Cardiovascular side effects of cancer treatments

Abstract
The cardiovascular side effects of cancer treatments remain a challenge in oncologic care. Patients with cancer and cancer survivors have an increased risk of adverse cardiovascular outcomes, including left ventricular (LV) dysfunction, heart failure (HF) and acute coronary events. The treatments most frequently associated with cardiovascular side effects include anthracyclines, monoclonal antibodies (mAbs) targeting the HER2 pathway, and small molecule tyrosine kinase inhibitors (TKIs), in particular vascular endothelial growth factor (VEGF)-signaling pathway (VSP) inhibitors. This review article focuses on the incidence and pathologic mechanisms of LV dysfunction observed with the most commonly implicated anticancer agents; summarises the existing clinical data on diagnosis, prevention and management of cardiac dysfunction related cancer therapeutics; and provides commentary on the cardiovascular risk associated with radiation, which may be used in conjunction with chemotherapy and biological therapies as part of multimodality cancer treatment.
Keywords: Anthracycline, cancer, cardio-oncology, cardiotoxicity, chemotherapy, doxorubicin, radiation, trastuzumab, tyrosine kinase inhibitor.
Original submitted: 23 May 2016; Revised submitted: 13 July 2016; Accepted for publication: 27 July 2016; Published online: 08 September 2016

Key points:

  • With advances in treatment and improved cancer survival rates cardiovascular safety of oncology therapeutics remains an important clinical challenge.
  • Novel targeted therapeutics are often combined with conventional chemotherapy and have the potential to increase cardiotoxic effects.
  • An increasing number of research studies is advancing prediction and treatment approaches to cardiotoxicity.

Introduction

The cardiovascular side effects of cancer treatments remain a challenge in oncologic care. Patients with cancer and cancer survivors have an increased risk of adverse cardiovascular outcomes, including left ventricular (LV) dysfunction, heart failure (HF) and acute coronary syndromes. These events are often a result of the cardiovascular toxicity of different cancer treatments and their synergism with cardiovascular risk factors and pre-existing cardiovascular disease.

LV dysfunction and HF have been linked to many cancer therapeutics and incidence reports vary widely depending on the agent, definition, diagnostic methods and population[1]
. The treatments most frequently associated with cardiovascular side effects include anthracyclines, monoclonal antibodies (mAbs) targeting the HER2 pathway and small molecule tyrosine kinase inhibitors (TKIs), in particular vascular endothelial growth factor (VEGF)-signaling pathway (VSP) inhibitors[2]
.

Anthracycline therapy and cancer treatment-related cardiac dysfunction

For decades, anthracyclines have been a core component of chemotherapy protocols for many malignancies. The UK Medicine and Healthcare products Regulatory Agency (MHRA), an executive agency of the Department of Health, the European Medicines Agency, a European Union agency for the evaluation of medicinal products, and the US Food and Drug Administration (FDA), the regulator for medicines in the United States, have similar labelling, which states that doxorubicin is indicated for several disseminated neoplastic conditions, including acute lymphoblastic leukaemia, breast carcinoma, ovarian carcinoma, sarcomas and malignant lymphoma[3]
,[4]
,[5]
. For more than 40 years it has been recognised that doxorubicin therapy causes dose-related cardiotoxicity, which commonly manifests as LV systolic dysfunction with or without clinical HF symptoms[6]
. Other anthracyclines also exhibit cumulative cardiotoxicity, consistent with a drug class effect. However, data supporting a specific relationship between dose and LV dysfunction are far less rigorous. Daunorubicin, epirubicin and idarubicin have been reported to be less cardiotoxic than doxorubicin but these studies remain limited and controversial[7]
,[8]
,[9]
. An additional challenge is that cardiotoxicity comparisons need to be made in oncologically equivalent dosages required to achieve similar myelosuppression levels (e.g. epirubicin has 0.67 relative myelosuppression potency compared with doxorubicin and its dose [mg/m2 per cycle] used in oncology protocols is usually about one third higher than the doxorubicin dose).

Historically, direct oxidative damage was given as the main explanation for the myocardial dysfunction observed after the use of anthracyclines[10]
; however, recently published data suggest that topoisomerase 2-beta-associated DNA damage[11]
and mitochondrial iron accumulation are key mechanisms leading to cellular injury and necrosis[12]
. This evidence highlights the relevance of multiple pathways, while simultaneously providing information on the protective mechanisms of agents such as dexrazoxane, as well as potential novel targets[13]
.

Genetic polymorphisms have also been studied, particularly in childhood cancer survivor cohorts, to understand the variability of doxorubicin-induced cardiotoxicity. Several gene variants involved in oxidative stress signalling and iron metabolism have been identified as potential markers of susceptibility[14]
,[15]
but prospective studies are awaited to lead implementation in clinical practice[16]
. These investigations continue to provide insights into new gene functions, such as the role of retinoic acid receptorγ (RARγ) gene; in a recent genome-wide association study, its nonsynonymous variant was found to be strongly associated with anthracycline-induced cardiotoxicity, which was explained by its actions on altering RARγ function and derepression of topoisomerase 2-beta[17]
.

The clinical paradigm of the history of doxorubicin cardiotoxicity has also been challenged. The established concept for many years was of discrete and unrelated presentations of early (during or within one year of completion of therapy) and late cardiotoxicity (occurring many years after the completion of treatment, without evident structural changes prior)[18]
. However, trials utilising early imaging demonstrated that the vast majority of incident anthracycline cardiotoxicity may be apparent within the first year of therapy with active echocardiography screening[19]
. Full understanding of the incidence and natural history of LV dysfunction and HF related to anthracyclines in survivors of adult cancers is limited by the lack of long-term cardiovascular data in these patients. Reports from adult survivor cohorts of paediatric malignancies indicate a high risk of long-term anthracycline-induced cardiotoxicity in children, and show that age at the time of cancer treatment, type of malignancy and dose of anthracycline play an important role[20]
. In contrast, acute clinical doxorubicin cardiotoxicity seems to be rare[21]
and recent reports implicate a pathologic mechanism, similar to stress-induced cardiomyopathy[22]
. A meta-analysis of 22,815 patients receiving anthracyclines with a median nine-year follow-up demonstrated that 6% of patients had cardiotoxicity with clinically apparent HF and 18% of patients had subclinical toxicity[23]
. In this meta-analysis, the greatest risk factor for cardiotoxicity was cumulative anthracycline dose, although chest radiotherapy, African-American ethnicity, very young or very old age, diabetes, hypertension, very high or very low body weight, and severe comorbidities were all additional independent risk factors on multivariate analysis. While left-sided HF symptoms and reduced LV systolic function remain the most common clinical presentations of anthracycline-induced cardiotoxicity, recent imaging studies reported impairment of right ventricular (RV) performance parameters after anthracycline treatment, suggesting global cardiotoxic effects on cardiac muscle[24]
,[25]
. However, the clinical relevance of these data needs to be determined in prospective studies with dedicated assessment of RV function and outcomes.

Defining anthracycline-induced cardiotoxicity

Earlier definitions for anthracycline-induced cardiotoxicity focused on the clinical signs and symptoms of HF. However, clinically asymptomatic decreases in LV ejection fraction (LVEF) have also been included in recent Common Terminology Criteria for Adverse Events (CTCAE) and clinical trials[26]
. At the same time, the pathogenesis of cardiomyopathy and the progression of HF is being increasingly investigated and the knowledge applied to the setting of cardiovascular toxicities of cancer therapies. The 2013 American College of Cardiology (ACC)/American Heart Association (AHA) HF guidelines recognise anthracycline-associated injury as a risk for development of HF (stage A) that may progress to structural changes and decline in heart function (stage B), leading to the development of signs and symptoms of HF (stages C and D)[27]
.

Biomarkers

The concepts of early myocardial injury and cardiotoxicity as progressive disease have also advanced the science of injury biomarkers as tools for risk stratification and therapy considerations. Biomarkers, in particular cardiac troponins, have been investigated to aid the identification of patients at risk of cardiomyopathy, target cardioprotective therapies and optimise cardiac monitoring. In patients receiving anthracycline-based chemotherapy, Cardinale et al. demonstrated that both early (within three days of doxorubicin infusion) and late (four weeks later) troponin elevations identified a group of patients at highest risk of cardiac events and persistent reduction in LV ejection fraction (EF) at three years[28]
. Similarly, Ky et al. reported changes in high sensitivity troponin that predicted development of cardiotoxicity in patients with breast cancer after anthracycline-containing chemotherapy[29]
.

Natriuretic peptides, measured by both B-type (BNP) and N-terminal proBNP (NT-proBNP) assays, demonstrated associations with the development of LV dysfunction following doxorubicin therapy in some studies[30]
,[31]
, but not others[32]
.

Several biomarkers are under investigation as predictors of cardiotoxicity, including but not limited to C-reactive protein (CRP), myeloperoxidase (MPO), growth differentiation factor 15 (GDF-15), placental growth factor (PlGF) and cardiac specific fatty acids[33]
. However, at present, they remain only an important research tool to understand anthracycline-associated cardiovascular injury and myocardial processes that may contribute to the development and progression of cancer-treatment associated cardiomyopathy. In the future, biomarker assays should be standardised and require validation in large prospective, multicentre clinical trials before they can be applied and successfully used to direct clinical cardio-oncology practice.

Cardiac imaging

Various modalities of cardiac imaging are used to monitor the occurrence of potential cardiotoxic effects during chemotherapy administration. Transthoracic echocardiography is the most commonly used. The majority of early studies of anthracycline-associated cardiotoxicity used multigated acquisition scan (MUGA) assessment of LVEF because this older technique has the advantage of higher-reproducibility of 3D assessment of LV volumes compared with 2D echocardiograms[34]
. Although radionuclide imaging continues to be used, in particular with patients with poor echocardiographic windows, advances in echocardiographic imaging and its comprehensive assessment of cardiac structure and function, including pericardial, valvular and RV pathology, have made echocardiography the most common choice for initial assessment in patients undergoing cancer therapies[35]
. Cardiac magnetic resonance (CMR) imaging is another important technique, since it is considered the reference standard for evaluation of ventricular volumes and function and may represent a valuable resource in patients with suspected changes in LV function if echocardiographic data are equivocal. In addition to higher accuracy in the assessment of LV function, CMR offers the advantage of tissue characterisation that may provide an insight into the aetiology of a cardiomyopathy[36]
.

While LVEF continues to represent the key cardiac function parameter used to define cardiotoxicity and make clinical decisions, there has also been a focus on identifying novel cardiac imaging parameters that could overcome the inherent limitations of LVEF (i.e. primarily late occurrence of LVEF decline after the onset of myocardial injury, ventricular load dependence, and lack of ability to reflect regional changes in cardiac function)[37]
. Myocardial deformation, or myocardial strain, that can be measured from standard 2D echocardiographic images using a speckle tracking technique, has been investigated in patients undergoing serial imaging while receiving cancer treatment[38]
. In these patients, absolute decreases in global longitudinal strain (expressed as a percentage of myocardial shortening in longitudinal direction during systole) predicted later decreases in LVEF, indicating that change in myocardial strain may represent an early marker of myocardial injury during imaging [38]
,[39]
. Based on these studies, the American Society of Echocardiography (ASE) and the European Association of Cardiovascular Imaging (EACVI) recommend utilising speckle-tracking echocardiography-derived strain imaging for the evaluation of subclinical LV systolic dysfunction in patients at risk of cancer-treatment related cardiomyopathy[40]
. The ASE/EACVI consensus document also emphasises the importance of comprehensive, standardised echocardiographic examination, including 3D measurements, with attention to test-retest variability in patients undergoing serial imaging and consideration of other techniques, such as CMR, when images are of limited quality.

Prevention and treatment

While monitoring for cardiotoxicity is essential for clinical care, preventing cardiotoxicity in the first place would be optimal. The mainstay of cardiotoxicity prevention has focused for many years on minimising the overall dose exposure to anthracyclines resulting in modifications of oncology regimens and alterations in dosing schedule[41]
. The US FDA drug labelling states that incident impaired myocardial function is dependent on total cumulative dose, with 1–2% at a dose of 300mg/m2, 3–5% at 400mg/m2, 5–8% at 450mg/m2, and 6–20% at 500mg/m2[3]
. While the European Medicines Agency does not report these data for epirubicin, which is more commonly used in European oncology clinical practices, its cardiotoxicity risk for oncologically equivalent dosages is considered to be similar to doxorubicin[7]
,[8]
,[9]
.

Slow infusion lasting longer than six hours (compared to bolus dosing)[42]
and liposomal preparation of doxorubicin have been shown to reduce the risk of developing heart failure[43]
and have been applied in the treatment of tumours often requiring larger cumulative doses, such as sarcomas. While bolus dosing of doxorubicin may lead to higher peak plasma levels and appears to increase the risk of cardiotoxicity in adults[7]
, there is no clear evidence for long-term cardioprotection with slower doxorubicin infusion in children[44]
. Liposomal doxorubicin has a much longer half-life than doxorubicin and a smaller volume of distribution, and therefore accumulates more selectively in abnormal, cancerous tissue[45]
,[46]
. Dexrazoxane is an intravenously-administered iron chelator that has been extensively studied for prevention of anthracycline-induced cardiotoxicity. A recent randomised, multi-year follow-up study of 537 paediatric and adolescent patients demonstrated a distinct difference in LV function and wall thickness in patients receiving dexrazoxane without compromising treatment efficacy[47]
. The Cochrane meta-analysis pooling of 1,619 patients enrolled in dexrazoxane-randomised trials demonstrated a decreased occurrence of HF in patients receiving dexrazoxane preparation prior to anthracycline treatment[48]
. The protective actions of dexrazoxane were attributed to its iron chelating effect and prevention of oxygen radical formation through reduction of doxorubicin-iron complex. However, novel mechanisms, in particular binding and inhibition of topoisomerase 2-beta, have been proposed to account for its cardioprotective effects[49]
,[50]
. Doxorubicin activity leads to increased levels of topoisomerase 2-beta-DNA covalent complexes and breakage of DNA strands[51]
. Variance in topoisomerase 2-beta levels and genetic polymorphisms, particularly in p53, Chk2 and Top2a, may predispose to sensitivity or resistance to doxorubicin therapy[52]
. Similarly, recent animal studies implicate dexrazoxane-induced depletion of topoisomerase 2-beta as a potential protective mechanism against anthracycline-induced cardiotoxicity[53]
.

Despite its apparent effectiveness, use of dexrazoxane has not been widely adopted in clinical practice because cost, need for intravenous administration, and questions of evidence of its benefit with specific lower anthracycline dose regimens represent barriers. It is worth noting that neither the 2008 American Society of Clinical Oncology (ASCO) nor the 2012 European Society of Medical Oncology (ESMO) guidelines recommend the routine use of dexrazoxane, while the 2013 ACC/AHA heart failure guidelines identify dexrazoxane as cardioprotective[27]
,[54]
,[55]
.

Another preventative approach for cancer treatment-related cardiomyopathy includes the use of cardiovascular agents approved to treat HF and coronary artery disease. Based on the concepts of early injury and progressive adverse LV remodelling, several studies have investigated the potential of neurohormonal blockade to prevent anthracycline-induced cardiomyopathy using angiotensin-converting enzyme (ACE) inhibitors, angiotensin receptor blockers (ARBs) and beta blockers (BBs)[56]
. The results of small studies using ACE inhibitors have been mixed, with some[57]
but not all trials demonstrating benefit[58]
,[59]
. Recent primary prevention trials using ARBs[60]
, BBs[61]
,[62]
or combination ACE inhibitors/BBs,[63]
concomitant with anthracycline treatment have demonstrated benefit, mostly measured by preserved LVEF, compared with patients receiving placebo. Similarly, preclinical studies and a small, randomised trial of 40 patients reported favourable effects of statins in patients receiving anthracycline therapy[64]
. Oral administration, availability and good safety data (extrapolated from other populations) make these agents favourable candidates for cardioprevention. However, their validation in larger, prospective, multicentre trials with longer follow-up is required to allow translation into clinical practice[65]
. This approach is currently being applied in the example of statins with a multicentre, National Cancer Institute-supported trial testing the hypothesis that, in patients receiving anthracycline therapy for breast cancer and lymphoma, prophylactic administration of atorvastatin attenuates changes in LV remodelling and prevents decrease in LVEF (NCT01988571).

Treatment of anthracycline-induced cardiotoxicity

The traditional treatment of patients with symptomatic HF has focused on avoiding further chemotoxicity and initiating standard HF therapy, including ACE inhibitors and BBs. The US FDA drug label for doxorubicin recommends discontinuation in patients who develop HF[3]
. Similarly, the 2012 European Society of Cardiology (ESC) HF guidelines recommend that patients who develop systolic dysfunction do not receive further anthracycline-based treatment[66]
. Management of patients with asymptomatic reduction in LVEF is an area of active investigation and requires individualisation with the patient, oncologists and cardiologists[67]
. One trial implemented active screening in 2,625 patients receiving anthracyclines, followed by initiation of HF pharmacotherapy for an LVEF decrease of more than ten points or to less than 50%, and demonstrated full recovery of LV function in 11% of patients and partial recovery in 71% of patients[19]
. These data suggest a benefit of cardiac imaging for early detection of cardiotoxicity in patients receiving anthracycline treatment that has not been part of clinical practice in the United States or Europe, with the exception of patients receiving HER2-targeted therapies. Notably, there is little evidence for treatment of anthracycline-induced cardiotoxicity in survivors of childhood cancer[68]
, and treatment with ACE inhibitors and BBs is extrapolated to all patients with reduced systolic function, although efficacy data in these populations remain limited.

HER2-targeted therapies and LV dysfunction

Development of trastuzumab, a humanised mAb that targets the HER2 receptor, has revolutionised treatment of HER2-positive breast cancer and improved survival in patients with metastatic and adjuvant disease[69]
,[70]
. However, cardiac dysfunction is the main concern with trastuzumab use and ASCO clinical practice guidelines list decreased LVEF or clinical congestive HF as the single most important contraindication to trastuzumab therapy[71]
, while ESMO guidelines similarly state that patient selection should be founded on baseline cardiac function[72]
. In the seminal trial in patients with metastatic breast cancer, cardiac dysfunction developed in 27% of participants on trastuzumab and doxorubicin, 13% on trastuzumab and paclitaxel, and only 1% on paclitaxel monotherapy, an unexpected and alarming finding. Despite this, trastuzumab was associated with improved overall survival[69]
. Although not fully determined, the proposed mechanism for the cardiotoxicity of trastuzumab includes functional blockade of the ErbB2/HER2 pathway, which impairs normal cellular repair[73]
. In animal models, ErbB2 overexpression may protect against doxorubicin cardiotoxicity, which provides hypothetical support for increased cardiac risk with concurrent doxorubicin and trastuzumab[74]
. This led to important changes in the design of subsequent clinical trials that resulted in the approval of trastuzumab for adjuvant treatment of HER2 positive breast cancer[75]
. In these studies, drug administration was changed (with trastuzumab given only after, never concomitantly with, anthracyclines), and monitoring of LVEF was mandated prior to and during one year of trastuzumab treatment. Patients with a decrease in LVEF below specified cut-offs were not eligible to continue trastuzumab therapy. In this new design setting, the rate of symptomatic HF was much lower, ranging from 3% to 6%, and most of the events were asymptomatic LV dysfunction, reported in around 17% of patients[70],
[
76]
,[77]
. Seven-year follow-up of one of the largest trials demonstrated that the majority of affected patients had recovery of LV function after holding or discontinuation of trastuzumab[78]
. A meta-analysis of 12,000 patients with early breast cancer noted relative risks of 5.1 and 1.8 for the development of congestive HF and LV dysfunction respectively, but improved breast cancer free survival and overall survival with trastuzumab therapy[79]
. Two other currently approved HER2 targeted therapies, pertuzumab and ado-trastuzumab emtansine, have similar recommendations for cardiac function monitoring and therapy holding or discontinuation in patients with LVEF below normal values.

Detection of HER2 therapy-associated cardiotoxicity

Cardiac imaging is central to the detection of HER2 therapy-associated cardiotoxicity. Following the design of adjuvant trials, routine LVEF assessment has been recommended, including a transthoracic echocardiogram every three months during the first year of trastuzumab therapy[80]
. Other modalities, including MUGA and CMR, can also be used to monitor LVEF, particularly in patients who have difficult echocardiographic windows. Superior accuracy in the assessment of LV volumes, visualisation of all cardiac structures and lack of radiation favour CMR when available[40]
. Given the importance of LVEF decreases, which may lead to discontinuation of HER2 targeted therapy, several studies have tried to identify early markers of LV function using imaging and serum biomarkers in patients receiving trastuzumab. Global longitudinal strain assessment using speckle tracking echocardiography has evolved to be one of the most promising research tools, together with changes in biomarkers, for prediction of the risk of future LVEF decline, but their role in clinical practice has yet to be established[29]
,[40]
.

Novel preventive strategies to reduce HER2 therapy-related cardiotoxicity

Strict cardiac eligibility criteria and regulated withholding of trastuzumab treatment based on the changes in LVEF have reduced trastuzumab cardiotoxicity and raised questions about potential compromises in cancer outcomes to lower the risk of asymptomatic LV dysfunction[81]
. Several ongoing and two recent published trials have tried to overcome this challenge using a primary prevention approach with initiation of BB or ACE inhibitor/ARB therapy concomitantly with trastuzumab treatment. In the PRADA study, the early breast cancer patients undergoing adjuvant epirubicin-containing chemotherapy followed by trastuzumab who had been randomised to the candesartan arm had a small but statistically significant attenuation of LVEF decrease at the end of cancer treatment compared with placebo-treated patients[60]
. Metoprolol did not affect LVEF changes in this study, in contrast to the similarly designed MANTICORE trial, in which a different BB, bisoprolol, had a small advantage over perindopril in preventing decreases in LVEF at the end of trastuzumab treatment[82]
,[83]
. Of interest, both of these studies used CMR-measurements for the detection of changes in LV volumes and LVEF changes that constituted outcomes. There were no HF events and LVEF declines were small, many within normal LV function range or only mildly reduced LVEF, suggesting that higher risk patients have not been included in the studies. Despite their shortcomings, these investigations represent an important step forward in conducting interdisciplinary cardio-oncology research and managing cardiotoxicity. They also highlight gaps in our knowledge and the need for future studies to address questions about biological differences between BBs and angiotensin-pathway inhibitors on trastuzumab-induced cardiac toxicity, confounding effects of anthracycline therapy (used in all patients in the PRADA trial[60]
, but only in a portion of patients in MANTICORE[82]
,[83]
), and strategies for improved risk stratification to identify patients who may receive the highest benefit of primary prevention[84]
.

Treatment of trastuzumab-related cardiac toxicity

Holding or stopping therapy, as well as the initiation of standard HF regimens, are the main approaches in the treatment of trastuzumab-related cardiac toxicity, according to the extent of LV decline and symptoms[85]
. The majority of patients who experience reduced systolic function related to trastuzumab therapy have subsequent improvement in cardiac function[86]
,[87]
. This LVEF recovery has also been observed with prolonged therapy in a 200-patient case series from MD Anderson (Texas, United States), in which 28% of patients developed cardiac toxicity with a median of 21 months of trastuzumab therapy. Of note, 94% of these patients had improved LVEF or symptoms of HF with discontinuation of trastuzumab and appropriate medical therapy[88]
.

Synergism of trastuzumab and anthracycline cardiotoxicity

Given the beneficial efficacy of the combination therapy of anthracyclines and trastuzumab on oncologic outcomes, their potential synergism on cardiac toxicity is of particular concern. Concurrent therapy may increase the risk of cardiac toxicity more than either trastuzumab or anthracycline therapy alone[75]
. Trastuzumab interferes with innate cardiac myocyte survival mechanisms intended to counter cellular stressors, such as those induced by anthracyclines[89]
,[90]
. High incidence of HF in the initial trastuzumab trials in metastatic HER2-positive breast cancer (in which patients were given concomitant anthracyclines and trastuzumab) was in a large part attributed to this mechanism and has resulted in a change to drug administration in subsequent trials. Currently, standard anthracycline-containing regimens for HER2-positive breast cancer typically include initial treatment with doxorubicin or epirubicin-based standard chemotherapy followed by an assessment of LVEF prior to continuation of trastuzumab. Importantly, the concern for synergistic toxicity and decreases in LVEF after anthracyclines has focused interest on the use of non-anthracycline based treatment in patients with HER2-positive breast cancer: the recently presented ten-year follow-up of the landmark BCIRG-006 trial indicated comparable effects of doxorubicin plus cyclophosphamide followed by docetaxel and trastuzumab (AC-TH) versus a non-anthracycline regimen of docetaxel plus carboplatin and trastuzumab (TCH) on disease-free and overall survival[91]
. At the same time, the TCH regimen was associated with a significant improvement in long-term cardiac safety leading to an updated ASCO guideline recommendation for its preferential use in patients who may be at higher risk of cardiotoxicity[92]
, while ESMO guidelines state that TCH is an alternative to anthracycline-based chemotherapy in patients at high risk of cardiac complications[72]
.

Novel targeted therapeutics and cardiovascular toxicity

Therapeutic targeting of kinases overexpressed in cancer cells using mAbs and small molecule kinase inhibitors has become an effective and standard treatment for many cancers, indicated by an increasing number of newly approved agents for clinical use[93]
. Cardiovascular effects of these targeted therapies initially came as a surprise, urgently prompting novel models and research investigating the role of different kinases on the heart and cardiovascular homeostasis[94]
,[95]
,[96]
. Both on-target (related to the intended target cancer kinase) and off-target (related to unintended kinase inhibition) actions may result in toxicities[97]
, which, coupled with the fact that a single small molecule inhibitor may target more than one kinase, has resulted in a wide spectrum of cardiovascular effects to be observed[98]
. At the same time, multitargeting characteristics have made these agents attractive therapeutic options for different cancers, often as part of distinct chemotherapeutic regimens, increasing the complexity of presentations, analysis and interpretation of cardiovascular phenomena. Categorisation based on the (main) cellular pathway inhibition offers one potential approach and inhibitors targeting the vascular endothelium signalling pathway (VSP) deserve a particular mention. Small molecule inhibitors in this category act by intercellular binding to, and blocking of, vascular endothelial growth receptors (VEGFRs) and include sunitinib, sorafenib, imatinib, nilotinib, axitinib, pazopanib, ponatinib, and lenvatinib, which are currently approved in the United States and Europe, vandetanib and cabozantinib, which are approved in the United States, and several other molecules being tested in clinical trials[97]
. Bevacizumab and ramucirumab are mAbs that target circulating VEGFA and extracellular VEGFR2, respectively. Cardiovascular toxicities, including LV dysfunction and HF, hypertension and arterial and venous thrombosis have all been associated with this class of agents. Similarly, nilotinib carries a black box warning for QT prolongation and sudden death[99]
. The exact mechanisms of these heterogeneous adverse effects remain the subject of active investigation but the interference with the numerous roles of VEGF signalling in cardiovascular homeostasis is likely to represent the critical component[94]
,[97]
.

VSP inhibitor-associated cardiomyopathy

Clinical trials using VSP inhibitors did not include routine screening for LV dysfunction or clinical HF and reports about cardiomyopathy and vascular toxicities are mostly based on retrospective studies. A meta-analysis of 6,935 patients treated with sunitinib for renal cell and non-renal cell carcinoma reported an overall incidence of 4.1% for symptomatic HF and 1.5% for high-grade HF[100]
. Single centre observational data that captured asymptomatic dysfunction found significantly higher incidence, with up to 28% of patients experiencing LVEF decline of at least 10%[101]
. The risk of cardiomyopathy appears to be increased with sorefenib[102]
and possibly imatinib[103]
, although industry data suggest that the incidence of imatinib induced cardiomyopathy is rare[104]
. Data regarding the cardiac dysfunction risk of other small molecule VSP pathway inhibitors remain scarce. In the absence of prospective cardiovascular follow-up with clearly defined baseline status and measured cardiac function parameters, presented numbers are likely to represent an underestimation of the true incidence of cardiomyopathy.

Despite shared propensity to block VSP signalling, different agents do not act the same, instead preferentially inhibiting multiple different kinases. In clinical practice this means that consideration of switching from one to another VSP inhibitor in case of cardiotoxicity may offer a viable option[105]
.

Vascular effects

The most commonly reported vascular effect of VSP inhibitors is systemic hypertension. Reported incidence of new or uncontrolled hypertension was 24% for bevacizumab, 36% for pazopanib, 22% for sunitinib and 23% for sorafenib[97]
. A subset of these patients, 7.0% of whom were treated with sunitinib and 5.7% with sorafenib, developed high-grade hypertension leading to drug dose reduction and/or discontinuation[106]
,[107]
. One of the proposed mechanisms of VSP inhibitor-induced hypertension includes functional (and/or structural) changes in the endothelium that result in decreased availability of nitric oxide as a consequence of VEGF inhibition[108]
. In this model, endothelial cardiovascular effects and hypertension develop as a result of an on-target effect of the cancer drug and may reflect the strength of VEGF inhibitory activity. It is of interest that development of hypertension has been associated with improved tumour response and outcomes, and has increasingly been recognised as a biomarker of sunitinib efficacy[109]
.

Detection, prevention and treatment of cardiovascular toxicities of novel targeted cancer therapies

In the

absence of prospective studies to elucidate the development of cardiomyopathy related to novel targeted therapies, early detection and prevention remain

challenging. While professional standards for treatment of these patients await development, it is reasonable to extrapolate from other populations at high risk of HF. In clinical practice a comprehensive cardiovascular exam prior to initiation of VSP inhibitors is recommended with consideration of a baseline echocardiogram, particularly in patients with cardiovascular risk factors[94]
. Patients with existing risk factors should be treated and followed closely and this is particularly true for patients with hypertension who may be at risk of worsening blood pressure control. New symptoms or signs of HF should prompt an evaluation and if HF is diagnosed, the VSP inhibitor should be stopped and HF treatment initiated. Successful re-challenges with a different VSP inhibitor have been reported[105]
but are still limited to individual cases and decisions should be made on an individual basis, in consultation with the oncologist and cardiologist, based on the potential of oncologic benefit and severity of cardiac prognosis.

In patients receiving VSP inhibitors, blood pressure should be monitored at least weekly during the first six weeks of therapy[110]
. Most patients with cancer treatment-induced hypertension respond appropriately to standard antihypertensive medications, including ACE inhibitors, ARBs, diuretics, dihydropyridine calcium-channel blockers and BBs, although discontinuation of cancer targeted therapy may be necessary until hypertension is properly controlled[111]
. Non-dihydropyridine calcium channel blockers, such as verapamil and diltiazem, interfere with the metabolism of certain TKIs via the CYP3A4 system and should be used cautiously[110]
. There are also limited case report level data that indicate refractory hypertension may respond well to treatment with nitric oxide donors, such as isosorbide dinitrate, presumably by targeting the underlying etiology of the hypertension[112]
.

Coronary artery disease

Incident or progressive coronary artery disease causing myocardial infarction is another feared adverse effect of some oncologic therapy. 5-fluorouracil (5-FU) cardiotoxicity, predominantly manifesting as coronary vasospasm, has been apparent for nearly three decades[113]
,[114]
. The mechanism of 5-FU cardiotoxicity remains unclear, but it is likely to be multifactorial and partially caused by endothelial injury and an increased risk of coronary vasospasm[115]
. Coronary vasospasm manifests as sudden onset chest pain with ST-elevation on electrocardiogram and can be effectively treated with vasodilators, such as calcium channel blockers, in addition to cessation of 5-FU therapy[116]
. Capecitabine, a prodrug of 5-FU, has also been associated with coronary vasospasm and more rarely coronary arterial thrombosis[117]
. Capecitabine-induced vasospasm, like 5-FU, responds to discontinuation and coronary vasodilator therapy. There is limited evidence for resumption of capecitabine therapy with concurrent secondary prophylaxis with diltiazem[118]
that continues to be used in clinical practice. A patient-tailored approach is important and there must be close collaboration between the oncologist and cardiologist to determine the benefit of continuation of 5-FU or capecitabine treatment (with attention to survival benefit) as well as risk of coronary ischaemia. It is important to exclude or, if found, treat coronary artery disease and aggressively control cardiovascular risk factors, such as hypercholesterolaemia, smoking and hypertension. Monitored 5-FU infusions can also be considered when oncological benefit and/or cardiac risks are high.

Radiation

Therapy using radiation represents a core component of multiple cancer treatment regimens, such as for Hodgkin’s lymphoma and early stage breast cancer, and its adverse cardiac effects have been known for decades[119]
. Radiation-induced heart disease can manifest as pericarditis, pericardial fibrosis, diffuse myocardial fibrosis, coronary artery disease and valvular disease[120]
. Unfortunately, even in an asymptomatic population, such radiation-induced heart disease is relatively common[121]
. The mechanism of radiation-induced cardiotoxicity is multifactorial and varies with the specific manifestation but inflammation and vascular lesions leading to cellular death have been proposed as the main mechanisms[122]
. Risk factors for radiation cardiotoxicity include earlier age of therapy and radiation dose[123]
.

The key to prevention has been modifications in radiation technique, including the deep inspiration breath-holding and intensity modulated radiation therapy that varies the radiation energy while treatment is delivered, in order to precisely contour the desired radiation distribution and avoid normal tissues[55]
. A recently published document by the European Association of Cardiovascular Imaging and the American Society of Echocardiography summarises recommendations for detection to include a yearly clinical history and physical exam[124]
. Targeted imaging and an echocardiogram should be considered five to ten years after radiation exposure with low threshold for stress test in high-risk patients[124]
. Additional echocardiography markers, strain rate imaging and diastolic dysfunction may detect subclinical cardiac dysfunction after radiation therapy and are recommended as part of a comprehensive exam[125]
,[126]
. Radiation-induced heart disease is associated with increased long-term mortality and extensive valvular and coronary calcifications, mediastinal fibrosis and radiation-related lung disease, which makes surgical management of these patient challenging[127]
,[128]
.

Conclusions

Cancer therapeutics can cause numerous cardiovascular toxicities, most notably LV dysfunction and HF, which can interfere with the efficacy of cancer treatment, decrease quality of life and adversely impact patient survival. With the advancement of the oncology and cardiology fields, opportunities to recognise and treat cardiovascular toxicities without compromising cancer or cardiovascular outcomes are increasing. Oncologists, cardiologists and other healthcare professionals, as well as patients, need to be aware of the potential effects of old and novel cancer therapeutics and formulate a plan for the baseline assessment and monitoring of symptoms. The rapid growth in novel cancer therapeutics and inherent complexity of balancing oncologic and cardiac care has created an important and frequently unmet clinical need[129]
. Professional societies are increasingly responding to this need; the American College of Cardiology, for example, recently pioneered a new Cardio-Oncology member section[129]
and the International Cardioncology Society and Canadian Cardiac Oncology Network published a call for multidisciplinary cardio-oncology training[130]
,[131]
. Hopefully, the increasing availability of cardio-oncology specialists will promote multidisciplinary care, from basic science to phase I trials of cardiovascular safety, through to large scale phase III clinical trials and most importantly to ongoing patient-focused clinical care.

Standard guideline-driven cardiovascular risk factor management and treatment for the identified cardiac pathology is an important start. However, at present, it is mostly based on extrapolation from non-cancer populations and validation studies are urgently needed. The implementation of comprehensive cardiovascular care along the continuum of cancer treatment from diagnosis to survivorship is critical to ultimately improve patient outcomes across a spectrum of cardio-oncology conditions (see Table 1). Specific cardiovascular monitoring with new targeted therapeutics and inclusion of cardiovascular phenotypes in oncology clinical trials and registries will be of paramount importance. Furthermore, individualisation and the identification of patients at high risk who may benefit from (primary) prevention strategies remains an important challenge that will be answered through further collaborative cardio-oncology trials.

Table 1: Approach to cardiovascular care across cancer treatment continuum*
Therapeutic agent/modality Pre-treatment During treatment Survivorship
AnthracyclineComprehensive cardivascular exam and LVEF assessment to assess for HF. Address other cardiac risk factors.No routine LVEF surveillance until more than 300mg/m2 of doxorubicin (or equivalent dose of epirubicin), then another assessment should be considered.Comprehensive cardiovascular exam and close attention to potential cardiovascular symptoms. Address other cardiac risk factors and consider an echocardiogram within one year if one or more cardiac risk factors are present[132]
.
TrastuzumabComprehensive cardiovascular exam and LVEF assessment to assess for HF. Address other cardiac risk factors.LVEF assessment while on treatment (current FDA recommendation is every three months while on therapy)[133]
.
Comprehensive cardiovascular exam and close attention to HF symptoms. Address other cardiac risk factors. LVEF assessment may be considered[133]
.
VEGF inhibitorScreening for hypertension.Monitoring and treatment of hypertension.Limited data.
Chest radiationComprehensive cardiovascular exam and LVEF assessment to assess for HF. Address other cardiac risk factors.Clinical monitoring for signs or symptoms of cardiovascular disease.Comprehensive cardiovascular exam and close attention to potential cardiovascular symptoms. Address other cardiac risk factors. Screening echocardiogram at five years for high-risk patients and ten years for others. Functional stress-test after five to ten years in high-risk patients[124]
.
*Reflects current clinical practice and recommendations of the authors. Cardiac risk factors: hypertension, dyslipidaemia, diabetes mellitus, family history of cardiomyopathy, age >65 years, history of other cardiovascular morbidities (i.e. atrial fibrillation or coronary artery disease). LVEF: Left ventricular ejection fraction, HF: Heart failure. 

Benjamin Kenigsberg is cardiology fellow, Umberto Campia is section director, vascular medicine and scientific lead in vascular research and Ana Barac is director, cardio-oncology programme at MedStar Heart and Vascular Institute, 110 Irving street, NW, Ste. 1F1218, Washington DC, United States. Correspondence to: 
ana.barac@medstar.net

Financial and conflicts of interests dislosure:

Ana Barac is a cardiology PI on an investigator-initiated study funded by Genentech Inc. The authors have no other relevant affiliations or financial involvement with any organisation or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed. No writing assistance was utilised in the production of this manuscript.

  • This article was amended on 14 September 2016 to fix an error in the references

Reading this article counts towards your CPD

You can use the following forms to record your learning and action points from this article from Pharmaceutical Journal Publications.

Your CPD module results are stored against your account here at The Pharmaceutical Journal. You must be registered and logged into the site to do this. To review your module results, go to the ‘My Account’ tab and then ‘My CPD’.

Any training, learning or development activities that you undertake for CPD can also be recorded as evidence as part of your RPS Faculty practice-based portfolio when preparing for Faculty membership. To start your RPS Faculty journey today, access the portfolio and tools at www.rpharms.com/Faculty

If your learning was planned in advance, please click:

If your learning was spontaneous, please click:

References

[1] Shelburne N, Adhikari B, Brell J et al. Cancer treatment-related cardiotoxicity: current state of knowledge and future research priorities. J Natl Cancer Inst 2014;106(9). doi: 10.1093/jnci/dju232

[2] Yeh ET & Bickford CL. Cardiovascular complications of cancer therapy: incidence, pathogenesis, diagnosis, and management. J Am Coll Cardiol 2009;53:2231–2247. doi: 10.1016/j.jacc.2009.02.050

[3] US Food and Drug Administration. Doxorubicin Hydrochloride for Injection, USP. Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/050467s070lbl.pdf (accessed 8 April 2016).

[4] Medicines and Healthcare Products Regulatory Agency (MHRA). Doxorubicin 2mg/ml concentrate for solution for infusion (doxorubicin hydrochloride) 50mg in 25ml. Available at: http://www.mhra.gov.uk/home/groups/par/documents/websiteresources/con2024436.pdf (accessed 31 July 2016).

[5] European Medicines Agency. Caelyx EPAR summary for the public. Available at: http://www.ema.europa.eu/docs/en_GB/document_library/EPAR_-_Summary_for_the_public/human/000089/WC500020173.pdf (accessed 31 July 2016).

[6] Lefrak EA, Pitha J, Rosenheim S et al. A clinicopathologic analysis of adriamycin cardiotoxicity. Cancer 1973;32(2):302-314. doi: 10.1002/1097-0142(197308)32:2<302::AID-CNCR2820320205>3.0.CO;2-2

[7] Smith LA, Cornelius VR, Plummer CJ et al. Cardiotoxicity of anthracycline agents for the treatment of cancer: systematic review and meta-analysis of randomised controlled trials. BMC Cancer 2010;10:337. doi: 10.1186/1471-2407-10-337

[8] van Dalen EC, Michiels EM, Caron HN et al. Different anthracycline derivates for reducing cardiotoxicity in cancer patients. Cochrane Database Syst Rev 2010;(5):CD005006. doi: 10.1002/14651858.cd005006.pub3

[9] Li X, Xu S, Tan Y & Chen J. The effects of idarubicin versus other anthracyclines for induction therapy of patients with newly diagnosed leukaemia. Cochrane Database Syst Rev 2015;6:CD010432. doi: 10.1002/14651858.cd010432.pub2

[10] Villani F, Galimberti M, Zunino F et al. Prevention of doxorubicin-induced cardiomyopathy by reduced glutathione. Cancer Chemother Pharmacol 1991;28(5):365-369. doi: 10.1007/BF00685691

[11] Zhang S, Liu X, Bawa-Khalfe T et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med 2012;18:1639–1642. doi: 10.1038/nm.2919

[12] Ichikawa Y, Ghanefar M, Bayeva M et al. Cardiotoxicity of doxorubicin is mediated through mitochondrial iron accumulation. J Clin Invest 2014;124(2):617-630. doi: 10.1172/JCI72931

[13] Vejpongsa P & Yeh ET. Prevention of anthracycline-induced cardiotoxicity: challenges and opportunities. J Am Coll Cardiol 2014;64(9):938-945. doi: 10.1016/j.jacc.2014.06.1167

[14] Reichwagen A, Ziepert M, Kreuz M et al. Association of NADPH oxidase polymorphisms with anthracycline-induced cardiotoxicity in the RICOVER-60 trial of patients with aggressive CD20(+) B-cell lymphoma. Pharmacogenomics 2015;16(4):361-372. doi: 10.2217/pgs.14.179

[15] Wang X, Liu W, Sun CL et al. Hyaluronan synthase 3 variant and anthracycline-related cardiomyopathy: a report from the children’s oncology group. J Clin Oncol 2014;32(7):647-653. doi: 10.1200/JCO.2013.50.3557

[16] Chow EJ, Chen Y, Kremer LC et al. Individual prediction of heart failure among childhood cancer survivors. J Clin Oncol 2015;33(5):394-402. doi: 10.1200/JCO.2014.56.1373

[17] Aminkeng F, Bhavsar AP, Visscher H et al; Canadian Pharmacogenomics Network for Drug Safety Consortium. A coding variant in RARG confers susceptibility to anthracycline-induced cardiotoxicity in childhood cancer. Nat Genet 2015;47(9):1079-1084. doi: 10.1038/ng.3374

[18] Grenier MA & Lipshultz SE. Epidemiology of anthracycline cardiotoxicity in children and adults. Semin Oncol 1998;25:72–85.

[19] Cardinale D, Colombo A, Bacchiani et al. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation 2015;131(22):1981-1988. doi: 10.1161/CIRCULATIONAHA.114.013777

[20] van Dalen EC, van der Pal HJ, Kok WE et al. Clinical heart failure in a cohort of children treated with anthracyclines: a long-term follow-up study. Eur J Cancer 2006;42(18):3191-3198. doi: 10.1016/j.ejca.2006.08.005

[21] Hayek ER, Speakman E & Rehmus E. Acute doxorubicin cardiotoxicity. N Engl J Med 2005;352:2456-2457. doi: 10.1056/NEJM200506093522321

[22] Fernandez SF, Basra M & Canty JM Jr. Takotsubo cardiomyopathy following initial chemotherapy presenting with syncope and cardiogenic shock – a case report and literature review. J Clinic Experiment Cardiol 2011;2:124. doi: 10.4172/2155-9880.1000124

[23] Lotrionte M, Biondi-Zoccai G, Abbate A et al. Review and meta-analysis of incidence and clinical predictors of anthracycline cardiotoxicity. Am J Cardiol 2013;112(12):1980-1984. doi: 10.1016/j.amjcard.2013.08.026

[24] Grover S, Leong DP, Chakrabarty A 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 2013;168(6):5465-5467. doi: 10.1016/j.ijcard.2013.07.246

[25] Murbraech K, Holte E, Broch K et al. Impaired right ventricular function in long-term lymphoma survivors. J Am Soc Echocardiogr 2016:29(6):528-536. doi: 10.1016/j.echo.2016.02.014

[26] National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) Version 4.0. 2010. Available at: http://evs.nci.nih.gov/ftp1/CTCAE/CTCAE_4.03_2010-06-14_QuickReference_5x7.pdf%20 (accessed 21 May 2016)

[27] Yancy CW, Jessup M, Bozkurt B et al; American College of Cardiology Foundation; American Heart Association Task on Practice Guidelines. 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(16):e147-239. doi: 10.1016/j.jacc.2013.05.019

[28] Cardinale D, Sandri MT, Colombo A et al. Prognostic value of troponin I in cardiac risk stratification of cancer patients undergoing high-dose chemotherapy. Circulation 2004;109:2749-2754. doi: 10.1161/01.CIR.0000130926.51766.CC

[29] Ky B, Putt M, Sawaya H et al. Early increases in multiple biomarkers predict subsequent cardiotoxicity in patients with breast cancer treated with doxorubicin, taxanes, and trastuzumab. J Am Coll Cardiol 2014;63(8):809-816. doi: 10.1016/j.jacc.2013.10.061

[30] Caram ME, Guo C, Leja M et al. Doxorubicin-induced cardiac dysfunction in unselected patients with a history of early-stage breast cancer. Breast Cancer Res Treat 2015;152(1):163-172. doi: 10.1007/s10549-015-3454-8

[31] Romano S, Fratini S, Ricevuto E et al. Serial measurements of NT-proBNP are predictive of not-high-dose anthracycline cardiotoxicity in breast cancer patients. Br J Cancer 2011;105(11):1663-1668. doi: 10.1038/bjc.2011.439

[32] Dodos F, Halbsguth T, Erdmann E et al. Usefulness of myocardial performance index and biochemical markers for early detection of anthracycline-induced cardiotoxicity in adults. Clin Res Cardiol 2008;97(5):318-326. doi: 10.1007/s00392-007-0633-6

[33] Tian S, Hirshfield KM, Jabbour SK et al. Serum biomarkers for the detection of cardiac toxicity after chemotherapy and radiation therapy in breast cancer patients. Front Oncol 2014;4:277. doi: 10.3389/fonc.2014.00277

[34] Alexander J, Dainiak N, Berger HJ et al. Serial assessment of doxorubicin cardiotoxicity with quantitative radionuclide angiocardiography. N Engl J Med 1979;300(6):278-283. doi: 10.1056/NEJM197902083000603

[35] Bloom MW, Hamo CE, Cardinale D et al. Cancer therapy-related cardiac dysfunction and heart failure. Part 1: definitions, pathophysiology, risk factors, and imaging. Circ Heart Fail 2016;9(1):e002661. doi: 10.1161/CIRCHEARTFAILURE.115.002661

[36] Vasu S & Hundley WG. Understanding cardiovascular injury after treatment for cancer: an overview of current uses and future directions of cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2013;15:66. doi: 10.1186/1532-429X-15-66

[37] Yu AF & Ky B. Roadmap for biomarkers of cancer therapy cardiotoxicity. Heart 2016;102(6):425-430. doi: 10.1136/heartjnl-2015-307894

[38] Thavendiranathan P, Poulin F, Lim KD et al. Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: a systematic review. J Am Coll Cardiol. 2014;63(25 Pt A):2751-2768. doi: 10.1016/j.jacc.2014.01.073

[39] Sawaya H, Sebag IA & Plana JC. Assessment of echocardiography and biomarkers for the extended prediction of cardiotoxicity in patients treated with anthracyclines, taxanes, and trastuzumab. Circ Cardiovasc Imaging 2012;5(5):596-603. doi: 10.1161/CIRCIMAGING.112.973321

[40] Plana JC, Galderisi M, Barac A et al. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging 2014;15(10):1063-1093. doi: 10.1093/ehjci/jeu192

[41] Conway A, McCarthy AL, Lawrence P et al. The prevention, detection and management of cancer treatment-induced cardiotoxicity: a meta-review. BMC Cancer 2015;15:366. doi: 10.1186/s12885-015-1407-6

[42] van Dalen EC, van der Pal HJ & Kremer LC. Different dosage schedules for reducing cardiotoxicity in people with cancer receiving anthracycline chemotherapy. Cochrane Database Syst Rev 2016;3:CD005008. doi: 10.1002/14651858.cd005008.pub4

[43] Yamaguchi N, Fujii T, Aoi S et al. Comparison of cardiac events associated with liposomal doxorubicin, epirubicin and doxorubicin in breast cancer: a Bayesian network meta-analysis. Eur J Cancer 2015;51(16):2314-2320. doi: 10.1016/j.ejca.2015.07.031

[44] Lipshultz SE, Miller TL, Lipsitz SR et al. Continuous versus bolus infusion of doxorubicin in children with ALL: long-term cardiac outcomes. Pediatrics 2012;130(6):1003-1011. doi: 10.1542/peds.2012-0727

[45] O’Brien ME, Wigler N, Inbar M et al. Reduced cardiotoxicity and comparable efficacy in a phase III trial of pegylated liposomal doxorubicin HCl (CAELYX/Doxil) versus conventional doxorubicin for first-line treatment of metastatic breast cancer. Ann Oncol 2004;15(3):440-449. doi: 10.1093/annonc/mdh097

[46] Lyass O, Uziely B, Ben-Yosef R et al. Correlation of toxicity with pharmacokinetics of pegylated liposomal doxorubicin (Doxil) in metastatic breast carcinoma. Cancer 2000;89(5):1037-1047. doi: 10.1002/1097-0142(20000901)89:5<1037::AID-CNCR13>3.0.CO;2-Z

[47] Asselin BL, Devidas M, Chen L et al. Cardioprotection and safety of dexrazoxane in patients treated for newly diagnosed T-cell acute lymphoblastic leukemia or advanced-stage lymphoblastic Non-Hodgkin lymphoma: a report of the children’s oncology group randomized trial pediatric oncology group 9404. J Clin Oncol 2016;34(8):854-862. doi: 10.1200/JCO.2015.60.8851

[48] van Dalen EC, Caron HN, Dickinson HO et al. Cardioprotective interventions for cancer patients receiving anthracyclines. Cochrane Database Syst Rev 2011;(6):CD003917. doi: 10.1002/14651858.cd003917.pub4

[49] Hasinoff BB, Takeda K, Ferrans VJ et al. The doxorubicin cardioprotective agent dexrazoxane (ICRF-187) induces endopolyploidy in rat neonatal myocytes through inhibition of DNA topoisomerase II. Anticancer Drugs 2002;13(3):255-258. doi: 10.1097/00001813-200203000-00007

[50] Rao VA. Iron chelators with topoisomerase-inhibitory activity and their anticancer applications. Antioxid Redox Signal 2013;18(8):930-955. doi: 10.1089/ars.2012.4877

[51] Nitiss JL. Targeting DNA topoisomerase II in cancer chemotherapy. Nat Rev Cancer 2009;9(5):338-350. doi: 10.1038/nrc2607

[52] Burgess DJ, Doles J, Zender L et al. Topoisomerase levels determine chemotherapy response in vitro and in vivo. Proc Natl Acad Sci USA 2008;105(26):9053-9058. doi: 10.1073/pnas.0803513105

[53] Deng S, Yan T, Jendrny C et al. Dexrazoxane may prevent doxorubicin-induced DNA damage via depleting both topoisomerase II isoforms. BMC Cancer 2014;14:842. doi: 10.1186/1471-2407-14-842

[54] Hensley ML, Hagerty KL, Kewalramani T et al. American Society of Clinical Oncology 2008 clinical practice guideline update: use of chemotherapy and radiation therapy protectants. J Clin Oncol 2009;27(1):127-145. doi: 10.1200/JCO.2008.17.2627

[55] Curigliano G, Cardinale D, Suter T et al; ESMO Guidelines Working Group. Cardiovascular toxicity induced by chemotherapy, targeted agents and radiotherapy: ESMO Clinical Practice Guidelines. Ann Oncol 2012;23(7):vii155-166. doi: 10.1093/annonc/mds293

[56] Witteles RM & Bosch X. Myocardial protection during cardiotoxic chemotherapy. Circulation 2015;132(19):1835-1845. doi: 10.1161/CIRCULATIONAHA.114.010486

[57] Janbabai G, Nabati M, Faghihinia M et al. Effect of enalapril on preventing anthracycline-induced cardiomyopathy. Cardiovasc Toxicol 2016. doi: 10.1007/s12012-016-9365-z

[58] Georgakopoulos P, Roussou P, Matsakas E et al. Cardioprotective effect of metoprolol and enalapril in doxorubicin-treated lymphoma patients: a prospective, parallel-group, randomized, controlled study with 36-month follow-up. Am J Hematol 2010;85(11):894-896. doi: 10.1002/ajh.21840

[59] Cardinale D, Colombo A, Sandri MT et al. Prevention of high- dose chemotherapy-induced cardiotoxicity in high-risk patients by angiotensin-converting enzyme inhibition. Circulation 2006;114:2474–2481. doi: 10.1161/CIRCULATIONAHA.106.635144

[60] Gulati G, Heck SL, Ree AH et al. Prevention of cardiac dysfunction during adjuvant breast cancer therapy (PRADA): a 2 × 2 factorial, randomized, placebo-controlled, double-blind clinical trial of candesartan and metoprolol. Eur Heart J 2016;37(21):1671-1680. doi: 10.1093/eurheartj/ehw022

[61] Kaya MG, Ozkan M, Gunebakmaz O et al. Protective effects of nebivolol against anthracycline-induced cardiomyopathy: a randomized control study. Int J Cardiol 2013;167:2306–2310. doi: 10.1016/j.ijcard.2012.06.023

[62] El-Shitany NA, Tolba OA, El-Shanshory MR et al. Protective effect of carvedilol on adriamycin-induced left ventricular dysfunction in children with acute lymphoblastic leukemia. J Card Fail 2012;18(8):607-613. doi: 10.1016/j.cardfail.2012.06.416

[63] Bosch X, Rovira M, Sitges M et al. Enalapril and carvedilol for preventing chemotherapy-induced left ventricular systolic dysfunction in patients with malignant hemopathies: the OVERCOME trial (preventiOn of left Ventricular dysfunction with Enalapril and caRvedilol in patients submitted to intensive ChemOtherapy for the treatment of Malignant hEmopathies). J Am Coll Cardiol 2013;61(23):2355-2362. doi: 10.1016/j.jacc.2013.02.072

[64] Acar Z, Kale A, Turgut M et al. Efficiency of atorvastatin in the protection of anthracycline-induced cardiomyopathy. J Am Coll Cardiol 2011;58(9):988-989. doi: 10.1016/j.jacc.2011.05.025

[65] Kalam K & Marwick TH. Role of cardioprotective therapy for prevention of cardiotoxicity with chemotherapy: a systematic review and meta-analysis. Eur J Cancer 2013;49(13):2900-2909. doi: 10.1016/j.ejca.2013.04.030

[66] McMurray JJ, Adamopoulos S, Anker SD et al; ESC Committee for Practice Guidelines. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012: The Task Force for the Diagnosis and Treatment of Acute and Chronic Heart Failure 2012 of the European Society of Cardiology. Developed in collaboration with the Heart Failure Association (HFA) of the ESC. Eur Heart J 2012;33(14):1787-1847. doi: 10.1093/eurheartj/ehs104

[67] Hamo CE, Bloom MW, Cardinale D et al. Cancer therapy-related cardiac dysfunction and heart failure. Part 2: prevention, treatment, guidelines, and future directions. Circ Heart Fail 2016;9(2):e002843. doi: 10.1161/CIRCHEARTFAILURE.115.002843

[68] Sieswerda E, van Dalen EC, Postma A et al. Medical interventions for treating anthracycline-induced symptomatic and asymptomatic cardiotoxicity during and after treatment for childhood cancer. Cochrane Database Syst Rev 2011;(9):CD008011. doi: 10.1002/14651858.cd008011.pub2

[69] Slamon DJ, Leyland-Jones B, Shak S et al. Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 2001;344:783-792. doi: 10.1056/NEJM200103153441101

[70] Slamon DJ, Eiermann W, Robert N et al. Adjuvant trastuzumab in HER2-positive breast cancer. N Engl J Med 2011;365:1273-1283. doi: 10.1056/NEJMoa0910383

[71] Giordano SH, Temin S, Kirshner JJ et al; American Society of Clinical Oncology. Systemic therapy for patients with advanced human epidermal growth factor receptor 2-positive breast cancer: American Society of Clinical Oncology clinical practice guideline. J Clin Oncol 2014;32(19):2078-2099. doi: 10.1200/JCO.2013.54.0948

[72] Senkus E, Kyriakides S, Penault-Llorca F et al; ESMO Guidelines Working Group. Primary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol 2013;24(6):vi7-23. doi: 10.1093/annonc/mdt284

[73] Wadugu B & Kühn B. The role of neuregulin/ErbB2/ErbB4 signaling in the heart with special focus on effects on cardiomyocyte proliferation. Am J Physiol Heart Circ Physiol 2012;302(11):H2139-2147. doi: 10.1152/ajpheart.00063.2012

[74] Belmonte F, Das S, Sysa-Shah P et al. ErbB2 overexpression upregulates antioxidant enzymes, reduces basal levels of reactive oxygen species, and protects against doxorubicin cardiotoxicity. Am J Physiol Heart Circ Physiol 2015;309(8):H1271-1280. doi: 10.1152/ajpheart.00517.2014

[75] Seidman A, Hudis C, Pierri MK et al. Cardiac dysfunction in the trastuzumab clinical trials experience. J Clin Oncol 2002;20(5):1215-1221. doi: 10.1200/JCO.20.5.1215

[76] Smith KL, Dang C & Seidman AD. Cardiac dysfunction associated with trastuzumab. Expert Opin Drug Saf 2006;5(5):619-629. doi: 10.1517/14740338.5.5.619

[77] Telli ML, Hunt SA, Carlson RW et al. Trastuzumab-related cardiotoxicity: calling into question the concept of reversibility. J Clin Oncol 2007;25(23):3525-3533. doi: 10.1200/JCO.2007.11.0106

[78] Romond EH, Jeong JH, Rastogi P et al. Seven-year follow-up assessment of cardiac function in NSABP B-31, a randomized trial comparing doxorubicin and cyclophosphamide followed by paclitaxel (ACP) with ACP plus trastuzumab as adjuvant therapy for patients with node-positive, human epidermal growth factor receptor 2-positive breast cancer. J Clin Oncol 2012;30(31):3792-3799. doi: 10.1200/JCO.2011.40.0010

[79] Moja L, Tagliabue L, Balduzzi S et al. Trastuzumab containing regimens for early breast cancer. Cochrane Database Syst Rev 2012;4:CD006243. doi: 10.1002/14651858.cd006243.pub2

[80] Mackey JR, Clemons M, Côté MA et al. Cardiac management during adjuvant trastuzumab therapy: recommendations of the Canadian Trastuzumab Working Group. Curr Oncol 2008;15(1):24-35. doi: 10.3747/co.2008.199

[81] Dang CT, Yu AF, Jones LW et al. Cardiac surveillance guidelines for trastuzumab-containing therapy in early-stage breast cancer: getting to the heart of the matter. J Clin Oncol 2016;34(10):1030-1033. doi: 10.1200/JCO.2015.64.5515

[82] Pituskin E, Haykowsky M, Mackey JR et al. Rationale and design of the Multidisciplinary Approach to Novel Therapies in Cardiology Oncology Research Trial (MANTICORE 101–Breast): a randomized, placebo-controlled trial to determine if conventional heart failure pharmacotherapy can prevent trastuzumab-mediated left ventricular remodeling among patients with HER2+ early breast cancer using cardiac MRI. BMC Cancer 2011;11:318. doi: 10.1186/1471-2407-11-318

[83] Pituskin E. Prophylactic beta blockade preserves left ventricular ejection fraction in HER2-overexpressing breast cancer patients receiving trastuzumab: Primary results of the MANTICORE randomized controlled trial. December 2015. Proceedings of the 36th Annual CTRC-AACR San Antonio Breast Cancer Symposium, San Antonio, TX.

[84] Barac A & Swain SM. Cardiac protection in HER2-targeted treatment: how should we measure new strategies? JAMA Oncol 2016. doi: 10.1001/jamaoncol.2016.0283

[85] Martín M, Esteva FJ, Alba E et al. Minimizing cardiotoxicity while optimizing treatment efficacy with trastuzumab: review and expert recommendations. Oncologist 2009;14(1):1-11. doi: 10.1634/theoncologist.2008-0137

[86] Perez EA, Suman VJ, Davidson NE et al. Cardiac safety analysis of doxorubicin and cyclophosphamide followed by paclitaxel with or without trastuzumab in the North Central Cancer Treatment Group N9831 adjuvant breast cancer trial. J Clin Oncol 2008;26(8):1231-1238. doi: 10.1200/JCO.2007.13.5467

[87] Russell SD, Blackwell KL, Lawrence J et al. Independent adjudication of symptomatic heart failure with the use of doxorubicin and cyclophosphamide followed by trastuzumab adjuvant therapy: a combined review of cardiac data from the National Surgical Adjuvant breast and Bowel Project B-31 and the North Central Cancer Treatment Group N9831 clinical trials. J Clin Oncol 2010;28(21):3416-3421. doi: 10.1200/JCO.2009.23.6950

[88] Guarneri V, Lenihan DJ, Valero V et al. Long-term cardiac tolerability of trastuzumab in metastatic breast cancer: the M.D. Anderson Cancer Center experience. J Clin Oncol 2006;24(25):4107-4115. doi: 10.1200/JCO.2005.04.9551

[89] Gianni L, Salvatorelli E & Minotti G. Anthracycline cardiotoxicity in breast cancer patients: synergism with trastuzumab and taxanes. Cardiovasc Toxicol 2007;7(2):67-71. doi: 10.1007/s12012-007-0013-5

[90] Rayson D, Richel D, Chia S et al. Anthracycline-trastuzumab regimens for HER2/neu-overexpressing breast cancer: current experience and future strategies. Ann Oncol 2008;19(9):1530-1539. doi: 10.1093/annonc/mdn292

[91] Slamon DJ, Eiermann W, Robert NJ et al: Ten-year follow-up of BCIRG-006 comparing doxorubicin plus cyclophosphamide followed by docetaxel with doxorubicin plus cyclophosphamide followed by docetaxel and trastuzumab with docetaxel, carboplatin and trastuzumab in HER2-positive early breast cancer patients. 2015 San Antonio Breast Cancer Symposium. Abstract S5-04. Presented on 11 December 2015.

[92] Denduluri N, Somerfield MR & Wolff AC. Selection of optimal adjuvant chemotherapy regimens for early breast cancer and adjuvant targeted therapy for HER2-positive breast cancers: An American Society of Clinical Oncology Guideline Adaptation of the Cancer Care Ontario Clinical Practice Guideline Summary. J Oncol Pract 2016;12(5):485-488. doi: 10.1200/JOP.2016.012344

[93] Zhang J, Yang PL & Gray NS. Targeting cancer with small molecule kinase inhibitors. Nat Rev Cancer 2009;9(1):28-39. doi: 10.1038/nrc2559

[94] Ky B, Vejpongsa P, Yeh ET et al. Emerging paradigms in cardiomyopathies associated with cancer therapies. Circ Res 2013;113(6):754-764. doi: 10.1161/CIRCRESAHA.113.300218

[95] Force T & Kerkelä R. Cardiotoxicity of the new cancer therapeutics–mechanisms of, and approaches to, the problem. Drug Discov Today 2008;13(17-18):778-784. doi: 10.1016/j.drudis.2008.05.011

[96] Barac A. Yet another player in the cardio-oncology conundrum?: deciphering the role of FLT3. J Am Coll Cardiol 2014;63(10):1020-1021. doi: 10.1016/j.jacc.2013.09.058

[97] Li W, Croce K, Steensma DP et al. Vascular and metabolic implications of novel targeted cancer therapies: focus on kinase inhibitors. J Am Coll Cardiol 2015;66(10):1160-1178. doi: 10.1016/j.jacc.2015.07.025

[98] Mellor HR, Bell AR, Valentin JP et al. Cardiotoxicity associated with targeting kinase pathways in cancer. Toxicol Sci 2011;120(1):14-32. doi: 10.1093/toxsci/kfq378

[99] Novartis Pharmaceuticals Corporation. Nilotinib prescribing information. Novartis Pharmaceuticals Corporation. 2007. Available at: http://www.pharma.us.novartis.com/product/pi/pdf/tasigna.pdf (accessed August 2016)

[100] Richards CJ, Je Y, Schutz FA et al. Incidence and risk of congestive heart failure in patients with renal and nonrenal cell carcinoma treated with sunitinib. J Clin Oncol 2011;29(25):3450-3456. doi: 10.1200/JCO.2010.34.4309

[101] Chu TF, Rupnick MA, Kerkela R et al. Cardiotoxicity associated with tyrosine kinase inhibitor sunitinib. The Lancet 2007;370:2011–2019. doi: 10.1016/S0140-6736(07)61865-0

[102] Schmidinger M, Zielinski CC, Vogl UM et al. Cardiac toxicity of sunitinib and sorafenib in patients with metastatic renal cell carcinoma. J Clin Oncol 2008;26(32):5204-5212. doi: 10.1200/JCO.2007.15.6331

[103] Kerkelä R, Grazette L, Yacobi R et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med 2006;12(8):908-916. doi: 10.1200/JCO.2007.15.6331

[104] Hatfield A, Owen S & Pilot PR. In reply to ‘Cardiotoxicity of the cancer therapeutic agent imatinib mesylate’. Nat Med 2007;13(1):13; author reply 15-6. doi: 10.1038/nm0107-13a

[105] Wong MK & Jarkowski A. Response to sorafenib after sunitinib-induced acute heart failure in a patient with metastatic renal cell carcinoma: case report and review of the literature. Pharmacotherapy 2009;29(4):473-478. doi: 10.1592/phco.29.4.473

[106] Zhu X, Stergiopoulos K & Wu S. Risk of hypertension and renal dysfunction with an angiogenesis inhibitor sunitinib: systematic review and meta-analysis. Acta Oncol 2009;48(1):9-17. doi: 10.1080/02841860802314720

[107] Wu S, Chen JJ, Kudelka A et al. Incidence and risk of hypertension with sorafenib in patients with cancer: a systematic review and meta-analysis. Lancet Oncol 2008;9:117–123. doi: 10.1016/S1470-2045(08)70003-2

[108] Robinson ES, Khankin EV, Choueiri TK et al. Suppression of the nitric oxide pathway in metastatic renal cell carcinoma patients receiving vascular endothelial growth factor-signaling inhibitors. Hypertension 2010;56:1131–1136. doi: 10.1161/HYPERTENSIONAHA.110.160481

[109] Donskov F, Michaelson MD, Puzanov I et al. Sunitinib-associated hypertension and neutropenia as efficacy biomarkers in metastatic renal cell carcinoma patients. Br J Cancer 2015;113(11):1571-1580. doi: 10.1038/bjc.2015.368

[110] Bair SM, Choueiri TK & Moslehi J. Cardiovascular complications associated with novel angiogenesis inhibitors: emerging evidence and evolving perspectives. Trends Cardiovasc Med 2013;23(4):104-113. doi: 10.1016/j.tcm.2012.09.008

[111] Kruzliak P, Novák J & Novák M. Vascular endothelial growth factor inhibitor-induced hypertension: from pathophysiology to prevention and treatment based on long-acting nitric oxide donors. Am J Hypertens 2014;27(1):3-13. doi: 10.1093/ajh/hpt201

[112] Kruzliak P, Kovacova G & Pechanova O. Therapeutic potential of nitric oxide donors in the prevention and treatment of angiogenesis-inhibitor-induced hypertension. Angiogenesis 2013;16:289–295. doi: 10.1007/s10456-012-9327-4

[113] Burger AJ & Mannino S. 5-Fluorouracil-induced coronary vasospasm. Am Heart J 1987;114(2):433-436. doi: 10.1016/0002-8703(87)90517-5

[114] Freeman NJ & Costanza ME. 5-Fluorouracil-associated cardiotoxicity. Cancer 1988;61(1):36-45. doi: 10.1002/1097-0142(19880101)61:1<36::AID-CNCR2820610108>3.0.CO;2-6

[115] Polk A, Vistisen K, Vaage-Nilsen M et al. A systematic review of the pathophysiology of 5-fluorouracil-induced cardiotoxicity. BMC Pharmacol Toxicol 2014;15:47. doi: 10.1186/2050-6511-15-47

[116] Rateesh S, Luis SA, Luis CR et al. Myocardial infarction secondary to 5-fluorouracil: not an absolute contraindication to rechallenge? Int J Cardiol 2014;172(2):e331-333. doi: 10.1016/j.ijcard.2013.12.274

[117] a Dzaye OD, Cleator S & Nihoyannopoulos P. Acute coronary artery thrombosis and vasospasm following capecitabine in conjunction with oxaliplatin treatment for cancer. BMJ Case Rep 2014.

[118] Ambrosy AP, Kunz PL, Fisher GA et al. Capecitabine-induced chest pain relieved by diltiazem. Am J Cardiol 2012;110(11):1623-1626. doi: 10.1016/j.amjcard.2012.07.026

[119] Cohn KE, Stewart JR, Fajardo LF et al. Heart disease following radiation. Medicine (Baltimore) 1967;46(3):281-298. doi: 10.1097/00005792-196705000-00003

[120] Darby SC, Cutter DJ, Boerma M et al. Radiation-related heart disease: current knowledge and future prospects. Int J Radiat Oncol Biol Phys 2010;76(3):656-665. doi: 10.1016/j.ijrobp.2009.09.064

[121] Heidenreich PA, Hancock SL, Lee BK et al. Asymptomatic cardiac disease following mediastinal irradiation. J Am Coll Cardiol 2003;42(4):743-749. doi: 10.1016/S0735-1097(03)00759-9

[122] Fajardo LF. The pathology of ionizing radiation as defined by morphologic patterns. Acta Oncol 2005;44(1):13-22. doi: 10.1080/02841860510007440

[123] Hooning MJ, Aleman BM, van Rosmalen AJ et al. Cause-specific mortality in long-term survivors of breast cancer: A 25-year follow-up study. Int J Radiat Oncol Biol Phys 2006;64(4):1081-1091. doi: 10.1016/j.ijrobp.2005.10.022

[124] Lancellotti P, Nkomo VT, Badano LP et al. Expert consensus for multi-modality imaging evaluation of cardiovascular complications of radiotherapy in adults: a report from the European Association of Cardiovascular Imaging and the American Society of Echocardiography. Eur Heart J Cardiovasc Imaging 2013;14(8):721-740. doi: 10.1093/ehjci/jet123

[125] Erven K, Florian A, Slagmolen P et al. Subclinical cardiotoxicity detected by strain rate imaging up to 14 months after breast radiation therapy. Int J Radiat Oncol Biol Phys 2013;85(5):1172-1178. doi: 10.1016/j.ijrobp.2012.09.022

[126] Heidenreich PA, Hancock SL, Vagelos RH et al. Diastolic dysfunction after mediastinal irradiation. Am Heart J 2005;150(5):977-982. doi: 10.1016/j.ahj.2004.12.026

[127] Wu W, Masri A, Popovic ZB et al. Long-term survival of patients with radiation heart disease undergoing cardiac surgery: a cohort study. Circulation 2013;127(14):1476-1485. doi: 10.1161/CIRCULATIONAHA.113.001435

[128] Bouillon K, Haddy N, Delaloge S et al. Long-term cardiovascular mortality after radiotherapy for breast cancer. J Am Coll Cardiol 2011;57(4):445-452. doi: 10.1016/j.jacc.2010.08.638

[129] Barac A, Murtagh G, Carver JR et al. Cardiovascular health of patients with cancer and cancer survivors: a roadmap to the next level. J Am Coll Cardiol 2015;65(25):2739-2746. doi: 10.1016/j.jacc.2015.04.059

[130] Lenihan DJ, Hartlage G, DeCara J et al. Cardio-oncology training: a proposal from the International Cardioncology Society and Canadian Cardiac Oncology Network for a new multidisciplinary specialty. J Card Fail 2016;22(6):465-471. doi: 10.1016/j.cardfail.2016.03.012

[131] Okwuosa TM & Barac A. Burgeoning cardio-oncology programs: challenges and opportunities for early career cardiologists/faculty firectors. J Am Coll Cardiol 2015;66(10):1193-1197. doi: 10.1016/j.jacc.2015.07.033

[132] National Comprehensive Cancer Network. Survivorship (Version 1.2016). Available at: https://www.nccn.org/professionals/physician_gls/pdf/survivorship.pdf. (accessed 8 July 2016)

[133] US Food and Drug Administration. HERCEPTIN® (trastuzumab) Intravenous Infusion. Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2010/103792s5250lbl.pdf%20 (accessed 8 July 2016)

Last updated
Citation
Clinical Pharmacist, CP, September 2016, Vol 8, No 9;8(9):DOI:10.1211/PJ.2016.20201651

You may also be interested in