Journal Information
Vol. 35. Issue 9.
Pages 485-494 (September 2016)
Visits
7820
Vol. 35. Issue 9.
Pages 485-494 (September 2016)
Review article
Open Access
Organization and implementation of a cardio-oncology program
Organização e implementação de uma consulta de cardio-oncologia
Visits
7820
Manuela Fiuzaa,
Corresponding author
manuela.fiuza@gmail.com

Corresponding author.
, Leonor Ribeirob, Andreia Magalhãesa, Ana Rita Sousab, Miguel N. Menezesa, Marília Jorgec, Luís Costab, Fausto J. Pintoa
a Serviço de Cardiologia, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte – EPE, Centro Académico Medicina de Lisboa / Centro Cardiovascular, Universidade de Lisboa, Lisboa, Portugal
b Serviço de Oncologia, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte – EPE, Centro Académico Medicina de Lisboa, Lisboa, Portugal
c Serviço de Radioterapia, Hospital de Santa Maria, Centro Hospitalar Lisboa Norte – EPE, Lisboa, Portugal
This item has received

Under a Creative Commons license
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (3)
Show moreShow less
Tables (5)
Table 1. Comparison of the European Society of Cardiology37 and National Cancer Institute38 guidelines according to the severity of hypertension.
Table 2. Steps in the monitoring of cardiac events during cancer therapy.
Table 3. Cardiotoxicity risk score based on cancer drugs used and patient-related risk factors.
Table 4. Recommendations for monitoring of cardiac events during cancer therapy.
Table 5. Criteria for referral for cardio-oncology consultation.
Show moreShow less
Abstract

Considerable advances in cancer therapies in recent decades have reshaped the prognosis of cancer patients. There are now estimated to be over 20 million cancer survivors in the USA and Europe, numbers unimaginable a few years ago. However, this increase in survival, along with the aging of the patient population, has been accompanied by a rise in adverse cardiovascular effects, particularly when there is a previous history of heart disease. The incidence of cardiotoxicity continues to grow, which can compromise the effectiveness of cancer therapy. Cardiotoxicity associated with conventional therapies, especially anthracyclines and radiation, is well known, and usually leads to left ventricular dysfunction. However, heart failure represents only a fraction of the cardiotoxicity associated with newer therapies, which have diverse cardiovascular effects. There are few guidelines for early detection, prevention and treatment of cardiotoxicity of cancer treatments, and no well-established tools for screening these patients. Echocardiography is the method of choice for assessment of patients before, during and after cancer treatment.

It therefore makes sense to adopt a multidisciplinary approach to these patients, involving cardiologists, oncologists and radiotherapists, collaborating in the development of new training modules, and performing clinical and translational research in a cardio-oncology program. Cardio-oncology is a new frontier in medicine and has emerged as a new medical subspecialty that concentrates knowledge, understanding, training and treatment of cardiovascular comorbidities, risks and complications in patients with cancer in a comprehensive approach to the patient rather than to the disease.

Keywords:
Cancer
Cardiotoxicity
Cardio-oncology
Chemotherapy
Radiation
Heart failure
Resumo

A taxa de sobrevivência dos doentes (dts) com cancro aumentou consideravelmente nas últimas décadas, havendo atualmente mais de 20 milhões de sobreviventes nos EUA e na Europa, números inimagináveis até há poucos anos. Para tal, muito contribuiu o aparecimento de novos fármacos (terapêuticas biológicas).

No entanto, estes benefícios na sobrevivência e o envelhecimento da população foram acompanhados de um aumento da taxa de efeitos adversos cardiovasculares, sobretudo se já havia doença cardíaca prévia. De facto, a incidência de cardiotoxicidade (CTX) tem sido continuamente mais evidente, comprometendo a eficácia das terapêuticas oncológicas (TO). São conhecidos os efeitos adversos cardíacos das TO tradicionais (antraciclinas e radioterapia torácica), como a insuficiência cardíaca. Contudo, esta representa apenas uma fração das manifestações de CTX, pois muitas das novas terapêuticas têm efeitos cardiovasculares diversos. As orientações clínicas existentes para fazer a deteção precoce, a prevenção e o tratamento da CTX dos tratamentos oncológicos, não abrangem todas as manifestações de CTX e ainda são poucas as ferramentas para a avaliação destes dts. A ecocardiografia é atualmente o método de escolha para avaliar os dts nas fases pré, durante e após a TO.

Dada a dimensão e relevância desta questão, faz todo o sentido falar de cardio-oncologia, uma nova subespecialidade médica. O número crescente de dts oncológicos com problemas cardíacos implica uma abordagem que deve ser partilhada entre cardiologistas, oncologistas e radioterapeutas.

Esta nova área do conhecimento médico deve também incluir uma componente formativa clínica, sendo também desejável a implementação de projetos de investigação clínicos e transacionais.

Palavras-chave:
Cancro
Cardiotoxicidade
Cardio-oncologia
Quimioterapia
Radioterapia
Insuficiência cardíaca
Full Text
Introduction

Cardiovascular disease and cancer together account for around 60% of deaths in the western world. In Portugal, data from the National Institute of Statistics for 2013 show that the leading cause of death is cardiovascular disease (29.5%), followed by cancer (24.3%).1

Nevertheless, survival rates for both diseases have increased in recent decades, as a result of significant advances in treatment. Five-year survival in the USA improved from 50% of patients diagnosed with cancer between 1975 and 1977 to 68% in those diagnosed between 1999 and 2005, and there are currently over 14 million cancer survivors, numbers unimaginable a few years ago.2,3 However, as survival improves, the late adverse cardiovascular effects of these therapies have become increasingly important.

It therefore makes sense to adopt a multidisciplinary approach to these patients, involving cardiologists, oncologists and radiotherapists in a cardio-oncology program. Interestingly, cardiovascular disease and cancer have risk factors in common, such as obesity and diabetes, and are often found in the same patient.

Cardiotoxicity is a common and well-known adverse effect of many conventional cancer therapies, especially anthracyclines and chest radiation, but may also occur with new biological therapies. It can affect survival and quality of life independently of cancer prognosis.

The most frequent adverse cardiovascular effects of cancer treatments include left ventricular dysfunction (symptomatic or asymptomatic), hypertension, arrhythmias, prolonged QT interval, thromboembolism and myocardial ischemia.4,5 Renal failure can also occur.

Unlike the cardiotoxicity associated with conventional cancer therapies (type I), that associated with biological therapies such as trastuzumab (type II) is usually reversible with discontinuation of treatment6,7 or treatable by medical therapy, such as hypertension associated with angiogenesis inhibitors such as sunitinib and bevacizumab.8,9 Furthermore, as therapeutic options evolve, conventional treatments are likely to be associated with one or more biological therapies, increasing the probability of cardiotoxicity.

Although cardiotoxicity associated with systemic cancer therapies is well understood, less is known about early and late cardiotoxicity due to new biological therapies or the early and late consequences of interactions between therapies.10 It is thus increasingly important to identify early biomarkers of cardiac involvement.11

The aim of this article is to provide a brief review of the state of the art and to recommend clinical practices that will improve the early detection and treatment of patients with cardiovascular complications arising from cancer therapies, by means of a multidisciplinary approach in a cardio-oncology program.

Anti-cancer drugsAnthracyclines

Anthracyclines (including doxorubicin, epirubicin, daunorubicin and idarubicin) are among the most commonly used drugs for chemotherapy and are especially effective in treating breast cancer and lymphoma. However, their effectiveness can be compromised by adverse cardiac effects, particularly heart failure (HF), which may appear early (weeks or months) or late (years) after treatment.

Recent research has shown that anthracyclines selectively inhibit the genetic expression of cardiac muscle. Doxorubicin binds to DNA by intercalating between specific bases and preventing the synthesis of DNA, RNA or both, thereby disrupting replication and transcription. The cardiac effects of the anthracyclines are associated with the inhibition of topoisomerase II and the formation of free oxygen radicals. These changes in genetic expression can lead to cardiomyocyte apoptosis and progressive loss of myofibrils in cardiac muscle.12,13

The main risk factor for the development of HF in anthracycline chemotherapy is the cumulative dose. The incidence of symptomatic cardiotoxicity with doxorubicin ranges between 5% for 400 mg/m2 to 48% for 700 mg/m2.14,15

It is estimated that more than half of patients treated with anthracyclines will develop cardiac changes within six years, and they have a five-fold greater probability of suffering HF than those not receiving these drugs.16

Other risk factors include older (>65 years) or younger (<18 years) age, female gender, hypertension, previous heart disease, diabetes, and previous chest radiation. Combined therapy with cyclophosphamide and taxanes, common in the treatment of breast cancer, can increase the risk of cardiotoxicity.17

Trastuzumab

Trastuzumab, a monoclonal antibody with a high affinity for the HER2 receptor, has changed the natural history of patients with HER2-positive breast cancer, which accounts for 25% of breast cancers and is associated with a worse prognosis. Treatment with trastuzumab increases survival by 33% and reduces the risk of recurrence by 50% in these patients.18,19 However, it is associated with cardiotoxicity, with a greater incidence of symptomatic and asymptomatic left ventricular dysfunction (ranging from 4% as an adjuvant to 27% in metastatic disease).

The cardiotoxicity found with trastuzumab differs from that associated with anthracyclines, since it is dose-independent and does not lead to the ultrastructural alterations typical of the latter.20

Proposed methods to reduce the cardiotoxicity of anthracyclines and trastuzumab include measurement of biomarkers such as troponin I and NT-proBNP, echocardiographic assessment of baseline left ventricular ejection fraction which is then monitored throughout treatment, and determination of ventricular global longitudinal strain (Figure 1). According to the results of these exams, cancer treatment may need to be suspended or discontinued and treatment for HF begun.21–24

Figure 1.

Decision algorithm for detection and monitoring of types I and II cardiotoxicity during chemotherapy with trastuzumab. CV: cardiovascular; LVEF: left ventricular ejection fraction. Adapted from Rashi et al.60

(0.38MB).

This type of cardiotoxicity, characterized by left ventricular dysfunction, should be treated using the same drugs used for HF of other etiologies (angiotensin-converting enzyme inhibitors, angiotensin receptor blockers and beta-blockers), in accordance with the European and American guidelines for HF.25–27

Angiogenesis inhibitors

Tumor growth is initially fed by nearby blood vessels, but when the tumor reaches a certain size, these vessels are no longer sufficient and in order to continue growing, the tumor must acquire the ability to generate new vessels, a process known as angiogenesis. To do so, tumor cells increase vascular pro-angiogenic factors such as endothelial growth factor (VEGF) and reduce angiogenesis-inhibiting factors.28–30

Hypertension is the adverse cardiovascular effect most frequently associated with VEGF inhibitors. The mechanisms behind VEGFI-induced hypertension are complex and multifactorial and are not fully understood. VEGF has vasodilatory effects by enhancing nitric oxide release; inhibition results in vasoconstriction, increased peripheral vascular resistance and hypertension. Other effects of VEGFI, such as endothelial dysfunction, vascular remodeling, arterial stiffness and capillary rarefaction, also appear to contribute significantly.32–34 A potent vasoconstrictor, endothelin-1 (ET-1), has also been implicated in VEGFI-induced hypertension. Its secretion appears to be increased in the presence of endothelial dysfunction, and there is evidence from clinical trials that raised ET-1 levels parallel increases in blood pressure (BP) in patients undergoing treatment with VEGFI.35

Virtually all patients treated with VEGFI have raised BP, and some develop hypertension; the greatest rise in BP is seen in the first cycle of VEGFI treatment. Hypertension is not an adverse effect of the treatment but a result of VEGFI-targeted treatment. This has raised the possibility that hypertension could be used as an indicator of the efficacy of the antiangiogenic response to VEGFI therapy and hence as a biomarker of good treatment outcome.30–32

As well as a dose-dependent rise in BP, angiogenesis inhibitors are also associated with increased risk of proteinuria. Some patients may develop glomerular disease or thrombotic glomerular microangiopathy leading to renal failure. These are reversible following immediate discontinuation of treatment.

There may be neurological complications in patients who develop hypertension, including posterior leukoencephalopathy syndrome, which is reversible by treatment discontinuation.

VEGFI-induced raised BP has a characteristic profile of rapid onset, within hours of beginning treatment, and systolic BP is more affected than diastolic. The incidence of hypertension is dose-related and is higher when multiple antiangiogenic agents are used in association.

The aim of optimal antihypertensive therapy is to enable VEGFI treatment to continue safely without altering the dose. To this end, a baseline cardiovascular assessment is recommended before therapy begins, including serial BP measurement. It is also important to assess renal function and proteinuria, since renal involvement can cause new-onset hypertension or worsen existing hypertension. The goal is not to exclude patients from such treatment but to assess their baseline risk and to monitor them closely. Antihypertensive therapy should aim to maintain BP below 140/90 mmHg, or 130/80 mmHg in the presence of diabetes or chronic renal failure (Table 1).33–36

Table 1.

Comparison of the European Society of Cardiology37 and National Cancer Institute38 guidelines according to the severity of hypertension.

NCI-CTC  Recommendations  ESC  Recommendations 
None  Optimal: <120/80 mmHg
Normal: SBP 120-129 and/or DBP 80-84 mmHg 
No intervention 
1 - Asymptomatic transient (<24 h) BP rise of >20 mmHg (diastolic) or >150/100 mmHg if BP previously normal  None  High normal: SBP 130-139 and/or DBP 85-89 mmHg  Lifestyle changes if >1 RF 
2 - Persistent recurrent rise of >20 mmHg (diastolic) or >150/100 mmHg if BP previously normal  Begin antihypertensive therapy (monotherapy)  Grade 1 HT: SBP 140-159 and/or DBP 90-99 mmHg  Lifestyle changes
Then add BP drugs targeting <140/90 mmHg if TOD, CKD stage 3 or diabetes 
3 - BP >160/100 mmHg  More aggressive antihypertensive therapy  Grade 2 HT: SBP 160-179 and/or DBP 100/109 mmHg  Lifestyle changes
Immediate BP drugs targeting <140/90 mmHg if >1 RF 
4 - Malignant HT, transient or permanent, neurological deficit, or hypertensive crisis  Urgent intervention  Grade 3 HT: ≥180/110  Lifestyle changes
Immediate BP drugs targeting <140/90 mmHg if symptomatic CVD, CKD stage ≥4 or diabetes with TOD 

BP: blood pressure; CKD: chronic kidney disease; CVD: cardiovascular disease; DBP: diastolic blood pressure; ESC: European Society of Cardiology guidelines (2013); HT: hypertension; NCI-CTC: National Cancer Institute Common Terminology Criteria for Adverse Events v. 4.0 (2010); RF: risk factor; SBP: systolic blood pressure; TOD: target organ damage.

Various antihypertensive drug classes are used to treat hypertension in cancer patients. All are effective, and no class has been shown to be superior to any other, but care must be taken with the non-dihydropyridine calcium channel blockers verapamil and diltiazem, which are also CYP3A4 inhibitors. Since endothelial nitric oxide is a putative mediator of angiogenesis, agents such as nitrates and nebivolol that increase nitric oxide levels are recommended for hypertension treatment in these patients.33

When therapy begins, regular BP assessment is recommended during the first treatment cycle and every 2-3 weeks thereafter; patients should be advised to measure their own BP at home. Assessment should be more frequent if patients are also taking drugs that increase the risk of hypertension such as anti-inflammatory agents or erythropoietin.

If systolic BP reaches ≥200 mmHg or diastolic BP reaches ≥100 mmHg, the dose of anticancer drugs should be reduced or treatment suspended. The dosage should be maintained at the highest level that the patient can tolerate, aiming to reduce the short-term risk of events associated with hypertension (stroke, myocardial infarction, or HF) while maintaining effective doses of antiangiogenic drugs.

There are no specific guidelines for VEGFI-induced hypertension, and so therapy should be based on the European Society of Cardiology (ESC) and National Cancer Institute guidelines37,38 (Figure 2).

Figure 2.

Initial assessment and monitoring of hypertension during therapy with angiogenesis inhibitors. AI: angiogenesis inhibitor; BP: blood pressure; CCB: calcium channel blocker; HT: hypertension. Adapted from Ederhy et al.36

(0.28MB).
Radiotherapy

Radiation of the heart can lead to radiation-induced heart disease (RIHD), which is related to cumulative dose (the product of the number of treatments and radiation dose) and can be exacerbated by chemotherapy, especially anthracyclines. Manifestations of RIHD can be acute but are usually only seen years after treatment. It has various adverse effects, including HF, coronary disease, pericarditis, valve disease and arrhythmias. It may also be aggravated by risk factors that are common to heart disease and cancer, such as obesity, sedentary lifestyles, diabetes, hypertension and smoking.

The dose-dependent increase in cardiovascular disease following chest radiation is well documented, especially with lymphoma and breast cancer (particularly of the left breast), and cardiovascular disease is the leading non-cancer cause of death in these patients. Studies analyzing the long-term risk/benefit ratio have shown that the positive effect of radiotherapy may in fact be partly canceled out by cardiac complications.39,40 However, these data are mostly retrospective and based on treatment protocols that are no longer used. The prevalence of RIHD is unknown with new radiotherapy protocols, which include planning the area to radiate by three-dimensional computed tomography (CT), using lower doses and reducing the size of the radiation field, in order to protect the heart. In breast cancer, for example, CT planning avoids including the heart in the radiation field, while excluding the internal mammary lymph nodes enables doses to the heart to be reduced. Another technique is fractionating the radiation dose; some studies have shown that the more the dose is fractionated, the lower the incidence of acute pericarditis and myocardial necrosis. These new protocols are intended to reduce the incidence of RIHD, but there are as yet no long-term follow-up data.

Ionizing radiation can damage virtually all cardiac structures. Its effects on the vascular system are seen at both microvascular and macrovascular levels. At the microvascular level, it leads to loss of endothelial cells, triggering an inflammatory response, vascular injury and ischemia. The resulting fibrosis appears to be due to the process of cell repair rather than to the direct effect of radiation. Macrovascular changes include accelerated arteriosclerosis and coronary artery obstruction, leading to acute coronary syndromes at younger ages.

Diffuse fibrosis after radiotherapy, which can be identified histologically in both myocardium and pericardium, can result in restrictive myocarditis and constrictive pericarditis.41–45

There are no guidelines for cardiac monitoring in these patients. To minimize the risk of RIHD, patients at high risk for cardiac events should be identified before beginning radiotherapy and, when appropriate, cardiac assessment should be repeated for the rest of their lives.

In patients who have undergone chest radiotherapy, cardiac function should be reassessed every 10 years, or every five years in those considered at high risk for RIHD (Figure 3).46

Figure 3.

Algorithm for patient management after chest radiotherapy. ACS: acute coronary syndrome; CAD: coronary artery disease; CMR: cardiac magnetic resonance; CT: computed tomography; HF: heart failure; LV: left ventricular; RIHD: radiation-induced heart disease; US: ultrasound. High-risk patients defined as having had anterior or left-side chest irradiation with ≥1 risk factors for RIHD. Adapted from Lancellotti et al.46

(0.27MB).
Cardio-oncology program

A cardio-oncology program should have three main components: (1) cardio-oncology clinic; (2) training; and (3) research.

Cardio-oncology clinicObjectives

  • -

    to provide specialized cardiological care to patients with cancer and a history of cardiovascular disease or who develop cardiac complications during cancer therapy;

  • -

    to optimize cardiac care in cancer patients undergoing potentially cardiotoxic therapy;

  • -

    to improve knowledge of cardiac complications of cancer treatments;

  • -

    to promote early detection of cardiotoxicity (using clinical, laboratory and imaging biomarkers, of which echocardiography is the most important) and to establish intervention strategies to optimize cardiological care;

  • -

    to improve patients’ prognosis through a multidisciplinary and integrated approach involving different health professionals (physicians, nurses and technicians).

The clinic's protocols for monitoring cardiotoxicity should include three steps (Table 2): assessment before beginning cancer therapy, particularly in patients with cardiovascular risk factors; assessment during treatment, in order to detect and treat cardiovascular complications promptly; and monitoring after treatment (Tables 3 and 4). The use of a risk score enables better identification of patients who should be referred for cardio-oncology consultation (Table 5).

Table 2.

Steps in the monitoring of cardiac events during cancer therapy.

Before therapy  - Risk assessment
- Identification and control of cardiovascular risk factors
- Personalization of cancer therapy to minimize cardiovascular risk 
During therapy  - Prompt detection of cardiotoxicity
- Identification and control of cardiovascular risk factors 
Following therapy  - Monitoring of symptoms and scheduled cardiovascular assessment
- Control of cardiovascular risk factors 
Table 3.

Cardiotoxicity risk score based on cancer drugs used and patient-related risk factors.

Medication-related risk  Patient-related risk factors 
High (risk score 4):
Anthracyclines; cyclophosphamide; trastuzumab 
- Age <15 or >65 years
- Female gender
- Hypertension, diabetes
- Cardiomyopathy, HF, CAD, PAD
- Prior anthracycline chemotherapy
Intermediate (risk score 2):
Docetaxel; pertuzumab; sunitinib; sorafenib 
Low (risk score 1):
Bevacizumab; dasatinib; imatinib; lapatinib 
Rare (risk score 0):
Etoposide; rituxumab; thalidomide 

CAD: coronary artery disease; HF: heart failure; PAD: peripheral arterial disease.

Risk categories by drug-related risk score plus number of patient-related risk factors: risk score >6: very high, 5-6: high, 3-4: intermediate; 1-2: low, 0: very low.

Table 4.

Recommendations for monitoring of cardiac events during cancer therapy.

Risk of cardiotoxicity  Patients  Monitoring 
High  Structural heart disease or very low LVEF  - Follow-up in cardio-oncology consultations
- Potentially cardiotoxic therapy only in exceptional circumstances 
Intermediate  Cardiovascular risk factors and LVEF >40%  Baseline assessment:
- ECG; blood tests (creatinine, HbA1c, lipids)
- Troponin, NT-proBNP
- Echocardiogram (LVEF and strain)
Assessment during treatment:
- Troponin in each cycle
- Echocardiogram (LVEF and strain)
- Consider cardiovascular therapy (beta-blockers, ACE inhibitors, statins)
Long-term assessment:
- ECG plus echocardiogram (strain) plus troponin and NT-proBNP at the end of treatment, after 6 and 12 months, and then every 3-4 years 
Low  Asymptomatic, no cardiovascular risk factors or structural heart disease  Baseline assessment:
- ECG; blood tests (creatinine, HbA1c, lipids)
- Troponin, NT-proBNP
- Echocardiogram (LVEF and strain)
Assessment during follow-up:
- ECG plus echocardiogram (LVEF and strain) at end of treatment 

ACE: angiotensin-converting enzyme; ECG: electrocardiogram; LVEF: left ventricular ejection fraction.

Adapted from Fernandez et al.50

Table 5.

Criteria for referral for cardio-oncology consultation.

Patients at high or intermediate risk for optimization of therapy (ACE inhibitors, beta-blockers, statins)
- Prior treatment with doxorubicin ≥300 mg/m2 and/or mediastinal radiotherapy ≥30 Gy
- Structural heart disease, HF, coronary disease or arrhythmias
- Uncontrolled hypertension, dyslipidemia or diabetes 
Alterations on ECG or baseline echocardiogram or during follow-up:
- Fall in LVEF of >10% with baseline LVEF ≥55%
- Abnormal GLS (>-19%) or >15% fall
- Positive troponin
- Chest pain, dyspnea, syncope, arrhythmias
- Hypertension refractory to therapy 

ACE: angiotensin-converting enzyme; ECG: electrocardiogram; GLS: global longitudinal strain; HF: heart failure.

Appropriate algorithms that are easy to apply in clinical practice are needed in order to enable prompt detection and monitoring of cardiotoxicity.47–50

Finally, continuous quality control should be implemented, possibly using the ‘plan, do, check, act’ (PDCA) method. The results will permit analysis of the quality of the program, evaluating therapeutic decisions, outcomes, safety profile, and patient satisfaction measured by questionnaires. There should also be recommendations on how to combat risk factors, including patient education. Records need to be kept and study and training plans, both undergraduate and postgraduate, should be prepared.55

Training

As cardio-oncology is a new frontier in medicine, it should also include a training component, both undergraduate and postgraduate, as recommended by the ESC. It is important that there should be a period of training in cardio-oncology for both oncologists and cardiologists, at a basic or advanced level according to individual options.51–58

Research

Clinical and translational research projects should be organized aimed at early identification of cardiotoxicity and of individual susceptibility to developing adverse cardiac effects.

Cardiotoxicity is an increasing concern in clinical and preclinical trials of new drugs. Adverse cardiac effects often result in discontinuation of cancer therapy. There is therefore a growing need for better prediction of the risk of cardiotoxicity of new drugs at an early stage in their investigation.59–61

Conclusion

Survival rates of cancer patients have increased, due to new therapies. However, this notable achievement may be overshadowed by the adverse effects of these therapies on the cardiovascular system. Manifestations of cardiotoxicity may be acute or late, months or years after the end of treatment. It can also exacerbate or unmask existing cardiac conditions.

The development of cardiovascular disease during cancer therapy may lead to changes in the therapeutic regime, such as alterations in dosage, duration of cycles, and temporary or permanent discontinuation of therapy, and can thus reduce its effectiveness.

The new medical subspecialty of cardio-oncology has arisen in response to the need to detect cardiovascular involvement as early as possible and to optimize cardiological treatment in cancer patients, both during therapy and long-term. The importance of a multidisciplinary approach is recognized by European and American medical societies and other organizations, and should become standard in the follow-up of these patients. Close collaboration between specialties will also help establish clinical guidelines and clinical and translational research protocols that can respond to the need to predict, prevent and treat cardiotoxicity.

Conflicts of interest

The authors have no conflicts of interest to declare.

References
[1]
Causas de morte 2013. Edição 2015. Editor: Instituto Nacional de Estatística, I.P. www.ine.pt.
[2]
A. Jemal, R. Siegel, J. Xu, et al.
Cancer statistics, 2010.
CA Cancer J Clin, 60 (2010), pp. 277-300
[3]
R. Siegel, C. DeSantis, K. Virgo, et al.
Cancer treatment and survivorship statistics, 2012.
CA Cancer J Clin, 62 (2012), pp. 220-241
[4]
E.T.H. Yeh, C.L. Bickford.
Cardiovascular complications of cancer therapy: incidence, pathogenesis, diagnosis, and management.
J Am Coll Cardiol, 53 (2009), pp. 2231-2247
[5]
G. Curigliano, E.L. Mayer, H.J. Burnstein.
Cardiac toxicity from systemic cancer therapy: a comprehensive review.
Prog Cardiovasc Dis, 53 (2010), pp. 94-104
[6]
M.S. Ewer, M.T. Vooletich, J.B. Durand, et al.
Reversibility of trastuzumab-related cardiotoxicity: new insights based on clinical course and response to medical treatment.
J Clin Oncol, 23 (2005), pp. 7820-7826
[7]
M.S. Ewer, S.M. Lippman, I.I. Type.
chemotherapy-related cardiac dysfunction: time to recognize a new entity.
J Clin Oncol, 23 (2005), pp. 2900-2902
[8]
T.F. Chu, M.A. Rupnick, R. Kerkela, et al.
Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib.
Lancet, 370 (2007), pp. 2011-2019
[9]
S.M. Gressett, S.R. Shah.
Intricacies of bevacizumab-induced toxicities and their management.
Ann Pharmacother, 43 (2009), pp. 490-501
[10]
S.E. Lipshultz, M.J. Adams, S.D. Colan, et al.
Long-term cardiovascular toxicity in children, adolescents, and young adults who receive cancer therapy: pathophysiology, course, monitoring, management, prevention and research directions: a scientific statement from the American Heart Association.
Circulation, 128 (2013), pp. 1927-1995
[11]
N. Fallah-Rad, J.R. Walker, A. Wassef, et al.
The utility of cardiac biomarkers, tissue velocity and strain imaging, and cardiac magnetic resonance imaging in predicting early left ventricular dysfunction in patients with human epidermal growth factor receptor II-positive breast cancer treated with adjuvant trastuzumab therapy.
J Am College Cardiol, 57 (2011), pp. 2263-2270
[12]
S. Zhang, X. Liu, T. Bawa-Khalfe, et al.
Identification of the molecular basis of doxorubicin-induced cardiotoxicity.
Nat Med, 18 (2012), pp. 1639-1642
[13]
MD Anderson Cancer Center. Cancer and the heart programs. Available at: www. cancerandtheheart.org [accessed 16.11.15].
[14]
S.M. Swain, F.S. Whaley, M.S. Ewer.
Congestive heart failure in patients treated with doxorubicin: a retrospective analysis of three trials.
Cancer, 97 (2003), pp. 2869-2879
[15]
L. Gianni, E.H. Herman, S.E. Lipshultz, et al.
Anthracycline cardiotoxicity: from bench to bedside.
J Clin Oncol, 26 (2008), pp. 3777-3784
[16]
S.E. Lipshultz, S.R. Lipsitz, S.E. Sallan, et al.
Chronic progressive cardiac dysfunction years after doxorubicin therapy for childhood acute lymphoblastic leukemia.
J Clin Oncol, 23 (2005), pp. 2629-2636
[17]
R.M. Witteles, M. Telli.
Underestimating cardiac toxicity in cancer trials: lessons learned?.
J Clin Oncol, 30 (2012), pp. 1916-1918
[18]
E.H. Romond, E.A. Perez, J. Bryant, et al.
Trastuzumab plus adjuvant chemotherapy for operable HER2-positive breast cancer.
N Engl J Med, 353 (2005), pp. 1673-1684
[19]
I. Smith, M. Procter, R.D. Gelber, et al.
2-year follow-up of trastuzumab after adjuvant chemotherapy in HER2-positive breast cancer: a randomised controlled trial.
[20]
M.L. Telli, S.A. Hunt, R.W. Carlson, et al.
Trastuzumab-related cardiotoxicity: calling into question the concept of reversibility.
J Clin Oncol, 25 (2007), pp. 3525-3533
[21]
S.A. Crone, Y.Y. Zhao, L. Fan, et al.
ErbB2 is essential in the prevention of dilated cardiomyopathy.
Nat Med, 8 (2002), pp. 459-465
[22]
D. Cardinale, M.T. Sandri, A. Colombo, et al.
Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy.
Circulation, 109 (2004), pp. 2749-2754
[23]
D. Cardinale, A. Colombo, R. Torrisi, et al.
Trastuzumab-induced cardiotoxicity: clinical and prognostic implications of troponin I evaluation.
J Clin Oncol, 28 (2010), pp. 3910-3916
[24]
M. Fiuza, A. Magalhaes.
Trastuzumab and cardiotoxicity.
Cardiotoxicity of oncologic treatments: InTech,
[25]
P. Ponikowski, A.A. Voors, S.D. Anker, et al.
ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure.
Eur Heart J, 37 (2016), pp. 2129-2200
[26]
C.W. Yancy, M. Jessup, B. Bozkurt, et al.
2013 ACCF/AHA guideline for the management of heart failure. A report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines.
Circulation, 128 (2013), pp. e240-e327
[27]
J.L. Hare, J.K. Brown, R. Leano, et al.
Use of myocardial deformation imaging to detect preclinical myocardial dysfunction before conventional measures in patients undergoing breast cancer treatment with trastuzumab.
Am Heart J, 158 (2009), pp. 294-301
[28]
X. Peng, L. Pentassuglia, D.B. Sawyer.
Emerging anticancer therapeutic targets and the cardiovascular system: is there cause for concern?.
Circ Res, 106 (2010), pp. 1022-1034
[29]
B.I. Levy.
Blood pressure as a potential biomarker of the efficacy angiogenesis inhibitor.
Ann Oncol, 20 (2009), pp. 200-203
[30]
J.-J. Mourad, G. des Guetz, H. Debbabi, et al.
Blood pressure rise following angiogenesis inhibition by bevacizumab. A crucial role for microcirculation.
Ann Oncol, 19 (2008), pp. 927-934
[31]
M.L. Maitland, G.L. Bakris, H.R. Black, et al.
Initial assessment, surveillance, and management of blood pressure in patients receiving vascular endothelial growth factor signaling pathway inhibitors.
J Natl Cancer Inst, 102 (2010), pp. 596-604
[32]
B. Nazer, B.D. Humphreys, J. Moslehi.
Effects of novel angiogenesis inhibitors for the treatment of cancer on the cardiovascular system: focus on hypertension.
Circulation, 124 (2011), pp. 1687-1691
[33]
L.E. Okamoto, A. Gamboa, C.A. Shibao, et al.
Nebivolol, but not metoprolol, lowers blood pressure in nitric oxide-sensitive human hypertension.
Hypertension, 64 (2014), pp. 1241-1247
[34]
N. Jesus-Gonzalez, E. Robinson, J. Moslehi, et al.
Management of antiangiogenic therapy-induced hypertension.
Hypertension, 60 (2012), pp. 607-615
[35]
F. Bohm, J. Pernow.
The importance of endothelin-1 for vascular dysfunction in cardiovascular disease.
Cardiovasc Res, 76 (2007), pp. 8-18
[36]
S. Ederhy, H. Izzedine, C. Massard, et al.
Cardiac side effects of molecular targeted therapies: towards a better dialogue between oncologists and cardiologists.
Crit Rev Oncol Hematol, 80 (2011), pp. 369-379
[37]
G. Mancia, R. Fagard, K. Narkiewicz, et al.
ESH/ESC Guidelines for the management of arterial hypertension. The Task Force for the management of arterial hypertension of the European Society of Hypertension (ESH) and of the European Society of Cardiology (ESC).
J Hypertens, 31 (2013), pp. 1281-1357
[38]
NCI Common Terminology Criteria for Adverse Events (v4.03: June 14, 2010).
[39]
N.K. Taunk, B.G. Haffty, J.B. Kostis, et al.
Radiation-induced heart disease: pathologic abnormalities and putative mechanisms.
Front Oncol, 5 (2015), pp. 39
[40]
S.C. Darby, D.J. Cutter, M. Boerma, et al.
Radiation-related heart disease: current knowledge and future prospects.
Int J Radiat Oncol Biol Phys, 76 (2010), pp. 656-665
[41]
S.C. Darby, M. Ewertz, P. McGale, et al.
Risk of ischemic heart disease in women after radiotherapy for breast cancer.
N Engl J Med, 368 (2013), pp. 987-998
[42]
G. Nilsson, L. Holmberg, H. Garmo, et al.
Distribution of coronary artery stenosis after radiation for breast cancer.
J Clin Oncol, 30 (2012), pp. 380-386
[43]
N.L. Weintraub, W.K. Jones, D. Manka.
Understanding radiation-induced vascular disease.
J Am Coll Cardiol, 55 (2010), pp. 1237-1239
[44]
S.W. Yusuf, S. Sami, I.N. Daher.
Radiation-induced heart disease: a clinical update.
Cardiol Res Pract, 2011 (2011), pp. 317659
[45]
C. Jaworski, J.A. Mariani, G. Wheeler, et al.
Cardiac complications of thoracic irradiation.
J Am Coll Cardiol, 61 (2013), pp. 2319-2328
[46]
P. Lancellotti, V.T. Nkomo, L.P. Badano, 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.
Eur Heart J Cardiovasc Imaging, 14 (2013), pp. 721-740
[47]
I.N. Daher, T.R. Daigle, N. Bhatia, et al.
The prevention of cardiovascular disease in cancer survivors.
Tex Heart Inst J, 39 (2012), pp. 190-198
[48]
J.C. Plana, M. Galderisi, A. Barac, 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 Echocardiog, 27 (2014), pp. 911-939
[49]
G. Curigliani, D. Cardinale, T. Suter, on behalf of the ESMO Guidelines Working Group, et al.
Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines.
Ann Oncol, 23 (2012), pp. vii55-vii66
[50]
T.L. Fernandez, S.O.R. Rodriguez, S.V. Lopez, et al.
Cardio-oncology.
Editor CTO, (2015),
[51]
M. Chavez-MacGregor, J. Niu, N. Zhang, et al.
Cardiac monitoring during adjuvant trastuzumab-based chemotherapy among older patients with breast cancer.
J Clin Oncol, 33 (2015), pp. 2176-2183
[52]
R.M. Witteles, X. Bosch.
Myocardial protection during cardiotoxic chemotherapy.
Circulation, 132 (2015), pp. 1835-1845
[53]
T. Eschenhagen, T. Force, M.S. Ewer, et al.
Cardiovascular side effects of cancer therapies: a position statement from the Heart Failure Association of the European Society of Cardiology.
Eur J Heart Fail, 13 (2011), pp. 1-10
[54]
J. Herrmann, A. Lerman, N.P. Sandhu, et al.
Evaluation and management of patients with heart disease and cancer: cardio-oncology.
Mayo Clin Proc, 89 (2014), pp. 1287-1306
[55]
S. Nancarrow, A. Booth, S. Ariss.
Ten principles of good interdisciplinary team work.
Hum Resour Health, 11 (2013), pp. 19
[56]
E.T. Yeh.
Onco-cardiology: the time has come.
Tex Heart Inst J, 38 (2011), pp. 246-247
[57]
S.G. Al-Kindi, G.H. Oliveira.
Prevalence of preexisting cardiovascular disease in patients with different types of cancer: the unmet need for onco-cardiology.
Mayo Clin Proc, (2015),
pii:S0025-6196(15)00738-7
[58]
T.C. Gillebert, N. Brooks, R. Fontes-Carvalho, et al.
ESC core curriculum for the general cardiologist (2013).
Eur Heart J, 34 (2013), pp. 2381-2411
[59]
J.L. Halperin, E.S. Williams, V. Fuster, et al.
ACC 2015 core cardiovascular training statement 4 (COCATS 4) (Revision of COCATS 3).
J Am Coll Cardiol, 65 (2015), pp. 1721-1723
[60]
E. Rashi, F. DePonti.
Cardiovascular toxicity of anticancer-targeted therapy: emerging issues in the era of cardio-oncology.
Intern Emerg Med, 7 (2012), pp. 113-131
[61]
H. Lal, K.L. Kolaja, T. Force.
Cancer genetics and the cardiotoxicity of the therapeutics.
J Am Coll Cardiol, 61 (2013), pp. 267-274

Please cite this article as: Fiuza M, Ribeiro L, Magalhães A, Sousa AR, Menezes MN, Jorge M, et al. Organização e implementação de uma consulta de cardio-oncologia. Rev Port Cardiol. 2016;35:485–494.

Copyright © 2016. Sociedade Portuguesa de Cardiologia
Idiomas
Revista Portuguesa de Cardiologia (English edition)
Article options
Tools
en pt

Are you a health professional able to prescribe or dispense drugs?

Você é um profissional de saúde habilitado a prescrever ou dispensar medicamentos

By checking that you are a health professional, you are stating that you are aware and accept that the Portuguese Journal of Cardiology (RPC) is the Data Controller that processes the personal information of users of its website, with its registered office at Campo Grande, n.º 28, 13.º, 1700-093 Lisbon, telephone 217 970 685 and 217 817 630, fax 217 931 095, and email revista@spc.pt. I declare for all purposes that the information provided herein is accurate and correct.