Next Article in Journal
Association between the Severity of Nonalcoholic Fatty Liver Disease and the Risk of Coronary Artery Calcification
Previous Article in Journal
Endocrinology in the Time of COVID-19: A Rapid Evolution of Knowledge and Care
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiotoxicity Associated with Chemotherapy Used in Gastrointestinal Tumours

by
Liliana Maria Radulescu
1,2,
Dan Radulescu
2,3,*,
Tudor-Eliade Ciuleanu
4,
Dana Crisan
3,5,
Elena Buzdugan
2,3,
Dragos-Mihai Romitan
2 and
Anca Dana Buzoianu
1
1
Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania
2
Department of Cardiology, Cluj Municipal Hospital, 400005 Cluj-Napoca, Romania
3
Department of Internal Medicine, Iuliu Hațieganu University of Medicine and Pharmacy, 400005 Cluj-Napoca, Romania
4
Department of Oncology, Iuliu Hatieganu University of Medicine and Pharmacy, 400015 Cluj-Napoca, Romania
5
Department of Internal Medicine, Cluj Municipal Hospital, 400139 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
Medicina 2021, 57(8), 806; https://doi.org/10.3390/medicina57080806
Submission received: 10 June 2021 / Revised: 25 July 2021 / Accepted: 4 August 2021 / Published: 6 August 2021
(This article belongs to the Section Cardiology)

Abstract

:
Cardiotoxicity is a well-recognised side effect of cancer-related therapies with a great impact on outcomes and quality of life in the cancer survivor population. The pathogenesis of chemotherapy-induced cardiotoxicity in patients with gastrointestinal cancers involves various molecular mechanisms, and the combined use of various chemotherapies augments the risk of each drug used alone. In terms of cardiotoxicity diagnosis, novel biomarkers, such as troponins, brain natriuretic peptide (BNP), myeloperoxidases and miRNAs have been recently assessed. Echocardiography is a noninvasive imaging method of choice for the primary assessment of chemotherapy-treated patients to generally evaluate the cardiovascular impact of these drugs. Novel echocardiography techniques, like three-dimensional and stress echocardiography, will improve diagnosis efficacy. Cardiac magnetic resonance (CMR) can evaluate cardiac morphology, function and wall structure. Corroborated data have shown the importance of CMR in the early evaluation of patients with gastrointestinal cancers, treated with anticancer drugs, but further studies are required to improve risk stratification in these patients. In this article, we review some important aspects concerning the cardiotoxicity of antineoplastic drugs used in gastrointestinal cancers. We also discuss the mechanism of cardiotoxicity, the role of biomarkers and the imaging methods used in its detection.

1. Introduction

Gastrointestinal (GI) cancer represents an important cause of cancer-related deaths being a major public health problem that encompasses cancers of the oesophagus, stomach, colon and rectum [1]. Colorectal cancer is the most commonly diagnosed GI cancer and comprises 9% of the global cancer burden, while the other GI cancers are not negligible at all [1,2].
As overall cancer-related deaths decline, chemotherapy-associated cardiotoxicity increases and has a great impact on the outcome and life-quality of cancer survivor patients. They have an important cardiovascular (CV) risk [3], and should be considered, as recommended by the ACC/AHA guidelines, as a stage A heart failure (HF) [4]. Lenneman et al. showed a rise in the number of heart transplants in patients with cardiomyopathy, due to anthracycline therapy [5]. Others have shown that the greatest risk is in those treated with a high dose of anthracyclines or when radiotherapy is associated. Moreover, patients treated with low-dose anthracyclines or trastuzumab alone, are also at high risk, especially if they are older (>60 years), have cardiac dysfunction, previous myocardial infarction, valvular heart disease, or multiple CV risk factors [6]. Therefore, active CV screening is strongly advised, especially in those who are at a high risk to develop CV disease [7,8].

2. Mechanism of Cardiotoxicity

Chemotherapy-induced cardiotoxicity is defined as a reduction in left ventricle (LV) ejection fraction (LVEF) below 50%, a reduction from baseline with >10% or a Global Longitudinal Strain (GLS) reduction of >15% from baseline [7]. It manifests as cardiac dysfunction, valve or pericardial disease, arrhythmias, pulmonary hypertension, or other vascular abnormalities. Doxorubicin is most frequently accounted for HF [9], while ibrutinib for arrhythmias [10], 5-fluorouracil (5-FU) for coronary spasm, vascular endothelial growth factor (VEGF) inhibitors for arterial hypertension, and paclitaxel for conduction abnormalities.
Cardiotoxicity was initially divided in two categories based on its reversibility, but this classification is no longer available, due to several mismatches [11]. Moreover, coronary artery disease may occur after treatment with tyrosine kinase inhibitors (TKIs), drugs that may increase the risk of intrastent thrombosis [12,13]. VEGF inhibitors prevent angiogenesis and were associated with decreased nitric oxide production, vasoconstriction and hypertension [12,14], thus potentiating endothelial dysfunction, atherosclerosis and thrombotic microangiopathy [15,16]. Other chemotherapeutics can cause atrial fibrillation, QT prolongation, ventricular arrhythmias and sudden cardiac death [17,18].

2.1. Anthracyclines

Anthracyclines may produce cardiotoxicity by interacting with both topoisomerase II (topoII) isoforms, topoIIα and topoII [19], and by enhancing cellular apoptosis and reactive oxidant species (ROS) synthesis. Genetic analysis identified genetic loci that are involved in cell uptake and carbonyl redox cycling, which also transport anthracyclines within the cells [20]. Concomitant drugs which are also transported by these proteins, such as digoxin, diltiazem, or verapamil, may increase the cytoplasmatic concentration of anthracyclines [21]. The most common compounds used in GI cancers are doxorubicin, daunorubicine, epirubicin and idarubicin.
Cardiac histopathological changes associated with anthracyclines are mainly represented by cytoplasmatic vacuolisation, myofibrillar and myocardial sarcomere disarray [22,23].
Late-onset cardiotoxicity is the most frequently encountered. Cardinale et al. have shown that 98% of cardiotoxicity cases occurred within the first year after treatment [24].

2.2. Fluoropyrimidines

Fluoropyrimidines may be cardiotoxic [25], even in patients without CV diseases [26]. Fluoropyrimidines (5-FU, capecitabine) is frequently used in solid tumours, as it is the standard treatment in colorectal cancer, and increase the radiosensitivity of tumours [27]. 5-FU acts as an S-phase antimetabolite, interfering with DNA synthesis, repair and elongation [28].
Being metabolised to 5-FU, capecitabine has similar cardiotoxicity as 5-FU [29]. Coronary spasm is another mechanism of fluoropyrimidines’ cardiotoxicity, which was suggested not to be endothelial-dependent [30,31]. Moreover, 5-FU increased the levels of endothelin-1 and von Willebrand factor [32]. 5-FU cardiotoxicity may also be secondary to enhanced synthesis of fluoroacetate and fluorocitrate, cellular apoptosis and ROS synthesis [25].
Therefore, fluoropyrimidines may cause chest pain, Takotsubo cardiomyopathy, Kounis syndrome, arrhythmias and cardiac arrest. The incidence of cardiac events in high-dose 5 FU treated patients may be as high as 7.6% [25,33,34].

2.3. Platinum-Based Compounds

Platinum-based compounds are important chemotherapies in gastrointestinal tumours, which is used in oesophageal cancer, colorectal cancer (FOLFOX). Cisplatin is another commonly used drug that is associated with significant toxicity [35], while carboplatin has fewer side effects. They interact with DNA and determine ROS production [36]. Additionally, cisplatin has been associated with cardiomyocyte apoptosis secondary to activating the transmembrane protein kinase RNA-like ER kinase pathway [37].
The incidence of thromboembolic side effects was as high as 10 to 20% in different studies. Endothelial dysfunction can also play a role in cisplatin-associated cardiotoxicity, which is associated with increased levels of von Willebrand factor [38]. Studies have shown an increased incidence of arterial hypertension and LV hypertrophy in patients treated with cisplatin, in a cumulative dose-dependent manner [39].

2.4. Human Epidermal Growth Factor Receptor 2 (HER2) Antagonists

Trastuzumab, a HER2 antagonist, has recently been proven beneficial in some HER2 positive gastric and colorectal cancers [40,41]. Trastuzumab cardiotoxicity was first described in breast cancer trials, especially when combined therapies were used [42]. Pertuzumab, another HER2 antagonist, used in gastric cancer, proved similar cardiotoxicity as trastuzumab, but without the same additive toxicity [43].
Neuregulin, an ErbB receptor found in the CV system, may be involved in HER2 therapy-induced cardiotoxicity. Neuregulin, ErbB2 and ErbB4 receptor tyrosine kinases have been documented to play important roles in cellular cross-communication and stress-response [44]. The disruption of ErbB2 expression may cause dilated cardiomyopathy [45], whereas the suppression of neuregulin/ErbB signal may prevent recovery from ischemic injury [46]. Moreover, anti-ErbB2 therapy may induce ROS and cell apoptosis, and as a consequence, increased levels of troponins I and cardiac myosin light chain-1 [47]. Recently, it has been documented that trastuzumab may provoke endothelial and mitochondrial dysfunction [48].

2.5. Immune Checkpoint Inhibitors (ICI)

ICI are a class of drugs that block the immune down-regulators, such as programmed cell death-1 (PD1), PD ligand-1 (PDL1), cytotoxic T-lymphocyte antigen 4 (CTLA-4) [49]. Nivolumab and pembrolizumab are anti-PD1 agents used in colorectal and gastric cancers. In murine models, CTLA-4 and PD1 exert cardioprotective effects. In turn, the blockage of CTLA-4 was associated with fatal myocarditis [50]. Some pericardial antigens may be activated by ICI, leading to pericarditis, however this is a less frequent side effect [51].
According to recent data, ICI-related myocarditis may occur in 1.14% of patients when the drugs are used alone and in 2.4% of patients if the drugs are used in combination therapy. Nonetheless, the risk of developing myocarditis is shown to be lower with anti-PD1 drugs, when compared to anti-PDL1 and anti-CTLA4 drugs [52]. Studies have shown that it usually appears after a median follow-up period of 2 to 3 months, but it can occur anytime during treatment, having a high mortality risk [52,53,54]. Additionally, Takotsubo syndrome related to ICI had been reported [55].

2.6. Angiogenesis Inhibitors

VEGFs are involved in angiogenesis, endothelial cell survival, vasomotricity and cardiac contractile function, and their inhibitors have been associated with HF and arterial hypertension [56]. Bevacizumab, an antibody-based anti-VEGF, was also associated with atherothrombotic events [57]. Moreover, some studies reported preeclampsia–like syndrome secondary to treatment with angiogenesis inhibitors [58]. Additionally, these compounds were associated with irreversible diastolic dysfunction [59].
It has been shown that these drugs can increase the risk of both ischemic and haemorrhagic stroke, especially when bevacizumab is used. This risk is also higher in patients with colorectal cancers, and it doubles when a double dose is used [60]. HF was encountered more frequently in those treated with TKIs, and it is caused by the tyrosine kinase, AMP-activated kinase and platelet-derived growth factor receptors inhibition [58,61].
VEGFs inhibitors associated cardiotoxicity is usually divided into two types: (1) On-target toxicity, which is due to the inhibition of CV-expressed kinases and it occurs mostly with imatinib treatment; and (2) off-target toxicity, encountered with sunitinib therapy, in which CV side effects are due to inhibition of non-targeted kinases [62].

3. Diagnosis Methods of Chemotherapy-Induced Cardiotoxicity in GI Cancers

The evaluation of cardiotoxicity has an important role in patients treated with cancer therapies. Detecting subclinical changes in cardiac function represents the subject of interest of many studies. Echocardiography and cardiac magnetic resonance imaging (CMR) is the most used techniques. Cardiac biomarkers are used to increase the sensitivity of imaging methods in detecting cardiotoxicity, mainly in the early stages [7,63].

3.1. Biomarkers

Biomarkers are considered to be a cost-effective alternative to other diagnostic techniques for screening subclinical changes in cardiac function. The most useful serum biomarkers used for the assessment of chemotherapy-based cardiotoxicity are represented by cardiac troponins (cTn) I/T, brain natriuretic peptide (BNP), C-reactive protein, myeloperoxidase (MPO), growth differentiation factor 15 (GDF-15), galectin-3, soluble interleukin 1 receptor-like 1 (ST2) and some microRNAs (miRNA). These can be used in the risk stratification and follow-up of cardiotoxicity and in evaluating cardio-protection. Current guidelines recommend using such biomarkers in patients treated with chemotherapy, by determining their value at baseline, during chemotherapy and after chemotherapy [7,64].

3.1.1. Troponins

Cardiac troponins I and T are organ-specific biomarkers used to diagnose myocardial injury, which are also reliable in chemotherapy-based cardiotoxicity. In a recently published meta-analysis, the significant link between cardiac troponins and chemotherapy was proved. Studies have found that cTn was able to detect early cardiac injury more accurately than natriuretic peptides [7]. Increased serum levels of cTn were frequently reported in patients treated with anthracyclines, even within the first 72 h of anthracycline treatment and at one month after therapy cessation [65]. Additionally, Jones et al. showed these biomarkers to be significantly increased after the fourth, the fifth and even after the sixth cycle of chemotherapy [66]. Garrone et al. found that high levels of cTn were positively correlated with the risk of developing HF [67], while other studies demonstrated that these biomarkers were predictors of early cardiac dysfunction, having important roles in detecting cardiotoxicity [68,69,70].
Furthermore, elevations in serum cTn were observed with trastuzumab treatment and were associated with cardiac dysfunction [70]. cTn has been shown to play important roles even in the prediction of LV dysfunction [68,71]. Furthermore, cTn may identify subclinical forms of ICI-based myocarditis, which are associated with a worse prognosis [52].

3.1.2. BNP

BNP, another serum biomarker, having an important role to diagnose chemotherapy-induced cardiotoxicity, is associated with increased LV filling pressures [72]. In patients with impaired LV function secondary to anthracycline therapy, Feola and al. documented significantly higher levels of BNP at baseline and at follow-up, in comparison with patients without LV dysfunction. Patients who develop cardiac side effects, due to chemotherapy, have higher BNP levels at baseline [73]. This biomarker has an important predictive value in documenting the development of chemotherapy-induced HF [74]. NT-proBNP levels are directly correlated with anthracycline treatment and are also associated with an increased 1-year mortality, as shown by Iuliis et al. [75].

3.1.3. Other Biomarkers of Use in Detecting Cardiotoxicity

MPO and GDF-15 are associated with impaired LV function and even with decreased LVEF [76]. miRNAs are also studied in patients treated with both anthracyclines and trastuzumab, and some studies have shown that several miRNAs are increased in the first 24 h after chemotherapy, but further studies are needed [77].

3.2. Echocardiography

Echocardiography is the main imaging method used in diagnosing cardiotoxicity, which comprehensively evaluates heart morphology and function [7,78]. LVEF is the most common marker of LV systolic function, which is assessed using the modified biplane Simpson’s method. Although, it has low sensitivity and high temporal variability [79,80], these shortcomings may be improved using contrast echocardiography [81]. Three-dimensional echocardiography (3DE) has increased accuracy in evaluating heart function, and the lowest variability [82]. Other echocardiography parameters, such as myocardial performance index (MPI), which can identify subclinical cardiotoxicity in patients treated with 5-FU and anthracyclines, and mitral annular plane systolic excursion (MAPSE), may also be used [83,84]. LVEF <55% was associated with a significantly increased risk to develop HF, due to HER2 antagonists and anthracyclines [85,86].
Diastolic dysfunction was the first to occur in most of the chemotherapy-treated patients. In some studies, the E/A ratio was impaired, due to doxorubicin, along with changes in isovolumetric relaxation time (IVRT), even after the first dose [87,88,89]. IVRT was also able to predict LVEF changes [87].
3DE has comparable accuracy with CMR and multi-gated acquisition scan (MUGA) in terms of LVEF evaluation [90,91]. The most sensitive 3DE markers of early cardiotoxicity, are represented by an LVEF <55%, and end-systolic volume index >29 mL/m2, and a global longitudinal strain (GLS) <−17.5%, having comparable efficacy with CMR [92]. LV strain is a novel technique used for monitoring cardiotoxicity, and a reduction in GLS of >15% is considered a marker of cardiotoxicity [64]. A 6-months decrease in GLS >15% may identify cardiac dysfunction in breast cancer patients at 12 months after stopping chemotherapy [70].
3DE speckle-tracking imaging (STI) overcomes the limitations of 2DE STI in terms of identifying early cardiac changes [93]. Global circumferential strain (GCS), and global radial strain are decreased early after anthracycline treatment, despite preserved LVEF. Global area strain curve (GAS) was also associated with subclinical LV changes [94]. In patients treated with trastuzumab, GLS significantly changed at 6-months after chemotherapy [95]. GLS was strongly associated with cardiac events among patients with ICI-related myocarditis [96].
During anthracycline therapy, changes in LV torsion, twisting and untwisting occurred earlier than an LVEF drop. Apical torsion, twisting and untwisting rates after chemotherapy, were correlated with prolonged IVRT, but didn’t predict LVEF decrease [97].
Echocardiography is used in detecting wall motion abnormalities, due to ischemia or evaluation of LV wall hypertrophy secondary to arterial hypertension in patients treated with fluoropyrimidines, platinum-based chemotherapy and anti-VEGFs. Stress echocardiography (SE) may be used to reveal subclinical changes in LV function [98]. In patients treated with doxorubicin, the sensitivity of resting LVEF may increase from 50% to 90% when SE is used [99]. SE can be used to assess stable coronary artery disease [100], and in those treated with 5-FU and bevacizumab. It can also evaluate LV’s contractile reserve, a 5-units decrease having a predictive value for future LVEF decline [101].

3.3. Cardiac Magnetic Resonance Imaging

CMR is used in evaluating cardiac structure and function, and it is the gold standard for accurate assessment of ventricles’ functions and volumes. It may detect myocardial oedema and fibrosis, and early changes in cardiac function [102]. Mild LV dysfunction occurs early during treatment with anthracyclines and trastuzumab with a 3% decrease in LVEF at one month and 5% at four months after stopping chemotherapy. Nearly all chemotherapy-treated patients developed RV dysfunction, an impairment that persisted during follow-up at twelve months, even though in some patients, LV function recovered at four months [103].
Myocardial oedema, inflammation and interstitial fibrosis, occur early before LV dysfunction and LVEF impairment [104]. Myocardial oedema can be detected by CMR, mostly on T2-weighted signal intensity [105]. Myocardial oedema and fibrosis are associated with higher mortality [106]. Steady-state free-precession (SSFP) CMR provides data on wall motion segmental and global changes, and may detect subtle changes [107]. Cine-CMR detects subtle LV morpho-functional abnormalities, even in patients treated with low dose anthracyclines. LV mass also changes during cancer therapy, mostly in anthracycline-treated patients [108]. Interestingly, a smaller LV mass correlated with the cumulative dose of anthracycline, a negative prognostic factor [90,109]. An LV mass less than 57 g/m2 was associated with a higher risk of HF and CV deaths [110]. RV dysfunction was also observed during anthracycline treatment or anthracycline-trastuzumab combination, and it was correlated with higher mortality [111,112].
Late-gadolinium enhancement (LGE) is used in evaluating myocardial fibrosis [113,114], and data from studies are contradictory [115]. LGE is infrequently observed in patients treated with trastuzumab and anthracyclines, and its absence doesn’t exclude myocardial fibrosis [116,117].
T1 mapping is used to quantify extracellular volume fraction (ECV), which indicates interstitial fibrosis [63]. On T1 weighted sequences, abnormal gadolinium accumulation is observed in cancer survivors [118], and may detect subclinical changes. Furthermore, ECV was significantly associated with histologically proven myocardial interstitial fibrosis [119,120]. Patients treated with anthracycline had higher ECV and associated diastolic dysfunction [116]. CMR can determine intracellular water lifetime using T1 mapping in order to properly characterise the myocardial tissue [121]. Anthracycline therapy determines a significant decrease of the LVEF, LV mass and intracellular water lifetime [119].
Early gadolinium enhancement (EGE) detects myocardial injury in patients with myocarditis [122], is included in the Lake Louise criteria. After anthracycline treatment, EGE significantly predicted a decline in LVEF [118]. CMR can also properly evaluate patients with ICI-related myocarditis and preserved LVEF, especially in combination with using LGE [123,124].
Furthermore, the myocardial consequences of arterial hypertension secondary to TKIs can be evaluated by CMR [64]. Vascular stiffness may be assessed by CMR, by aortic phase-contrast imaging. Pulse wave velocity (PWV) increases after anthracycline administration, which is associated with LV dysfunction and CV adverse effects, but its impact on the outcome of cancer patients needs further studies [108]. Lastly, T2 relaxation time is another marker of early LV dysfunction, which occurs sooner than ECV [125], but the studies are just at the beginning.

4. Risk Stratification and Surveillance during and after Chemotherapy

An important aspect in evaluating patients treated with chemotherapy is establishing their baseline cardiovascular risk and their risk of developing cardiotoxicity [126]. This approach will allow for a universal standard of care and a surveillance plan [63,64,78] (Figure 1). According to their risk level (low, medium, high, very high), we could establish the risk of developing cardiotoxicity, which is <2%, 2–9%, 10–19%, ≥20%, respectively [7].
According to the recently published position paper by the European Society of Cardiology (ESC), risk factors that should be considered are represented by previous cardiovascular diseases, co-existing medical disease, demographic traits and lifestyle factors that are accounted as cardiovascular risk factors, elevated serum biomarkers before initiation of chemotherapy and previous chemotherapy. High risk patients are considered those with one or more high risk factors or patients with medium risk factors with a sum of >5 points. Very high risk patients are considered those with one or more very high risk factors [7].
Very high risk factors are represented by pre-existing heart failure or cardiomyopathy in patients treated with anthracycline, HER-2 targeted therapies and VEGF inhibitors.
High risk factors are represented by ischemic cardiomyopathy (myocardial infarction, coronary artery by-pass grapht, stable angina), severe valvular heart disease, low LVEF at baseline (<50%), age >80 years, previous chemotherapy (anthracycline or HER-2 therapies) and radiotherapy to the left side of the chest or mediastinum [7].
Serum biomarkers are used in surveillance during and after chemotherapy, and according to ESC position paper in high risk CV patients treated with anthracycline BNP/NT-proBNP, cTn should be measured at baseline, before cycles 2, 4 and 6 or before every cycle and at 3 and 6 months following chemotherapy or at 12 months after the final cycle. In low and medium CV risk patients, these biomarkers should be determined at baseline, before the fifth cycle and at 12 months after chemotherapy. In patients treated with HER-2 targeted therapies for gastric cancer, serum biomarkers should be measured at baseline, before every cycle for 3–6 months and then every 3 months for the remaining treatment in the first year. Following chemotherapy serum biomarkers are measured only if the patient is symptomatic [126].
According to the British Society of Echocardiography and British Society of Cardio-Oncology guidelines published recently regarding the frequency of echocardiography during chemotherapy, high risk patients treated with anthracycline, should be evaluated every two cycles or every cycle if the dose of doxorubicin is >240 mg/m2. In patients treated with HER-2 targeted therapy, echocardiography should be done every two or three cycles for three months, then to every fourth cycle in the first year [78].

5. Future Directions

There are still many uncertainties in evaluating patients undergoing cancer therapy, especially in detecting subclinical changes. Some new imaging techniques have been investigated to detect early cardiotoxicity, such as 3DE STI and new CMR techniques, which proved to have promising results. With the development of these new imaging techniques, there is a need for more clinical trials, for better cancer treated patients’ surveillance, to improve their outcome. Resorting to cardiac imaging methods and cardiac biomarkers may help to detect subclinical changes in cardiac function, while the damage is still reversible and cardio-protection methods are effective.

Author Contributions

L.M.R., D.R. and A.D.B. researched data for the article and wrote the article, T.-E.C., D.C., E.B., D.-M.R. discussed the content of the article, and L.M.R., D.R., T.-E.C., A.D.B. reviewed and edited before submission. All authors have read and agreed to the published version of the manuscript.

Funding

This article did not receive any external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

This research paper was supported by the institutional doctoral fellowship of the Iuliu Hatieganu University of Medicine and Pharmacy of Cluj-Napoca.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Arnold, M.; Abnet, C.C.; Neale, R.E.; Vignat, J.; Giovannucci, E.L.; McGlynn, K.A.; Bray, F. Global Burden of 5 Major Types of Gastrointestinal Cancer. Gastroenterology 2020, 159, 335–349.e15. [Google Scholar] [CrossRef]
  2. Rawla, P.; Sunkara, T.; Barsouk, A. Epidemiology of colorectal cancer: Incidence, mortality, survival, and risk factors. Gastroenterol. Rev. 2019, 14, 89–103. [Google Scholar] [CrossRef]
  3. Lipshultz, S.E.; Adams, M.J.; Colan, S.D.; Constine, L.S.; Herman, E.H.; Hsu, D.T.; Hudson, M.M.; Kremer, L.C.; Landy, D.C.; Miller, T.L.; et al. Long-term Cardiovascular Toxicity in Children, Adolescents, and Young Adults Who Receive Cancer Therapy: Pathophysiology, Course, Monitoring, Management, Prevention, and Research Directions. Circulation 2013, 128, 1927–1995. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E.; Drazner, M.H.; Fonarow, G.C.; Geraci, S.A.; Horwich, T.; Januzzi, J.L.; et al. 2013 ACCF/AHA Guideline for the Management of Heart Failure. J. Am. Coll. Cardiol. 2013, 62, e147–e239. [Google Scholar] [CrossRef] [Green Version]
  5. Lenneman, A.J.; Wang, L.; Wigger, M.; Frangoul, H.; Harrell, F.E.; Silverstein, C.; Sawyer, D.B.; Lenneman, C.G. Heart Transplant Survival Outcomes for Adriamycin-Dilated Cardiomyopathy. Am. J. Cardiol. 2013, 111, 609–612. [Google Scholar] [CrossRef] [Green Version]
  6. Armenian, S.H.; Lacchetti, C.; Barac, A.; Carver, J.; Constine, L.S.; Denduluri, N.; Dent, S.; Douglas, P.S.; Durand, J.-B.; Ewer, M.; et al. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2017, 35, 893–911. [Google Scholar] [CrossRef]
  7. Čelutkienė, J.; Pudil, R.; López-Fernández, T.; Grapsa, J.; Nihoyannopoulos, P.; Bergler-Klein, J.; Cohen-Solal, A.; Farmakis, D.; Tocchetti, C.G.; Haehling, S.; et al. Role of cardiovascular imaging in cancer patients receiving cardiotoxic therapies: A position statement on behalf of the Heart Failure Association (HFA), the European Association of Cardiovascular Imaging (EACVI) and the Cardio-Oncology Council of the European Society of Cardiology (ESC). Eur. J. Heart Fail. 2020, 22, 1504–1524. [Google Scholar] [CrossRef]
  8. 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] [PubMed]
  9. Fu, X.; Kong, L.; Tang, M.; Zhang, J.; Zhou, X.; Li, G.; Wang, H.; Fu, F. Protective effect of ocotillol against doxorubicin-induced acute and chronic cardiac injury. Mol. Med. Rep. 2014, 9, 360–364. [Google Scholar] [CrossRef] [Green Version]
  10. Curigliano, G.; Cardinale, D.; Dent, S.; Criscitiello, C.; Aseyev, O.; Lenihan, D.; Cipolla, C.M. Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management. CA Cancer J. Clin. 2016, 66, 309–325. [Google Scholar] [CrossRef] [Green Version]
  11. Hamo, C.E.; Bloom, M.W. Getting to the Heart of the Matter: An Overview of Cardiac Toxicity Related to Cancer Therapy. Clin. Med. Insights Cardiol. 2015, 9 (Suppl. 2). [Google Scholar] [CrossRef] [PubMed]
  12. Moslehi, J.J.; Deininger, M. Tyrosine Kinase Inhibitor–Associated Cardiovascular Toxicity in Chronic Myeloid Leukemia. J. Clin. Oncol. 2015, 33, 4210–4218. [Google Scholar] [CrossRef] [PubMed]
  13. Gori, T.; Polimeni, A.; Indolfi, C.; Räber, L.; Adriaenssens, T.; Münzel, T. Predictors of stent thrombosis and their implications for clinical practice. Nat. Rev. Cardiol. 2019, 16, 243–256. [Google Scholar] [CrossRef]
  14. Douxfils, J.; Haguet, H.; Mullier, F.; Chatelain, C.; Graux, C.; Dogné, J.-M. Association Between BCR-ABL Tyrosine Kinase Inhibitors for Chronic Myeloid Leukemia and Cardiovascular Events, Major Molecular Response, and Overall Survival. JAMA Oncol. 2016, 2, 625. [Google Scholar] [CrossRef]
  15. Hadzijusufovic, E.; Albrecht-Schgoer, K.; Huber, K.; Hoermann, G.; Grebien, F.; Eisenwort, G.; Schgoer, W.; Herndlhofer, S.; Kaun, C.; Theurl, M.; et al. Nilotinib-induced vasculopathy: Identification of vascular endothelial cells as a primary target site. Leukemia 2017, 31, 2388–2397. [Google Scholar] [CrossRef] [Green Version]
  16. Latifi, Y.; Moccetti, F.; Wu, M.; Xie, A.; Packwood, W.; Qi, Y.; Ozawa, K.; Shentu, W.; Brown, E.; Shirai, T.; et al. Thrombotic microangiopathy as a cause of cardiovascular toxicity from the BCR-ABL1 tyrosine kinase inhibitor ponatinib. Blood 2019, 133, 1597–1606. [Google Scholar] [CrossRef] [Green Version]
  17. Fradley, M.G.; Gliksman, M.; Emole, J.; Viganego, F.; Rhea, I.; Welter-Frost, A.; Armanious, M.; Lee, D.H.; Walko, C.; Shah, B.; et al. Rates and Risk of Atrial Arrhythmias in Patients Treated with Ibrutinib Compared with Cytotoxic Chemotherapy. Am. J. Cardiol. 2019, 124, 539–544. [Google Scholar] [CrossRef] [PubMed]
  18. Guha, A.; Derbala, M.H.; Zhao, Q.; Wiczer, T.E.; Woyach, J.A.; Byrd, J.C.; Awan, F.T.; Addison, D. Ventricular Arrhythmias Following Ibrutinib Initiation for Lymphoid Malignancies. J. Am. Coll. Cardiol. 2018, 72, 697–698. [Google Scholar] [CrossRef]
  19. Mordente, A.; Meucci, E.; Martorana, G.E.; Tavian, D.; Silvestrini, A. Topoisomerases and Anthracyclines: Recent Advances and Perspectives in Anticancer Therapy and Prevention of Cardiotoxicity. Curr. Med. Chem. 2017, 24. [Google Scholar] [CrossRef]
  20. Aminkeng, F.; Ross, C.J.D.; Rassekh, S.R.; Rieder, M.J.; Bhavsar, A.P.; Sanatani, S.; Bernstein, D.; Hayden, M.R.; Amstutz, U.; Carleton, B.C. Pharmacogenomic screening for anthracycline-induced cardiotoxicity in childhood cancer. Br. J. Clin. Pharmacol. 2017, 83, 1143–1145. [Google Scholar] [CrossRef] [Green Version]
  21. Couture, L.; Nash, J.A.; Turgeon, J. The ATP-Binding Cassette Transporters and Their Implication in Drug Disposition: A Special Look at the Heart. Pharmacol. Rev. 2006, 58, 244–258. [Google Scholar] [CrossRef]
  22. Orogo, A.M.; Gonzalez, E.R.; Kubli, D.A.; Baptista, I.L.; Ong, S.-B.; Prolla, T.A.; Sussman, M.A.; Murphy, A.N.; Gustafsson, Å.B. Accumulation of mitochondrial DNA mutations disrupts cardiac progenitor cell function and reduces survival. J. Biol. Chem. 2017, 292, 11348. [Google Scholar] [CrossRef] [Green Version]
  23. Lipshultz, S.E.; Cochran, T.R.; Franco, V.I.; Miller, T.L. Treatment-related cardiotoxicity in survivors of childhood cancer. Nat. Rev. Clin. Oncol. 2013, 10, 697–710. [Google Scholar] [CrossRef]
  24. Cardinale, D.; Colombo, A.; Bacchiani, G.; Tedeschi, I.; Meroni, C.A.; Veglia, F.; Civelli, M.; Lamantia, G.; Colombo, N.; Curigliano, G.; et al. Early Detection of Anthracycline Cardiotoxicity and Improvement with Heart Failure Therapy. Circulation 2015, 131, 1981–1988. [Google Scholar] [CrossRef] [Green Version]
  25. Kanduri, J.; More, L.A.; Godishala, A.; Asnani, A. Fluoropyrimidine-Associated Cardiotoxicity. Cardiol. Clin. 2019, 37, 399–405. [Google Scholar] [CrossRef]
  26. Wacker, A.; Lersch, C.; Scherpinski, U.; Reindl, L.; Seyfarth, M. High Incidence of Angina pectoris in Patients Treated with 5-Fluorouracil. Oncology 2003, 65, 108–112. [Google Scholar] [CrossRef] [Green Version]
  27. Bartelink, H.; Roelofsen, F.; Eschwege, F.; Rougier, P.; Bosset, J.F.; Gonzalez, D.G.; Peiffert, D.; van Glabbeke, M.; Pierart, M. Concomitant radiotherapy and chemotherapy is superior to radiotherapy alone in the treatment of locally advanced anal cancer: Results of a phase III randomized trial of the European Organization for Research and Treatment of Cancer Radiotherapy and Gastro. J. Clin. Oncol. 1997, 15, 2040–2049. [Google Scholar] [CrossRef]
  28. Curtin, N.J.; Harris, A.L.; Aherne, G.W. Mechanism of cell death following thymidylate synthase inhibition: 2′-deoxyuridine-5′-triphosphate accumulation, DNA damage, and growth inhibition following exposure to CB3717 and dipyridamole. Cancer Res. 1991, 51, 2346–2352. [Google Scholar]
  29. Layoun, M.E.; Wickramasinghe, C.D.; Peralta, M.V.; Yang, E.H. Fluoropyrimidine-Induced Cardiotoxicity: Manifestations, Mechanisms, and Management. Curr. Oncol. Rep. 2016, 18, 35. [Google Scholar] [CrossRef] [Green Version]
  30. Atar, A.; Korkmaz, M.E.; Ozin, B. Two cases of coronary vasospasm induced by 5-fluorouracil. Anadolu Kardiyol. Derg. Anatol. J. Cardiol. 2010, 10, 461–462. [Google Scholar] [CrossRef]
  31. Mosseri, M.; Fingert, H.J.; Varticovski, L.; Chokshi, S.; Isner, J.M. In vitro evidence that myocardial ischemia resulting from 5-fluorouracil chemotherapy is due to protein kinase C-mediated vasoconstriction of vascular smooth muscle. Cancer Res. 1993, 53, 3028–3033. [Google Scholar] [PubMed]
  32. Jensen, S.A.; Sørensen, J.B. 5-Fluorouracil-based therapy induces endovascular injury having potential significance to development of clinically overt cardiotoxicity. Cancer Chemother. Pharmacol. 2012, 69, 57–64. [Google Scholar] [CrossRef]
  33. Lker, D.; Muammer Karaayvaz, M. Reduced Antioxidant Defense Capacity in Myocardial Tissue from Guinea Pigs Treated with 5-fluorouracil. J. Toxicol. Environ. Heal. Part A 2000, 59, 585–589. [Google Scholar] [CrossRef]
  34. Karabay, C.; Gecmen, C.; Aung, S.; Guler, A.; Candan, O.; Batgerel, U.; Kalayci, A.; Kirma, C. Is 5-fluorouracil-induced vasospasm a Kounis syndrome? A diagnostic challenge. Perfusion 2011, 26, 542–545. [Google Scholar] [CrossRef]
  35. Dugbartey, G.J.; Peppone, L.J.; de Graaf, I.A.M. An integrative view of cisplatin-induced renal and cardiac toxicities: Molecular mechanisms, current treatment challenges and potential protective measures. Toxicology 2016, 371, 58–66. [Google Scholar] [CrossRef] [Green Version]
  36. Santandreu, F.M.; Roca, P.; Oliver, J. Uncoupling protein-2 knockdown mediates the cytotoxic effects of cisplatin. Free Radic. Biol. Med. 2010, 49, 658–666. [Google Scholar] [CrossRef]
  37. Ma, H.; Jones, K.R.; Guo, R.; Xu, P.; Shen, Y.; Ren, J. Cisplatin compromises myocardial contractile function and mitochondrial ultrastructure: Role of endoplasmic reticulum stress. Clin. Exp. Pharmacol. Physiol. 2010, 37, 460–465. [Google Scholar] [CrossRef]
  38. Pretnar-Oblak, J.; Zaletel, M.; Jagodic, M.; Zaletel, M. Thrombosis of Internal Carotid Artery after Cisplatin-Based Chemotherapy. Eur. Neurol. 2007, 57, 109–110. [Google Scholar] [CrossRef] [PubMed]
  39. Glendenning, J.L.; Barbachano, Y.; Norman, A.R.; Dearnaley, D.P.; Horwich, A.; Huddart, R.A. Long-term neurologic and peripheral vascular toxicity after chemotherapy treatment of testicular cancer. Cancer 2010. [Google Scholar] [CrossRef] [PubMed]
  40. Yu, A.F.; Yadav, N.U.; Lung, B.Y.; Eaton, A.A.; Thaler, H.T.; Hudis, C.A.; Dang, C.T.; Steingart, R.M. Trastuzumab interruption and treatment-induced cardiotoxicity in early HER2-positive breast cancer. Breast Cancer Res. Treat. 2015, 149, 489–495. [Google Scholar] [CrossRef] [Green Version]
  41. Takegawa, N.; Yonesaka, K. HER2 as an Emerging Oncotarget for Colorectal Cancer Treatment After Failure of Anti-Epidermal Growth Factor Receptor Therapy. Clin. Colorectal Cancer 2017, 16, 247–251. [Google Scholar] [CrossRef]
  42. Slamon, D.J.; Leyland-Jones, B.; Shak, S.; Fuchs, H.; Paton, V.; Bajamonde, A.; Fleming, T.; Eiermann, W.; Wolter, J.; Pegram, M.; et al. Use of Chemotherapy plus a Monoclonal Antibody against HER2 for Metastatic Breast Cancer That Overexpresses HER2. N. Engl. J. Med. 2001, 344, 783–792. [Google Scholar] [CrossRef]
  43. Swain, S.M.; Baselga, J.; Kim, S.-B.; Ro, J.; Semiglazov, V.; Campone, M.; Ciruelos, E.; Ferrero, J.-M.; Schneeweiss, A.; Heeson, S.; et al. Pertuzumab, Trastuzumab, and Docetaxel in HER2-Positive Metastatic Breast Cancer. N. Engl. J. Med. 2015, 372, 724–734. [Google Scholar] [CrossRef] [Green Version]
  44. Odiete, O.; Hill, M.F.; Sawyer, D.B. Neuregulin in Cardiovascular Development and Disease. Circ. Res. 2012, 111, 1376–1385. [Google Scholar] [CrossRef] [Green Version]
  45. Crone, S.A.; Zhao, Y.-Y.; Fan, L.; Gu, Y.; Minamisawa, S.; Liu, Y.; Peterson, K.L.; Chen, J.; Kahn, R.; Condorelli, G.; et al. ErbB2 is essential in the prevention of dilated cardiomyopathy. Nat. Med. 2002, 8, 459–465. [Google Scholar] [CrossRef] [PubMed]
  46. Hedhli, N.; Huang, Q.; Kalinowski, A.; Palmeri, M.; Hu, X.; Russell, R.R.; Russell, K.S. Endothelium-Derived Neuregulin Protects the Heart Against Ischemic Injury. Circulation 2011, 123, 2254–2262. [Google Scholar] [CrossRef] [Green Version]
  47. Mohan, N.; Shen, Y.; Endo, Y.; ElZarrad, M.K.; Wu, W.J. Trastuzumab, but not pertuzumab, dysregulates HER2 signaling to mediate inhibition of autophagy and increase in reactive oxygen species production in human cardiomyocytes. Mol. Cancer Ther. 2016, 15, 1321–1331. [Google Scholar] [CrossRef] [Green Version]
  48. Kitani, T.; Ong, S.-G.; Lam, C.K.; Rhee, J.-W.; Zhang, J.Z.; Oikonomopoulos, A.; Ma, N.; Tian, L.; Lee, J.; Telli, M.L.; et al. Human-Induced Pluripotent Stem Cell Model of Trastuzumab-Induced Cardiac Dysfunction in Patients with Breast Cancer. Circulation 2019, 139, 2451–2465. [Google Scholar] [CrossRef]
  49. Robert, C.; Thomas, L.; Bondarenko, I.; O’Day, S.; Weber, J.; Garbe, C.; Lebbe, C.; Baurain, J.-F.; Testori, A.; Grob, J.-J.; et al. Ipilimumab plus Dacarbazine for Previously Untreated Metastatic Melanoma. N. Engl. J. Med. 2011, 364, 2517–2526. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Love, V.A.; Grabie, N.; Duramad, P.; Stavrakis, G.; Sharpe, A.; Lichtman, A. CTLA-4 Ablation and Interleukin-12–Driven Differentiation Synergistically Augment Cardiac Pathogenicity of Cytotoxic T Lymphocytes. Circ. Res. 2007, 101, 248–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Yun, S.; Vincelette, N.D.; Mansour, I.; Hariri, D.; Motamed, S. Late Onset Ipilimumab-Induced Pericarditis and Pericardial Effusion: A Rare but Life Threatening Complication. Case Rep. Oncol. Med. 2015, 2015, 1–5. [Google Scholar] [CrossRef] [PubMed]
  52. 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]
  53. Escudier, M.; Cautela, J.; Malissen, N.; Ancedy, Y.; Orabona, M.; Pinto, J.; Monestier, S.; Grob, J.-J.; Scemama, U.; Jacquier, A.; et al. Clinical Features, Management, and Outcomes of Immune Checkpoint Inhibitor–Related Cardiotoxicity. Circulation 2017, 136, 2085–2087. [Google Scholar] [CrossRef] [PubMed]
  54. Moslehi, J.J.; Salem, J.-E.; Sosman, J.A.; Lebrun-Vignes, B.; Johnson, D.B. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet 2018, 391, 933. [Google Scholar] [CrossRef] [Green Version]
  55. Lees, C.; Yazdan-Ashoori, P.; Jerzak, K.J.; Gandhi, S. Takotsubo Cardiomyopathy During Anti-HER2 Therapy for Metastatic Breast Cancer. Oncologist 2019, 24, e80. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Ylä-Herttuala, S.; Rissanen, T.T.; Vajanto, I.; Hartikainen, J. Vascular Endothelial Growth Factors. J. Am. Coll. Cardiol. 2007, 49, 1015–1026. [Google Scholar] [CrossRef] [Green Version]
  57. Sugrue, M.M.; Yi, J.; Purdie, D.; Dong, W.; Grothey, A.; Kozloff, M. Serious arterial thromboembolic events (sATE) in patients (pts) with metastatic colorectal cancer (mCRC) treated with bevacizumab (BV): Results from the BRiTE registry. J. Clin. Oncol. 2007, 25, 4136. [Google Scholar] [CrossRef]
  58. Chintalgattu, V.; Ai, D.; Langley, R.R.; Zhang, J.; Bankson, J.A.; Shih, T.L.; Reddy, A.K.; Coombes, K.R.; Daher, I.N.; Pati, S.; et al. Cardiomyocyte PDGFR-β signaling is an essential component of the mouse cardiac response to load-induced stress. J. Clin. Investig. 2010, 120, 472–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Thavendiranathan, P.; Wintersperger, B.J.; Flamm, S.D.; Marwick, T.H. Cardiac MRI in the Assessment of Cardiac Injury and Toxicity from Cancer Chemotherapy. Circ. Cardiovasc. Imaging 2013, 6, 1080–1091. [Google Scholar] [CrossRef]
  60. Zuo, P.-Y.; Chen, X.-L.; Liu, Y.-W.; Xiao, C.-L.; Liu, C.-Y. Increased Risk of Cerebrovascular Events in Patients with Cancer Treated with Bevacizumab: A Meta-Analysis. PLoS ONE 2014, 9, e102484. [Google Scholar] [CrossRef] [Green Version]
  61. Zhang, P.; Hu, X.; Xu, X.; Fassett, J.; Zhu, G.; Viollet, B.; Xu, W.; Wiczer, B.; Bernlohr, D.A.; Bache, R.J.; et al. AMP Activated Protein Kinase-α2 Deficiency Exacerbates Pressure-Overload–Induced Left Ventricular Hypertrophy and Dysfunction in Mice. Hypertension 2008, 52, 918–924. [Google Scholar] [CrossRef] [Green Version]
  62. Biomarkers and surrogate endpoints: Preferred definitions and conceptual framework. Clin. Pharmacol. Ther. 2001, 69, 89–95. [CrossRef] [PubMed]
  63. Makavos, G.; Ikonomidis, I.; Palios, J.; Rigopoulos, A.; Katogiannis, K.; Parissis, J.; Paraskevaidis, I.; Noutsias, M. Cardiac imaging in cardiotoxicity: A focus on clinical practice. Heart Fail. Rev. 2020. [Google Scholar] [CrossRef]
  64. Luis Zamorano, J.; Lancellotti, P.; Rodriguez Muñ oz, 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 The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur. Heart J. 2016, 36, 2768–2801. [Google Scholar] [CrossRef] [PubMed]
  65. 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] [Green Version]
  66. 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]
  67. Garrone, O.; Crosetto, N.; Lo Nigro, C.; Catzeddu, T.; Vivenza, D.; Monteverde, M.; Merlano, M.; Feola, M. Prediction of Anthracycline Cardiotoxicity after Chemotherapy by Biomarkers Kinetic Analysis. Cardiovasc. Toxicol. 2012, 12, 135–142. [Google Scholar] [CrossRef]
  68. 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] [Green Version]
  69. Kitayama, H.; Kondo, T.; Sugiyama, J.; Kurimoto, K.; Nishino, Y.; Kawada, M.; Hirayama, M.; Tsuji, Y. High-sensitive troponin T assay can predict anthracycline- and trastuzumab-induced cardiotoxicity in breast cancer patients. Breast Cancer 2017, 24, 774–782. [Google Scholar] [CrossRef] [PubMed]
  70. 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] [Green Version]
  71. Morris, P.G.; Chen, C.; Steingart, R.; Fleisher, M.; Lin, N.; Moy, B.; Come, S.; Sugarman, S.; Abbruzzi, A.; Lehman, R.; et al. Troponin I and C-Reactive Protein Are Commonly Detected in Patients with Breast Cancer Treated with Dose-Dense Chemotherapy Incorporating Trastuzumab and Lapatinib. Clin. Cancer Res. 2011, 17, 3490–3499. [Google Scholar] [CrossRef] [Green Version]
  72. Kirchhof, P.; Benussi, S.; Kotecha, D.; Ahlsson, A.; Atar, D.; Casadei, B.; Castella, M.; Diener, H.-C.; Heidbuchel, H.; Hendriks, J.; et al. 2016 ESC Guidelines for the management of atrial fibrillation developed in collaboration with EACTS. Eur. Heart J. 2016, 37, 2893–2962. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. 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]
  74. Skovgaard, D.; Hasbak, P.; Kjaer, A. BNP Predicts Chemotherapy-Related Cardiotoxicity and Death: Comparison with Gated Equilibrium Radionuclide Ventriculography. PLoS ONE 2014, 9, e96736. [Google Scholar] [CrossRef]
  75. 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]
  76. 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] [Green Version]
  77. Frères, P.; Bouznad, N.; Servais, L.; Josse, C.; Wenric, S.; Poncin, A.; Thiry, J.; Moonen, M.; Oury, C.; Lancellotti, P.; et al. Variations of circulating cardiac biomarkers during and after anthracycline-containing chemotherapy in breast cancer patients. BMC Cancer 2018, 18. [Google Scholar] [CrossRef] [PubMed]
  78. Dobson, R.; Ghosh, A.K.; Ky, B.; Marwick, T.; Stout, M.; Harkness, A.; Steeds, R.; Robinson, S.; Oxborough, D.; Adlam, D.; et al. British Society for Echocardiography and British Cardio-Oncology Society guideline for transthoracic echocardiographic assessment of adult cancer patients receiving anthracyclines and/or trastuzumab. Echo. Res. Pract. 2021, 8, G1–G18. [Google Scholar] [CrossRef]
  79. Ponikowski, P.; Voors, A.A.; Anker, S.D.; Bueno, H.; Cleland, J.G.F.; Coats, A.J.S.; Falk, V.; González-Juanatey, J.R.; Harjola, V.-P.; Jankowska, E.A.; et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure. Eur. Heart J. 2016, 37, 2129–2200. [Google Scholar] [CrossRef] [PubMed]
  80. Lang, R.M.; Badano, L.P.; Mor-Avi, V.; Afilalo, J.; Armstrong, A.; Ernande, L.; Flachskampf, F.A.; Foster, E.; Goldstein, S.A.; Kuznetsova, T.; et al. Recommendations for Cardiac Chamber Quantification by Echocardiography in Adults: An Update from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur. Hear. J. Cardiovasc. Imaging 2015, 16, 233–271. [Google Scholar] [CrossRef]
  81. Yu, E.H.C.; Sloggett, C.E.; Iwanochko, R.M.; Rakowski, H.; Siu, S.C. Feasibility and Accuracy of Left Ventricular Volumes and Ejection Fraction Determination by Fundamental, Tissue Harmonic, and Intravenous Contrast Imaging in Difficult-to-Image Patients. J. Am. Soc. Echocardiogr. 2000, 13, 216–224. [Google Scholar] [CrossRef] [PubMed]
  82. Thavendiranathan, P.; Grant, A.D.; Negishi, T.; Plana, J.C.; Popović, Z.B.; Marwick, T.H. Reproducibility of echocardiographic techniques for sequential assessment of left ventricular ejection fraction and volumes: Application to patients undergoing cancer chemotherapy. J. Am. Coll. Cardiol. 2013, 61, 77–84. [Google Scholar] [CrossRef] [Green Version]
  83. Turan, T.; Agac, M.T.; Aykan, A.Ç.; Kul, S.; Akyüz, A.R.; Gökdeniz, T.; Gül, İ.; Cengiz, E.; Boyacı, F.; Erkan, H.; et al. Usefulness of Heart-Type Fatty Acid-Binding Protein and Myocardial Performance Index for Early Detection of 5-Fluorouracil Cardiotoxicity. Angiology 2017, 68, 52–58. [Google Scholar] [CrossRef] [PubMed]
  84. Eidem, B.W.; Sapp, B.G.; Suarez, C.R.; Cetta, F. Usefulness of the myocardial performance index for early detection of anthracycline-induced cardiotoxicity in children. Am. J. Cardiol. 2001, 87, 1120–1122. [Google Scholar] [CrossRef]
  85. Yu, A.F.; Moskowitz, C.S.; Lee Chuy, K.; Yang, J.; Dang, C.T.; Liu, J.E.; Oeffinger, K.C.; Steingart, R.M. Cardiotoxicity Surveillance and Risk of Heart Failure During HER2 Targeted Therapy. JACC Cardio Oncol. 2020, 2, 166–175. [Google Scholar] [CrossRef]
  86. Kim, E.K.; Cho, J.; Kim, J.-Y.; Chang, S.-A.; Park, S.-J.; Choi, J.O.; Lee, S.C.; Ahn, J.S.; Park, S.W.; Im, Y.-H.; et al. Early Decline in Left Ventricular Ejection Fraction Can Predict Trastuzumab-Related Cardiotoxicity in Patients with Breast Cancer: A Study Using 13 Years of Registry Data. Cancer Res. Treat. 2019, 51, 727–736. [Google Scholar] [CrossRef]
  87. Stoddard, M.F.; Seeger, J.; Liddell, N.E.; Hadley, T.J.; Sullivan, D.M.; Kupersmith, J. Prolongation of isovolumetric relaxation time as assessed by Doppler echocardiography predicts doxorubicin-induced systolic dysfunction in humans. J. Am. Coll. Cardiol. 1992, 20, 62–69. [Google Scholar] [CrossRef] [Green Version]
  88. Tassan-Mangina, S.; Codorean, D.; Metivier, M.; Costa, B.; Himberlin, C.; Jouannaud, C.; Blaise, A.M.; Elaerts, J.; Nazeyrollas, P. Tissue Doppler imaging and conventional echocardiography after anthracycline treatment in adults: Early and late alterations of left ventricular function during a prospective study. Eur. J. Echocardiogr. 2006, 7, 141–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Lange, S.A.; Ebner, B.; Wess, A.; Kögel, M.; Gajda, M.; Hitschold, T.; Jung, J. Echocardiography signs of early cardiac impairment in patients with breast cancer and trastuzumab therapy. Clin. Res. Cardiol. 2012, 101, 415–426. [Google Scholar] [CrossRef]
  90. Armstrong, G.T.; Plana, J.C.; Zhang, N.; Srivastava, D.; Green, D.M.; Ness, K.K.; Daniel Donovan, F.; Metzger, M.L.; Arevalo, A.; Durand, J.-B.; et al. Screening Adult Survivors of Childhood Cancer for Cardiomyopathy: Comparison of Echocardiography and Cardiac Magnetic Resonance Imaging. J. Clin. Oncol. 2012, 30, 2876–2884. [Google Scholar] [CrossRef]
  91. Walker, J.; Bhullar, N.; Fallah-Rad, N.; Lytwyn, M.; Golian, M.; Fang, T.; Summers, A.R.; Singal, P.K.; Barac, I.; Kirkpatrick, I.D.; et al. Role of Three-Dimensional Echocardiography in Breast Cancer: Comparison with Two-Dimensional Echocardiography, Multiple-Gated Acquisition Scans, and Cardiac Magnetic Resonance Imaging. J. Clin. Oncol. 2010, 28, 3429–3436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  92. Toro-Salazar, O.H.; Ferranti, J.; Lorenzoni, R.; Walling, S.; Mazur, W.; Raman, S.V.; Davey, B.T.; Gillan, E.; O’Loughlin, M.; Klas, B.; et al. Feasibility of Echocardiographic Techniques to Detect Subclinical Cancer Therapeutics–Related Cardiac Dysfunction among High-Dose Patients When Compared with Cardiac Magnetic Resonance Imaging. J. Am. Soc. Echocardiogr. 2016, 29, 119–131. [Google Scholar] [CrossRef] [PubMed]
  93. Kleijn, S.A.; Aly, M.F.A.; Terwee, C.B.; van Rossum, A.C.; Kamp, O. Three-Dimensional Speckle Tracking Echocardiography for Automatic Assessment of Global and Regional Left Ventricular Function Based on Area Strain. J. Am. Soc. Echocardiogr. 2011, 24, 314–321. [Google Scholar] [CrossRef] [Green Version]
  94. Miyoshi, T.; Tanaka, H.; Kaneko, A.; Tatsumi, K.; Matsumoto, K.; Minami, H.; Kawai, H.; Hirata, K. Left ventricular endocardial dysfunction in patients with preserved ejection fraction after receiving anthracycline. Echocardiography 2014, 31, 848–857. [Google Scholar] [CrossRef]
  95. Mor-Avi, V.; Patel, M.B.; Maffessanti, F.; Singh, A.; Medvedofsky, D.; Zaidi, S.J.; Mediratta, A.; Narang, A.; Nazir, N.; Kachenoura, N.; et al. Fusion of Three-Dimensional Echocardiographic Regional Myocardial Strain with Cardiac Computed Tomography for Noninvasive Evaluation of the Hemodynamic Impact of Coronary Stenosis in Patients with Chest Pain. J. Am. Soc. Echocardiogr. 2018, 31, 664–673. [Google Scholar] [CrossRef]
  96. Zhang, L.; Awadalla, M.; Mahmood, S.S.; Groarke, J.D.; Nohria, A.; Liu, S.; Hassan, M.Z.O.; Cohen, J.V.; Jones-O’Connor, M.; Murphy, S.P.T.; et al. Late Gadolinium Enhancement in Patients with Myocarditis from Immune Checkpoint Inhibitors. J. Am. Coll. Cardiol. 2019, 73, 675. [Google Scholar] [CrossRef]
  97. Motoki, H.; Koyama, J.; Nakazawa, H.; Aizawa, K.; Kasai, H.; Izawa, A.; Tomita, T.; Miyashita, Y.; Kumazaki, S.; Takahashi, M.; et al. Torsion analysis in the early detection of anthracycline-mediated cardiomyopathy. Eur. Heart J. Cardiovasc. Imaging 2012, 13, 95–103. [Google Scholar] [CrossRef]
  98. Tan, T.C.; Scherrer-Crosbie, M. Assessing the Cardiac Toxicity of Chemotherapeutic Agents: Role of Echocardiography. Curr. Cardiovasc. Imaging Rep. 2012, 5, 403–409. [Google Scholar] [CrossRef] [Green Version]
  99. McKillop, J.H.; Bristow, M.R.; Goris, M.L.; Billingham, M.E.; Bockemuehl, K. Sensitivity and specificity of radionuclide ejection fractions in doxorubicin cardiotoxicity. Am. Heart J. 1983, 106, 1048–1056. [Google Scholar] [CrossRef]
  100. Douglas, P.S.; Carr, J.J.; Cerqueira, M.D.; Cummings, J.E.; Gerber, T.C.; Mukherjee, D.; Taylor, A.J. Developing an Action Plan for Patient Radiation Safety in Adult Cardiovascular Medicine. J. Am. Coll. Cardiol. 2012, 59, 1833–1847. [Google Scholar] [CrossRef] [Green Version]
  101. Civelli, M.; Cardinale, D.; Martinoni, A.; Lamantia, G.; Colombo, N.; Colombo, A.; Gandini, S.; Martinelli, G.; Fiorentini, C.; Cipolla, C.M. Early reduction in left ventricular contractile reserve detected by dobutamine stress echo predicts high-dose chemotherapy-induced cardiac toxicity. Int. J. Cardiol. 2006, 111, 120–126. [Google Scholar] [CrossRef]
  102. Friedrich, M.G.; Abdel-Aty, H.; Taylor, A.; Schulz-Menger, J.; Messroghli, D.; Dietz, R. The Salvaged Area at Risk in Reperfused Acute Myocardial Infarction as Visualized by Cardiovascular Magnetic Resonance. J. Am. Coll. Cardiol. 2008, 51, 1581–1587. [Google Scholar] [CrossRef] [Green Version]
  103. Nousiainen, T.; Vanninen, E.; Jantunen, E.; Puustinen, J.; Remes, J.; Rantala, A.; Hartikainen, J. Comparison of echocardiography and radionuclide ventriculography in the follow-up of left ventricular systolic function in adult lymphoma patients during doxorubicin therapy. J. Intern. Med. 2001, 249, 297–303. [Google Scholar] [CrossRef] [Green Version]
  104. Ferreira de Souza, T.; Quinaglia, A.C.; Silva, T.; Osorio Costa, F.; Shah, R.; Neilan, T.G.; Velloso, L.; Nadruz, W.; Brenelli, F.; Sposito, A.C.; et al. Anthracycline Therapy is Associated with Cardiomyocyte Atrophy and Preclinical Manifestations of Heart Disease. JACC Cardiovasc. Imaging 2018, 11, 1045–1055. [Google Scholar] [CrossRef] [PubMed]
  105. Carbone, I.; Childs, H.; Aljizeeri, A.; Merchant, N.; Friedrich, M.G. Importance of Reference Muscle Selection in Quantitative Signal Intensity Analysis of T2-Weighted Images of Myocardial Edema Using a T2 Ratio Method. Biomed Res. Int. 2015, 2015, 1–9. [Google Scholar] [CrossRef] [PubMed]
  106. Farhad, H.; Staziaki, P.V.; Addison, D.; Coelho-Filho, O.R.; Shah, R.V.; Mitchell, R.N.; Szilveszter, B.; Abbasi, S.A.; Kwong, R.Y.; Scherrer-Crosbie, M.; et al. Characterization of the Changes in Cardiac Structure and Function in Mice Treated with Anthracyclines Using Serial Cardiac Magnetic Resonance Imaging. Circ. Cardiovasc. Imaging 2016, 9. [Google Scholar] [CrossRef] [Green Version]
  107. Sarwar, A.; Shapiro, M.D.; Abbara, S.; Cury, R.C. Cardiac Magnetic Resonance Imaging for the Evaluation of Ventricular Function. Semin. Roentgenol. 2008, 43, 183–192. [Google Scholar] [CrossRef] [PubMed]
  108. Drafts, B.C.; Twomley, K.M.; D’Agostino, R.; Lawrence, J.; Avis, N.; Ellis, L.R.; Thohan, V.; Jordan, J.; Melin, S.A.; Torti, F.M.; et al. Low to Moderate Dose Anthracycline-Based Chemotherapy Is Associated with Early Noninvasive Imaging Evidence of Subclinical Cardiovascular Disease. JACC Cardiovasc. Imaging 2013, 6, 877–885. [Google Scholar] [CrossRef] [Green Version]
  109. Neilan, T.G.; Pena-Herrera, D.; Coelho-Filho, O.R.; Jerosch-Herold, M.; Moslehi, J.; Kwong, R. Left ventricular mass by cardiac magnetic resonance imaging and adverse cardiovascular outcomes in patients treated with anthracycline-based chemotherapy. J. Cardiovasc. Magn. Reson. 2012, 14, O30. [Google Scholar] [CrossRef] [Green Version]
  110. Jordan, J.H.; Castellino, S.M.; Meléndez, G.C.; Klepin, H.D.; Ellis, L.R.; Lamar, Z.; Vasu, S.; Kitzman, D.W.; Ntim, W.O.; Brubaker, P.H.; et al. Left Ventricular Mass Change After Anthracycline Chemotherapy. Circ. Hear. Fail. 2018, 11. [Google Scholar] [CrossRef]
  111. Calleja, A.; Poulin, F.; Khorolsky, C.; Shariat, M.; Bedard, P.L.; Amir, E.; Rakowski, H.; McDonald, M.; Delgado, D.; Thavendiranathan, P. Right Ventricular Dysfunction in Patients Experiencing Cardiotoxicity during Breast Cancer Therapy. J. Oncol. 2015, 2015, 1–10. [Google Scholar] [CrossRef] [PubMed]
  112. Ghio, S.; Guazzi, M.; Scardovi, A.B.; Klersy, C.; Clemenza, F.; Carluccio, E.; Temporelli, P.L.; Rossi, A.; Faggiano, P.; Traversi, E.; et al. Different correlates but similar prognostic implications for right ventricular dysfunction in heart failure patients with reduced or preserved ejection fraction. Eur. J. Heart Fail. 2017, 19, 873–879. [Google Scholar] [CrossRef] [Green Version]
  113. Cojan-Minzat, B.O.; Zlibut, A.; Agoston-Coldea, L. Non-ischemic dilated cardiomyopathy and cardiac fibrosis. Heart Fail. Rev. 2020. [Google Scholar] [CrossRef] [PubMed]
  114. Cojan-Minzat, B.O.; Zlibut, A.; Muresan, I.D.; Cionca, C.; Horvat, D.; Kiss, E.; Revnic, R.; Florea, M.; Ciortea, R.; Agoston-Coldea, L. Left Ventricular Geometry and Replacement Fibrosis Detected by cMRI Are Associated with Major Adverse Cardiovascular Events in Nonischemic Dilated Cardiomyopathy. J. Clin. Med. 2020, 9, 1997. [Google Scholar] [CrossRef]
  115. Ylänen, K.; Poutanen, T.; Savikurki-Heikkilä, P.; Rinta-Kiikka, I.; Eerola, A.; Vettenranta, K. Cardiac Magnetic Resonance Imaging in the Evaluation of the Late Effects of Anthracyclines Among Long-Term Survivors of Childhood Cancer. J. Am. Coll. Cardiol. 2013, 61, 1539–1547. [Google Scholar] [CrossRef] [Green Version]
  116. Neilan, T.G.; Coelho-Filho, O.R.; Shah, R.V.; Feng, J.H.; Pena-Herrera, D.; Mandry, D.; Pierre-Mongeon, F.; Heydari, B.; Francis, S.A.; Moslehi, J.; et al. Myocardial Extracellular Volume by Cardiac Magnetic Resonance Imaging in Patients Treated with Anthracycline-Based Chemotherapy. Am. J. Cardiol. 2013, 111, 717–722. [Google Scholar] [CrossRef] [Green Version]
  117. Lawley, C.; Wainwright, C.; Segelov, E.; Lynch, J.; Beith, J.; Mccrohon, J. Pilot study evaluating the role of cardiac magnetic resonance imaging in monitoring adjuvant trastuzumab therapy for breast cancer. Asia. Pac. J. Clin. Oncol. 2012, 8, 95–100. [Google Scholar] [CrossRef]
  118. Wassmuth, R.; Lentzsch, S.; Erdbruegger, U.; Schulz-Menger, J.; Doerken, B.; Dietz, R.; Friedrich, M.G. Subclinical cardiotoxic effects of anthracyclines as assessed by magnetic resonance imaging—A pilot study. Am. Heart J. 2001, 141, 1007–1013. [Google Scholar] [CrossRef]
  119. Coelho-Filho, O.R.; Shah, R.V.; Neilan, T.G.; Mitchell, R.; Moreno, H.; Kwong, R.; Jerosch-Herold, M. Cardiac Magnetic Resonance Assessment of Interstitial Myocardial Fibrosis and Cardiomyocyte Hypertrophy in Hypertensive Mice Treated with Spironolactone. J. Am. Heart Assoc. 2014, 3. [Google Scholar] [CrossRef] [Green Version]
  120. Vita, T.; Gräni, C.; Abbasi, S.A.; Neilan, T.G.; Rowin, E.; Kaneko, K.; Coelho-Filho, O.; Watanabe, E.; Mongeon, F.-P.; Farhad, H.; et al. Comparing CMR Mapping Methods and Myocardial Patterns Toward Heart Failure Outcomes in Nonischemic Dilated Cardiomyopathy. JACC Cardiovasc. Imaging 2019, 12, 1659–1669. [Google Scholar] [CrossRef]
  121. Coelho-Filho, O.R.; Shah, R.V.; Mitchell, R.; Neilan, T.G.; Moreno, H.; Simonson, B.; Kwong, R.; Rosenzweig, A.; Das, S.; Jerosch-Herold, M. Quantification of Cardiomyocyte Hypertrophy by Cardiac Magnetic Resonance. Circulation 2013, 128, 1225–1233. [Google Scholar] [CrossRef] [Green Version]
  122. Friedrich, M.G.; Sechtem, U.; Schulz-Menger, J.; Holmvang, G.; Alakija, P.; Cooper, L.T.; White, J.A.; Abdel-Aty, H.; Gutberlet, M.; Prasad, S.; et al. Cardiovascular Magnetic Resonance in Myocarditis: A JACC White Paper. J. Am. Coll. Cardiol. 2009, 53, 1475–1487. [Google Scholar] [CrossRef] [Green Version]
  123. Zhang, L.; Awadalla, M.; Mahmood, S.S.; Nohria, A.; Hassan, M.Z.O.; Thuny, F.; Zlotoff, D.A.; Murphy, S.P.; Stone, J.R.; Golden, D.L.A.; et al. Cardiovascular magnetic resonance in immune checkpoint inhibitor-associated myocarditis. Eur. Heart J. 2020, 41, 1733–1743. [Google Scholar] [CrossRef] [PubMed]
  124. Ganatra, S.; Neilan, T.G. Immune Checkpoint Inhibitor-Associated Myocarditis. Oncologist 2018, 23, 879–886. [Google Scholar] [CrossRef] [Green Version]
  125. Galán-Arriola, C.; Lobo, M.; Vílchez-Tschischke, J.P.; López, G.J.; de Molina-Iracheta, A.; Pérez-Martínez, C.; Agüero, J.; Fernández-Jiménez, R.; Martín-García, A.; Oliver, E.; et al. Serial Magnetic Resonance Imaging to Identify Early Stages of Anthracycline-Induced Cardiotoxicity. J. Am. Coll. Cardiol. 2019, 73, 779–791. [Google Scholar] [CrossRef]
  126. 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 theHeart Failure Association and the Cardio-Oncology Council of the European Socie. Eur. J. Heart Fail. 2020, 22, 1966–1983. [Google Scholar] [CrossRef]
Figure 1. Standard of care and a surveillance plan.
Figure 1. Standard of care and a surveillance plan.
Medicina 57 00806 g001
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Radulescu, L.M.; Radulescu, D.; Ciuleanu, T.-E.; Crisan, D.; Buzdugan, E.; Romitan, D.-M.; Buzoianu, A.D. Cardiotoxicity Associated with Chemotherapy Used in Gastrointestinal Tumours. Medicina 2021, 57, 806. https://doi.org/10.3390/medicina57080806

AMA Style

Radulescu LM, Radulescu D, Ciuleanu T-E, Crisan D, Buzdugan E, Romitan D-M, Buzoianu AD. Cardiotoxicity Associated with Chemotherapy Used in Gastrointestinal Tumours. Medicina. 2021; 57(8):806. https://doi.org/10.3390/medicina57080806

Chicago/Turabian Style

Radulescu, Liliana Maria, Dan Radulescu, Tudor-Eliade Ciuleanu, Dana Crisan, Elena Buzdugan, Dragos-Mihai Romitan, and Anca Dana Buzoianu. 2021. "Cardiotoxicity Associated with Chemotherapy Used in Gastrointestinal Tumours" Medicina 57, no. 8: 806. https://doi.org/10.3390/medicina57080806

Article Metrics

Back to TopTop