Publish in this journal
Journal Information
Vol. 70. Issue 6.
Pages 487-495 (June 2017)
Visits
Not available
Vol. 70. Issue 6.
Pages 487-495 (June 2017)
Review article
Full text access
Emerging Cardiac Imaging Modalities for the Early Detection of Cardiotoxicity Due to Anticancer Therapies
Nuevas técnicas de imagen cardiaca en la detección precoz de cardiotoxicidad secundaria a tratamientos oncológicos
Visits
12862
Teresa López-Fernándeza,
Corresponding author
tlfernandez8@gmail.com

Corresponding author: Servicio de Cardiología, Hospital Universitario La Paz. P.° de la Castellana 261, planta 1, 28046 Madrid, Spain.
, Paaladinesh Thavendiranathanb
a Servicio de Cardiología, Unidad de Imagen Cardiaca, Unidad de Cardio-Oncología, Hospital Universitario La Paz, IdiPaz, Madrid, Spain
b Peter Munk Cardiac Center, Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
Related content
Virginia Pubul, Irene Casáns, Santiago Aguadé, Francisco Javier de Haro
This item has received
Article information
Abstract
Full Text
Bibliography
Download PDF
Statistics
Figures (4)
Show moreShow less
Tables (3)
Table 1. Imaging Methods for Assessment of Cardiotoxicity
Table 2. Tips and Tricks for 3D Echocardiography Left Ventricular Ejection Fraction Measurement
Table 3. Tips and Tricks for Myocardial Strain Measurement
Show moreShow less
Abstract

The undeniable advances in the field of oncology have finally led to a decrease in overall cancer-related mortality. However, this population of long-term cancer survivors is now facing a shift toward a substantial increase in cardiovascular morbidity and mortality. Because the development of overt cardiotoxicity can be associated with poor outcomes, preclinical identification of cardiac toxicity is important. This will promote early instauration of treatments to prevent overt heart dysfunction and allow oncologists to continue cancer therapy in an uninterrupted manner. Surveillance strategies for the early detection of cardiac injury include cardiac imaging and biomarkers during treatment. In this review, we outline existing cardiac imaging modalities to detect myocardial changes in patients undergoing cancer treatment and in survivors, and their strengths and limitations.

Keywords:
Cardiotoxicity
Chemotherapy
Heart failure
Echocardiography
Abbreviations:
2DE
3DE
CMR
CT
CTRCD
CVD
GLS
LVEF
Resumen

Los indudables progresos en el campo de la oncología han disminuido la mortalidad secundaria al cáncer. Sin embargo, esta población de larga supervivencia se enfrenta ahora a un aumento de la morbimortalidad cardiovascular. Dado que la aparición de cardiotoxicidad se asocia con mal pronóstico, identificarla en una fase subclínica es importante para promover el inicio precoz de tratamientos cardioprotectores y evitar interrupciones del tratamiento oncológico. Las estrategias de detección precoz de la cardiotoxicidad incluyen el uso de biomarcadores y técnicas de imagen cardiaca. Este artículo revisa las técnicas de imagen disponibles, con sus ventajas y limitaciones, para detectar alteraciones precoces de la función miocárdica de pacientes en tratamiento antitumoral y en supervivientes al cáncer.

Palabras clave:
Cardiotoxicidad
Quimioterapia
Insuficiencia cardiaca
Ecocardiografía
Full Text
INTRODUCTION

Advances in the early detection and treatment of malignancies have resulted in a 20% decline in cancer mortality.1 However, cardiovascular disease (CVD) has become an important competing risk for morbidity and mortality in cancer survivors.2 For example, in breast cancer survivors older than 66 years who survived more than 5 years, CVD exceeds breast cancer as the leading cause of death.2 This heighted risk of CVD is due to a combination of shared risk factors for cancer and CVD, the direct impact of cancer therapy on the cardiovascular system, and the gap in the cardiac care of patients with cancer.3–5 Optimization of preexisting conditions before treatment is important, but is unlikely to be sufficient as a sole strategy to prevent CVD. Particularly for the prevention of cancer therapy-related heart failure (HF), strategies to identify early myocardial injury are needed so that targeted therapy can be instituted to prevent overt HF. Cardiac imaging and serum biomarkers have been demonstrated to be key strategies in identifying early myocardial injury. Serum biomarkers have shown tremendous promise but have some limitations, including the lack of clarity on the best biomarker to use, the timing of measurements, and the thresholds to define abnormality.6 There has been increasing enthusiasm for the use of cardiac imaging to detect cardiac injury as it provides direct assessment of myocardial function.7 In this review, we outline existing cardiac imaging modalities to detect myocardial changes in patients undergoing cancer treatment and in survivors and their strengths and limitations. We also provide some practical suggestions for clinicians involved in the cardiac care of patients with cancer.

3D ECHOCARDIOGRAPHYControversies in the Definition of Cardiotoxicity

Different definitions of cardiotoxicity have been used historically with practical implications for how patients are managed.8 The strongest controversy concerns the definition of cancer therapy-related cardiac dysfunction (CTRCD) both in clinical trials and consensus documents.3,9 In the modern era, overt clinical HF and cardiac death occur in 5% to 6% of treated patients.10,11 Asymptomatic deterioration in left ventricular ejection fraction (LVEF), associated with a higher incidence of symptomatic HF is documented in as many as 20% of patients depending on the cancer treatment.12–14

All definitions of CTRCD are based on a serial decline in LVEF. Unfortunately, there are no universal threshold changes to define CTRCD. The American Society of Echocardiography Consensus document defines CTRCD as a LVEF drop ≥ 10% to a value of < 53%,15 based on new American Society of Echocardiography (ASE) and European Association of Cardiovascular Imaging (EACVI) recommendations for chamber quantification.16 These recommendations have been endorsed by the Canadian Cardiovascular Consensus Statement.17 Recently, the European Society of Cardiology 2016 position paper considers the lower limit of normal LVEF by echocardiography as 50%, in line with the definition commonly used in registries and trials in patients with cancer.18

Independent of normal reference ranges, relative changes between baseline and follow-up LVEF are needed to appropriately identified CTRCD. If LVEF decreases > 10% to a value below the lower limit of normal, angiotensin-converting enzyme inhibitors (or angiotensin receptor blockers) in combination with beta-blockers are recommended (unless contraindicated) to prevent further left ventricular (LV) dysfunction or symptomatic HF.15,17,18 If LVEF decreases > 10%, to a value that does not drop below the lower limit of normal, patients should undergo repeat assessment of LVEF promptly. In addition, these patients should be considered for advance echo imaging monitoring to avoid delays in HF therapy initiation.15,17,18

This article focuses on the ASE definition of CTRCD (lower limit of normal considered as an LVEF < 53%) to improve the early detection and prompt therapy of cardiotoxicity, which are crucial for substantial recovery of cardiac function.19,20 In fact, Wang et al.11 demonstrated that even normal LVEF within 5 points of the lower limits of normal was associated with a near-to-3 fold increase in the rate of cardiac events in patient treated with anthracyclines

We Need Precise LVEF Measurements: Pros and Cons of 3DE-LVEF

In cancer patients, serial evaluation of LVEF must be reliable enough to identify true changes in ventricular function leading to subsequent clinical and therapeutic decisions.21 While the imaging modality for monitoring should be based on local institutional expertise, 2-dimensional echocardiography (2DE) is increasingly used due to its widespread availability and safety. This modality allows for characterization of systolic and diastolic function, pulmonary pressures, valvular function, right ventricular function, and the pericardium. Digital storage of images is advisable to allow visual comparison in doubtful cases. The recommended method for 2DE-LVEF quantification is the modified Simpson's biplane method.16 However, due to various factors (reader experience, geometrical assumptions or suboptimal endocardial border definition), 2DE-LVEF has low sensitivity for the detection of small changes in LV function and has a reported test-re-test variability ranging from 9% to 10.8%, which is higher than the threshold used to define CTRCD.22 Contrast agents and automated contour detection may be used to reduce variability. In fact, in the study by Cannesson et al.,23 automated 2DE-LVEF measurements had lower interobserver variability (3.4%) than the manual biplane method (9.8%).

Three-dimensional echocardiography (3DE) provides a compelling alternative with many advantages similar to cardiac magnetic resonance (CMR) imaging.16 It increases the ability to detect smaller changes in LVEF, with a higher reproducibility than 2DE when compared with CMR.24 Three-dimensional echocardiography volume measurements are independent of LV geometric assumptions or apical foreshortening (Table 1). The reduced observer and test-re-test variability in 3DE is at least partially attributable to the automated endocardial tracing.25 In a recent study, Thavendiranathan et al.26 followed up 56 women undergoing chemotherapy by 2DE and 3DE at 3 monthly intervals for 1 year to determine the technique with the least variability. The use of noncontrast 3DE provides lower temporal variability than 2D-LVEF (5.8 vs 9.8%), which is of the utmost importance in these patients.

Table 1.

Imaging Methods for Assessment of Cardiotoxicity

  Advantages  Limitations 
2DE-LVEF  Low cost
Availability
High temporal resolution
Wealth of published data 
Apex frequently foreshortened
Endocardial dropout
Blind to shape distortions not visualized in the apical 2- and 4-chamber planes
Low detection of subclinical toxicity 
3DE-LVEF  No geometrical assumption
Unaffected by foreshortening
More accurate and reproducible than 2DE-LVEF 
Lower temporal resolution
Fewer published data on normal values
Image quality dependent
Low detection of subclinical toxicity 
GLS  More accurate and reproducible than 2D-LVEF
Ability to identify subclinical toxicity
Reproducible when performed by trained operators 
Availability
Vendor- and software-specific
Influenced by loading conditions
Lack of long-term randomized clinical trials 
CMR  Most accurate method to measure LVEF
Provides assessment of myocardial tissue changes moving the field from depending on functional measures of cardiotoxicity to recognizing the underlying pathological changes 
Lack of availability
Several contraindications
Low temporal resolution
Limited data on its use in cardio-oncology 
Cardiac CT  Noninvasive method to assess CAD
High sensitivity and negative predictive value for CAD 
Radiation exposure
Reduced specificity for obstructive CAD especially in the context of calcified plaque 

2DE, 2-dimensional echocardiography; 3DE, 3-dimensional echocardiography; CAD, coronary artery disease; CMR, cardiac magnetic resonance; CT, computed tomography; GLS, global longitudinal strain; LVEF, left ventricular ejection fraction.

The reproducibility of 3DE may be of particular importance in patients with low normal LVEF. In 114 adult survivors of childhood malignancies treated with chest radiation and/or anthracyclines, 16 (14%) were found to have LVEF < 50% by CMR as the reference standard, but 10 of the 16 were misclassified by 2DE as having preserved LVEF by the biplane method.27 On average, 2DE overestimated LVEF by 5% (mean LVEF 56% in CMR, 55% in 3DE, and 61% in 2DE by biplane) and had wider ranges and limits of agreement. 3DE-measured LVEF was the most sensitive echocardiography parameter to identify a LVEF < 55% with CMR. Based on these results, it was suggested that 2DE-LVEF at the lower limits of normal (range of 50%-59%) warrants particular attention and may require further cardiac evaluation to rule out cardiac dysfunction.28

The ASE, the EACVI, the European Society of Cardiology and the Canadian Cardiovascular Consensus Statement recommend serial imaging with calculated LVEF by the best method available in an echocardiography laboratory.15,17,18 Today 3DE is the preferred technique for the longitudinal monitoring of LVEF in cancer patients.26,29 Fully automated software decreases 3DE-LVEF measurement variability, is timesaving and will help facilitate the integration of 3DE into clinical practice.30 Operator expertise, standardized approaches, and quality improvement initiatives within the echocardiography laboratory are required to achieve the superiority of 3DE-LVEF.16,24 The latter is particularly important given that changes as small as 10% in LVEF are commonly used to define CTRCD and to initiate cardioprotective therapies.15,17,18

Tips and Tricks for Daily Practice

Image acquisition in 3DE is similar to 2DE with a 1 to 2minute acquisition time from the apical position (Table 2).16,25 Two to 3 volume datasets should be obtained for analysis with the best dataset used for analysis. Volume rates should be greater than 20 volumes per second. Analysis is typically performed offline with semi- or fully automated border identification to delineate the endocardium and epicardium. To integrate 3DE measurements into clinical practice, fully automated software is the logical next step (Figure 1).30

Table 2.

Tips and Tricks for 3D Echocardiography Left Ventricular Ejection Fraction Measurement

Optimization of: 
Image quality and volume rate 
Optimize 2D depth, gain, focus, and endocardial border definition 
Optimize 3D sector width and depth to include the entire LV 
Two to 3 volume datasets should be obtained for analysis 
Volume rates should be > 20 volumes/s 
ECG tracking 
Detection of software analysis landmarks 
Segmentation of cardiac cycle 
Semi- or fully-automated border identification 
Ensure that the location of the apex and the mitral annulus is appropriate 
Evaluate tracing quality of the endocardial border 

2D, 2-dimensional; 3D, 3-dimensional; ECG, electrocardiogram; LV, left ventricle.

Figure 1.

Three-dimensional echocardiography (3DE) left ventricular ejection fraction with a fully automated software. A: single-beat full-volume 3DE data sets of the entire left ventricle, performed in the apical 4-chamber view window (B). Users can edit the location of the endocardial border if needed. Final ED (C) and ES frame (D). E: 3D left ventricular volumes and EF. AP2, apical 2-chamber view; AP3, apical 3-chamber view; AP4, apical 4-chamber view; ED, end-diastolic; EF, ejection fraction; ES, end-systolic; HR, heart rate; LA left atrium; LV, left ventricle; SV, stroke volume.

(0.51MB).
MYOCARDIAL STRAINDeformation Imaging vs LVEF

Most efforts to reduce cardiotoxicity are focused on the early diagnosis and treatment of LV dysfunction. However, LVEF is an insensitive measure of early myocardial dysfunction.13 When a patient develops LV dysfunction, particularly with anthracyclines, myocardial damage is established and the chance of recovery, even with evidence-based cardiac therapy, decreases with time.21,31 Whether the same concerns exist with other cancer therapy agents remains to be determined. Emerging data suggest that cardiac biomarkers and new echo techniques may have more sensitivity for the early detection of cardiotoxicity.6,15,17,18

The heart has a helical structure composed of 3 layers of myocardial fibers. Left ventricular systolic function is a coordinated action between them (longitudinal contraction, circumferential shortening, and radial thickening) and ejection fraction predominantly evaluates radial function. Technologies such as speckle-tracking echocardiography (STE) have enhanced the noninvasive assessment of myocardial deformation from conventional 2DE images and provide accurate information in the early phases of myocardial diseases.32 The measurement of deformation is commonly referred to as myocardial strain. Global longitudinal strain (GLS) is the most commonly studied parameter to detect preclinical disease.33 It is highly reproducible when performed by trained operators (inter- and intraobserver variability < 4%),34 but normal ranges are vendor- and software-dependent.35,36 Recognizing the critical need for standardization of strain imaging, the EACVI/ASE invited technical representatives from all interested vendors to participate in a concerted effort to reduce intervendor variability of strain measurement.37 Recently, a study compared head-to-head GLS measurements using 7 different STE software packages, in a group of volunteers with an average LVEF of 60%. Global longitudinal strain values ranged from −18.0% to −21.5%. The inter- and intraobserver reproducibility of GLS proved to be comparable with or superior to that of LVEF (interobserver relative mean errors were 5.4% to 8.6%, while the intraobserver relative mean errors were 4.9% to 7.3%). The absolute difference between vendors for GLS was up to 3.7% strain units (P < .001), lower than for LVEF.38 Although the current recommendation is to use the same vendor for serial surveillance,15,17,18 improvements in the standardization of measures will soon lead to follow-up of patients with different vendor echocardiography machines.39

A growing body of literature supports the use of myocardial strain analysis in patients receiving cancer therapy for baseline evaluation, treatment monitoring, and surveillance of cancer survivors.40

Baseline Cardiotoxicity Risk Assessment

Several studies have shown the usefulness of STE-derived strain, as a predictor of outcome in HF patients.33 In cancer patients, GLS has been demonstrated to be superior to LVEF in cardiotoxicity prediction. Prechemotherapy GLS was independently associated with cardiac events at a median follow-up of 4 years. A GLS absolute value < 17.5% was associated with a 6-fold increase in cardiac death or symptomatic HF.41 Prechemotherapy GLS has also been demonstrated to be an effective tool to stratify cardiotoxicity risk in patients with a baseline LVEF between 50% and 59%.42 Recently, circumferential strain was also demonstrated to be strongly predictive of CTRCD.43 These studies open new lines of investigation to noninvasively identify patients at high risk for symptomatic HF before cancer treatment.

Monitoring During Cancer Treatment

In chemotherapy-treated patients, GLS detects early myocardial dysfunction and predicts CTRCD.40 Although data regarding its ability to predict long-term CTRCD are still lacking, several studies have demonstrated its usefulness in the short-term.44,45 The degree of change in GLS that predicted subsequent cardiotoxicity differed between studies and ranged from 10% to 15%.40 An early study demonstrated that in 45 women with breast cancer treated with trastuzumab/anthracyclines, a relative reduction in GLS of 10% at 3 months predicted subsequent cardiotoxicity (defined as an LVEF < 50% at 6 months).46 In another study of 81 consecutive women prospectively treated with trastuzumab (most also received anthracyclines), 24 (30%) developed cardiotoxicity (defined as a > 10% decline in LVEF from baseline at 12 months). The strongest predictor of cardiotoxicity was a relative decrease in GLS > 11% at 6 months (area under the curve, 0.84; 95% confidence interval, 8.3%-14.6%).47 A combination of biomarkers and GLS increased accuracy for cardiotoxicity diagnosis and if both were negative they identified a group at relatively low risk for CTRCD (negative predictive value of any of these markers for LV dysfunction at follow-up between 91% to 97%).45,47

A relative reduction in GLS of > 15% is the threshold defined by ASE to identify subclinical LV dysfunction, whereas a change of < 8% appears not to be of clinical significance. To avoid a false-positive diagnosis of CTRCD, the abnormal value should be confirmed by a repeat study performed 2 to 3 weeks later15 (Figure 2).

Figure 2.

Cancer treatment monitoring. A 67-year-old woman with HER2+ breast cancer. The patient had a previous history of smoking and mild hypertension. A tumorectomy was performed and she was subsequently treated with anthracycline-based therapy and trastuzumab. At 3 months’ follow-up, an asymptomatic decrease in GLS and 2DE-LVEF was documented. Under enalapril and carvedilol therapy, cardiac dysfunction improves without treatment interruption, but may not fully recover to baseline. 2DE, 2-dimensional echocardiography; ANT, anterior; AP2 L. Strain, apical 2-chamber view longitudinal strain; AP3 L. Strain, apical 3-chamber view longitudinal strain; AP4 L. Strain, apical 4-chamber view longitudinal strain; EDV, end-diastolic volume; EF, ejection fraction; ESV, end-systolic volume; F/U, follow-up; GLS, global longitudinal strain; INF, inferior; LAT, lateral; LVEF, left ventricular ejection fraction; SD, standard deviation; SEPT, septal.

(0.45MB).

There is no solid evidence on the most appropriate clinical management when an isolated fall in strain is the only abnormality. Preliminary data support the use of beta-blockers in preventing CTRCD in cancer patients experiencing a significant drop in GLS during treatment.48 However, this will need to be proven prospectively. If a strain-based approach is shown to affect clinical outcomes, this would change the way patients receiving potentially cardiotoxicity cancer therapy are followed up in the future, given that this approach has been shown to be cost-effective.49

Assessment in Survivors

Cardiac follow-up should be undertaken in cancer survivors who have received heart radiation or chemotherapy to enable the detection of cardiovascular toxicities, ideally in the asymptomatic phase of the disease.50 In survivors of childhood cancer, the most common abnormality found in echo follow-up was a significant reduction in the GLS with preserved LVEF (28% of patients with abnormal GLS and LVEF > 50%).5 In the HF population, GLS improves risk stratification especially in patients with LVEF in the low normal or mild depressed ranges.33 Further studies are needed to determine whether this concept applies to cancer survivors and whether a strain-based intervention alters long-term clinical outcomes.

Tips and Tricks for Daily Practice

Global longitudinal strain is a sensitive and robust measure to detect subclinical myocardial dysfunction; however, there are no standard guidelines for measuring it adequately, which could contribute to interobserver variability. Experience and training affect the precision and validity of GLS measurement.51 Recently Negishi et al.52 published a set of instructions to help improve reader uniformity in an international multicenter trial of the incremental value of myocardial strain for the detection and management of cardiotoxicity (SUCCOUR ACTRN12614000341628). Table 3 summarizes the steps for STE image acquisition and tips and tricks for daily practice. As the range of normality for different techniques is not interchangeable, the same method should be used to assess GLS during follow-up. Digital storage of images is advisable to allow visual comparison in doubtful cases.

Table 3.

Tips and Tricks for Myocardial Strain Measurement

Optimization of: 
2D image quality 
Frame rate > 50 Hz 
Gain 
Focus 
Endocardial border definition for the entire cardiac cycle 
ECG and endocardial border tracking 
Detection of software analysis landmarks 
Aortic valve closure: segmentation of cardiac cycle to ensure measurement of peak systolic strain 
Adjustment of ROI 
Appropriate location of the apex 
Avoid the inclusion of the papillary muscle 
Location of the mitral annulus requires careful attention to avoid an apparent reduction of regional strain related to atrial tissue analysis 
The ROI should not include the left ventricular outflow tract 
Adequate ROI thickness includes the myocardium but not the pericardium 
ROI width is too thin: overestimate GLS 
ROI is too wide: deformation is reduced 

2D, 2-dimensional; ECG, electrocardiogram; GLS, global longitudinal strain; ROI, region of interest.

CARDIAC MAGNETIC RESONANCE IMAGING

Applications of CMR in cardio-oncology include the accurate measurement of LVEF and myocardial tissue characterization.

Cardiac Magnetic Resonance Measured LVEF

Cardiac magnetic resonance is considered the reference standard for the measurement of ventricular volumes and ejection fraction.53 This is due to its accuracy and superior interobserver, intraobserver, and test-re-test variability.54 Routine use of Cardiac Magnetic Resonance in cardio-oncology is not feasible outside certain selected centers due to the lack of widespread accessibility. However, if available, it would provide a more reliable assessment of small changes in left and right ventricular volumes and function.55,56 Detection of these small changes may represent early markers of myocardial injury. However, this theory has not been prospectively tested. The current practical application of CMR-measured ventricular function in cardio-oncology include: a) measurement of LVEF when there is either a discrepancy between LVEF measurement and clinical symptoms or when there is disagreement between other imaging modalities, and b) identification of subclinical cardiomyopathy in cancer survivors.27

Cardiac Magnetic Resonance Tissue Characterization

In addition to accurate LVEF assessment, CMR has the unique ability to detect and quantify pathological myocardial changes noninvasively, making it an invaluable tool in the diagnosis of cardiomyopathies.57–59 Cardiac magnetic resonance techniques for myocardial tissue characterization include early gadolinium enhancement, T2 and T1 weighted imaging or mapping, and late gadolinium enhancement imaging.60 The early gadolinium enhancement technique allows assessment of myocardial inflammation based on the fact that inflammation is associated with myocardial hyperemia and capillary leak.58,61 The increased blood volume in the inflamed areas leads to higher concentration of gadolinium based contrast agents (GBCA) during the early vascular phase. This can be detected by measuring the signal intensity ratio on pre- and early postcontrast T1 weighted fast spin echo images.58,62 Another consequence of myocardial injury is an increase in the permeability of cell membranes followed by a loss of cell membrane integrity.62 This results in intracellular edema initially followed by interstitial edema. These cellular changes can be identified by qualitative T2 weighted or quantitative T2 mapping sequences (Figure 3) with some growing interest in using precontrast T1 mapping sequences as well.57,63–66 An increased T2 tissue signal, or quantitative T2 or T1 values are felt to represent myocardial edema.57,63–70 More recently, a combined use of pre- and postcontrast T1 mapping has been used to calculate the myocardial extracellular volume (ECV) fraction (Figure 3).71 This is based on the fact that with an expansion of the extracellular space due to fibrosis or interstitial edema, the accumulation of extracellular GBCA would be larger, affecting myocardial T1 values.72,73 Considering respective pre-GBCA T1 values, corresponding T1 values of the blood pool and the patient's hematocrit, the ECV fraction can be calculated. Although primarily described to assess diffuse interstitial myocardial fibrosis, myocardial interstitial edema can also increase ECV.72 Finally, late gadolinium enhancement imaging can be used for the identification of replacement fibrosis (scar).74 Gadolinium based contrast agent will accumulate into the expanded extracellular space (due to scar) and reduce the T1 values in this area. This amount of scar can then be identified by T1-weighted inversion recovery.75

Figure 3.

Tissue characterization techniques with cardiac magnetic resonance. A and B: short axis T2 maps obtained before ecancer therapy and after anthracycline treatment demonstrating a regional increase in T2 values suggesting myocardial edema. C: extracellular volume map showing marginally elevated extracellular volume in a patient after cancer therapy.

(0.4MB).

A recent systematic review has outlined the limited literature on the use of CMR tissue characterization in cardio-oncology.76 Existing animal model studies demonstrate that edema detected using T1 and T2 weighted imaging can identify the earliest signs of cardiotoxicity.77,78 A clinical study of 46 women with breast cancer treated with anthracyclines+/-trastuzumab, demonstrated myocardial edema using T2 weighted imaging in 49% of the patients at 1 to 4 months during therapy. Patients with edema were more likely to have persistent reduction in right ventricular function at follow-up.57 Limited data also suggest that with anthracyclines an early increase in early gadolinium enhancement identifies patients who subsequently have a reduction in LVEF. Several studies have also examined myocardial tissue changes in cancer survivors and have demonstrated an increase in myocardial ECV, suggesting diffuse fibrosis. In pediatric cancer survivors the increase in ECV was associated with poor exercise tolerance.79,80 Despite these interesting data, the clinical relevance of these myocardial tissue changes during cancer therapy and in survivors is undefined.

Tips and Tricks for Daily Practice

Cardiac Magnetic Resonance is the modality of choice for accurate measurement of LV volumes and function. Its routine application in clinical practice to follow up patients receiving cancer therapy is not practical at present. Its current clinical role is when there is discrepancy in LV function between 2 different modalities, and decisions regarding continuing cancer therapy is contingent on accurate LVEF determine. Myocardial tissue characterization is a potential unique application of CMR particularly for understanding the pathophysiological myocardial changes that underpin the functional changes seen during and after cancer therapy. Several ongoing studies are currently exploring the clinical value of myocardial tissue characterization in patients receiving cancer therapy

CARDIAC COMPUTED TOMOGRAPHY

There are limited data on the use of cardiac computed tomography (CT) in the assessment and management of patients receiving cardiotoxic cancer therapy.81 Although functional assessment can be performed with cardiac CT, it is limited by significantly lower temporal resolution and would not be considered a primary modality for this purpose. Its primary use includes the detection or exclusion of coronary artery disease and pericardial disease. Cardiac CT may have a role in pretreatment risk assessment via the demonstration of coronary calcification to identify subclinical coronary disease in patients with established risk factors. In fact, in the Multiethnic Study of Atherosclerosis (MESA), there was an increased prevalence of coronary atherosclerosis in patients with a new diagnosis of cancer.82 Identification of such risk factors can promote the use of evidence-based therapies, such as statins prior to initiation of cardiotoxic cancer therapy.

Certain chemotherapeutics, such as antimetabolites, antimicrotuble agents, and tyrosine kinase inhibitors, have been associated with the development of coronary disease and ischemia.83 In addition, radiation therapy to the chest is associated with the development of coronary artery disease.84 Cardiac CT has excellent diagnostic performance in the detection of coronary disease and is well known for its high negative predictive value (Figure 4). In fact, several studies have suggested that the CT coronary angiogram may be an ideal technique for the early detection of radiation-induced coronary artery disease in pediatric cancer survivors.85,86

Figure 4.

Coronary computed tomography angiography. A: a mixed plaque (calcified and noncalcified) in the proximal left anterior descending coronary artery. B and C: the right coronary artery and the circumflex artery (CX), respectively, with no any atherosclerosis.

(0.25MB).

Certain chemotherapies, as well as radiation therapy, are associated with pericardial disease. Cardiac CT provides exquisite assessment of pericardial effusion, thickening, and pericardial calcification. When pericardial disease is suspected, after physiological assessment using echocardiography or CMR, cardiac CT is an excellent adjunct modality. The use of cardiac CT in the assessment of pericardial disease in general has been previously reviewed.87

Tips and Tricks for Daily Practice

Cardiac CT does not have a routine role in cardio-oncology, but its potential use is in the assessment of subclinical or clinical coronary artery disease and in the investigation of potential pericardial disease.

CONCLUSIONS

Patients, who undergo cancer treatments, should be considered to be at high risk of cardiovascular complications; the most frequent of which is cardiac dysfunction and HF. Conclusive data regarding the optimal surveillance scheme are lacking and no evidence-based recommendations can be firmly established. Because resources are limited, each center should design its own surveillance algorithm depending on the availability and expertise of each technique. In experienced laboratories, the use of 3DE-LVEF and GLS should be emphasized for serial surveillance during cancer treatment. 3DE is more accurate and reproducible than 2DE for the measurement of LVEF and has the best temporal reproducibility during cancer therapy. The use of GLS to identify subclinical myocardial injury should be implemented particularly in high risk patients. Routine use of CMR to follow up patients receiving cancer therapy is not practical at present. It effective use is when there are discrepancies between different modalities and cancer treatment needs to be withheld. The potential use of cardiac CT is in the assessment of subclinical or clinical coronary artery disease and in the investigation of potential pericardial disease. Depending on the modality and frequency of the surveillance strategy feasible at each center, local cardio-oncology teams should reach a consensus on how to manage patients with suspected CTRCD or subclinical myocardial dysfunction, in order to standardize decisions such as when to start cardioprotective drugs or withhold life-saving cancer therapy.

CONFLICTS OF INTEREST

None declared.

References
[1]
K. Miller, R.L. Siegel, C.C. Lin, et al.
Cancer Treatment and Survivorship Statistics, 2016.
CA Cancer J Clin., 66 (2016), pp. 271-289
[2]
H. Abdel-Qadir, P.C. Austin, D.S. Lee, et al.
A population-based study of cardiovascular mortality following early-stage breast cancer.
JAMA Cardiol, 2 (2017), pp. 88-93
[3]
G. Curigliano, D. Cardinale, S. Dent, C. Criscitiello, O. Aseyev, D. Lenihan.
Cardiotoxicity of anticancer treatments: Epidemiology, detection, and management.
CA Cancer J Clin., 66 (2016), pp. 309-325
[4]
N. Haddy, S. Diallo, C. l-Fayech, et al.
Cardiac diseases following childhood cancer treatment cohort study.
Circulation., 133 (2016), pp. 31-38
[5]
G.T. Armstrong, V.M. Joshi, K.K. Ness, et al.
Comprehensive echocardiographic detection of treatment related cardiac dysfunction in adult survivors of childhood cancer: Results from the St. Jude lifetime cohort study.
J Am Coll Cardiol., 65 (2015), pp. 2511-2522
[6]
C. Henri, T. Heinonen, J.C. Tardif.
The role of biomarkers in decreasing risk of cardiac toxicity after cancer therapy.
Biomark Cancer., 8 (2016), pp. 39-45
[7]
T. López-Fernández, D. Saura, J.F. Rodríguez-Palomares, S. Aguadé-Bruix, L. Pérez de Isla, J. Barba-Cosials.
Cardiac imaging 2015: A selection of topical issues.
Rev Esp Cardiol., 69 (2016), pp. 286-293
[8]
G. Curigliano, D. Cardinale, T. Suter, et al.
Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines.
Annals of Oncology., 23 Suppl 7 (2012), pp. vii155-vii166
[9]
J. Herrmann, A. Lerman, N.P. Sandhu, H.R. Villarraga, S.L. Mulvagh, M. Kohli.
Evaluation and management of patients with heart disease and cancer: Cardio-oncology.
Mayo Clin Proc., 89 (2014), pp. 1287-1306
[10]
P. Thavendiranathan, H. Abdel-Qadir, H.D. Fischer, X. Camacho, E. Amir, P.C. Austin.
Breast cancer therapy-related cardiac dysfunction in adult women treated in routine clinical practice: A population-based cohort study.
J Clin Oncol., 34 (2016), pp. 2239-2246
[11]
L. Wang, T.C. Tan, E.F. Halpern, et al.
Major cardiac events and the value of echocardiographic evaluation in patients receiving anthracycline-based chemotherapy.
Am J Cardiol., 116 (2015), pp. 442-446
[12]
J.B. Echouffo-Tcheugui, S. Ergou, J. Butler, C.W. Yancy, G.C. Fonarow.
Assessing the risk of progression from asymptomatic left ventricular dysfunction to overt heart failure: Asystematic overview and meta-analysis.
JACC Heart Fail., 4 (2016), pp. 237-248
[13]
D. Cardinale, A. Colombo, G. Bacchiani, et al.
Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy.
Circulation., 131 (2015), pp. 1981-1988
[14]
P.S. Hall, L.C. Harshman, S. Srinivas, R.M. Witteles.
The frequency and severity of cardiovascular toxicity from targeted therapy in advanced renal cell carcinoma patients.
JACC Heart Fail., 1 (2013), pp. 72-78
[15]
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 ASE and the EACVI.
J Am Soc Echocardiogr., 27 (2014), pp. 911-939
[16]
R.M. Lang, L.P. Badano, V. Mor-Avi, et al.
Recommendations for cardiac chamber quantification by echocardiography in adults: An update from the ASE and the EACVI.
J Am Soc Echocardiogr., 28 (2015), pp. 1-39
[17]
S.A. Virani, S. Dent, C. Brezden-Masley, et al.
Canadian Cardiovascular Society Guidelines for evaluation and management of cardiovascular complications of cancer therapy.
Can J Cardiol., 32 (2016), pp. 831-841
[18]
J.L. Zamorano, P. Lancellotti, D. Rodriguez Muñoz, et al.
2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines.
Eur Heart J., 37 (2016), pp. 2768-2801
[19]
P. Ponikowski, A.A. Voors, S.D. Anker, et al.
2016 ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure.
Eur Heart J., 37 (2016), pp. 2129-2200
[20]
M. Piepoli, W. Hoes, S. Agewall, et al.
2016 European Guidelines on cardiovascular disease prevention in clinical practice.
Eur Heart J., 37 (2016), pp. 2315-2381
[21]
D. Cardinale, A. Colombo, G. Lamantia, et al.
Anthracycline-induced cardiomyopathy: Clinical relevance and response to pharmacologic therapy.
J Am Coll Cardiol., 55 (2010), pp. 213-220
[22]
J.E. Otterstad, G. Froeland, M. St John Sutton, I. Holme.
Accuracy and reproducibility of biplane two-dimensional echocardiographic measurements of left ventricular dimensions and function.
Eur Heart J., 18 (1997), pp. 507-513
[23]
M. Cannesson, M. Tanabe, M.S. Suffoletto, et al.
A novel two-dimensional echocardiographic image analysis system using artificial intelligence-learned pattern recognition for rapid automated ejection fraction.
J Am Coll Cardiol., 49 (2007), pp. 217-226
[24]
C. Jenkins, S. Moir, J. Chan, D. Rakhit, B. Haluska, T.H. Marwick.
Left ventricular volume measurement with echocardiography: A comparison of left ventricular opacification, three-dimensional echocardiography, or both with magnetic resonance imaging.
Eur Heart J., 30 (2009), pp. 98-106
[25]
R.M. Lang, L.P. Badano, W. Tsang, et al.
EAE/ASE recommendations for image acquisition and display using three-dimensional echocardiography.
Eur Heart J Cardiovasc Imaging., 13 (2012), pp. 1-46
[26]
P. Thavendiranathan, A.D. Grant, T. Negishi, J.C. Plana, Z.B. Popovic, T.H. Marwick.
Reproducibility of echocardiographic techniques for sequential assessment of left ventricular ejection fraction and volumes: Application to patients undergoing cancer chemotherapy.
J Am Coll Cardiol., 61 (2013), pp. 77-84
[27]
G.T. Armstrong, J.C. Plana, N. Zhang, et al.
Screening adult survivors of childhood cancer for cardiomyopathy: Comparison of echocardiography and cardiac magnetic resonance imaging.
J Clin Oncol., 30 (2012), pp. 2876-2884
[28]
O.H. Toro-Salazar, J. Ferranti, R. Lorenzoni, et al.
Feasibility of echocardiographic techniques to detect subclinical cancer therapeutics-related cardiac dysfunction among high-dose patients when compared with cardiac magnetic resonance imaging.
J Am Soc Echocardiogr., 29 (2016), pp. 119-131
[29]
J. Walker, N. Bhullar, N. Fallah-Rad, et al.
Role of three-dimensional echocardiography in breast cancer: Comparison with two-dimensional echocardiography, multiple-gated acquisition scans, and cardiac magnetic resonance imaging.
J Clin Oncol., 28 (2010), pp. 3429-3436
[30]
W. Tsang, I.S. Salgo, D. Medvedofsky, et al.
Transthoracic 3D echocardiographic left heart chamber quantification using an automated adaptive analytics algorithm.
J Am Coll Cardiol Imaging., 9 (2016), pp. 769-782
[31]
G.H. Oliveira, S. Mukerji, A.V. Hernandez, et al.
Incidence, predictors, and impact on survival of left ventricular systolic dysfunction and recovery in advanced cancer patients.
Am J Cardiol., 113 (2014), pp. 1893-1898
[32]
V. Mor-Avi, R.M. Lang, L.P. Badano, et al.
Current and evolving echocardiographic techniques for the quantitative evaluation of cardiac mechanics: ASE/EAE consensus statement on methodology and indications endorsed by the japanese society of echocardiography.
J Am Soc Echocardiogr., 24 (2011), pp. 277-313
[33]
M. Cikes, S.D. Solomon.
Beyond ejection fraction: An integrative approach for assessment of cardiac structure and function in heart failure.
Eur Heart J., 37 (2016), pp. 1642-1650
[34]
S. Cheng, M.G. Larson, E.L. McCabe, et al.
Reproducibility of speckle-tracking-based strain measures of left ventricular function in a community-based study.
J Am Soc Echocardiogr., 26 (2013), pp. 1258-1266.e2
[35]
T. Yingchoncharoen, S. Agarwal, Z.B. Popović, T.H. Marwick.
Normal ranges of left ventricular strain: A meta-analysis.
J Am Soc Echocardiogr., 26 (2013), pp. 185-191
[36]
K. Takigiku, M. Takeuchi, C. Izumi, et al.
Normal range of left ventricular 2-dimensional strain: Japanese ultrasound speckle tracking of the left ventricle (JUSTICE) study.
Circ J., 76 (2012), pp. 2623-2632
[37]
J. Voigt, G. Pedrizzetti, P. Lysyansky, et al.
Definitions for a common standard for 2D speckle tracking echocardiography: Consensus document of the EACVI/ASE/Industry Task Force to standardize deformation imaging.
Eur Heart J Cardiovascular Imaging., 16 (2015), pp. 1-11
[38]
K.E. Farsalinos, A.M. Daraban, S. Unlü, J.D. Thomas, L.P. Badano, J.U. Voigt.
Head-to-head comparison of global longitudinal strain measurements among nine different vendors. The EACVI/ASE inter-vendor comparison study.
J Am Soc Echocardiogr., 28 (2015), pp. 1171-1181
[39]
H. Yang, T.H. Marwick, N. Fukuda, et al.
Improvement in strain concordance between two major vendors after the strain standardization initiative.
J Am Soc Echocardiogr., 28 (2015), pp. 642-648
[40]
P. Thavendiranathan, F. Poulin, K.D. Lim, J.C. Plana, A. Woo, T.H. Marwick.
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., 63 (2014), pp. 2751-2768
[41]
M.T. Ali, E. Yucel, S. Bouras, et al.
Myocardial strain is associated with adverse clinical cardiac events in patients treated with anthracyclines.
J Am Soc Echocardiogr., 29 (2016), pp. 522-527
[42]
M. Negareh, T.C. Tan, M. Ali, E.F. Halpern, L. Wang, M. Scherrer-Crosbie.
Echocardiographic parameters of left ventricular size and function as predictors of symptomatic heart failure in patients with a left ventricular ejection fraction of 50-59% treated with anthracyclines.
Eur Heart J Cardiovasc Imaging., 16 (2015), pp. 977-984
[43]
H.K. Narayan, B. French, A.M. Khan, et al.
Noninvasive measures of ventricular-arterial coupling and circumferential strain predict cancer therapeutics-related cardiac dysfunction.
JACC Cardiovasc Imaging., 9 (2016), pp. 1131-1141
[44]
I.B. Rhea, S. Uppuluri, S. Sawada, B.P. Schneider, H. Feigenbaum.
Incremental prognostic value of echocardiographic strain and its association with mortality in cancer patients.
J Am Soc Echocardiogr., 28 (2015), pp. 667-673
[45]
H. Sawaya, I.A. Sebag, J.C. Plana, et al.
Assessment of echocardiography and biomarkers for the extended prediction of cardiotoxicity in patients treated with anthracyclines, taxanes, and trastuzumab.
Circ Cardiovasc Imaging., 5 (2012), pp. 596-603
[46]
H. Sawaya, I.A. Sebag, J.C. Plana, et al.
Early detection and prediction of cardiotoxicity in chemotherapy-treated patients.
Am J Cardiol., 107 (2011), pp. 1375-1380
[47]
K. Negishi, T. Negishi, J.L. Hare, B.A. Haluska, J.C. Plana, T.H. Marwick.
Independent and incremental value of deformation indices for prediction of trastuzumab-induced cardiotoxicity.
J Am Soc Echocardiogr., 26 (2013), pp. 493-498
[48]
K. Negishi, T. Negishi, B.A. Haluska, J.L. Hare, J.C. Plana, T.H. Marwick.
Use of speckle strain to assess left ventricular responses to cardiotoxic chemotherapy and cardioprotection.
Eur Heart J Cardiovasc Imaging., 15 (2014), pp. 324-331
[49]
M.T. Nolan, J.C. Plana, P. Thavendiranathan, L. Shaw, L. Si, T.H. Marwick.
Cost-effectiveness of strain-targeted cardioprotection for prevention of chemotherapy-induced cardiotoxicity.
Int J Cardiol., 212 (2016), pp. 336-345
[50]
D.A. Mulrooney, G.T. Armstrong, S. Huang, et al.
Cardiac outcomes in adult survivors of childhood cancer exposed to cardiotoxic therapy: A cross-sectional study from the St. Jude lifetime cohort.
Ann Intern Med., 164 (2016), pp. 93-101
[51]
T. Negishi, K. Negishi, P. Thavendiranathan, G.Y. Cho, B.A. Popescu, D. Vinereanu.
Effect of experience and training on the concordance and precision of strain measurements.
JACC Cardiovasc Imaging., (2016),
pii: S1936-878X(16)306209
[52]
N. Negishi, T. Negishi, K. Kurosawa, et al.
Practical guidance in echocardiographic assessment of global longitudinal strain.
JACC Imaging., 8 (2015), pp. 489-492
[53]
S. Valbuena-López, R. Hinojar, V.O. Puntmann.
Cardiovascular magnetic resonance in cardiology practice: A concise guide to image acquisition and clinical interpretation.
Rev Esp Cardiol., 69 (2016), pp. 202-210
[54]
K. Ylanen, T. Poutanen, P. Savikurki-Heikkila, I. Rinta-Kiikka, A. Eerola, K. Vettenranta.
Cardiac magnetic resonance imaging in the evaluation of the late effects of anthracyclines among long-term survivors of childhood cancer.
J Am Coll Cardiol., 61 (2013), pp. 1539-1547
[55]
B.C. Drafts, K.M. Twomley, R. D’Agostino, et al.
Low to moderate dose anthracycline-based chemotherapy is associated with early noninvasive imaging evidence of subclinical cardiovascular disease.
JACC Cardiovasc Imaging., 6 (2013), pp. 877-885
[56]
S. Grover, D.P. Leong, A. Chakrabarty, et al.
Left and right ventricular effects of anthracycline and trastuzumab chemotherapy: A prospective study using novel cardiac imaging and biochemical markers.
Int J Cardiol., 168 (2013), pp. 5465-5467
[57]
P. Thavendiranathan, M. Walls, S. Giri, et al.
Improved detection of myocardial involvement in acute inflammatory cardiomyopathies using T2 mapping.
Circ Cardiovasc Imaging., 5 (2012), pp. 102-110
[58]
M.G. Friedrich, U. Sechtem, J. Schulz-Menger, et al.
International Consensus Group on Cardiovascular Magnetic Resonance in myocarditis: A JACC white paper.
J Am Coll Cardiol., 53 (2009), pp. 1475-1487
[59]
K.J. Giesbrandt, C.W. Bolan, B.P. Shapiro, W.D. Edwards, P.J. Mergo.
Diffuse diseases of the myocardium: MRI-pathologic review of nondilated cardiomyopathies.
AJR Am J Roentgenol., 200 (2013), pp. W266-W273
[60]
J. Sanz, G. LaRocca, J.G. Mirelis.
Myocardial mapping with cardiac magnetic resonance: The diagnostic value of novel sequences.
Rev Esp Cardiol., 69 (2016), pp. 849-861
[61]
H. Paajanen, R.C. Brasch, U. Schmiedl, M. Ogan.
Magnetic resonance imaging of local soft tissue inflammation using gadolinium-DTPA.
Acta Radiol., 28 (1987), pp. 79-83
[62]
X. Gao, Y. Luo, Y. Wang, et al.
Prostate stem cell antigen-targeted nanoparticles with dual functional properties: In vivo imaging and cancer chemotherapy.
Int J Nanomedicine., 7 (2012), pp. 4037-4051
[63]
S. Giri, S. Shah, H. Xue, et al.
Myocardial T2 mapping with respiratory navigator and automatic nonrigid motion correction.
Magn Reson Med., 68 (2012), pp. 1570-1578
[64]
S. Giri, Y.C. Chung, A. Merchant, et al.
T2 quantification for improved detection of myocardial edema.
J Cardiovasc Magn Reson., 11 (2009), pp. 56
[65]
D. Verhaert, P. Thavendiranathan, S. Giri, et al.
Direct T2 quantification of myocardial edema in acute ischemic injury.
JACC Cardiovasc Imaging., 4 (2011), pp. 269-278
[66]
A.A. Usman, K. Taimen, M. Wasielewski, et al.
Cardiac magnetic resonance T2 mapping in the monitoring and follow-up of acute cardiac transplant rejection: A pilot study.
Circ Cardiovasc Imaging., 5 (2012), pp. 782-790
[67]
V.M. Ferreira, S.K. Piechnik, E. Dall’Armellina, et al.
Non-contrast T1-mapping detects acute myocardial edema with high diagnostic accuracy: A comparison to T2-weighted cardiovascular magnetic resonance.
J Cardiovasc Magn Reson., 14 (2012), pp. 42
[68]
C.B. Higgins, R. Herfkens, M.J. Lipton, et al.
Nuclear magnetic resonance imaging of acute myocardial infarction in dogs: Alterations in magnetic relaxation times.
Am J Cardiol., 52 (1983), pp. 184-188
[69]
M.I. Zia, N.R. Ghugre, K.A. Connelly, et al.
Thrombus aspiration during primary percutaneous coronary intervention is associated with reduced myocardial edema, hemorrhage, microvascular obstruction and left ventricular remodeling.
J Cardiovasc Magn Reson., 14 (2012), pp. 19
[70]
M.I. Zia, N.R. Ghugre, K.A. Connelly, et al.
Characterizing myocardial edema and hemorrhage using quantitative T2 and T2* mapping at multiple time intervals post ST-segment elevation myocardial infarction.
Circ Cardiovasc Imaging., 5 (2012), pp. 566-572
[71]
H. Arheden, M. Saeed, C.B. Higgins, et al.
Measurement of the distribution volume of gadopentetate dimeglumine at echo-planar MR imaging to quantify myocardial infarction: Comparison with 99mTc-DTPA autoradiography in rats.
[72]
P. Kellman, J.R. Wilson, H. Xue, et al.
Extracellular volume fraction mapping in the myocardium, part 2: Initial clinical experience.
J Cardiovasc Magn Reson., 14 (2012), pp. 64
[73]
M. Ugander, P.S. Bagi, O.J. Booker, et al.
Edema by T2-weighted imaging in salvaged myocardium is extraceullular, not intracellular.
J Cardiovasc Magn Reson, 13 Suppl 1 (2011), pp. P70
[74]
R.J. Kim, E. Wu, A. Rafael, et al.
The use of contrast-enhanced magnetic resonance imaging to identify reversible myocardial dysfunction.
N Engl J Med., 343 (2000), pp. 1445-1453
[75]
J. Vogel-Claussen, C.E. Rochitte, K.C. Wu, et al.
Delayed enhancement mr imaging: Utility in myocardial assessment.
Radiographics., 26 (2006), pp. 795-810
[76]
P. Thavendiranathan, B.J. Wintersperger, S.D. Flamm, T.H. Marwick.
Cardiac MRI in the assessment of cardiac injury and toxicity from cancer chemotherapy: A systematic review.
Circ Cardiovasc Imaging., 6 (2013), pp. 1080-1091
[77]
Y. Cottin, C. Ribuot, V. Maupoil, et al.
Early incidence of adriamycin treatment on cardiac parameters in the rat.
Can J Physiol Pharmacol., 72 (1994), pp. 140-145
[78]
J.C. Lightfoot, R.B. D’Agostino Jr., C.A. Hamilton, et al.
Novel approach to early detection of doxorubicin cardiotoxicity by gadolinium-enhanced cardiovascular magnetic resonance imaging in an experimental model.
Circ Cardiovasc Imaging., 3 (2010), pp. 550-558
[79]
E.B. Tham, M.J. Haykowsky, K. Chow, et al.
Diffuse myocardial fibrosis by T1-mapping in children with subclinical anthracycline cardiotoxicity: Relationship to exercise capacity, cumulative dose and remodeling.
J Cardiovasc Magn Reson., 15 (2013), pp. 48
[80]
J.H. Jordan, S. Vasu, T.M. Morgan, et al.
Anthracycline-associated T1 mapping characteristics are elevated independent of the presence of cardiovascular comorbidities in cancer survivors.
Circ Cardiovasc Imaging., (2016),
http://dx.doi.org/10.1161/CIRCIMAGING.115.004325
[81]
J.D. Groarke, P.L. Nguyen, A. Nohria, R. Ferrari, S. Cheng, J. Moslehi.
Cardiovascular complications of radiation therapy for thoracic malignancies: The role for non-invasive imaging for detection of cardiovascular disease.
Eur Heart J., 35 (2014), pp. 612-623
[82]
M.C. Whitlock, J. Yeboah, G.L. Burke, H. Chen, H.D. Klepin, W.G. Hundley.
Cancer and its association with the development of coronary artery calcification: An assessment from the multi-ethnic study of atherosclerosis.
J Am Heart Assoc., (2015),
http://dx.doi.org/10.1161/JAHA.115.002533
[83]
E.T. Yeh, H.M. Chang.
Oncocardiology-past, present, and future: A review.
JAMA Cardiol., 1 (2016), pp. 1066-1072
[84]
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
[85]
L.A. Daniels, A.D. Krol, M.A. De Graaf, et al.
Screening for coronary artery disease after mediastinal irradiation in Hodgkin lymphoma survivors: Phase II study of indication and acceptance.
Ann Oncol., 25 (2014), pp. 1198-1203
[86]
S. Kupeli, T. Hazirolan, A. Varan, et al.
Evaluation of coronary artery disease by computed tomography angiography in patients treated for childhood Hodgkin's lymphoma.
J Clin Oncol., 28 (2010), pp. 1025-1030
[87]
D. Verhaert, R.S. Gabriel, D. Johnston, B.W. Lytle, M.Y. Desai, A.L. Klein.
The role of multimodality imaging in the management of pericardial disease.
Circ Cardiovasc Imaging., 3 (2010), pp. 333-343
Copyright © 2017. Sociedad Española de Cardiología
Idiomas
Revista Española de Cardiología (English Edition)

Subscribe to our newsletter

View newsletter history
Article options
Tools