
DM
EGFR inhibitors
BRAF inhibitors Bleeding
MEK inhibitors Bleeding
Immuno-checkpoint inhibitors Myopericarditis
List of abbreviations: DM, diabetes mellitus; EMA, European Medicines Agency; FDA, Food and Drug Administration; HF, heart failure; HTN, hypertension; PE, pulmonary embolism; PH, pulmonary hypertension; VTE, venous thromboembolism.
Table 2. Examples of increased levels of serum biomarkers during specific oncological therapies.
Table 2. Examples of increased levels of serum biomarkers during specific oncological therapies.
Cancer TreatmentBiomarker of CardiotoxicityReferences
Anthracycline
Bevacizumab
Cyclosporine A
Isoprenaline
-
miR-146a, miR-1, miR-133a and miR-208
-
troponin I (>80 ng/L)
-
NT-proBNP (>100 ng/L)
Skála, M. et al. Arch Toxicol, 2019 [98];
Cardinale, D. et al. Circulation, 2004 [84];
Horie, T. et al. Cardiovascular Research, 2010 [99].
Immune checkpoint inhibitors
-
TnTc (>1.5 ng/L)
-
NPs (no clear cut-off)
Mahmood, S.S. et al. Journal of the American College of Cardiology, 2018 [100].
CAR-T cell therapies
-
TnTc (no clear cut-off)
Lee, D.W. et al. Blood, 2014 [101].
Proteasome inhibitors (bortezomib, carfilzomib and ixazomib)
-
NPs (no clear cut-off)
Cornell, R.F. et al. Journal of Clinical
Oncology, 2019 [89].
Trastuzumab
-
hsCRP (>3 mg)
-
NPs (no clear cut-off)
Onitilo, A.A., et al. Breast Cancer Research and Treatment, 2012 [102].
List of abbreviations: miR-146a, microRNA 146a; miR-1, microRNA-1; miR-133a, microRNA 133a-1; miR-208, microRNA-208; NT-proBNP, N-terminal pro-B-type natriuretic peptide; TnTc, cardiac troponin T; NPs, natriuretic peptides; CAR-T cell, chimeric antigen receptor T-cell; hsCRP, high-sensitivity C-reactive protein.
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

Attanasio, U.; Di Sarro, E.; Tricarico, L.; Di Lisi, D.; Armentaro, G.; Miceli, S.; Fioretti, F.; Deidda, M.; Correale, M.; Novo, G.; et al. Cardiovascular Biomarkers in Cardio-Oncology: Antineoplastic Drug Cardiotoxicity and Beyond. Biomolecules 2024, 14, 199. https://doi.org/10.3390/biom14020199

AMA Style

Attanasio U, Di Sarro E, Tricarico L, Di Lisi D, Armentaro G, Miceli S, Fioretti F, Deidda M, Correale M, Novo G, et al. Cardiovascular Biomarkers in Cardio-Oncology: Antineoplastic Drug Cardiotoxicity and Beyond. Biomolecules. 2024; 14(2):199. https://doi.org/10.3390/biom14020199

Chicago/Turabian Style

Attanasio, Umberto, Elena Di Sarro, Lucia Tricarico, Daniela Di Lisi, Giuseppe Armentaro, Sofia Miceli, Francesco Fioretti, Martino Deidda, Michele Correale, Giuseppina Novo, and et al. 2024. "Cardiovascular Biomarkers in Cardio-Oncology: Antineoplastic Drug Cardiotoxicity and Beyond" Biomolecules 14, no. 2: 199. https://doi.org/10.3390/biom14020199

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