Next Article in Journal
Clinical Outcome of Discordant Empirical Therapy and Risk Factors Associated to Treatment Failure in Children Hospitalized for Urinary Tract Infections
Previous Article in Journal
Ultra-Processed Foods Are the Major Sources of Total Fat, Saturated and Trans-Fatty Acids among Tunisian Preschool and School Children: A Cross-Sectional Study
Previous Article in Special Issue
Pediatric Cardio-Oncology Medicine: A New Approach in Cardiovascular Care
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current State of Pediatric Cardio-Oncology: A Review

1
Children’s Wisconsin, Milwaukee, WI 53226, USA
2
Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
*
Author to whom correspondence should be addressed.
Children 2022, 9(2), 127; https://doi.org/10.3390/children9020127
Submission received: 11 October 2021 / Revised: 23 November 2021 / Accepted: 30 November 2021 / Published: 19 January 2022
(This article belongs to the Special Issue Cardio-Oncology in Children)

Abstract

:
The landscape of pediatric oncology has dramatically changed over the course of the past several decades with five-year survival rates surpassing 80%. Anthracycline therapy has been the cornerstone of many chemotherapy regimens for pediatric patients since its introduction in the 1960s, and recent improved survival has been in large part due to advancements in chemotherapy, refinement of supportive care treatments, and development of novel therapeutics such as small molecule inhibitors, chimeric antigen receptor T-cell therapy, and immune checkpoint inhibitors. Unfortunately, many cancer-targeted therapies can lead to acute and chronic cardiovascular pathologies. The range of cardiotoxicity can vary but includes symptomatic or asymptotic heart failure, arrhythmias, coronary artery disease, valvar disease, pericardial disease, hypertension, and peripheral vascular disease. There is lack of data guiding primary prevention and treatment strategies in the pediatric population, which leads to substantial practice variability. Several important future research directions have been identified, including as they relate to cardiac disease, prevention strategies, management of cardiovascular risk factors, risk prediction, early detection, and the role of genetic susceptibility in development of cardiotoxicity. Continued collaborative research will be key in advancing the field. The ideal model for pediatric cardio-oncology is a proactive partnership between pediatric cardiologists and oncologists in order to better understand, treat, and ideally prevent cardiac disease in pediatric oncology patients.

1. Introduction

The landscape of pediatric oncology has dramatically changed over the course of the past several decades, with five-year survival rates surpassing 80%. Improved survival has been in large part due to advancements in chemotherapy, refinement of supportive care treatments, and development of novel therapeutics, such as chimeric antigen receptor T-cell therapy (CAR-T) and immune checkpoint inhibitors (ICI) [1,2,3]. However, with improved survival rates, a five- to six-fold increase in cardiovascular disease risk has been observed, and cardiovascular disease is now the leading non-cancer cause of death. The range of cardiotoxicity can vary, but includes symptomatic or asymptotic heart failure, arrhythmias, coronary artery disease, valvar disease, pericardial disease, hypertension, and peripheral vascular disease [4,5,6]. Many patients will be asymptomatic for prolonged periods and may present for care at a late stage of disease if not appropriately screened early.

1.1. Mechanisms of Cardiac Toxicity

1.1.1. Conventional Chemotherapy

Anthracycline therapy has been the cornerstone of many chemotherapy regimens for pediatric patients, since its introduction in the 1960s. Cardiotoxicity is the main dose limiting side effect that was reported a decade after its first use [7,8,9,10]. While the mechanisms of anthracycline cardiotoxicity are multifaceted, one key pathway is through interaction with topoisomerase 2β, which leads to nuclear DNA damage, mitochondrial dysfunction, and formation of reactive oxygen species [11,12,13,14]. Acute onset cardiotoxicity caused by an anthracycline is rare; it is defined as occurring within one week of administration of the anthracycline and is often reversible with discontinuation. Early onset chronic cardiotoxicity occurs within one year of administration, and late onset chronic cardiotoxicity presents greater than one year after administration of an anthracycline. For the chronic forms, disease is generally progressive. The current definition of high dose anthracycline exposure within the Children’s Oncology Group (COG) is a doxorubicin equivalent of 250 mg/m2. However, there are reports of children developing cardiovascular disease with doses as low at 60 mg/m2 [15,16]. Importantly, recent data have demonstrated that certain accepted dosing equivalencies for mediations like mitoxantrone may actually underestimate the cardiotoxic effect of such therapeutics on survivors of childhood cancer [17].
Non-anthracycline chemotherapy agents are not always thought of as cardiotoxic. However, there is a growing body of evidence demonstrating that alkylating agents (e.g., cyclophosphamide), microtubule inhibitors, proteasome inhibitors, platinum-based drugs, and antimetabolites contribute to cardiovascular disease, which can manifest as ventricular dysfunction, ischemia, venous thromboembolism, arrhythmia, and QT prolongation (Table 1) [18]. Therefore, all patients undergoing cancer therapy are at an increased risk of developing cardiotoxic side effects, regardless of the treatment modality utilized [19,20,21,22].

1.1.2. Radiation

Radiation therapy is associated with cardiotoxicity through direct or indirect exposure of cardiovascular structures to the radiation field, dependent on the type and location of a cancer [23]. This is likely due to the initiation of an inflammatory cascade, generation of fibrosis, and development of endothelial dysfunction. Clinically, this may manifest as pericardial disease, coronary artery disease, calcification of the aortic root, conduction system abnormalities, valvar tissue injury (in severe cases leading to aortic and mitral valve stenosis), cerebrovascular disease, peripheral vascular disease, and heart failure [6,24,25]. While there is no known safe dose of radiation, high risk radiation has been accepted as >30 Gy of total exposure and >15 Gy for direct cardiac exposure [26]. The reduction in exposure to cardiac radiation from the 1970s to the 1990s has led to a significant decrease in heart failure and late-term coronary artery disease in adult survivors of pediatric cancers [27]. Alternatively, utilizing proton therapy may spare the heart from radiation exposure, which could in turn reduce the risk of cardiotoxicity [23]. There is literature suggesting that different areas and structures of the heart are able to withstand varying amounts of total doses of radiation before overall clinical change is seen. Novel research will be needed to advance this area, to further reduce cardiotoxity as well [25].
Intensity-modulated radiation therapy is a novel approach to directing radiation effect to desired fields (i.e., neoplasms), while sparing unaffected tissue. A recent prospective clinical trial determined that whole lung radiation, using intensity-modulated radiation therapy, with the goal of sparing the cardiac field, was feasible and offered similar cancer outcomes but lower doses of total Gy to the heart [28]. These data will be incorporated into the next generation of the Children’s Oncology Group Wilms Tumor Clinical Trials [29]. These studies will allow for the long-term follow-up of both oncologic and cardiac effects, to determine efficacy and safety.

1.1.3. Chimeric Antigen Receptor T-Cell Therapy

The advent of CAR-T therapy has increased remission rates for refractory or relapsed acute lymphocytic leukemia. CAR-T therapy utilizes genetically engineered T-cells to target specific cancer antigens [30,31,32]. However, a major, and potentially fatal, complication of CAR-T therapy is cytokine release syndrome (CRS). This syndrome is defined by a triad of fevers, hypotension, and hypoxia with multi-organ involvement, driven by high levels of inflammatory cytokines (IL-6, TNF-alpha, IL-10, and IFN-Y). CRS can range in severity from mild to severe. This cascade of events can lead to cardiovascular dysfunction, including tachycardia, heart failure, and even death. The mechanism of action is unclear, but it is hypothesized that IL-6 plays a role similar to that in sepsis-related cardiomyopathy. This is confounded by patients having previously received cardiotoxic therapies such as anthracyclines and radiation prior to undergoing treatment with CAR-T therapy. However, the timing of a cardiac event following CAR-T is somewhat predictable, with most occurring just under a week following the infusion [33,34,35,36].
It is imperative that clinicians monitor for CRS and be conscientious that CRS may lead to serious and devastating cardiovascular injuries. Echocardiogram, ECG, and cardiac biomarkers should be obtained in the setting of progressive and severe CRS. Treatment with Tocilizumab, an anti-IL 6 receptor antagonist, may reverse CRS and prevent long-term cardiovascular complications [37]. Inotropic and vasoactive agents may be required to support patients with ventricular dysfunction and hypotension. Unfortunately, ventricular dysfunction can persist after CAR-T cell therapy [38,39].

1.1.4. Immune Checkpoint Inhibitors

Therapy with ICI has changed the landscape of cancer treatment. In recent years, ICI have become more prevalent and at times are used as a front line therapy for a subset of pediatric cancers, which has helped to advance cure rates [40,41]. By definition, ICI are monoclonal antibodies that alter the patient’s immune response to cancer, leading to cell blockade or apoptosis. Cytotoxic T-lymphocyte associated antigen-4 (CTLA-4) and the programmed cell protein-1 (PD-1) pathways are the two most common targets utilized with ICI [42]. Both of these pathways are critical in T-cell regulation; thus, altering the pathway has autoimmune side effects that may affect every organ [43].
Cardiac related adverse effects of ICIs are becoming more frequent, and it is necessary to monitor patients closely throughout therapy. The most common cardiac related events are myocarditis, pericarditis, vasculitis, and arrhythmias. While myocarditis from ICI is a rare occurrence, it has a mortality rate of 40%. This high rate of mortality is thought to be a direct result of T-cell dysregulation targeting the heart, which can translate into clinical symptoms of arrhythmias, congestive heart failure, pneumonitis, or myositis. Vasculitis fatality rate is reported at 6% and can manifest with arthritis and rashes [44].

1.1.5. Small Molecule Inhibitors

Targeted cancer therapies include tyrosine kinase inhibitors, vascular growth factor inhibitors, human epidermal growth factor-2 targeted therapies, and platelet-derived growth factor inhibitors. Currently, over 20 small molecule tyrosine kinase inhibitors, such as sorafenib and imatinib, are available for clinical use [45,46,47]. These drugs are well suited for cancer therapy, given their impact on cellular proliferation, differentiation, and survival, particularly in malignancies. Tyrosine kinase inhibitors inhibit cancer cell proliferation by competing through ATP binding sites, thereby reducing the tyrosine kinase phosphorylation leading to cell dysregulation [48,49,50,51]. Their benefits in pediatric cancers have been established [52,53,54]. Although these medications affect tyrosine kinase pathways in the myocardium, there are currently limited data on their role in cardiotoxicity. Kinase inhibitors have been associated with ventricular dysfunction, hypertension, pulmonary hypertension, and thromboembolism [55,56,57,58].

1.1.6. Targeted Antibody Therapy

Targeted antibody therapy can be used to disrupt molecular pathways, similarly to those affected by small molecule kinase inhibitors [59,60,61]. Given the overlap in the mechanism of action, these monoclonal antibodies can produce similar cardiotoxic effects [62]. Trastuzumab has a primary role in the treatment of human epidermal growth factor 2 positive (HER2+) breast cancers. It is a murine monoclonal antibody that stops the proliferation of overexpression of HER2+ cells. It has been found to work in synergy with many traditional chemotherapy agents commonly used to treat breast cancer [63]. While it is currently the standard of care in this patient population, the most frequent adverse outcome with use of trastuzumab is cardiotoxicity [64]. In pediatric patients, antibody therapies have demonstrated effectiveness in various cancers [59,60,61,65]. These monoclonal antibodies can also cause cardiotoxicity, but clinical trials have shown that standard heart failure therapies offer protection [66].

1.2. Cardioprotection and Prevention

1.2.1. Alternative Anthracycline Dosing Strategies and Derivatives

To mitigate anthracycline mediated cardiotoxicity, alternative dosing strategies have been utilized. In adult patients, increasing anthracycline infusion duration to longer than 6 hours may reduce the risk of subclinical cardiac injury, when compared to a shorter time of administration. Due to limited data in pediatric patients, these data cannot be extrapolated to this population. There was no difference in cardiotoxicity seen for those receiving single peak doses >60 mg/m2 of doxorubicin, when compared to <60 mg/m2 [67]. A liposomal formulation of doxorubicin was developed in order to allow clinicians to overcome the lifetime cumulative dose maximum. This formulation encapsulates the doxorubicin with a phospholipid bilayer of methoxypolyethylene glycol [68]. Liposomal doxorubicin allows a longer half-life, but with decreased cardiac side-effects, and is the only derivative definitively shown to decrease cardiotoxicity [69].

1.2.2. Dexrazoxane

Dexrazoxane (Zinecard) is an EDTA derivative that acts as an iron chelator. It was first approved by the U.S. Food and Drug Administration in 1991 for prevention of cardiomyopathy associated with doxorubicin in breast cancer patients. In 2014, it was designated as an orphan drug for prevention of cardiomyopathy in pediatric and adolescent patients receiving anthracycline therapy. Dexrazoxane has been shown to be cardio-protective by multiple groups. Its primary mechanism of action is to prevent mitochondriopathy by chelating myocardial iron, preventing it from coupling with anthracyclines and reducing the formation of superoxide free radicals. Data support the cardioprotective effects of dexrazoxane, as manifested by improved troponins, natriuretic peptides, and function by echocardiography [70,71,72,73]. Despite the potential benefit, dexrazoxane has not routinely been utilized, due to concerns over its impact on anthracycline treatment effect and the risk of secondary malignancies. The risk of secondary malignancy or decreased efficacy of anthracyclines against the primary cancer are reasons dexrazoxane has not been widely incorporated into pediatric care. Some studies have shown dexrazoxane to be safe in these regards, [74,75,76,77], while others suggest a statistically borderline increase in risk [78]. COG now mandates the use of dexrazoxane in children who have a life time cumulative dose greater than >150 mg/m2 of anthracyclines or any dose of anthracyclines with concomitant radiation use [79].

1.2.3. Exercise and Modifiable Risk Factors

Patients who have cancer and undergo cancer treatment are more likely to have modifiable cardiovascular risk factors, such as hypertension, diabetes, and obesity. Importantly, pre-existing cardiovascular risk factors are strong predictors for development of anthracycline- and radiation-related cardiotoxicity [80]. In addition, the incidence of medical frailty, as defined by five domains (walking limitations, low energy, exhaustion, low lean mass, and weakness) is significantly higher in survivors of pediatric cancer than in sibling controls [81]. Structured exercise demonstrates improvement in mortality, cancer progression, cancer recurrence, health-related quality of life, cardiovascular risk factors, and frailty in a dose-dependent manner [82,83,84,85,86,87]. Routine exercise in adults has been shown to improve cardiovascular function, immune function, body composition, chemotherapy completion rates, and reported markers of mental health. Several studies in adult survivors of pediatric cancers, as well as limited studies in pediatric patients, have likewise demonstrated decreases in cardiovascular-related and total mortality, often in a dose-dependent fashion [85,88,89]. As such, the American Cancer Society has established the ‘Moving Through Cancer’ initiative, with the mission ‘to ensure that that all individuals living with and beyond cancer are assessed, advised, referred to, and supported to engage in appropriate exercise and rehabilitation programming as the standard of care.’ [90].
Aerobic activity is generally considered safe for survivors of pediatric cancer and is advised as part of a ‘heart healthy lifestyle’. Traditionally, patients were advised to avoid isometric/weightlifting activities. However, recent guidelines from the National Comprehensive Cancer Network include recommendations regarding strength training activity for patients with normal ventricular function. There are exercise guidelines for cancer survivors published by the American College of Sports Medicine, although these are primarily adult focused [91]. COG only cautions against such activities for individuals with ventricular dysfunction. For patients who wish to participate in competitive sports, standard guidelines for athletic participation should be followed and ongoing monitoring by a cardiologist is recommended. In 2019 the American Heart Association released a Scientific Statement about cardio-oncology rehabilitation exercise (CORE) programs, including a safety checklist prior to engaging in CORE, components of CORE, and recommendations on how a patient should engage with various rehabilitation services [80]. Finally, clinicians should consider performing an assessment of physical activity when a patient is seen and provide an ‘exercise prescription’ that is safe and effective [92]. A systematic review demonstrated that adherence to such recommendations was improved by goal setting and instruction on how to perform the activities, and there were only a small number of adverse events [93]. Exercise interventions by telehealth have also shown good compliance and limited adverse events [94].

1.2.4. Other Cardioprotective Strategies under Investigation

Remote ischemic conditioning using intermittent limb ischemia-reperfusion is a novel approach in the cancer community. Animal models demonstrated significantly reduced anthracycline cardiac toxicity with utilization of remote ischemic preconditioning [95,96,97]. Currently, clinical studies are ongoing to assess the feasibility and efficacy of remote ischemic conditioning in humans [98].
The COG ALTE1621 study is a multi-center, prospective, randomized, placebo-controlled trial intended to determine if low-dose carvedilol can prevent left ventricular remodeling and dysfunction in survivors of pediatric cancer. The goal enrollment is 250 individuals diagnosed at <21-years-old and treated with high-dose anthracyclines (>300 mg/m2), who will be followed for a period of 2 years. Participants will undergo scheduled assessments with echocardiographic and serum biomarkers [99].

2. Screening and Surveillance

Childhood cancer survivors are a unique group of patients, who require a collaborative approach to optimize their care. COG has published survivorship guidelines that provide broad health counseling for potential late side effects, including carotid artery disease and cardiac toxicity (cardiomyopathy, heart failure, and valve disease), with referral to Cardiology if concerns arise.

2.1. Risk Prediction

Data from the Childhood Cancer Survivor Study (CCSS) and other studies identified factors that increase the risk of developing cardiac toxicity including: younger patient age, African American race, female sex, total anthracycline dose, concomitant radiation exposure, underlying heart disease, pre-modern radiation protocols, and time since treatment (Table 2) [5,100].
According to the National Comprehensive Cancer Network (www.nccn.org accessed 10 October 2021), patients that have undergone cancer treatment should be considered American College of Cardiology/American Heart Association stage A heart failure (no structural abnormality, but at risk to develop heart failure) [101]. Based on both patient and treatment risk factors from the CCSS data (Table 2), an online risk calculator (https://ccss.stjude.org/tools-documents/calculators-other-tools/ccss-cardiovascular-risk-calculator.html accessed on 10 October 2021) was created to predict risk of heart failure, ischemic heart disease, and stroke by age 50 years in survivors of pediatric cancers [102,103,104]. No specific surveillance or treatment recommendations are made by this risk calculator.

2.2. Surveillance Guidelines

Most of the existing guidelines regarding monitoring for the development of cardiotoxicity are established for adult patients, with limited discussion of adult survivors of pediatric cancer [105,106,107,108]. Unfortunately, there are no standardized guidelines for pediatric patients during therapy, and there are variations between protocols [67]. Adult studies revealed that up to 10% of patients can develop subclinical and asymptomatic ventricular dysfunction during induction therapy with anthracycline administration. A delay in recognition and initiation of treatment of just 1–2 months may produce adverse long-term outcomes [109,110]. Recent data in pediatric patients undergoing treatment for acute myeloid leukemia showed that early cardiac toxicity was significantly associated with reduction in event-free survival and overall survival over a 5-year follow up [111].
COG has produced surveillance recommendations for patients that have completed cancer therapy (www.survivorshipguidelines.org accessed on 10 October 2021). These guidelines recommend an annual history and physical exam; lab work, including a lipid profile and glucose every 2 years; ECG during initial evaluation and then as necessary; and echocardiogram every 2–5 years based on risk factors. There are currently no recommendations for pediatric-specific imaging protocols or recommendations regarding the use of serum biomarkers, although efforts to develop such guidelines are underway. In addition to COG, other organizations have also created surveillance guidelines for childhood cancer (Table 3).
In summarizing and interpreting several separate recommendations, the International Late Effects of Childhood Cancer Guideline Harmonization Group recommends screening of left ventricular function with echocardiography as the preferred method, no later than 2 years after completion of anthracycline and/or radiation therapy. Repeat ECG and echocardiogram are recommended every 5 years thereafter, unless dictated otherwise by clinical status. More frequent and lifelong screening can be considered in high-risk survivors [113].
More recently developed imaging modalities such as 3-D echocardiography and myocardial strain assessment have been found to be more sensitive in identifying myocardial changes prior to changes of ejection fraction and shortening fraction [3,114,115,116]. Studies in adult cohorts have assessed the overall utility of myocardial strain in the patient with cancer, with ongoing debate as to the benefits of early detection of ventricular dysfunction weighed against the risk of modifying therapy for a change in strain when the ejection fraction remains normal [117,118]. Cardiac MRI or radionuclide angiography may be reasonable when echocardiography is not technically feasible or optimal. Cardiac MRI is used frequently in pediatric centers, while radionuclide studies are much less common than in adult patients. Cardiac biomarkers may be incorporated in conjunction with imaging but should not be used in isolation. Modalities such as stress echocardiography, exercise testing, and ambulatory rhythm monitoring are not included in the guidelines but are often considered and utilized based on clinical needs.

3. Therapeutic Approaches

3.1. Medical Heart Failure Therapy

Treatment of adult patients who develop heart failure should be directed by standard heart failure guidelines and supplemented by cardio-oncology-specific guidelines regarding changes in imaging, serum biomarkers, symptoms, and chemotherapy exposure risk stratification [119]. Standard medical management in adults includes use of ACE inhibitors, beta blockers, and statins [106,109,120,121]. Starting therapy in the first few months after the development of ventricular dysfunction can lead to improvements in systolic function in the vast majority of patients [109,110]. There are limited comparable data in the pediatric population. Guidelines for management of pediatric cardiomyopathy and heart failure exist, but they do not specifically discuss the cardio-oncology population [122]. ACE inhibitors can decrease left ventricular wall stress and improve subclinical markers of cardiac dysfunction in children. However, the long-term therapeutic effects are unclear [123,124]. Moreover, Lipshultz suggests that the long-term phenotype in survivors of pediatric cancer is that of ‘inadequate ventricular mass with chronic afterload excess associated with progressive contractile deficit and possibly reduced cardiac output and restrictive cardiomyopathy’, the so-called ‘Grinch syndrome’ in which treatment with an ACE inhibitor may be inappropriate [125]. A Cochrane database in 2016 showed no improvement in survival or development of heart failure in the limited number of studies that looked at various treatments, including one of enalapril with 135 survivors of pediatric cancers with asymptomatic LV dysfunction [126]. Sacubitril-valsartan (Entresto) has been studied and shown to have benefit in adult patients with cardiotoxicity, but it has not yet been studied in pediatric patients for this purpose [127].
Two recent surveys of practitioners who care for pediatric cardio-oncology patients found that the majority (>80%) use ACE inhibitors to treat ventricular dysfunction. Conversely, the addition of beta blockers varied between the two studies, with one survey reporting a 20% utilization rate and the other study up to 70%. Only one of the studies reported on the use of aldosterone antagonists, at approximately 50% [128,129].
Once therapies are started, it is unclear if and when they can be discontinued if function returns to normal. The TRED-HF study demonstrated higher rate of relapse of ventricular dysfunction after cessation of medical therapy when compared to patients maintained on medication [130]. Unfortunately, there are not stronger data to suggest a universal practice in this regard. Discontinuation of therapy should be made on an individual basis with the understanding that function may deteriorate.

3.2. Implantable Cardiac Defibrillators and Cardiac Resynchronization Therapy

Indications for an implantable cardiac defibrillator and cardiac resynchronization therapy are similar to other disease processes that cause heart failure and cardiomyopathy. Adult patients with cancer are less likely to receive an implantable cardiac defibrillator compared to other heart failure patients. There is a paucity of data on the efficacy of cardiac resynchronization therapy in cancer survivors, particularly involving pediatric patients [131].

3.3. Advanced Heart Failure Therapy

For some patients, standard oral therapies may become insufficient for the management of cancer treatment-mediated heart failure. In those cases, inotropic infusions, mechanical circulatory support, and even heart transplantation could be considered.

3.4. Heart Transplantation

The first reports of transplantation for anthracycline-induced heart failure in pediatric patients date back to the early 1990s [132,133]. Based on the International Society for Heart and Lung Transplantation guidelines, listing criteria for heart transplantation in patients with a cancer diagnosis should take into account a variety of factors, including type of neoplasm, response to therapies, risk of recurrence, and presence or absence of metastases. Active neoplasm and ongoing cancer treatment with chemotherapy/radiation are absolute contraindications to transplantation at most centers [134]. There is no defined time from the onset of remission to listing for heart transplantation.
For appropriately selected pediatric patients, there is no difference in long-term outcomes after transplant when compared to dilated cardiomyopathy [135,136]. In patients transplanted after a primary oncological diagnosis, there is concern for disease recurrence or increased risk of secondary cancers, related to the immunosuppression necessary to maintain a transplanted heart. However, data from the Pediatric Heart Transplant Society, representing 1985 transplants, demonstrated that all malignancies were due to post-transplant lymphoproliferative disorder, with no difference in malignancy rates in anthracycline-induced cardiomyopathy recipients [137,138,139].

3.5. Mechanical Circulatory Support

Patients that cannot wait for transplantation or are inappropriate for listing due to ongoing cancer therapy may be candidates for mechanical circulatory support. Short- or medium-term support strategies can be utilized as a bridge to recovery in the setting of temporary or reversible dysfunction [140]. Long-term support can be used as a bridge to transplantation or as destination therapy for those that are not transplant candidates. Several case reports and two small cohort studies have described the use of a left ventricular assist device as a bridge to recovery in adult patients with anthracycline-induced cardiomyopathy. Survival was similar to other causes of ventricular dysfunction. However, in one study, there was a higher need for subsequent right ventricular support in the anthracycline-induced cardiomyopathy group [141,142,143,144]. Data in pediatric patients are currently limited to a single case report [145].

4. Conclusions

Many cancer-targeted therapies can lead to acute and chronic cardiovascular pathologies. Unfortunately, there is lack of data guiding primary prevention and treatment strategies in the pediatric population, which leads to substantial practice variability. Several important future research directions have been identified, including those related to cardiac disease, prevention, management of risk factors, risk prediction, early detection, and the role of genetic susceptibility in development of cardiotoxicity. A combination of cohort studies and randomized controlled trials will be key in answering these important questions [3]. The future state within pediatric cardio-oncology should shift to a more proactive stance, to promote continued partnership between pediatric cardiologists and oncologists, in order to better understand, treat, and, ideally, prevent cardiac disease in pediatric oncology patients. Collaboration amongst specialties and across centers will provide critical data to further advance the rapidly growing field of cardio-oncology.

Author Contributions

Conceptualization A.R. and T.D.R.; writing review editing M.B., A.R. and T.D.R. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Miller, K.D.; Siegel, R.L.; Lin, C.C.; Mariotto, A.B.; Kramer, J.L.; Rowland, J.H.; Stein, K.D.; Alteri, R.; Jemal, A. Cancer treatment and sur-vivorship statistics, 2016. CA Cancer J. Clin. 2016, 66, 271–289. [Google Scholar] [CrossRef] [Green Version]
  2. Ward, E.; DeSantis, C.; Robbins, A.; Kohler, B.; Jemal, A. Childhood and adolescent cancer statistics, 2014. CA Cancer J. Clin. 2014, 64, 83–103. [Google Scholar] [CrossRef]
  3. Leerink, J.M.; de Baat, E.C.; Feijen, E.A.M.; Bellersen, L.; van Dalen, E.C.; Grotenhuis, H.B.; Kapusta, L.; Kok, W.E.M.; Loonen, J.; van der Pal, H.J.H. Cardiac Disease in Childhood Cancer Survivors: Risk Prediction, Prevention, and Surveillance: JACC CardioOncology State-of-the-Art Review. JACC CardioOncol. 2020, 2, 363–378. [Google Scholar] [CrossRef] [PubMed]
  4. Bloom, M.W.; Hamo, C.E.; Cardinale, D.; Ky, B.; Nohria, A.; Baer, L.; Skopicki, H.; Lenihan, D.J.; Gheorghiade, M.; Lyon, A.R. Cancer Therapy-Related Cardiac Dysfunction and Heart Failure: Part 1: Definitions, Pathophysiology, Risk Factors, and Imaging. Circ. Heart Fail. 2016, 9, e002661. [Google Scholar] [CrossRef] [Green Version]
  5. Mulrooney, D.A.; Yeazel, M.W.; Leisenring, W.M.; Kawashima, T.; Mertens, A.C.; Mitby, P.; Stovall, M.; Donaldson, S.S.; Green, D.M.; Sklar, C.A.; et al. Cardiac outcomes in a cohort of adult survivors of childhood and adolescent cancer: Retrospective analysis of the Childhood Cancer Survivor Study cohort. BMJ 2009, 339, b4606. [Google Scholar] [CrossRef] [Green Version]
  6. Lipshultz, S.E.; Adams, M.J.; Colan, S.D.; Constine, L.S.; Herman, E.H.; Hsu, D.T.; Hudson, M.M.; Kremer, L.C.; Landy, D.C.; Miller, T.L. 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 2013, 128, 1927–1995. [Google Scholar] [CrossRef] [Green Version]
  7. Octavia, Y.; Tocchetti, C.G.; Gabrielson, K.L.; Janssens, S.; Crijns, H.J.; Moens, A.L. Doxorubicin-induced cardiomyopathy: From molecular mechanisms to therapeutic strategies. J. Mol. Cell. Cardiol. 2012, 52, 1213–1225. [Google Scholar] [CrossRef] [Green Version]
  8. Lefrak, E.A.; Piťha, J.; Rosenheim, S.; Gottlieb, J.A. A clinicopathologic analysis of adriamycin cardiotoxicity. Cancer 1973, 32, 302–314. [Google Scholar] [CrossRef]
  9. Rinehart, J.J.; Lewis, R.P.; Balcerzak, S.P. Adriamycin Cardiotoxicity in Man. Ann. Intern. Med. 1974, 81, 475–478. [Google Scholar] [CrossRef] [PubMed]
  10. Bristow, M.R.; Mason, J.W.; Billingham, M.E.; Daniels, J.R. Doxorubicin Cardiomyopathy: Evaluation by Phonocardiography, Endomyocardial Biopsy, and Cardiac Catheterization. Ann. Intern. Med. 1978, 88, 168–175. [Google Scholar] [CrossRef] [PubMed]
  11. Lipshultz, S.E.; Cochran, T.R.; Franco, V.I.; Miller, T.L. Treatment-related cardiotoxicity in survivors of childhood cancer. Nat. Rev. Clin. Oncol. 2013, 10, 697–710. [Google Scholar] [CrossRef]
  12. Burridge, P.W.; Li, Y.F.; Matsa, E.; Wu, H.; Ong, S.G.; Sharma, A.; Holmstrom, A.; Chang, A.C.; Coronado, M.J.; Ebert, A.D. Human induced pluripotent stem cell-derived cardio-myocytes recapitulate the predilection of breast cancer patients to doxorubicin-induced cardiotoxicity. Nat. Med. 2016, 22, 547–556. [Google Scholar] [CrossRef] [Green Version]
  13. Moslehi, J.; Amgalan, D.; Kitsis, R.N. Grounding Cardio-Oncology in Basic and Clinical Science. Circulation 2017, 136, 3–5. [Google Scholar] [CrossRef]
  14. Vejpongsa, P.; Yeh, E.T. Prevention of anthracycline-induced cardiotoxicity: Challenges and opportunities. J. Am. Coll Cardiol. 2014, 64, 938–945. [Google Scholar] [CrossRef] [Green Version]
  15. Ganame, J.; Mertens, L.; Eidem, B.W.; Claus, P.; D’Hooge, J.; Havemann, L.M.; McMahon, C.J.; Elayda, M.A.A.; Vaughn, W.K.; Towbin, J.A.; et al. Regional myocardial deformation in children with hypertrophic cardiomyopathy: Morphological and clinical correlations. Eur. Heart J. 2007, 28, 2886–2894. [Google Scholar] [CrossRef] [Green Version]
  16. Ganame, J.; Claus, P.; Eyskens, B.; Uyttebroeck, A.; Renard, M.; D’Hooge, J.; Gewillig, M.; Bijnens, B.; Sutherland, G.R.; Mertens, L. Acute Cardiac Functional and Morphological Changes After Anthracycline Infusions in Children. Am. J. Cardiol. 2007, 99, 974–977. [Google Scholar] [CrossRef]
  17. Feijen, E.A.M.; Leisenring, W.M.; Stratton, K.L.; Ness, K.K.; van der Pal, H.J.H.; van Dalen, E.C.; Armstrong, G.T.; Aune, G.J.; Green, D.M.; Hudson, M.M. Derivation of Anthracycline and An-thraquinone Equivalence Ratios to Doxorubicin for Late-Onset Cardiotoxicity. JAMA Oncol. 2019, 5, 864–871. [Google Scholar] [CrossRef]
  18. de Boer, R.A.; Aboumsallem, J.P.; Bracun, V.; Leedy, D.; Cheng, R.; Patel, S.; Rayan, D.; Zaharova, S.; Rymer, J.; Kwan, J.M. A new classification of cardio-oncology syndromes. Cardiooncology 2021, 7, 24. [Google Scholar] [CrossRef]
  19. Ness, K.K.; Plana, J.C.; Joshi, V.M.; Luepker, R.V.; Durand, J.B.; Green, D.M.; Partin, R.E.; Santucci, A.K.; Howell, R.M.; Srivastava, D.K.; et al. Exercise Intolerance, Mortality, and Organ System Impairment in Adult Survivors of Childhood Cancer. J. Clin. Oncol. 2020, 38, 29–42. [Google Scholar] [CrossRef]
  20. Babiker, H.M.; McBride, A.; Newton, M.; Boehmer, L.M.; Drucker, A.G.; Gowan, M.; Cassagnol, M.; Camenisch, T.D.; Anwer, F.; Hollands, J.M. Cardiotoxic effects of chemotherapy: A review of both cytotoxic and molecular targeted oncology therapies and their effect on the cardiovascular system. Crit. Rev. Oncol. 2018, 126, 186–200. [Google Scholar] [CrossRef]
  21. Ryan, T.D.; Nagarajan, R.; Godown, J. Pediatric Cardio-Oncology: Development of Cancer Treatment-Related Cardiotoxicity and the Therapeutic Approach to Affected Patients. Curr. Treat. Options Oncol. 2019, 20, 56. [Google Scholar] [CrossRef]
  22. Yeh, E.T.; Bickford, C.L. Cardiovascular Complications of Cancer Therapy: Incidence, Pathogenesis, Diagnosis, and Management. J. Am. Coll. Cardiol. 2009, 53, 2231–2247. [Google Scholar] [CrossRef] [Green Version]
  23. Bergom, C.; Bradley, J.A.; Ng, A.K.; Samson, P.; Robinson, C.; Lopez-Mattei, J.; Mitchell, J.D. Past, Present, and Future of Radia-tion-Induced Cardiotoxicity: Refinements in Targeting, Surveillance, and Risk Stratification. JACC CardioOncol. 2021, 3, 343–359. [Google Scholar] [CrossRef]
  24. Mitchell, J.D.; Cehic, D.A.; Morgia, M.; Bergrom, C.; Toohey, J.; Guerrero, P.A.; Ferencik, M.; Kikuchi, R.; Carver, J.R.; Zaha, V.G. Cardiovascular Manifestations From Therapeutic Radiation: A Multidisciplinary Expert Consensus Statement From the International Car-dio-Oncology Society. JACC CardioOncol. 2021, 3, 360–380. [Google Scholar] [CrossRef]
  25. Banfill, K.; Giuliani, M.; Aznar, M.; Franks, K.; McWilliam, A.; Schmitt, M.; Sun, F.; Vozenin, M.C.; Finn, C.F. Cardiac Toxicity of Thoracic Radiotherapy: Existing Evidence and Future Directions. J. Thorac. Oncol. 2020, 16, 216–227. [Google Scholar] [CrossRef]
  26. Darby, S.C.; Cutter, D.J.; Boerma, M.; Constine, L.S.; Fajardo, L.F.; Kodama, K.; Mabuchi, K.; Marks, L.B.; Mettler, F.A.; Pierce, L.J.; et al. Radiation-Related Heart Disease: Current Knowledge and Future Prospects. Int. J. Radiat. Oncol. 2010, 76, 656–665. [Google Scholar] [CrossRef] [Green Version]
  27. Mulrooney, D.A.; Hyun, G.; Ness, K.K.; Ehrhardt, M.J.; Yasui, Y.; Duprez, D.; Howell, R.M.; Leisenring, W.; Constine, L.S.; Tonorezos, E.; et al. Major cardiac events for adult survivors of childhood cancer diagnosed between 1970 and 1999: Report from the Childhood Cancer Survivor Study cohort. BMJ 2020, 368, l6794. [Google Scholar] [CrossRef] [Green Version]
  28. Kalapurakal, J.A.; Gopalakrishnan, M.; Walterhouse, D.O.; Rigsby, C.; Rademaker, A.; Helenowski, I.; Kessel, S.; Morano, K.; Laurie, F.; Ulin, K.; et al. Cardiac-Sparing Whole Lung IMRT in Patients With Pediatric Tumors and Lung Metastasis: Final Report of a Prospective Multicenter Clinical Trial. Int. J. Radiat. Oncol. 2018, 103, 28–37. [Google Scholar] [CrossRef]
  29. Pater, L.; Melchior, P.; Rube, C.; Cooper, B.T.; McAleer, M.F.; Kalapurakal, J.A.; Paulino, A.C. Wilms tumor. Pediatr. Blood Cancer 2021, 68 (Suppl. S2), e28257. [Google Scholar] [CrossRef]
  30. Mahadeo, K.M.; The Pediatric Acute Lung Injury and Sepsis Investigators (PALISI) Network; Khazal, S.J.; Abdel-Azim, H.; Fitzgerald, J.C.; Taraseviciute, A.; Bollard, C.M.; Tewari, P.; Duncan, C.; Traube, C.; et al. Management guidelines for paediatric patients receiving chimeric antigen receptor T cell therapy. Nat. Rev. Clin. Oncol. 2018, 16, 45–63. [Google Scholar] [CrossRef] [Green Version]
  31. Grupp, S.A.; Kalos, M.; Barrett, D.; Aplenc, R.; Porter, D.L.; Rheingold, S.R.; Teachey, D.T.; Chew, A.; Hauck, B.; Wright, J.F. Chimeric antigen receptor-modified T cells for acute lymphoid leukemia. N. Engl. J. Med. 2013, 368, 1509–1518. [Google Scholar] [CrossRef] [Green Version]
  32. Maude, S.L.; Frey, N.; Shaw, P.A.; Aplenc, R.; Barrett, D.M.; Bunin, N.J.; Chew, A.; Gonzalez, V.E.; Zheng, Z.; Lacey, S.F.; et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N. Engl. J. Med. 2014, 371, 1507–1517. [Google Scholar] [CrossRef] [Green Version]
  33. Lee, D.W.; Santomasso, B.D.; Locke, F.L.; Ghobadi, A.; Turtle, C.J.; Brudno, J.N.; Maus, M.V.; Park, J.H.; Mead, E.; Pavletic, S. ASTCT Consensus Grading for Cytokine Release Syndrome and Neurologic Toxicity Associated with Im-mune Effector Cells. Biol. Blood Marrow Transpl. 2019, 25, 625–638. [Google Scholar] [CrossRef] [Green Version]
  34. Alvi, R.M.; Frigault, M.J.; Fradley, M.G.; Jain, M.; Mahmood, S.S.; Awadalla, M.; Lee, D.H.; Zlotoff, D.A.; Zhang, L.; Drobni, Z.D.; et al. Cardiovascular Events Among Adults Treated With Chimeric Antigen Receptor T-Cells (CAR-T). J. Am. Coll. Cardiol. 2019, 74, 3099–3108. [Google Scholar] [CrossRef]
  35. Lefebvre, B.; Kang, Y.; Smith, A.M.; Frey, N.V.; Carver, J.R.; Scherrer-Crosbie, M. Cardiovascular Effects of CAR T Cell Therapy: A Retrospective Study. JACC CardioOncol. 2020, 2, 193–203. [Google Scholar] [CrossRef]
  36. Pathan, N.; Hemingway, C.; Alizadeh, A.A.; Stephens, A.C.; Boldrick, J.C.; Oragui, E.; McCabe, C.; Welch, S.B.; Whitney, A.; O’Gara, P.; et al. Role of interleukin 6 in myocardial dysfunction of meningococcal septic shock. Lancet 2004, 363, 203–209. [Google Scholar] [CrossRef] [Green Version]
  37. Shalabi, H.; Sachdev, V.; Kulshreshtha, A.; Cohen, J.W.; Yates, B.; Rosing, D.R.; Sidenko, S.; Delbrook, C.; Mackall, C.; Wiley, B.; et al. Impact of cytokine release syndrome on cardiac function following CD19 CAR-T cell therapy in children and young adults with hematological malignancies. J. Immunother. Cancer 2020, 8, e001159. [Google Scholar] [CrossRef]
  38. Burstein, D.S.; Maude, S.; Grupp, S.; Griffis, H.; Rossano, J.; Lin, K. Cardiac Profile of Chimeric Antigen Receptor T Cell Therapy in Children: A Single-Institution Experience. Biol. Blood Marrow Transpl. 2018, 24, 1590–1595. [Google Scholar] [CrossRef] [Green Version]
  39. Fitzgerald, J.C.; Weiss, S.L.; Maude, S.L.; Barrett, D.M.; Lacey, S.F.; Melenhorst, J.J.; Shaw, P.; Berg, R.A.; June, C.H. Cytokine Release Syndrome After Chimeric Antigen Receptor T Cell Therapy for Acute Lympho-blastic Leukemia. Crit. Care Med. 2017, 45, e124–e131. [Google Scholar] [CrossRef]
  40. Kabir, T.; Chauhan, A.; Anthony, L.; Hildebrandt, G.C. Immune Checkpoint Inhibitors in Pediatric Solid Tumors: Status in 2018. Ochsner J. 2018, 18, 370–376. [Google Scholar] [CrossRef] [Green Version]
  41. Bosse, K.R.; Majzner, R.G.; Mackall, C.L.; Maris, J.M. Immune-Based Approaches for the Treatment of Pediatric Malignancies. Annu. Rev. Cancer Biol. 2020, 4, 353–370. [Google Scholar] [CrossRef] [Green Version]
  42. Balanescu, D.V.; Donisan, T.; Palaskas, N.; Lopez-Mattei, J.; Kim, P.Y.; Buja, L.M.; McNamara, D.M.; Kobashigawa, J.A.; Durand, J.-B.; Iliescu, C.A. Immunomodulatory treatment of immune checkpoint inhibitor-induced myocarditis: Pathway toward precision-based therapy. Cardiovasc. Pathol. 2020, 47, 107211. [Google Scholar] [CrossRef]
  43. Rubio-Infante, N.; Ramírez-Flores, Y.A.; Castillo, E.C.; Lozano, O.; García-Rivas, G.; Torre-Amione, G. Cardiotoxicity associated with immune checkpoint inhibitor therapy: A meta-analysis. Eur. J. Heart Fail. 2021, 23, 1739–1747. [Google Scholar] [CrossRef]
  44. Salem, J.E.; Manouchehri, A.; Moey, M.; Lebrun-Vignes, B.; Bastarache, L.; Pariente, A.; Gobert, A.; Spano, J.P.; Balko, J.M.; Bonaca, M.P. Cardiovascular toxicities associated with immune checkpoint inhibitors: An observa-tional, retrospective, pharmacovigilance study. Lancet Oncol. 2018, 19, 1579–1589. [Google Scholar] [CrossRef]
  45. Moslehi, J.J. Cardiovascular Toxic Effects of Targeted Cancer Therapies. N. Engl. J. Med. 2016, 375, 1457–1467. [Google Scholar] [CrossRef] [PubMed]
  46. Jiao, Q.; Bi, L.; Ren, Y.; Song, S.; Wang, Q.; Wang, Y. Advances in studies of tyrosine kinase inhibitors and their acquired re-sistance. Molecular Cancer 2018, 17, 36. [Google Scholar] [CrossRef] [PubMed]
  47. Adamson, P.C. Improving the outcome for children with cancer: Development of targeted new agents. CA Cancer J. Clin. 2015, 65, 212–220. [Google Scholar] [CrossRef] [Green Version]
  48. Krause, D.S.; Van Etten, R.A. Tyrosine Kinases as Targets for Cancer Therapy. N. Engl. J. Med. 2005, 353, 172–187. [Google Scholar] [CrossRef] [Green Version]
  49. Kantarjian, H.; Sawyers, C.; Hochhaus, A.; Guilhot, F.; Schiffer, C.; Gambacorti-Passerini, C.; Niederwieser, D.; Resta, D.; Capdeville, R.; Zoellner, U. Hematologic and cytogenetic responses to imatinib mesylate in chronic myelogenous leu-kemia. N. Engl. J. Med. 2002, 346, 645–652. [Google Scholar] [CrossRef]
  50. Druker, B.J.; Talpaz, M.; Resta, D.J.; Peng, B.; Buchdunger, E.; Ford, J.M.; Lydon, N.B.; Kantarjian, H.; Capdeville, R.; Ohno-Jones, S.; et al. Efficacy and Safety of a Specific Inhibitor of the BCR-ABL Tyrosine Kinase in Chronic Myeloid Leukemia. N. Engl. J. Med. 2001, 344, 1031–1037. [Google Scholar] [CrossRef] [Green Version]
  51. Druker, B.J.; Guilhot, F.; O’Brien, S.G.; Gathmann, I.; Kantarjian, H.M.; Gattermann, N.; Deininger, M.W.; Silver, R.T.; Goldman, J.M.; Stone, R.M.; et al. Five-Year Follow-up of Patients Receiving Imatinib for Chronic Myeloid Leukemia. N. Engl. J. Med. 2006, 355, 2408–2417. [Google Scholar] [CrossRef]
  52. Gross, A.M.; Wolters, P.L.; Dombi, E.; Baldwin, A.; Whitcomb, P.; Fisher, M.J.; Weiss, B.; Kim, A.; Bornhorst, M.; Shah, A.C.; et al. Selumetinib in Children with Inoperable Plexiform Neurofibromas. N. Engl. J. Med. 2020, 382, 1430–1442. [Google Scholar] [CrossRef]
  53. Ronsley, R.; Hounjet, C.D.; Cheng, S.; Rassekh, S.R.; Duncan, W.J.; Dunham, C.; Gardiner, J.; Ghag, A.; Ludemann, J.P.; Wensley, D. Trametinib therapy for children with neurofibromatosis type 1 and life-threatening plex-iform neurofibroma or treatment-refractory low-grade glioma. Cancer Med. 2021, 10, 3556–3564. [Google Scholar] [CrossRef]
  54. Dombi, E.; Baldwin, A.; Marcus, L.J.; Fisher, M.J.; Weiss, B.; Kim, A.; Whitcomb, P.; Martin, S.; Aschbacher-Smith, L.E.; Rizvi, T.A.; et al. Activity of Selumetinib in Neurofibromatosis Type 1–Related Plexiform Neurofibromas. N. Engl. J. Med. 2016, 375, 2550–2560. [Google Scholar] [CrossRef]
  55. Chaar, M.; Kamta, J.; Ait-Oudhia, S. Mechanisms, monitoring, and management of tyrosine kinase inhibitors-associated car-diovascular toxicities. Onco. Targets Ther. 2018, 11, 6227–6237. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Jain, D.; Russell, R.R.; Schwartz, R.G.; Panjrath, G.S.; Aronow, W. Cardiac Complications of Cancer Therapy: Pathophysiology, Identification, Prevention, Treatment, and Future Directions. Curr. Cardiol. Rep. 2017, 19, 36. [Google Scholar] [CrossRef] [PubMed]
  57. Chu, T.F.; Rupnick, M.A.; Kerkela, R.; Dallabrida, S.M.; Zurakowski, D.; Nguyen, L.; Woulfe, K.; Pravda, E.; Cassiola, F.; Desai, J. Cardiotox-icity associated with tyrosine kinase inhibitor sunitinib. Lancet 2007, 370, 2011–2019. [Google Scholar] [CrossRef] [Green Version]
  58. Ewer, M.S.; Suter, T.M.; Lenihan, D.J.; Niculescu, L.; Breazna, A.; Demetri, G.D.; Motzer, R.J. Cardiovascular events among 1090 cancer patients treated with sunitinib, interferon, or placebo: A comprehensive adjudicated database analysis demonstrating clin-ically meaningful reversibility of cardiac events. Eur. J. Cancer 2014, 50, 2162–2170. [Google Scholar] [CrossRef] [PubMed]
  59. Ladenstein, R.; Pötschger, U.; Valteau-Couanet, D.; Luksch, R.; Castel, V.; Yaniv, I.; Laureys, G.; Brock, P.; Michon, J.M.; Owens, C.; et al. Interleukin 2 with anti-GD2 antibody ch14.18/CHO (dinutuximab beta) in patients with high-risk neuroblastoma (HR-NBL1/SIOPEN): A multicentre, randomised, phase 3 trial. Lancet Oncol. 2018, 19, 1617–1629. [Google Scholar] [CrossRef]
  60. Grill, J.; Massimino, M.; Bouffet, E.; Azizi, A.; McCowage, G.; Cañete, A.; Saran, F.; Le Deley, M.-C.; Varlet, P.; Morgan, P.; et al. Phase II, Open-Label, Randomized, Multicenter Trial (HERBY) of Bevacizumab in Pediatric Patients With Newly Diagnosed High-Grade Glioma. J. Clin. Oncol. 2018, 36, 951–958. [Google Scholar] [CrossRef]
  61. Chisholm, J.C.; Merks, J.H.; Casanova, M.; Bisogno, G.; Orbach, D.; Gentet, J.-C.; Defachelles, A.-S.; Chastagner, P.; Lowis, S.; Ronghe, M.; et al. Open-label, multicentre, randomised, phase II study of the EpSSG and the ITCC evaluating the addition of bevacizumab to chemotherapy in childhood and adolescent patients with metastatic soft tissue sarcoma (the BERNIE study). Eur. J. Cancer 2017, 83, 177–184. [Google Scholar] [CrossRef]
  62. Chen, Z.; Ai, D. Cardiotoxicity associated with targeted cancer therapies. Mol. Clin. Oncol. 2016, 4, 675–681. [Google Scholar] [CrossRef] [Green Version]
  63. Slamon, D.; Eiermann, W.; Robert, N.; Pienkowski, T.; Martin, M.; Press, M.; Mackey, J.; Glaspy, J.; Chan, A.; Pawlicki, M.D.; et al. Adjuvant Trastuzumab in HER2-Positive Breast Cancer. N. Engl. J. Med. 2011, 365, 1273–1283. [Google Scholar] [CrossRef] [Green Version]
  64. Mazzotta, M.; Krasniqi, E.; Barchiesi, G.; Pizzuti, L.; Tomao, F.; Barba, M.; Vici, P. Long-Term Safety and Real-World Effectiveness of Trastuzumab in Breast Cancer. J. Clin. Med. 2019, 8, 254. [Google Scholar] [CrossRef] [Green Version]
  65. Zhukova, N.; Rajagopal, R.; Lam, A.; Coleman, L.; Shipman, P.; Walwyn, T.; Williams, M.; Sullivan, M.; Campbell, M.; Bhatia, K.; et al. Use of bevacizumab as a single agent or in adjunct with traditional chemotherapy regimens in children with unresectable or progressive low-grade glioma. Cancer Med. 2018, 8, 40–50. [Google Scholar] [CrossRef] [PubMed]
  66. Gulati, G.; Heck, S.L.; Ree, A.H.; Hoffmann, P.; Schulz-Menger, J.; Fagerland, M.W.; Gravdehaug, B.; von Knobelsdorff-Brenkenhoff, F.; Bratland, A.; Storas, T.H. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): A 2 x 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol. Eur. Heart J. 2016, 37, 1671–1680. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Markman, M.; Ec, V.D.; Hj, V.D.P.; Lc, K. Faculty Opinions recommendation of Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy. Cochrane Database Syst. Rev. 2019, 3, CD005008. [Google Scholar] [CrossRef]
  68. Franco, Y.L.; Vaidya, T.R.; Ait-Oudhia, S. Anticancer and cardio-protective effects of liposomal doxorubicin in the treatment of breast cancer. Breast Cancer Targets Ther. 2018, 10, 131–141. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. van Dalen, E.C.; Michiels, E.M.; Caron, H.N.; Kremer, L.C. Different anthracycline derivates for reducing cardiotoxicity in cancer patients. Cochrane Database Syst. Rev. 2010, 4, CD005006. [Google Scholar]
  70. Lipshultz, S.E.; Rifai, N.; Dalton, V.M.; Levy, D.E.; Silverman, L.B.; Lipsitz, S.R.; Colan, S.D.; Asselin, B.L.; Barr, R.D.; Clavell, L.A. The effect of dexrazoxane on myocardial injury in doxoru-bicin-treated children with acute lymphoblastic leukemia. N. Engl. J. Med. 2004, 351, 145–153. [Google Scholar] [CrossRef] [PubMed]
  71. Lipshultz, S.E.; Miller, T.L.; Scully, R.E.; Lipsitz, S.R.; Rifai, N.; Silverman, L.B.; Colan, S.D.; Neuberg, D.S.; Dahlberg, S.E.; Henkel, J.M. Changes in cardiac biomarkers during doxoru-bicin treatment of pediatric patients with high-risk acute lymphoblastic leukemia: Associations with long-term echocardio-graphic outcomes. J. Clin. Oncol. 2012, 30, 1042–1049. [Google Scholar] [CrossRef]
  72. Lipshultz, S.E.; Scully, R.E.; Lipsitz, S.R.; Sallan, S.E.; Silverman, L.B.; Miller, T.L.; Barry, E.V.; Asselin, B.L.; Athale, U.; Clavell, L.A. Assessment of dexrazoxane as a cardioprotectant in doxorubicin-treated children with high-risk acute lymphoblastic leukaemia: Long-term follow-up of a prospective, randomised, multicentre trial. Lancet Oncol. 2010, 11, 950–961. [Google Scholar] [CrossRef] [Green Version]
  73. Wexler, L.H. Ameliorating anthracycline cardiotoxicity in children with cancer: Clinical trials with dexrazoxane. Semin. Oncol. 1998, 25, 86–92. [Google Scholar]
  74. Kopp, L.M.; Womer, R.B.; Schwartz, C.L.; Ebb, D.H.; Franco, V.I.; Hall, D.; Barkauskas, D.A.; Krailo, M.D.; Grier, H.E.; Meyers, P.A. Effects of dexrazoxane on doxorubicin-related cardiotoxicity and second malignant neoplasms in children with osteosarcoma: A report from the Chil-dren’s Oncology Group. Cardiooncology 2019, 5, 15. [Google Scholar] [CrossRef] [Green Version]
  75. Seif, A.E.; Walker, D.M.; Li, Y.; Huang, Y.-S.V.; Kavcic, M.; Torp, K.; Bagatell, R.; Fisher, B.T.; Aplenc, R. Dexrazoxane exposure and risk of secondary acute myeloid leukemia in pediatric oncology patients. Pediatr. Blood Cancer 2014, 62, 704–709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Chow, E.J.; Asselin, B.L.; Schwartz, C.L.; Doody, D.R.; Leisenring, W.M.; Aggarwal, S.; Scott Baker, K.; Bhatia, S.; Constine, L.S.; Freyer, D.R. Late Mortality After Dexrazoxane Treatment: A Report From the Children’s Oncology Group. J. Clin. Oncol. 2015, 33, 2639. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. Lipshultz Steven, E.; Law Yuk, M.; Asante-Korang, A.; Austin Eric, D.; Dipchand Anne, I.; Everitt Melanie, D.; Hsu Daphne, T.; Lin Kimberly, Y.; Price Jack, F.; Wilkinson James, D. Cardiomyopathy in Children: Classification and Di-agnosis: A Scientific Statement From the American Heart Association. Circulation 2019, 140, e9–e68. [Google Scholar] [CrossRef]
  78. Shaikh, F.; Dupuis, L.L.; Alexander, S.; Gupta, A.; Mertens, L.; Nathan, P.C. Cardioprotection and Second Malignant Neoplasms Associated With Dexrazoxane in Children Receiving Anthracycline Chemotherapy: A Systematic Review and Meta-Analysis. J. Natl. Cancer Inst. 2015, 108, djv357. [Google Scholar] [CrossRef] [Green Version]
  79. Lipshultz, S.E.; Franco, V.I.; Sallan, S.E.; Adamson, P.C.K.; Steiner, R.; Swain, S.M.; Gligorov, J.; Minotti, G. Dexrazoxane for reducing anthracycline-related cardiotoxicity in children with cancer: An update of the evidence. Prog. Pediatr. Cardiol. 2014, 36, 39–49. [Google Scholar] [CrossRef]
  80. Gilchrist, S.C.; Barac, A.; Ades, P.A.; Alfano, C.M.; Franklin, B.A.; Jones, L.W.; La Gerche, A.; Ligibel, J.A.; Lopez, G.; Madan, K. Cardio-Oncology Rehabilitation to Manage Cardiovascular Outcomes in Cancer Patients and Survivors: A Scientific State-ment From the American Heart Association. Circulation 2019, 139, e997–e1012. [Google Scholar] [CrossRef]
  81. Hayek, S.; Gibson, T.M.; Leisenring, W.; Guida, J.; Gramatges, M.M.; Lupo, P.J.; Howell, R.M.; Oeffinger, K.C.; Bhatia, S.; Edelstein, K.; et al. Prevalence and Predictors of Frailty in Childhood Cancer Survivors and Siblings: A Report From the Childhood Cancer Survivor Study. J. Clin. Oncol. 2020, 38, 232–247. [Google Scholar] [CrossRef]
  82. Vainshelboim, B.; Chen, Z.; Lima, R.M.; Myers, J. Cardiorespiratory Fitness, Smoking Status, and Risk of Incidence and Mortality From Cancer: Findings From the Veterans Exercise Testing Study. J. Phys. Act. Health 2019, 16, 1098–1104. [Google Scholar] [CrossRef] [Green Version]
  83. Cormie, P.; Zopf, E.M.; Zhang, X.; Schmitz, K.H. The Impact of Exercise on Cancer Mortality, Recurrence, and Treatment-Related Adverse Effects. Epidemiol. Rev. 2017, 39, 71–92. [Google Scholar] [CrossRef]
  84. Mishra, S.I.; Scherer, R.W.; Snyder, C.; Geigle, P.M.; Berlanstein, D.R.; Topaloglu, O. Exercise interventions on health-related quality of life for people with cancer during active treatment. Cochrane Database Syst. Rev. 2012, 8, CD008465. [Google Scholar] [CrossRef] [PubMed]
  85. Jones, L.W.; Liu, Q.; Armstrong, G.T.; Ness, K.K.; Yasui, Y.; Devine, K.; Tonorezos, E.; Soares-Miranda, L.; Sklar, C.A.; Douglas, P.S. Exercise and risk of major cardiovascular events in adult survivors of childhood hodgkin lym-phoma: A report from the childhood cancer survivor study. J. Clin. Oncol. 2014, 32, 3643–3650. [Google Scholar] [CrossRef]
  86. Hutchins, K.K.; Siddeek, H.; Franco, V.I.; Lipshultz, S.E. Prevention of cardiotoxicity among survivors of childhood cancer. Br. J. Clin. Pharmacol. 2016, 83, 455–465. [Google Scholar] [CrossRef]
  87. Chen, J.J.; Wu, P.-T.; Middlekauff, H.R.; Nguyen, K.-L. Aerobic exercise in anthracycline-induced cardiotoxicity: A systematic review of current evidence and future directions. Am. J. Physiol. Circ. Physiol. 2017, 312, H213–H222. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Scott, J.M.; Li, N.; Liu, Q.; Yasui, Y.; Leisenring, W.; Nathan, P.C.; Gibson, T.; Armenian, S.H.; Nilsen, T.S.; Oeffinger, K.C.; et al. Association of Exercise With Mortality in Adult Survivors of Childhood Cancer. JAMA Oncol. 2018, 4, 1352–1358. [Google Scholar] [CrossRef] [PubMed]
  89. Davis, N.L.; Tolfrey, K.; Jenney, M.; Elson, R.; Stewart, C.; Moss, A.D.; Cornish, J.M.; Stevens, M.C.G.; Crowne, E.C. Combined resistance and aerobic exercise intervention improves fitness, insulin resistance and quality of life in survivors of childhood haemo-poietic stem cell transplantation with total body irradiation. Pediatr Blood Cancer 2020, 67, e28687. [Google Scholar] [CrossRef] [PubMed]
  90. Schmitz, K.H.; Stout, N.L.; Mpp, M.M.; Campbell, A.; Schwartz, A.L.; Grimmett, C.; Meyerhardt, J.A.; Do, J.M.S. Moving through cancer: Setting the agenda to make exercise standard in oncology practice. Cancer 2020, 127, 476–484. [Google Scholar] [CrossRef]
  91. Campbell, K.L.; Winters-Stone, K.M.; Wiskemann, J.; May, A.M.; Schwartz, A.L.; Courneya, K.S.; Zucker, D.S.; Matthews, C.E.; Ligibel, J.A.; Gerber, L.H.; et al. Exercise Guidelines for Cancer Survivors: Consensus Statement from International Multidisciplinary Roundtable. Med. Sci. Sports Exerc. 2019, 51, 2375–2390. [Google Scholar] [CrossRef] [Green Version]
  92. Schmitz, K.H.; Campbell, A.M.; Stuiver, M.M.; Pinto, B.M.; Schwartz, A.L.; Morris, G.S.; Ligibel, J.A.; Cheville, A.; Galvão, D.A.; Alfano, C.M.; et al. Exercise is medicine in oncology: Engaging clinicians to help patients move through cancer. CA Cancer J. Clin. 2019, 69, 468–484. [Google Scholar] [CrossRef] [Green Version]
  93. Turner, R.R.; Steed, L.; Quirk, H.; Greasley, R.U.; Saxton, J.M.; Taylor, S.J.; Rosario, D.J.; Thaha, M.A.; Bourke, L. Interventions for pro-moting habitual exercise in people living with and beyond cancer. Cochrane Database Syst. Rev. 2018, 9, CD010192. [Google Scholar]
  94. Morrison, K.S.; Paterson, C.; Toohey, K. The Feasibility of Exercise Interventions Delivered via Telehealth for People Affected by Cancer: A Rapid Review of the Literature. Semin. Oncol. Nurs. 2020, 36, 151092. [Google Scholar] [CrossRef] [PubMed]
  95. He, Q.; Wang, F.; Ryan, T.D.; Chalasani, M.; Redington, A.N. Repeated Remote Ischemic Conditioning Reduces Doxorubi-cin-Induced Cardiotoxicity. JACC CardioOncol. 2020, 2, 41–52. [Google Scholar] [CrossRef]
  96. Galán-Arriola, C.; Villena-Gutiérrez, R.; Higuero-Verdejo, M.; Díaz-Rengifo, I.; Pizarro, G.; López, G.J.; de Molina-Iracheta, A.; Pérez-Martínez, C.; García, R.D.; González-Calle, D.; et al. Remote ischaemic preconditioning ameliorates anthracycline-induced cardiotoxicity and preserves mitochondrial integrity. Cardiovasc. Res. 2020, 117, 1132–1143. [Google Scholar] [CrossRef]
  97. Gertz, Z.M.; Cain, C.; Kraskauskas, D.; Devarakonda, T.; Mauro, A.G.; Thompson, J.; Samidurai, A.; Chen, Q.; Gordon, S.W.; Lesnefsky, E.J.; et al. Remote Ischemic Pre-Conditioning Attenuates Adverse Cardiac Remodeling and Mortality Following Doxorubicin Administration in Mice. JACC CardioOncol. 2019, 1, 221–234. [Google Scholar] [CrossRef]
  98. Chung, R.; Maulik, A.; Hamarneh, A.; Hochhauser, D.; Hausenloy, D.J.; Walker, J.M.; Yellon, D.M. Effect of Remote Ischaemic Con-ditioning in Oncology Patients Undergoing Chemotherapy: Rationale and Design of the ERIC-ONC Study—A Single-Center, Blinded, Randomized Controlled Trial. Clin. Cardiol. 2016, 39, 72–82. [Google Scholar] [CrossRef] [PubMed]
  99. Armenian, S.H.; Hudson, M.M.; Chen, M.H.; Colan, S.D.; Lindenfeld, L.; Mills, G.; Siyahian, A.; Gelehrter, S.; Dang, H.; Hein, W.; et al. Rationale and design of the Children’s Oncology Group (COG) study ALTE1621: A randomized, placebo-controlled trial to determine if low-dose carvedilol can prevent anthracycline-related left ventricular remodeling in childhood cancer survivors at high risk for developing heart failure. BMC Cardiovasc. Disord. 2016, 16, 187. [Google Scholar] [CrossRef] [Green Version]
  100. Mulrooney, D.A.; Armstrong, G.T.; Huang, S.; Ness, K.K.; Ehrhardt, M.J.; Joshi, V.M.; Plana, J.C.; Soliman, E.Z.; Green, D.M.; Srivastava, D. Cardiac Outcomes in Adult Survivors of Childhood Cancer Exposed to Cardiotoxic Therapy: A Cross-sectional Study. Ann. Intern. Med. 2016, 164, 93–101. [Google Scholar] [CrossRef] [Green Version]
  101. Denlinger, C.S.; Sanft, T.; Baker, K.S.; Broderick, G.; Demark-Wahnefried, W.; Friedman, D.L.; Goldman, M.; Hudson, M.; Khakpour, N.; King, A. Survivorship, Version 2.2018, NCCN Clinical Practice Guidelines in Oncology. J. Natl. Compr. Cancer Netw. 2018, 16, 1216–1247. [Google Scholar] [CrossRef]
  102. Chow, E.J.; Chen, Y.; Kremer, L.C.; Breslow, N.E.; Hudson, M.M.; Armstrong, G.T.; Border, W.L.; Feijen, E.A.M.; Green, D.M.; Meacham, L.R.; et al. Individual Prediction of Heart Failure Among Childhood Cancer Survivors. J. Clin. Oncol. 2015, 33, 394–402. [Google Scholar] [CrossRef]
  103. Chow, E.J.; Chen, Y.; Hudson, M.M.; Feijen, E.A.M.; Kremer, L.C.; Border, W.L.; Green, D.M.; Meacham, L.R.; Mulrooney, D.A.; Ness, K.K.; et al. Prediction of Ischemic Heart Disease and Stroke in Survivors of Childhood Cancer. J. Clin. Oncol. 2018, 36, 44–52. [Google Scholar] [CrossRef] [PubMed]
  104. Chen, Y.; Chow, E.J.; Oeffinger, K.C.; Border, W.L.; Leisenring, W.M.; Meacham, L.R.; A Mulrooney, D.; A Sklar, C.; Stovall, M.; Robison, L.L.; et al. Traditional Cardiovascular Risk Factors and Individual Prediction of Cardiovascular Events in Childhood Cancer Survivors. J. Natl. Cancer Inst. 2019, 112, 256–265. [Google Scholar] [CrossRef]
  105. Carver, J.R.; Szalda, D.; Ky, B. Asymptomatic cardiac toxicity in long-term cancer survivors: Defining the population and rec-ommendations for surveillance. Semin. Oncol. 2013, 40, 229–238. [Google Scholar] [CrossRef] [Green Version]
  106. Zamorano, J.L.; Lancellotti, P.; Rodriguez Munoz, D.; Aboyans, V.; Asteggiano, R.; Galderisi, M.; Habib, G.; Lenihan, D.J.; Lip, G.Y.; Lyon, A.R. 2016 ESC Position Paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: The Task Force for cancer treatments and cardiovascular toxicity of the European Society of Cardiology (ESC). Eur. J. Heart Fail. 2017, 19, 9–42. [Google Scholar] [CrossRef] [PubMed]
  107. Armenian, S.H.; Lacchetti, C.; Barac, A.; Carver, J.; Constine, L.S.; Denduluri, N.; Dent, S.; Douglas, P.S.; Durand, J.-B.; Ewer, M.; et al. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J. Clin. Oncol. 2017, 35, 893–911. [Google Scholar] [CrossRef]
  108. Barac, A.; Murtagh, G.; Carver, J.R.; Chen, M.H.; Freeman, A.M.; Herrmann, J.; Iliescu, C.; Ky, B.; Mayer, E.L.; Okwuosa, T.M. Cardiovascular Health of Patients With Cancer and Cancer Survivors: A Roadmap to the Next Level. J. Am. Coll. Cardiol. 2015, 65, 2739–2746. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Cardinale, D.; Colombo, A.; Bacchiani, G.; Tedeschi, I.; Meroni, C.A.; Veglia, F.; Civelli, M.; Lamantia, G.; Colombo, N.; Curigliano, G. Early Detection of Anthracycline Cardiotoxicity and Improvement With Heart Failure Therapy. Circulation 2015, 131, 1981–1988. [Google Scholar] [CrossRef] [Green Version]
  110. Cardinale, D.; Colombo, A.; Lamantia, G.; Colombo, N.; Civelli, M.; De Giacomi, G.; Rubino, M.; Veglia, F.; Fiorentini, C.; Cipolla, C.M. Anthracycline-Induced Cardiomyopathy: Clinical Relevance and Response to Pharmacologic Therapy. J. Am. Coll. Cardiol. 2010, 55, 213–220. [Google Scholar] [CrossRef] [Green Version]
  111. Getz, K.D.; Sung, L.; Ky, B.; Gerbing, R.B.; Leger, K.J.; Leahy, A.B.; Sack, L.; Woods, W.G.; Alonzo, T.; Gamis, A. Occurrence of Treatment-Related Cardiotoxicity and Its Impact on Outcomes Among Children Treated in the AAML0531 Clinical Trial: A Report From the Children’s Oncology Group. J. Clin. Oncol. 2019, 37, 12–21. [Google Scholar] [CrossRef] [PubMed]
  112. Sieswerda, E.; Postma, A.; van Dalen, E.C.; van der Pal, H.J.; Tissing, W.J.; Rammeloo, L.A.; Kok, W.E.; van Leeuwen, F.E.; Caron, H.N.; Kremer, L.C. The Dutch Childhood On-cology Group guideline for follow-up of asymptomatic cardiac dysfunction in childhood cancer survivors. Ann. Oncol. 2012, 23, 2191–2198. [Google Scholar] [CrossRef] [PubMed]
  113. Armenian, S.H.; Hudson, M.M.; Mulder, R.L.; Chen, M.H.; Constine, L.S.; Dwyer, M.; Nathan, P.C.; Tissing, W.J.; Shankar, S.; Sieswerda, E. International Late Effects of Childhood Cancer Guideline Harmonization G. Recommendations for cardiomyopathy surveillance for survivors of child-hood cancer: A report from the International Late Effects of Childhood Cancer Guideline Harmonization Group. Lancet Oncol. 2015, 16, e123–e136. [Google Scholar] [CrossRef] [Green Version]
  114. Thavendiranathan, P.; Poulin, F.; Lim, K.D.; Plana, J.C.; Woo, A.; Marwick, T.H. Use of myocardial strain imaging by echocardiog-raphy for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: A systematic review. J. Am. Coll. Cardiol. 2014, 63, 2751–2768. [Google Scholar] [CrossRef] [Green Version]
  115. Pignatelli, R.H.; Ghazi, P.; Reddy, S.C.-B.; Thompson, P.; Cui, Q.; Castro, J.; Okcu, M.F.; Jefferies, J.L.; Ghazi, P.; Jefferies, L.J. Abnormal Myocardial Strain Indices in Children Receiving Anthracycline Chemotherapy. Pediatr. Cardiol. 2015, 36, 1610–1616. [Google Scholar] [CrossRef]
  116. Tuzovic, M.; Wu, P.-T.; Bs, S.K.; Nguyen, K.-L. Natural history of myocardial deformation in children, adolescents, and young adults exposed to anthracyclines: Systematic review and meta-analysis. Echocardiography 2018, 35, 922–934. [Google Scholar] [CrossRef]
  117. Thavendiranathan, P.; Negishi, T.; Somerset, E.; Negishi, K.; Penicka, M.; Lemieux, J.; Aakhus, S.; Miyazaki, S.; Shirazi, M.; Galderisi, M.; et al. Strain-Guided Management of Potentially Cardiotoxic Cancer Therapy. J. Am. Coll. Cardiol. 2020, 77, 392–401. [Google Scholar] [CrossRef]
  118. Moslehi, J.J.; Witteles, R.M. Global Longitudinal Strain in Cardio-Oncology. J. Am. Coll. Cardiol. 2021, 77, 402–404. [Google Scholar] [CrossRef]
  119. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E.; Drazner, M.H.; Fonarow, G.C.; Geraci, S.A.; Horwich, T.; Januzzi, J.L. American Heart Association Task Force on Practice G. 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. J. Am. Coll. Cardiol. 2013, 62, e147–e239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Kalay, N.; Basar, E.; Ozdogru, I.; Er, O.; Cetinkaya, Y.; Dogan, A.; Oguzhan, A.; Eryol, N.K.; Topsakal, R.; Ergin, A.; et al. Protective Effects of Carvedilol Against Anthracycline-Induced Cardiomyopathy. J. Am. Coll. Cardiol. 2006, 48, 2258–2262. [Google Scholar] [CrossRef] [Green Version]
  121. Seicean, S.; Seicean, A.; Plana, J.C.; Budd, G.T.; Marwick, T.H. Effect of statin therapy on the risk for incident heart failure in patients with breast cancer receiving anthracycline chemotherapy: An observational clinical cohort study. J. Am. Coll. Cardiol. 2012, 60, 2384–2390. [Google Scholar] [CrossRef]
  122. Kirk, R.; Dipchand, A.I.; Rosenthal, D.N.; Addonizio, L.; Burch, M.; Chrisant, M.; Dubin, A.; Everitt, M.; Gajarski, R.; Mertens, L. The International Society for Heart and Lung Trans-plantation Guidelines for the management of pediatric heart failure: Executive summary [Corrected]. J. Heart Lung Transpl. 2014, 33, 888–909. [Google Scholar] [CrossRef]
  123. Lipshultz, S.E.; Lipsitz, S.R.; Sallan, S.E.; Simbre, V.C.; Shaikh, S.L.; Mone, S.M.; Gelber, R.D.; Colan, S.D. Long-term enalapril therapy for left ventricular dysfunction in doxorubicin-treated survivors of childhood cancer. J. Clin. Oncol. 2002, 20, 4517–4522. [Google Scholar] [CrossRef]
  124. Silber, J.H.; Cnaan, A.; Clark, B.J.; Paridon, S.M.; Chin, A.J.; Rychik, J.; Hogarty, A.N.; Cohen, M.I.; Barber, G.; Rutkowski, M. Enalapril to prevent cardiac function decline in long-term survivors of pediatric cancer ex-posed to anthracyclines. J. Clin. Oncol. 2004, 22, 820–828. [Google Scholar] [CrossRef]
  125. Lipshultz, S.E.; Lipsitz, S.R.; Sallan, S.E.; Dalton, V.M.; Mone, S.M.; Gelber, R.D.; Colan, S.D. Chronic progressive cardiac dysfunction years after doxorubicin therapy for childhood acute lymphoblastic leukemia. J. Clin. Oncol. 2005, 23, 2629–2636. [Google Scholar] [CrossRef] [PubMed]
  126. Cheuk, D.K.; Sieswerda, E.; van Dalen, E.C.; Postma, A.; Kremer, L.C. Medical interventions for treating anthracycline-induced symptomatic and asymptomatic cardiotoxicity during and after treatment for childhood cancer. Cochrane Database Syst. Rev. 2016, CD008011. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  127. Martín-Garcia, A.; López-Fernández, T.; Mitroi, C.; Chaparro-Muñoz, M.; Moliner, P.; Martin-Garcia, A.C.; Martinez-Monzonis, A.; Castro, A.; Lopez-Sendon, J.L.; Sanchez, P.L. Effectiveness of sacubitril–valsartan in cancer patients with heart failure. ESC Heart Fail. 2020, 7, 763–767. [Google Scholar] [CrossRef] [Green Version]
  128. Kenney, L.B.; Ames, B.; Margossian, R.; Moss, K.; Michaud, A.L.; Williams, D.N.; Nohria, A. Regional practice norms for the care of childhood cancer survivors at risk for cardiomyopathy: A Delphi study. Pediatr. Blood Cancer 2019, 66, e27868. [Google Scholar] [CrossRef]
  129. Ryan, T.D.; Border, W.L.; Baker-Smith, C.; Barac, A.; Bock, M.J.; Canobbio, M.M.; Choueiter, N.F.; Chowdhury, D.; Gambetta, K.E.; Glickstein, J.S.; et al. The landscape of cardiovascular care in pediatric cancer patients and survivors: A survey by the ACC Pediatric Cardio-Oncology Work Group. Cardio-Oncology 2019, 5, 16. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  130. Halliday, B.P.; Wassall, R.; Lota, A.S.; Khalique, Z.; Gregson, J.; Newsome, S.; Jackson, R.; Rahneva, T.; Wage, R.; Smith, G.; et al. Withdrawal of pharmacological treatment for heart failure in patients with recovered dilated cardiomyopathy (TRED-HF): An open-label, pilot, randomised trial. Lancet 2018, 393, 61–73. [Google Scholar] [CrossRef] [Green Version]
  131. Bianco, C.M.; Al-Kindi, S.G.; Oliveira, G.H. Advanced Heart Failure Therapies for Cancer Therapeutics-Related Cardiac Dys-function. Heart Fail Clin 2017, 13, 327–336. [Google Scholar] [CrossRef]
  132. Arico, M.; Pedroni, E.; Nespoli, L.; Vigano, M.; Porta, F.; Burgio, G.R. Long term survival after heart transplantation for doxorubicin induced cardiomyopathy. Arch. Dis. Child. 1991, 66, 985–986. [Google Scholar] [CrossRef] [Green Version]
  133. McManus, R.P.; O’Hair, D.P. Pediatric heart transplantation for doxorubicin-induced cardiomyopathy. J. Heart Lung Transpl. 1992, 11, 375–376. [Google Scholar]
  134. Mehra, M.R.; Canter, C.E.; Hannan, M.M.; Semigran, M.J.; Uber, P.A.; Baran, D.A.; Danziger-Isakov, L.; Kirklin, J.K.; Kirk, R.; Kushwaha, S.S.; et al. The 2016 International Society for Heart Lung Transplantation listing criteria for heart transplantation: A 10-year update. J. Heart Lung Transpl. 2016, 35, 1–23. [Google Scholar] [CrossRef]
  135. Musci, M.; Loebe, M.; Grauhan, O.; Weng, Y.; Hummel, M.; Lange, P.; Hetzer, R. Heart transplantation for doxorubicin-induced congestive heart failure in children and adolescents. Transpl. Proc. 1997, 29, 578–579. [Google Scholar] [CrossRef]
  136. Bock, M.J.; Pahl, E.; Rusconi, P.G.; Boyle, G.J.; Parent, J.J.; Twist, C.J.; Kirklin, J.K.; Pruitt, E.; Bernstein, D. Cancer recurrence and mortality after pediatric heart transplantation for anthracycline cardiomyopathy: A report from the Pediatric Heart Transplant Study (PHTS) group. Pediatr. Transpl. 2017, 21, e12923. [Google Scholar] [CrossRef] [PubMed]
  137. Ward, K.M.; Binns, H.; Chin, C.; Webber, S.A.; Canter, C.E.; Pahl, E. Pediatric heart transplantation for anthracycline cardiomyo-pathy: Cancer recurrence is rare. J. Heart Lung Transpl. 2004, 23, 1040–1045. [Google Scholar] [CrossRef] [PubMed]
  138. Mangat, J.S.; Rao, K.; Kingston, J.; Veys, P.; Amrolia, P.; Burch, M. Early Pediatric Anthracycline Cardiotoxicity: Managed by Serial Heart and Bone Marrow Transplantation. J. Heart Lung Transpl. 2007, 26, 658–660. [Google Scholar] [CrossRef] [PubMed]
  139. Menon, N.M.; Katsanis, E.; Khalpey, Z.; Whitlow, P. Pediatric secondary chronic myeloid leukemia following cardiac transplan-tation for anthracycline-induced cardiomyopathy. Pediatr Blood Cancer 2015, 62, 166–168. [Google Scholar] [CrossRef]
  140. Cavigelli-Brunner, A.; Schweiger, M.; Knirsch, W.; Stiasny, B.; Klingel, K.; Kretschmar, O.; Hubler, M. VAD as bridge to recovery in anthracycline-induced cardiomyopathy and HHV6 myocarditis. Pediatrics 2014, 134, e894–e899. [Google Scholar] [CrossRef] [Green Version]
  141. Sayin, O.A.; Ozpeker, C.; Schoenbrodt, M.; Oz, F.; Borgermann, J.; Gummert, J.; Morshuis, M. Ventricular assist devices in patients with chemotherapy-induced cardiomyopathy: New modalities. Acta Cardiol. 2015, 70, 430–434. [Google Scholar] [CrossRef]
  142. Appel, J.M.; Sander, K.; Hansen, P.B.; Moller, J.E.; Krarup-Hansen, A.; Gustafsson, F. Left ventricular assist device as bridge to re-covery for anthracycline-induced terminal heart failure. Congest Heart Fail 2012, 18, 291–294. [Google Scholar] [CrossRef] [PubMed]
  143. Thomas, G.R.; McDonald, M.A.; Day, J.; Ross, H.J.; Delgado, D.H.; Billia, F.; Butany, J.W.; Rao, V.; Amir, E.; Bedard, P.L. A Matched Cohort Study of Patients With End-Stage Heart Failure from Anthracycline-Induced Cardiomyopathy Re-quiring Advanced Cardiac Support. Am. J. Cardiol. 2016, 118, 1539–1544. [Google Scholar] [CrossRef] [PubMed]
  144. Oliveira, G.H.; Dupont, M.; Naftel, D.; Myers, S.L.; Yuan, Y.; Tang, W.H.; Gonzalez-Stawinski, G.; Young, J.B.; Taylor, D.O.; Starling, R.C. Increased need for right ventricular support in patients with chemotherapy-induced cardiomyopathy undergoing mechan-ical circulatory support: Outcomes from the INTERMACS Registry (Interagency Registry for Mechanically Assisted Circula-tory Support). J. Am. Coll. Cardiol. 2014, 63, 240–248. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Krasnopero, D.; Asante-Korang, A.; Jacobs, J.P.; Stapleton, S.; Carapellucci, J.; Dotson, M.; Stapleton, G. Case report and review of the literature: The utilisation of a ventricular assist device as bridge to recovery for anthracycline-induced ventricular dysfunction. Cardiol. Young 2017, 28, 471–475. [Google Scholar] [CrossRef] [PubMed]
Table 1. Cancer therapies associated with cardiovascular toxicity.
Table 1. Cancer therapies associated with cardiovascular toxicity.
Treatment AgentPotential Cardiovascular Toxicity
AnthracyclinesVentricular dysfunction/heart failure
RadiationVentricular dysfunction/heart failure
Valvular disease
Pericardial disease
Ischemic vascular disease/coronary artery disease Arrhythmias
Tyrosine kinase and Vascular endothelial growth factor inhibitorsVentricular dysfunction/heart failure
Hypertension
Pulmonary hypertension
Ischemic vascular disease/coronary artery disease
Thromboembolism
QT Prolongation
HER2-targeted agentsVentricular dysfunction/heart failure
Immune checkpoint inhibitorsMyocarditis
Arrhythmia
CAR-T cell therapyVentricular dysfunction
Cytokine release syndrome-related hypotension
Alkylating agentsVentricular dysfunction/heart failure
Thromboembolism
Platinum-based agentsVentricular dysfunction/heart failure
Ischemic vascular disease/coronary artery disease
Thromboembolism
Proteasome inhibitorsVentricular dysfunction/heart failure
AntimetabolitesIschemic vascular disease/coronary artery disease
Microtubule inhibitorsArrhythmia
Ischemic vascular disease/coronary artery disease
Other
Thalidomide and analogs
arsenic
Arrhythmia; Thromboembolism
QT Prolongation
Table 2. Patient and treatment risk factors in the development of cancer treatment-related cardiotoxicity in patients treated for pediatric cancer.
Table 2. Patient and treatment risk factors in the development of cancer treatment-related cardiotoxicity in patients treated for pediatric cancer.
Risk Factors
Patient-RelatedTreatment-Related
Younger age (especially <5 years of age)Total cumulative anthracycline dose **
Female genderChest radiation ***
African American raceTime since treatment
Trisomy 21Pre-modern radiation protocols (before 1975)
Cardiovascular risk factors (hypertension, hyperlipidemia, diabetes, obesity)Concomitant therapy with cyclophosphamide, bleomycin, vincristine, amsacrine, mitoxantrone, immunotherapy
Underlying heart disease (congenital heart disease, cardiomyopathy)
Genetic factors *
* Multiple genotypes identified as risk factors. ** Dose cut-off frequently cited as >250 mg/m2 doxorubicin equivalent. *** Dose cut-off frequently cited as >15–30 Gy chest radiation.
Table 3. Resources providing information and/or guidance for cardiovascular care of survivors of pediatric cancers.
Table 3. Resources providing information and/or guidance for cardiovascular care of survivors of pediatric cancers.
Resource
American Heart Association Scientific Statement on Pediatric, Adolescent, and Young Adult Long-Term Survivors [6]
Children’s Oncology Group (www.childrensoncologygroup.org)
National Comprehensive Cancer Network (nccn.org)
Dutch Childhood Oncology Group [112]
Scottish Intercollegiate Guidelines Network (www.sign.ac.uk)
UK Children’s Cancer and Leukaemia Group (www.cclg.org.uk)
International Late Effects of Childhood Cancer Guideline Harmonization Group [113]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Brickler, M.; Raskin, A.; Ryan, T.D. Current State of Pediatric Cardio-Oncology: A Review. Children 2022, 9, 127. https://doi.org/10.3390/children9020127

AMA Style

Brickler M, Raskin A, Ryan TD. Current State of Pediatric Cardio-Oncology: A Review. Children. 2022; 9(2):127. https://doi.org/10.3390/children9020127

Chicago/Turabian Style

Brickler, Molly, Alexander Raskin, and Thomas D. Ryan. 2022. "Current State of Pediatric Cardio-Oncology: A Review" Children 9, no. 2: 127. https://doi.org/10.3390/children9020127

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop