Skip to main content
Top
Gepubliceerd in: Netherlands Heart Journal 11/2018

Open Access 23-08-2018 | Review Article

Cardio-oncology: an overview on outpatient management and future developments

Auteurs: A. J. Teske, M. Linschoten, J. A. M. Kamphuis, W. R. Naaktgeboren, T. Leiner, E. van der Wall, J. Kuball, A. van Rhenen, P. A. Doevendans, M. J. Cramer, F. W. Asselbergs

Gepubliceerd in: Netherlands Heart Journal | Uitgave 11/2018

Abstract

Recent advances in the early detection and treatment of cancer have led to increasing numbers of cancer survivors worldwide. Nonetheless, despite major improvements in the outcome of these patients, long-term side effects of radio- and chemotherapy affect both patient survival and quality of life, independent of the oncological prognosis. Chemotherapy-related cardiac dysfunction is one of the most notorious short-term side effects of anticancer treatment, occurring in ~10% of patients. Progression to overt heart failure carries a strikingly poor prognosis with a 2-year mortality rate of 60%. Early detection of left ventricular damage by periodic monitoring and prompt initiation of heart failure treatment is key in improving cardiovascular prognosis. To meet the growing demand for a specialised interdisciplinary approach for the prevention and management of cardiovascular complications induced by cancer treatment, a new discipline termed cardio-oncology has evolved. However, an uniform, multidisciplinary approach is currently lacking in the Netherlands. This overview provides an introduction and comprehensive summary of this emerging discipline and offers a practical strategy for the outpatient management of this specific patient population.

Introduction

Advances in the early detection and treatment of cancer have led to increasing numbers of cancer survivors worldwide [1, 2]. Nonetheless, despite this substantial progress, long-term side effects of anticancer treatment can affect patient survival and quality of life considerably. Chemotherapy-related cardiac dysfunction (CTRCD) is one of the most notorious short-term side effects of anticancer treatment, occurring in ~10% of patients [3]. To meet the growing demand for a specialised interdisciplinary approach for the prevention and management of cardiovascular complications, a new discipline termed cardio-oncology has emerged since the late 1990s [4]. Cardiovascular toxicity due to chemo- and radiotherapy manifests itself in many other forms beyond myocardial dysfunction including, for example, hypertension, arrhythmias and valvular and coronary artery disease; these forms of toxicity fall outside the scope of this review [5]. However, the main focus of this overview is on the direct cardiotoxic effects of chemotherapy on cardiomyocyte survival. In this article, we aim to provide the clinical cardiologist, haematologists, and oncologists with an overview of this emerging discipline and share our current knowledge regarding the practical implementation of risk stratification, screening, and treatment of CTRCD. Suggested further reading on the following topics, as well as those that fall outside the scope of this overview, are summarised in Tab. 1.
Table 1
Suggested further reading
Ref. no
Author
Year
Topic
Description
[6]
Rochette
2015
Pathophysiology
Cardiotoxic mechanisms of anthracyclines and trastuzumab
[7]
Lenneman
2016
Pathophysiology
Overview of most common anticancer treatments and their mechanism of cardiotoxicity
[8]
Moslehi
2016
Targeted cancer therapy
Overview of cardiovascular toxicity of new targeted (non-anthracycline) cancer therapies
[9]
Curigliano
2012
Definitions/management
ESMO oncology guidelines on cardiac monitoring, referral, and therapy
[10]
Christenson
2015
Early detection
Biomarkers
Overview of circulating biomarkers in predicting chemotherapy-induced cardiac toxicity
[11]
Thavendiranathan
2014
Early detection Echocardiography
Echocardiographic myocardial deformation in the early detection of cardiotoxicity
[12]
Thavendiranathan
2013
Early detection
CMR
The role of cardiac magnetic resonance in the detection of cardiotoxicity
[13]
Plana
2014
Imaging
ESC position paper on non-invasive imaging modalities in cardio-oncology
[14]
Herrmann
2014
Risk stratification and management
Practical aspects regarding cardio-oncology care, including an outline of a risk assessment tool
[15]
Zamorano
2016
Risk stratification and management
ESC position paper on cancer treatments and cardiovascular toxicity
[16]
Lancellotti
2013
Radiotherapy
Consensus paper on imaging and management of cardiovascular complications of radiotherapy
[5]
Naaktgeboren
2017
Long-term outcome
Overview on long-term outcome after anticancer treatment (chemo- and radiotherapy)
[17]
Dalen
2011
Prevention
Cochrane review on cardioprotective interventions for cancer patients receiving anthracyclines
[18]
Kalam
2013
Prevention
Systematic review on cardioprotective therapy for prevention of cardiotoxicity with chemotherapy
[19]
Johnson
2017
Training
Paper exploring training programs for medical specialists in cardio-oncology

Scope of the problem

The incidence of CTRCD is determined by multiple factors, of which the most important involve the administered chemotherapeutic agent(s) and, in the case of anthracyclines, the cumulative dose. Additionally, specific patient characteristics have been shown to be associated with a higher risk of CTRCD. Anthracyclines and trastuzumab (Herceptin) are among the most widely prescribed agents associated with serious cardiotoxicity.
Anthracyclines (e. g. doxorubicin) are a cornerstone in the treatment of numerous haematological and solid malignancies. In a large meta-analysis pooling data from 18 studies involving a total of almost 50,000 patients undergoing treatment with anthracyclines, the incidence of clinically overt and subclinical cardiotoxicity was reported in 6.3% (3.2–9.3%) and 17.9% (11.6–24.2%) of patients respectively [20]. End-stage heart failure was observed in 2–4% of patients and carries a strikingly poor prognosis, with a 2-year mortality rate of up to 60% [21, 22].
Several attempts have been undertaken to reduce the incidence of anthracycline-induced cardiotoxicity. A dose-dependent relationship with heart failure led to restrictions in the administered cumulative dose. Other initiatives have involved the generation of numerous anthracycline analogues (e. g. epirubicin), concomitant administration of cardioprotective drugs (e. g. dexrazoxane), liposomal drug formulations, the application of prolonged infusion regimens to reduce peak plasma dose, and consecutive administration of other cardiotoxic drugs (i. e. trastuzumab), since simultaneous administration dramatically increases CTRCD incidence [18, 2328]. Nevertheless, due to fear of impaired antitumour efficacy, the implementation of several of the above-mentioned preventive actions has been limited in clinical practice. Hence, anthracycline-related cardiotoxicity still remains a significant clinical problem [29].
Trastuzumab is administered in breast cancer patients with human epidermal growth factor receptor 2 (HER2) positive tumours [30]. A meta-analysis found an overall incidence of a left ventricular ejection fraction (LVEF) decline in 11.2% of patients (RR 1.83, 90% CI 1.36–2.47) [31]. Importantly, the prognosis of trastuzumab-induced cardiotoxicity is generally more favourable when compared to anthracycline-induced cardiotoxicity, with a recovery of LVEF after timely cessation of trastuzumab administration in a majority of patients [32].

Definition

Multiple definitions of CTRCD have been proposed in the literature to date, although a consensus is currently still lacking [33]. The most widely adapted definition is a decrease in LVEF of more than 10 percentage points to a value below the lower limit of normal, irrespective of symptoms. The American Society of Echocardiography and the European Association of Cardiovascular Imaging (EACVI) define an LVEF of 53% on echocardiography as the lower limit of normal [13]. Subclinical CTRCD is defined as a global longitudinal strain (GLS) with >15% relative reduction from baseline with preservation of LVEF [13]. Unfortunately, this definition does not cover other signs of cardiotoxicity, such as the detection of cardiac troponin release.

Pathophysiology

The pathophysiological mechanisms leading to CTRCD are complex, incompletely elucidated, and differ among chemotherapeutic agents [7, 8]. Traditionally, for agents that have a direct effect on cardiomyocytes, two types of cardiotoxicity have been proposed [24].
Type I cardiotoxicity is characterised by irreversible damage and related to the cumulative administered dose. Anthracyclines are most well-known for their association with type I cardiotoxicity. Mechanisms believed to play a role in this type of cardiotoxicity are multifactorial and involve (1) the generation of excess reactive oxygen species, (2) accumulation of toxic anthracycline metabolites that interfere with calcium handling and thereby disrupt sarcomere structure and function, (3) interaction with transcription factor topoisomerase-2β, and (4) mitochondrial dysfunction [6, 34, 35].
Type II cardiac damage is believed to cause temporary, reversible dysfunction in a dose-independent manner [32]. Trastuzumab is a classical type II agent, which binds to the HER2 receptor and thereby inhibits downstream associated signalling cascades. It is conceivable that the inhibition of these pathways plays a central role in trastuzumab-associated cardiotoxicity, but the exact mechanism still remains to be discovered [36].
Although the subdivision into type I and type II cardiotoxicity is arbitrary, as persistent LV dysfunction has also been observed in patients treated solely with type II agents, this subdivision is nevertheless still widely applied [37].

Risk stratification

Cardiovascular management of patients receiving cardiotoxic treatment is based on the timely recognition of those at high risk of developing CTRCD. Accurate risk stratification is crucial to enable an effective pre-selection of patients that should be referred to a cardio-oncological team prior to, or in an early phase of, anticancer treatment.
Not all chemotherapeutic agents are (equally) cardiotoxic, as shown in Supplementary Table 1. In this table, we subdivided a subset of the most commonly used chemotherapeutic agents into four categories, based on the incidence of LV dysfunction reported in the literature (group 1: <1%, group 2: 1–5%, group 3: 5–10% and group 4: >10%). It should be noted that the applied definition of LV dysfunction varies between the studies, and that the incidence is an approximation of risk.
One of the most important risk factors for CTRCD in agents that cause type I cardiotoxicity is the administered dose. In patients that require more intensive regimens or have a history of previous malignancy for which they were treated with type I agents, it is important to take the cumulative dose into consideration. However, some patients develop CTRCD even with doses far below the maximum cumulative dose [23]. Therefore, a tolerated and ‘safe’ dose seems to be highly dependent on the presence of patient-related risk factors.
Patient-related risk factors that have been identified thus far include female gender, black race, exposure to cardiotoxic drugs at a young or old age (<15 and >65 years), previous or concomitant chest radiation therapy, obesity and classical cardiovascular risk factors including hypertension and diabetes mellitus [15, 20, 38]. Having ≥3 of these risk factors has been associated with a 5–6 times higher risk of cardiotoxic side effects compared to patients without any risk factors [20]. Nonetheless, in the absence of all these known determinants, some patients still develop severe CTRCD, indicating that unknown factors contribute to individual susceptibility. It is conceivable that the individual genetic profile plays a considerable role in modulating individual risk [39].

Risk prediction models

A few risk prediction models have been published in the literature thus far [14, 40, 41]. The Cardiotoxicity Risk Score (CRS) proposed by the Mayo Clinic takes both patient and treatment risk factors into account (Tab. 2; [14]). This model addresses the a priori risk of developing CTRCD. Patients with a CRS score of ≥4 could benefit from cardiological consultation during and after chemotherapeutic treatment, and those with high risk scores should be closely monitored during and after treatment. However, this risk model, as well as the other models, has not been validated in a prospective setting and its real clinical value remains to be determined. In the absence of validation, the European Society of Cardiology (ESC) therefore currently does not advise the use of a particular model in the position paper on cancer treatments and cardiovascular toxicity released in 2016 [15]. Instead, the committee stresses the importance of clinical judgement in individual risk assessment, which includes clinical history, physical examination and evaluation of cardiac function pre-chemotherapy.
Table 2
Cardiotoxicity Risk Score (CRS)
Medication-related risk a
Examples b
High (risk score 4)
Anthracyclines; trastuzumab; cyclophosphamide; 5‑fluorouracil
Intermediate (risk score 2)
Pertuzumab; vinblastine; capecitabine; ponatinib
Low (risk score 1)
Bevacizumab; imatinib
Rare (risk score 0)
Carboplatin; fludarabine; paclitaxel; rituximab
Patient-related risk factors (1 point per item)
– Cardiomyopathy or heart failure
– Coronary artery disease or equivalent (including peripheral artery disease)
– Hypertension
– Diabetes mellitus
– Prior or concurrent anthracyclines
– Prior or concurrent chest irradiation
– Age <15 years or >65 years
– Female gender
Overall risk by CRS and intensity of monitoring
>6
5–6
3–4
1–2
0
Very high
High
Intermediate
Low
Very low
 
aThe highest medication-related risk score (e. g. 4, 2, 1 or 0) is used for calculation of the CRS
bSee the supplementary table for each separate agent and/or regime. Adapted from: [14]

Early detection of myocardial damage

Circulating biomarkers

Due to the minimal invasiveness, limited costs, and low inter-observer variability, biomarkers constitute an appealing approach to aid in the early detection of subclinical cardiotoxicity (Fig. 1). Most studies have assessed the potential of classical cardiac biomarkers, i. e. cardiac troponin (cTn) and N‑terminal pro-B-type natriuretric peptide (NT-proBNP) [10]. Troponins seem to be the most promising candidates, both in patients treated with anthracyclines and various agents used for targeted-therapy (e. g. trastuzumab) [10, 42]. Nevertheless, repeated sampling is currently necessary to detect cTn elevations, as the optimal timing to reach maximal sensitivity has not yet been established [42].

Multiple-gated acquisition scan

Since the 1970s the mainstay imaging modality for the screening and monitoring of cardiac function in oncology patients has been the multiple-gated acquisition scan [43]. Unfortunately, the only measurement that can be derived from these scans is the LVEF, which is less sensitive for early detection of CTRCD. Another important concern is the radiation exposure (~5–10 mSv/scan) in patients undergoing serial assessments. For example, the Dutch Guidelines for Breast Cancer recommend that patients receiving trastuzumab therapy should undergo cardiac evaluation to determine the LVEF before the start of treatment and subsequently once every 3 months during treatment [44]. The cumulative radiation exposure in patients treated with this agent for 1 year thereby equals ~25–50 mSv, which is comparable to 250–500 chest radiographs or 4–8 CT angiography procedures.

(Strain) echocardiography

Echocardiography is the most suitable imaging modality for the evaluation of patients in preparation for, during, and after cancer therapy, because of its wide availability, easy repeatability, versatility, lack of radiation exposure, and safety in patients with concomitant renal disease. Furthermore, echocardiography allows a comprehensive evaluation of most cardiac structures and multiple parameters besides the LVEF. It has been recommended as the first line screening tool to assess cardiac function in this specific patient population [13]. In particular, the measurement of LVEF by 3D echocardiography has been shown to be feasible and accurate with an error of <5% (compared to a 10% variation in biplane LVEF calculation) [45]. Echocardiographically derived GLS calculates the systolic deformation of the myocardium by a commercially available speckle tracking algorithm (Fig. 2; [46]). This parameter reflects contractile function and is well validated in healthy subjects and in a variety of myocardial disease states. The GLS has been shown to be the single best parameter to predict CTRCD, as a decrease of this parameter is often seen before a relevant reduction of LVEF is observed [11].

Cardiac magnetic resonance

Cardiac magnetic resonance (CMR) is considered the reference standard for the assessment of ventricular function [47]. Due to its superiority in myocardial tissue characterisation, CMR is suitable to detect early tissue damage following chemotherapy. Early changes in tissue composition include myocardial oedema with an increase in LV mass, inflammation, and decrease in myocardial strain [12]. Within months after initiation of therapy LV end-systolic volume increases, and with T1 mapping techniques diffuse interstitial fibrosis, a hallmark of anthracycline-induced cardiotoxicity, can be detected and quantified (Fig. 3a–c). The absence of focal fibrotic lesions results in a lack of late gadolinium enhancement, although contrast-enhanced CMR can be used to exclude other causes of myocardial dysfunction in these patients, such as myocardial infarction. Although unique insights can be obtained with MRI, due to costs and availability CMR is presently not suitable as the first-line imaging technique for regular follow-up imaging.

Therapeutic interventions

With the exclusion of cancer patients in all high-impact heart failure intervention randomised controlled trials regarding the efficacy of, for example, ACE inhibitors and beta-blockers, the response rate of patients with CTRCD to conventional heart failure therapy has not been thoroughly investigated and evidence-based decision-making on optimal treatment is lacking [48].
In a single-centre study by Cardinale et al. (n = 215), treatment response in patients with a decline of LVEF to ≤45% was highly dependent on the timing of treatment initiation [49]. The response rate was the highest (64% responders) among patients with CTRCD that received heart failure treatment <2 months after detection of LV impairment and decreased to only 7% after 4–6 months. Remarkably, no response was observed in patients with CTRCD that received treatment ≥6 months after the last chemotherapeutic cycle (Fig. 4a). Hence, detecting the development of CRTCD as soon as possible guides the optimal timing of treatment initiation, since this seems to be particularly crucial for treatment response. However, in a follow-up study by the same group, a large proportion of patients did not show recovery of cardiac function despite early initiation of conventional heart failure treatment. Only 11% showed full recovery to a mean LVEF of 61%. Cardiac function was partially restored in 71% of the patients, to a mean LVEF of 54%. Notably, 18% of patients did not respond to treatment, with a mean LVEF at the end of the study of 38% (Fig. 4b; [3]). Cardiac outcome for partial and non-responders is significantly worse, including heart failure requiring hospitalisation and cardiac-related death (Fig. 4c; [49]).
Currently, the only practice guidelines on the management of cardiovascular toxicity induced by anticancer treatment have been released by the European Society of Medical Oncology (ESMO) [9]. Guidelines from the ESC and the American Heart Association are still lacking, albeit the ESC recently released the first position paper on cancer treatments and cardiovascular toxicity [15].
The evidence for the use of cardioprotective agents to counteract LVEF decline in patients treated with anthracyclines is marginal, with the exception of dexrazoxane [17, 50]. Cardioprotective agents to prevent trastuzumab-related cardiotoxicity are largely unexplored. The results of the first randomised controlled trials investigating the cardioprotective effect of candesartan and carvedilol were recently published, showing no protection against LVEF decline [51].

Roadmap towards outpatient management

Collaboration between the Departments of Cardiology, Radiology, Haematology and Oncology resulted in a specialised cardio-oncology healthcare pathway, which was launched at the University Medical Centre Utrecht, the Netherlands early in 2015. The aim of this initiative is to improve cardiac outcome in oncology patients by (1) identifying patients at high risk of developing CTRCD before chemotherapeutic treatment is initiated, (2) screen and monitor high-risk patients to enable early detection of (subclinical) cardiac dysfunction which (3) facilitates early treatment initiation in order to improve overall cardiovascular outcome. Patients are monitored up to 1 year after the end of chemotherapy, as a majority of the patients described in the literature develop CTRCD within this time frame [3]. It should be noted that long-term follow-up data in these patients are scarce. Our in-house protocol is delineated below (Fig. 5).

Registration procedure

Patients are referred by the oncologist/haematologist based on in-hospital protocols on cardio-oncology referral. Main indications for referral are planned treatment with cardiotoxic agents, cardiac evaluation before (autologous or allogenic) stem-cell transplantation (SCT), or patients presenting with complaints suggestive of underlying cardiac disease (e. g. heart failure, ischaemia, or arrhythmias). The baseline risk is determined based on the Cardiotoxicity Risk Score (CRS) (Tab. 2). This risk score has been slightly adjusted from its initial publication [14] regarding the risk attributed to the different chemotherapeutic agents (see Supplementary table). Albeit this risk model has not been prospectively validated, we have chosen to incorporate it in our in-house protocol to ensure objective and uniform risk assessment and limit inter-physician variability. All patients pre-SCT as well as high-risk patients (CRS ≥ 4) that will have to undergo treatment with cardiotoxic agents are seen at our cardio-oncology outpatient clinic. Childhood cancer survivors and ex-Hodgkin patients are referred to the LATER and BETER [Dutch for ‘better’] outpatient clinic respectively [52].

Initial cardiac evaluation

Preferentially, the first cardiac evaluation is performed before chemotherapeutic treatment is initiated. This first assessment involves exploration of the cardiac and oncological history (including a detailed assessment of known cardiovascular risk factors as well as past and planned chemotherapeutic regimens), physical examination, laboratory analysis (including kidney function, NT-proBNP and cTn), and an ECG. Furthermore, a complete echocardiographic evaluation is performed to determine cardiac dimensions, valvular function, LVEF (preferably 3D), GLS, diastolic function, right ventricular (RV) function and RV systolic pressure. Reference values can be obtained from the current EACVI guidelines [13]. In short, an LVEF of >53% on 2D/3D echocardiography and a GLS of −19.7% (−20.4% to −18.9%) are considered normal.

Individual follow-up

The interval of follow-up for patients who are treated with agents associated with type I cardiotoxicity is determined by the baseline CRS (Fig. 5). The follow-up duration at the cardio-oncology clinic is typically up to 1 year after the last cycle of chemotherapy. After this period, further cardiac assessment takes place at the BETER outpatient clinic if the patient is found to be eligible [52]. Patients treated with trastuzumab are seen pre-chemotherapy, once every 3 months during treatment, and 1 year after the end of treatment. In asymptomatic patients that develop subclinical CTRCD, the monitoring interval is intensified as these subjects are at high risk for developing heart failure.

Additional examinations

CMR is recommended in patients with clinical CTRCD and can be considered in patients with subclinical CTRCD. Measurements include LVEF, RVEF, delayed enhancement imaging, T1rho-mapping, and determination of GLS. Ischaemia detection using adenosine stress imaging is performed if there is a history of coronary artery disease, suspected ischaemia, or ≥2 cardiovascular risk factor(s) (smoking, diabetes, hypertension, hypercholesterolaemia and a positive family history) in men >40 years and post-menopausal women. Patients with severe CTRCD (LVEF < 45%) or a positive family history for cardiomyopathies are offered a referral for genetic counselling as part of our local policy, as genetic variants in cardiomyopathy-associated genes may have predisposed these patients to CTRCD [53, 54].

Management of patients with CTRCD

Multidisciplinary meetings attended by the cardio-oncologist and oncologist/haematologist take place to discuss cases with (subclinical) CTRCD. At these meetings, the effect of modification(s) to the chemotherapeutic regimen to decrease cardiovascular toxicity is weighed against the consequences of these alterations for the oncological prognosis. Furthermore, in patients that have LV dysfunction prior to the initiation of cancer treatment, the optimal treatment regimen is chosen through shared decision-making.
Type I cardiac dysfunction
In patients with clinical CTRCD, treatment with ACE inhibition (preferentially enalapril) and a beta-blocker (preferentially carvedilol) is indicated. In patients with contraindications for ACE inhibition, an angiotensin-receptor blocker can be considered. Patients that show signs/symptoms of congestion receive loop diuretics. These recommendations are based on two single-centre studies [3, 49]. The value of other agents or the optimal dose are currently unknown. At the moment, there is no evidence that treatment with other heart failure drugs (e. g. aldosterone antagonists) has any added value.
Type II cardiac dysfunction
Upon development of type II cardiac dysfunction, the causal chemotherapeutic agent(s) should be discontinued immediately. After 3–4 weeks, the LVEF and GLS are re-assessed. If LV function has recovered, a re-challenge with the same chemotherapeutic agent(s) can be attempted under strict cardiological monitoring. Even though there are currently no evidence-based recommendations for heart failure medication in patients with this type of cardiotoxicity, treatment according to the ESC and ESMO guidelines should be considered if there is a persistent decline in LVEF and signs/symptoms of decompensation [9, 48].
Subclinical cardiac dysfunction
At this point in time, there is no evidence that initiation of heart failure medication in patients with subclinical cardiac dysfunction improves outcome. To prevent unnecessary treatment, we have decided to wait with initiation of heart failure treatment until the patient develops signs of clinical CTRCD, during the intensified follow-up.

Future perspectives

Despite the advantages in our understanding of this specific heart-failure entity with regard to the underlying pathophysiological mechanisms, improving diagnostic accuracy, and implementation of specific therapeutic interventions, there are still several unresolved issues and challenges within the field of cardio-oncology. To detect opportunities for improvement at this moment in time, the routinely provided cardiovascular care in oncology patients prior to the cardio-oncology era has to be investigated. This includes the frequency at which baseline cardiac function is assessed, the incidence of cardiovascular complications, referral patterns, treatment initiation- and response. We acknowledge that the (adjusted) CRS is probably insufficient to accurately identify high-risk patients. Development of sophisticated algorithms, which can be applied in the clinical field, will be an important focus of future trials and registries in order to optimise resources and pursue a cost-effective health care system. Risk stratification models need prospective validation and further improvement by the identification of additional (genetic) risk factors. Furthermore, personalised chemotherapeutic regimens, with their increasing complexity, go hand in hand with the need to establish interactions between agents and the combined effect on the cardiovascular system. Early detection of subclinical cardiac damage and dysfunction seems essential to optimise the treatment response rate; therefore, suitable biomarkers as well as the timing of biomarker sampling and echocardiographic monitoring need to be investigated. Ongoing therapeutic trials (Tab. 3) will shed more light on the potential of conventional heart failure treatment in this population as well as the optimal timing of treatment initiation. The establishing of specialised cardio-oncology units across the Netherlands will speed the development of this field, optimising the cost-benefit ratio of chemotherapeutic treatment with the potential to improve both oncological and cardiac outcome [19, 55]. Furthermore, we will launch the ONCOR prospective multicentre registry in the near future, in which we aim to collect information on patients visiting cardio-oncology units across the Netherlands. Information from this registry will enable further national and international studies to improve the prognosis of this patient population.
Table 3
Ongoing clinical trials on the treatment of cardiotoxicity
Location
NCT number
Title
Intervention
Start date
 
USA
NCT02943590
STOP-CA (Statins TO Prevent the Cardiotoxicity from Anthracyclines)
Atorvastatin or placebo
January 2017
Recruiting
USA
NCT02674204
STOP Heart Disease in Breast Cancer Survivors Trial
Atorvastatin or placebo
May 2016
Recruiting
USA
NCT02096588
Detection and Prevention of Anthracycline-Related Cardiac Toxicity with Concurrent Simvastatin
Simvastatin or placebo
May 2014
Active, not recruiting
Canada
NCT03186404
Statins for the Primary Prevention of Heart Failure in Patients Receiving Anthracycline Pilot Study
Atorvastatin or placebo
July 2017
Not yet recruiting
UK
NCT03265574
PROACT: Can We Prevent Chemotherapy-Related Heart Damage in Patients with Breast Cancer?
Enalapril or placebo
September 2017
Not yet recruiting
Italy
NCT01968200
Prevention of Anthracycline-Induced Cardiotoxicity
Enalapril
December 2012
Active, not recruiting
USA
NCT02177175
Carvedilol for the Prevention of Anthracycline/Anti-HER2 Therapy Associated Cardiotoxicity among Women with HER2-Positive Breast Cancer Using Myocardial Strain Imaging for Early Risk Stratification
Carvedilol or placebo
June 2014
Active, not recruiting
Brazil
NCT01724450
Carvedilol Effect in Preventing Chemotherapy-Induced Cardiotoxicity
Carvedilol or placebo
June 2012
Recruiting
USA
NCT02717507
Carvedilol in Preventing Heart Failure in Childhood Cancer Survivors
Carvedilol or placebo
April 2016
Recruiting
USA
NCT01347970
Pharmacologic Reversal of Ventricular Remodeling in Childhood Cancer Survivors at Risk for Congestive Heart Failure (PREVENT-CHF): A Phase IIB Randomized Placebo-Controlled Trial
Carvedilol or placebo
May 2012
Active, not recruiting
Italy
NCT02236806
Cardiotoxicity Prevention in Breast Cancer Patients Treated with Anthracyclines and/or Trastuzumab
Bisoprolol or ramipril or placebo
July 2015
Recruiting
Canada
NCT01016886
Multidisciplinary Approach to Novel Therapies in Cardiology Oncology Research
Perindopril or bisoprolol or placebo
September 2010
Active, not recruiting
USA
NCT01009918
Lisinopril or Coreg CR® in Reducing Side Effects in Women with Breast Cancer Receiving Trastuzumab
Carvedilol or lisinopril or placebo
March 2010
Active, not recruiting

Acknowledgements

We would like to thank Eefke Petersen, Denise Buter, and Roel de Weijer of the Department of Haematology and Rhodé Bijlsma and Alexander de Graeff of the Department of Medical Oncology for their logistical support in our cardio-oncology healthcare pathway at the University Medical Centre Utrecht.

Conflict of interest

A.J. Teske, M. Linschoten, J.A.M. Kamphuis, W.R. Naaktgeboren, T. Leiner, E. van der Wall, J. Kuball, A. van Rhenen, P.A. Doevendans, M.J. Cramer and F.W. Asselbergs declare that they have no competing interests.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Onze productaanbevelingen

Netherlands Heart Journal

Het Netherlands Heart Journal wordt uitgegeven in samenwerking met de Nederlandse Vereniging voor Cardiologie en de Nederlandse Hartstichting. Het tijdschrift is Engelstalig en wordt gratis beschikbaa ...

Literatuur
1.
go back to reference de Moor JS, Mariotto AB, Parry C, et al. Cancer survivors in the United States: prevalence across the survivorship trajectory and implications for care. Cancer Epidemiol Biomarkers Prev. 2013;22:561–70.CrossRef de Moor JS, Mariotto AB, Parry C, et al. Cancer survivors in the United States: prevalence across the survivorship trajectory and implications for care. Cancer Epidemiol Biomarkers Prev. 2013;22:561–70.CrossRef
2.
go back to reference Verdecchia A, Francisci S, Brenner H, et al. Recent cancer survival in Europe: a 2000–02 period analysis of EUROCARE-4 data. Lancet Oncol. 2007;8:784–96.CrossRef Verdecchia A, Francisci S, Brenner H, et al. Recent cancer survival in Europe: a 2000–02 period analysis of EUROCARE-4 data. Lancet Oncol. 2007;8:784–96.CrossRef
3.
go back to reference Cardinale D, Colombo A, Bacchiani G, et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation. 2015;131:1981–8.CrossRef Cardinale D, Colombo A, Bacchiani G, et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation. 2015;131:1981–8.CrossRef
4.
go back to reference Cardinale D. A new frontier: cardio-oncology. Cardiologia. 1996;41:887–91.PubMed Cardinale D. A new frontier: cardio-oncology. Cardiologia. 1996;41:887–91.PubMed
5.
go back to reference Naaktgeboren WR, Linschoten M. de Graeff, et al. Cardiovascular health in adult cancer survivors. Maturitas. 2017;105:37–45.CrossRef Naaktgeboren WR, Linschoten M. de Graeff, et al. Cardiovascular health in adult cancer survivors. Maturitas. 2017;105:37–45.CrossRef
6.
go back to reference Rochette L, Guenancia C, Gudjoncik A, et al. Anthracyclines/trastuzumab: new aspects of cardiotoxicity and molecular mechanisms. Trends Pharmacol Sci. 2015;36:326–48.CrossRef Rochette L, Guenancia C, Gudjoncik A, et al. Anthracyclines/trastuzumab: new aspects of cardiotoxicity and molecular mechanisms. Trends Pharmacol Sci. 2015;36:326–48.CrossRef
7.
go back to reference Lenneman CG, Cardio-oncology SDB. an update on cardiotoxicity of cancer-related treatment. Circ Res. 2016;118:1008–20.CrossRef Lenneman CG, Cardio-oncology SDB. an update on cardiotoxicity of cancer-related treatment. Circ Res. 2016;118:1008–20.CrossRef
8.
go back to reference Moslehi JJ. Cardiovascular toxic effects of targeted cancer therapies. N Engl J Med. 2016;375:1457–67.CrossRef Moslehi JJ. Cardiovascular toxic effects of targeted cancer therapies. N Engl J Med. 2016;375:1457–67.CrossRef
9.
go back to reference Curigliano G, Cardinale D, Suter T, et al. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2012;23(Suppl 7):vii155–66.CrossRef Curigliano G, Cardinale D, Suter T, et al. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol. 2012;23(Suppl 7):vii155–66.CrossRef
10.
go back to reference Christenson ES, James T, Agrawal V, Park BH. Use of biomarkers for the assessment of chemotherapy-induced cardiac toxicity. Clin Biochem. 2015;48:223–35.CrossRef Christenson ES, James T, Agrawal V, Park BH. Use of biomarkers for the assessment of chemotherapy-induced cardiac toxicity. Clin Biochem. 2015;48:223–35.CrossRef
11.
go back to reference Thavendiranathan P, Poulin F, Lim KD, et al. Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: a systematic review. J Am Coll Cardiol. 2014;63:2751–68.CrossRef Thavendiranathan P, Poulin F, Lim KD, et al. Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: a systematic review. J Am Coll Cardiol. 2014;63:2751–68.CrossRef
12.
go back to reference Thavendiranathan P, Wintersperger BJ, Flamm SD, et al. Cardiac MRI in the assessment of cardiac injury and toxicity form cancer chemotherapy: a systematic review. Circ Cardiovasc Imaging. 2013;6:1080–91.CrossRef Thavendiranathan P, Wintersperger BJ, Flamm SD, et al. Cardiac MRI in the assessment of cardiac injury and toxicity form cancer chemotherapy: a systematic review. Circ Cardiovasc Imaging. 2013;6:1080–91.CrossRef
13.
go back to reference Plana JC, Galderisis M, Barac A, 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;27:911–39.CrossRef Plana JC, Galderisis M, Barac A, 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;27:911–39.CrossRef
14.
go back to reference Herrmann J, Lerman A, Sandhu NP, Villarraga HR, Mulvagh SL, Kohli M. Evaluation and management of patients with heart disease and cancer: cardio-oncology. Mayo Clin Proc. 2014;89:1287–306.CrossRef Herrmann J, Lerman A, Sandhu NP, Villarraga HR, Mulvagh SL, Kohli M. Evaluation and management of patients with heart disease and cancer: cardio-oncology. Mayo Clin Proc. 2014;89:1287–306.CrossRef
15.
go back to reference Zamorano JL, Lancellotti P, Rodriguez Muñoz D, 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;37:2768–801.CrossRef Zamorano JL, Lancellotti P, Rodriguez Muñoz D, 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;37:2768–801.CrossRef
16.
go back to reference Lancellotti P, Nkomo VT, Badano LP, et al. Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26:1013–32.CrossRef Lancellotti P, Nkomo VT, Badano LP, et al. Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography. J Am Soc Echocardiogr. 2013;26:1013–32.CrossRef
17.
go back to reference van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev. 2011;6:CD3917. van Dalen EC, Caron HN, Dickinson HO, Kremer LC. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev. 2011;6:CD3917.
18.
go back to reference Kalam K, Marwick TH. Role of cardioprotective therapy for prevention of cardiotoxicity with chemotherapy: a systematic review and meta-analysis. Eur J Cancer. 2013;49:2900–9.CrossRef Kalam K, Marwick TH. Role of cardioprotective therapy for prevention of cardiotoxicity with chemotherapy: a systematic review and meta-analysis. Eur J Cancer. 2013;49:2900–9.CrossRef
19.
go back to reference Johnson MN, Steingart R, Carver J. How to develop a cardio-oncology fellowship. Heart Fail Clin. 2017;13:361–6.CrossRef Johnson MN, Steingart R, Carver J. How to develop a cardio-oncology fellowship. Heart Fail Clin. 2017;13:361–6.CrossRef
20.
go back to reference Lotrionte M, Biondi-Zoccai G, Abbate A, et al. Review and meta-analysis of incidence and clinical predictors of anthracycline cardiotoxicity. Am J Cardiol. 2013;112:1980–4.CrossRef Lotrionte M, Biondi-Zoccai G, Abbate A, et al. Review and meta-analysis of incidence and clinical predictors of anthracycline cardiotoxicity. Am J Cardiol. 2013;112:1980–4.CrossRef
21.
go back to reference Yeh ETH. Cardiovascular complications of cancer therapy: diagnosis, pathogenesis, and management. Circulation. 2004;109:3122–31.CrossRef Yeh ETH. Cardiovascular complications of cancer therapy: diagnosis, pathogenesis, and management. Circulation. 2004;109:3122–31.CrossRef
22.
go back to reference Felker GM, Thompson RE, Hare JM, et al. Underlying causes and long-term survival in patients with initially unexplained cardiomyopathy. N Engl J Med. 2000;342:1077–84.CrossRef Felker GM, Thompson RE, Hare JM, et al. Underlying causes and long-term survival in patients with initially unexplained cardiomyopathy. N Engl J Med. 2000;342:1077–84.CrossRef
23.
go back to reference Von Hoff DD, Layard MW, Basa P, et al. Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med. 1979;91:710–7.CrossRef Von Hoff DD, Layard MW, Basa P, et al. Risk factors for doxorubicin-induced congestive heart failure. Ann Intern Med. 1979;91:710–7.CrossRef
24.
go back to reference Ewer MS, Ewer SM. Cardiotoxicity of anticancer treatments. Nat Rev Cardiol. 2015;12:547–58.CrossRef Ewer MS, Ewer SM. Cardiotoxicity of anticancer treatments. Nat Rev Cardiol. 2015;12:547–58.CrossRef
25.
go back to reference Minotti G, Menna P, Salvatorelli E, Cairo G, Anthracyclines GL. molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004;56:185–229.CrossRef Minotti G, Menna P, Salvatorelli E, Cairo G, Anthracyclines GL. molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol Rev. 2004;56:185–229.CrossRef
26.
go back to reference Rafiyath SM, Rasul M, Lee B, Wei G, Lamba G, Liu D. Comparison of safety and toxicity of liposomal doxorubicin vs. conventional anthracyclines: a meta-analysis. Exp Hematol Oncol. 2012;1:10.CrossRef Rafiyath SM, Rasul M, Lee B, Wei G, Lamba G, Liu D. Comparison of safety and toxicity of liposomal doxorubicin vs. conventional anthracyclines: a meta-analysis. Exp Hematol Oncol. 2012;1:10.CrossRef
27.
go back to reference van Dalen EC, van der Pal HJ, Kremer LC. Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy. Cochrane Database Syst Rev. 2016;3:CD5008.PubMed van Dalen EC, van der Pal HJ, Kremer LC. Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy. Cochrane Database Syst Rev. 2016;3:CD5008.PubMed
28.
go back to reference Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92.CrossRef Slamon DJ, Leyland-Jones B, Shak S, et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783–92.CrossRef
30.
go back to reference Senkus E, Kyriakides S, Ohno S, et al. Primary breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;5:8–30.CrossRef Senkus E, Kyriakides S, Ohno S, et al. Primary breast cancer: ESMO clinical practice guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2015;5:8–30.CrossRef
31.
go back to reference Mantarro S, Rossi M, Bonifazi M, et al. Risk of severe cardiotoxicity following treatment with trastuzumab: a meta-analysis of randomized and cohort studies of 29,000 women with breast cancer. Intern Emerg Med. 2016;11:123–40.CrossRef Mantarro S, Rossi M, Bonifazi M, et al. Risk of severe cardiotoxicity following treatment with trastuzumab: a meta-analysis of randomized and cohort studies of 29,000 women with breast cancer. Intern Emerg Med. 2016;11:123–40.CrossRef
32.
go back to reference Ewer MS, Vooletich MT, Durand JB, et al. Reversibility of trastuzumab-related cardiotoxicity: new insights based on clinical course and response to medical treatment. J Clin Oncol. 2005;23:7820–6.CrossRef Ewer MS, Vooletich MT, Durand JB, et al. Reversibility of trastuzumab-related cardiotoxicity: new insights based on clinical course and response to medical treatment. J Clin Oncol. 2005;23:7820–6.CrossRef
33.
go back to reference Bloom MW, Hamo CE, Cardinale D, et al. Cancer therapy-related cardiac dysfunction and heart failure: Part 1: definitions, pathophysiology, risk factors, and imaging. Circ Heart Fail. 2016;9:e2661.CrossRef Bloom MW, Hamo CE, Cardinale D, et al. Cancer therapy-related cardiac dysfunction and heart failure: Part 1: definitions, pathophysiology, risk factors, and imaging. Circ Heart Fail. 2016;9:e2661.CrossRef
34.
go back to reference Mushlin PS, Cusack BJ, Boucek RJ Jr, Andrejuk T, Li X, Olson RD. Time-related increases in cardiac concentrations of doxorubicinol could interact with doxorubicin to depress myocardial contractile function. Br J Pharmacol. 1993;110:975–82.CrossRef Mushlin PS, Cusack BJ, Boucek RJ Jr, Andrejuk T, Li X, Olson RD. Time-related increases in cardiac concentrations of doxorubicinol could interact with doxorubicin to depress myocardial contractile function. Br J Pharmacol. 1993;110:975–82.CrossRef
35.
go back to reference Zhang S, Liu X, Bawa-Khalfe T, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 2012;18:1639–42.CrossRef Zhang S, Liu X, Bawa-Khalfe T, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med. 2012;18:1639–42.CrossRef
36.
go back to reference De Keulenaer GW, Doggen K, Lemmens K. The vulnerability of the heart as a pluricellular paracrine organ: lessons from unexpected triggers of heart failure in targeted ErbB2 anticancer therapy. Circ Res. 2010;106:35–46.CrossRef De Keulenaer GW, Doggen K, Lemmens K. The vulnerability of the heart as a pluricellular paracrine organ: lessons from unexpected triggers of heart failure in targeted ErbB2 anticancer therapy. Circ Res. 2010;106:35–46.CrossRef
37.
go back to reference Telli ML, Hunt SA, Carlson RW, Guardino AE. Trastuzumab-related cardiotoxicity: calling into question the concept of reversibility. J Clin Oncol. 2007;25:3525–33.CrossRef Telli ML, Hunt SA, Carlson RW, Guardino AE. Trastuzumab-related cardiotoxicity: calling into question the concept of reversibility. J Clin Oncol. 2007;25:3525–33.CrossRef
38.
go back to reference Krischer JP, Epstein S, Cuthbertson DD, Goorin AM, Epstein ML, Lipshultz SE. Clinical cardiotoxicity following anthracycline treatment for childhood cancer: the Pediatric Oncology Group experience. J Clin Oncol. 1997;15:1544–52.CrossRef Krischer JP, Epstein S, Cuthbertson DD, Goorin AM, Epstein ML, Lipshultz SE. Clinical cardiotoxicity following anthracycline treatment for childhood cancer: the Pediatric Oncology Group experience. J Clin Oncol. 1997;15:1544–52.CrossRef
39.
go back to reference Linschoten M, Teske AJ, Cramer MJ, van der Wall E, Asselbergs FW. Chemotherapy-related cardiac dysfunction—a systematic review of genetic variants modulating individual risk. Circ Genom Precis. Med. 2018;11:e1753. Linschoten M, Teske AJ, Cramer MJ, van der Wall E, Asselbergs FW. Chemotherapy-related cardiac dysfunction—a systematic review of genetic variants modulating individual risk. Circ Genom Precis. Med. 2018;11:e1753.
40.
go back to reference Dranitsaris G, Rayson D, Vincent M, et al. The development of a predictive model to estimate cardiotoxic risk for patients with metastatic breast cancer receiving anthracyclines. Breast Cancer Res Treat. 2008;107:443–50.CrossRef Dranitsaris G, Rayson D, Vincent M, et al. The development of a predictive model to estimate cardiotoxic risk for patients with metastatic breast cancer receiving anthracyclines. Breast Cancer Res Treat. 2008;107:443–50.CrossRef
41.
go back to reference Ezaz G, Long JB, Gross CP, Chen J. Risk prediction model for heart failure and cardiomyopathy after adjuvant trastuzumab therapy for breast cancer. J Am Heart Assoc. 2014;3:e472.CrossRef Ezaz G, Long JB, Gross CP, Chen J. Risk prediction model for heart failure and cardiomyopathy after adjuvant trastuzumab therapy for breast cancer. J Am Heart Assoc. 2014;3:e472.CrossRef
42.
go back to reference Cardinale D, Biasillo G, Salvatici M, Sandri MT, Cipolla CM. Using biomarkers to predict and to prevent cardiotoxicity of cancer therapy. Expert Rev Mol Diagn. 2017;17:245–56.CrossRef Cardinale D, Biasillo G, Salvatici M, Sandri MT, Cipolla CM. Using biomarkers to predict and to prevent cardiotoxicity of cancer therapy. Expert Rev Mol Diagn. 2017;17:245–56.CrossRef
43.
go back to reference Gottdiener JS, Mathisen DJ, Borer JS, et al. Doxorubicin cardiotoxicity: assessment of late left ventricular dysfunction by radionuclide cineangiography. Ann Intern Med. 1981;94:430–5.CrossRef Gottdiener JS, Mathisen DJ, Borer JS, et al. Doxorubicin cardiotoxicity: assessment of late left ventricular dysfunction by radionuclide cineangiography. Ann Intern Med. 1981;94:430–5.CrossRef
45.
go back to reference Thavendiranathan P, Grant AD, Negishi T, et al. 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.CrossRef Thavendiranathan P, Grant AD, Negishi T, et al. 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.CrossRef
46.
go back to reference Teske AJ, De Boeck BW, Melman PG, Sieswerda GT, Doevendans PA, Cramer MJ. Echocardiographic quantification of myocardial function using tissue deformation imaging, a guide to image acquisition and analysis using tissue Doppler and speckle tracking. Cardiovasc Ultrasound. 2007;5:27. Teske AJ, De Boeck BW, Melman PG, Sieswerda GT, Doevendans PA, Cramer MJ. Echocardiographic quantification of myocardial function using tissue deformation imaging, a guide to image acquisition and analysis using tissue Doppler and speckle tracking. Cardiovasc Ultrasound. 2007;5:27.
47.
go back to reference Grothues F, Smith GC, Moon JC, et al. Comparison of interstudy reproducibility of cardiovascular magnetic resonance with two-dimensional echocardiography in normal subjects and in patients with heart failure or left ventricular hypertrophy. Am J Cardiol. 2002;90:29–34.CrossRef Grothues F, Smith GC, Moon JC, et al. Comparison of interstudy reproducibility of cardiovascular magnetic resonance with two-dimensional echocardiography in normal subjects and in patients with heart failure or left ventricular hypertrophy. Am J Cardiol. 2002;90:29–34.CrossRef
48.
go back to reference Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail. 2016;18:891–975.CrossRef Ponikowski P, Voors AA, Anker SD, et al. 2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure: The Task Force for the diagnosis and treatment of acute and chronic heart failure of the European Society of Cardiology (ESC). Developed with the special contribution of the Heart Failure Association (HFA) of the ESC. Eur J Heart Fail. 2016;18:891–975.CrossRef
49.
go back to reference Cardinale D, Colombo A, Lamantia G, et al. Anthracycline-induced cardiomyopathy: clinical relevance and response to pharmacologic therapy. J Am Coll Cardiol. 2010;55:213–20.CrossRef Cardinale D, Colombo A, Lamantia G, et al. Anthracycline-induced cardiomyopathy: clinical relevance and response to pharmacologic therapy. J Am Coll Cardiol. 2010;55:213–20.CrossRef
50.
go back to reference Avila MS, Ayub-Ferreira SM, de Barros Wanderley MR Jr, et al. Carvedilol for prevention of chemotherapy-related cardiotoxicity: the CECCY trial. J Am Coll Cardiol. 2018;71:2281–90.CrossRef Avila MS, Ayub-Ferreira SM, de Barros Wanderley MR Jr, et al. Carvedilol for prevention of chemotherapy-related cardiotoxicity: the CECCY trial. J Am Coll Cardiol. 2018;71:2281–90.CrossRef
51.
go back to reference Boekhout AH, Gietema JA, Milojkovic Kerklaan B, et al. Angiotensin II-receptor inhibition with candesartan to prevent trastuzumab-related cardiotoxic effects in patients with early breast cancer: a randomized clinical trial. Jama Oncol. 2016;2:1030–7.CrossRef Boekhout AH, Gietema JA, Milojkovic Kerklaan B, et al. Angiotensin II-receptor inhibition with candesartan to prevent trastuzumab-related cardiotoxic effects in patients with early breast cancer: a randomized clinical trial. Jama Oncol. 2016;2:1030–7.CrossRef
52.
go back to reference Dekker N, van ‘t Veer MB, Aleman BM, et al. The BETER survivorship care initiative for Hodgkin lymphoma; a tailored survivorship care for late effects of treatment. Ned Tijdschr Geneeskd. 2015;159:A9269.PubMed Dekker N, van ‘t Veer MB, Aleman BM, et al. The BETER survivorship care initiative for Hodgkin lymphoma; a tailored survivorship care for late effects of treatment. Ned Tijdschr Geneeskd. 2015;159:A9269.PubMed
53.
go back to reference Wasielewski M, van Spaendonck-Zwarts KY, Westerink ND, et al. Potential genetic predisposition for anthracycline-associated cardiomyopathy in families with dilated cardiomyopathy. Open Heart. 2014;1:e116.CrossRef Wasielewski M, van Spaendonck-Zwarts KY, Westerink ND, et al. Potential genetic predisposition for anthracycline-associated cardiomyopathy in families with dilated cardiomyopathy. Open Heart. 2014;1:e116.CrossRef
54.
go back to reference Linschoten M, Teske AJ, Baas AF, et al. Truncating titin (TTN) variants in chemotherapy-induced cardiomyopathy. J Card Fail. 2017;23:476.CrossRef Linschoten M, Teske AJ, Baas AF, et al. Truncating titin (TTN) variants in chemotherapy-induced cardiomyopathy. J Card Fail. 2017;23:476.CrossRef
55.
go back to reference Shee K, Kono AT, D’Anna SP, et al. Maximizing the benefit-cost ratio of anthracyclines in metastatic breast cancer: case report of a patient with a complete response to high-dose doxorubicin. Case Rep Oncol. 2016;9:840–6.CrossRef Shee K, Kono AT, D’Anna SP, et al. Maximizing the benefit-cost ratio of anthracyclines in metastatic breast cancer: case report of a patient with a complete response to high-dose doxorubicin. Case Rep Oncol. 2016;9:840–6.CrossRef
Metagegevens
Titel
Cardio-oncology: an overview on outpatient management and future developments
Auteurs
A. J. Teske
M. Linschoten
J. A. M. Kamphuis
W. R. Naaktgeboren
T. Leiner
E. van der Wall
J. Kuball
A. van Rhenen
P. A. Doevendans
M. J. Cramer
F. W. Asselbergs
Publicatiedatum
23-08-2018
Uitgeverij
Bohn Stafleu van Loghum
Gepubliceerd in
Netherlands Heart Journal / Uitgave 11/2018
Print ISSN: 1568-5888
Elektronisch ISSN: 1876-6250
DOI
https://doi.org/10.1007/s12471-018-1148-7

Andere artikelen Uitgave 11/2018

Netherlands Heart Journal 11/2018 Naar de uitgave