Cardiotoxicity
Cardiotoxicity
doi:10.1093/annonc/mdq295
Cardiotoxicity
I. Brana & J. Tabernero*
Medical Oncology Department. Vall d’Hebron University Hospital, Barcelona, Spain
Cancer treatment has improved extraordinarily in recent years. The development of targeted therapies has widened
the cardiotoxic spectrum of antineoplastic drugs. Optimum management of cardiovascular disease before and during
antineoplastic treatment is essential to reduce morbidity and mortality in cancer patients. This article reviews the
incidence and characteristics of cardiotoxic effects of antineoplastic drugs with special focus on the
pathophysiological mechanisms. It also emphasizes the importance of early detection and correction of cardiovascular
risk factors and the relevance of close cardiac monitoring during antineoplastic treatment in order to reduce
cardiotoxicity.
Key words: adverse effects, antineoplastic agents, arrhythmia, chemically induced heart diseases, heart failure, QTc
prolongation
symposium
‘toxicity that affects the heart’. This definition includes a direct the induction of apoptosis, mitochondrial DNA damage,
article
effect of the drug on the heart but also an indirect effect due to changes in ATP production, downregulation of mRNA
enhancement of haemodynamic flow alterations or due to expression for sarcoplasmic reticulum calcium ATPase [5].
thrombotic events [1]. Cardiotoxicity has a rising relevance as Early interventional treatment with angiotensin-converting
a consequence of the global improvement in cancer enzyme (ACE) inhibitors and b-adrenergic blocking drugs has
management, which leads to better survival and, therefore, shown a clear benefit in adult patients with decreased left
adverse effects of treatments have significant consequences on ventricle ejection function (LVEF) not specifically caused by
patient outcome [2–4]. New targeted therapies have widened anthracycine therapy [4]. The potential role of preventive
the cardiotoxic spectrum of antineoplastic drugs. interventions, such as dexrazoxane treatment, will be discussed
The aim of this article is to describe the incidence and the later in this article.
underlying mechanisms of cardiotoxicity induced by
antineoplastic drugs focusing on targeted therapies, as well as
targeted therapies
strategies to prevent and treat this spectrum of toxic effects.
Drugs will be classified based on their main cardiotoxic effect: trastuzumab. Trastuzumab is a humanized monoclonal
left ventricle dysfunction, rhythm disturbances and ischaemia. antibody against the HER2 tyrosine kinase receptor. The
incidence of LVEF decrease or asymptomatic heart failure (HF)
is 7% (Table 1), but it can rise to 13% when trastuzumab is
left ventricle dysfunction administered with concurrent paclitaxel and to 27% with
anthracyclines concurrent anthracyclines [6, 7].
The risk factors described for the development of
Anthracycline-associated cardiotoxicity (AAC) can be divided
trastuzumab-induced cardiotoxicity include age >50 years,
into three forms: immediate pericarditis–myocarditis
borderline LVEF before trastuzumab treatment, history of
syndrome, an early-onset chronic progressive form and a late-
cardiovascular disease, cardiovascular risk factors such as
onset chronic progressive form. The latter two forms are the
diabetes, dislipidemia or elevated body mass index (>30),
most frequently observed [4, 5].
sequence in which chemotherapy is administered and prior
The main risk factor for AAC is the cumulative dose [5].
treatment with anthracyclines (cumulative doses >300 mg/m2)
Other described factors include administration schedule,
[4, 7–10]. Possibly, genetic background and immune status
mediastinal radiotherapy, combination therapy, old age or age
may also have an influence in patient susceptibility to
<4 years, female gender, ethnicity, hypertension, previous
trastuzumab-induced cardiotoxicity [10].
cardiovascular disease, chromosomal abnormalities and liver
Some differences can be established between anthracycline-
disease [5].
and trastuzumab-induced cardiotoxicity: trastuzumab-induced
cardiotoxicity is at least reversible while anthracycline-induced
*Correspondence to: Dr J. Tabernero. Email: jtabernero@vhebron.net is not; there are no ultrastructural changes in cardiomyocytes in
ª The Author 2010. Published by Oxford University Press on behalf of the European Society for Medical Oncology.
All rights reserved. For permissions, please email: journals.permissions@oxfordjournals.org
symposium article Annals of Oncology
Table 1. Safety with trastuzumab in the HERA trial study [6]. its phosphorylation and therefore inactivating it. Nilotinib has
a similar inhibition spectrum but its potency at inhibiting BCR-
Adverse events Trastuzumab, Observation, P ABL is 20-fold greater. Dasatinib also inhibits Src family kinases
n (%) n (%) and can inhibit phosphorylated and unphosphorylated ABL,
(N = 1677) (N = 1710) which gives it a greater potency (100-fold greater) [10].
Patients with grade 3/4 AEs 132 (7.9) 75 (4.4) <0.001 Kerkela et al. [3] initially described 10 cases of congestive
Fatal AEs 6 (0.4) 3 (0.2) 0.34 heart failure in patients with chronic myelogenous leukaemia
Cardiac events treated with imatinib. Later on, a retrospective review of the six
Death 9 (0.5) 1 (0.06) 1.0 registration trials was conducted and the incidence of HF was
Severe CHF 29 (1.7) 0 0.002 established as 0.5% out of 2327 patients [18]. The major
Symptomatic CHF 113 (7.1) 1 (0.06) <0.001 limitation of this study is the absence of pre-planned cardiac
Decrease in LVEF (>10%) 32 (2.2) <0.001 monitoring. During a phase III dose-optimization study with
dasatinib in patients with chronic myeloid leukaemia the
AE, adverse event; CHF, cardiac heart failure.
incidence of HF was 4% [4].
The proposed mechanism of cardiotoxicity [3] of these
trastuzumab-induced cardiotoxicity while in anthracycline compounds is related to the activation of the endoplasmic
cardiotoxicity changes such as vacuolization on cardiomyocytes reticulum (ER) stress response, a mechanism that protects cells
or loss of cardiomyocytes are present [10, 11]. by shutting down general protein translation while
pathophysiology. HER2 has a relevant role in embryonic upregulating the expression of specific protective stress
heart development. It forms heterodimers with HER4 after the response genes. This phenomenon is mediated by PKR like ER
union of neuregulin 1 with HER4 (neuregulins are peptide kinase (PERK), which phosphorylates the transcription factor
ligands of HER3 and HER4 expressed only in cardiac tissue). eIF2a and activates the IRE1 signalling pathway. Imatinib also
HER2–HER4 heterodimerization leads to its leads to an increase in PKCd expression. Ultimately, prolonged
autophosphorylation, which activates several signalling ER stress response and PKCd expression lead to apoptosis. It
pathways such as Src–FAK, which increases cell–cell contacts has been recently indicated that ER stress response and
and mechanical coupling [12], or phosphatidylinositol 3-kinase apoptosis induced by imatinib may occur at concentrations
(PI3K)–Akt and mitogen-activated protein kinase (MAPK), higher than the therapeutic concentration [19].
which promote cardiomyocite proliferation, survival and sunitinib. Sunitinib is an oral inhibitor of vascular endothelial
contractile function [13]. HER2, HER4 and neuregulin 1 have growth factor recptor (VEGFR) 1–3, PDGFRa/b, KIT, FMS-
been shown to be indispensable for heart development as related tyrosine kinase 3 (FLT3), colony-stimulating factor 1
deletion of one of them is lethal in mice embryos [10]. receptor (CSF1R) and rearranged during transfection (RET).
Lapatinib, an oral dual kinase inhibitor of epidermal growth The incidence of HF related to sunitinib in clinical trials ranges
factor receptor (EGFR) and HER2, has been shown to produce from 4% to 11% [20, 21], but the incidence in the general
a low rate of cardiotoxicity (1.6%) [14]. Several mechanisms population may be higher (up to 33.8% in renal cancer
have been described to explain the difference in cardiotoxicity patients) [22, 23]. The main cause of these discrepant rates may
rates between trastuzumab and lapatinib [10]. First, an be related to the higher number of co-morbidities in the general
immune-mediating effect can be raised as trastuzumab has population (higher rate of hypertension and previous cardiac
been shown to stimulate antibody-dependent cell-mediated ischaemia).
toxicity against HER2 tumour cells in vitro [15]. Second, the The mechanism of sunitinib-induced cardiotoxicity is an off-
pharmacokinetic difference in the duration or extent of target effect due to ribosomal S6kinase inhibition producing
inhibition of HER2 by trastuzumab or lapatinib could partially ATP depletion that activates the intrinsic apoptotic pathway. It
explain these differences, although some studies indicate that is also known that VEGF signalling is necessary to obtain an
the kinetics of HER2-signalling inhibition is similar after adequate response of cardiomyocytes to pressure load in those
a single dose of either agent [10]. A third mechanism has been patients with hypertension, which may explain the higher rate
studied with various anti-HER2 antibodies in rat of HF in this population [10].
cardiomyocyte models. The binding of anti-HER2 antibodies to
HER2 also triggers BCL-XL downregulation and BCL-XS
activation, which leads to loss of mitochondrial membrane other targeted therapies
potential, reduction in ATP levels, cytochrome C release and bortezomib. The incidence of HF with bortezomib, a
caspase activation. The intrinsic resistance of cardiomyocytes to proteasome inhibitor, is 5%. The underlying mechanism
apoptosis induced by the BCL pathway may explain the lack of of HF is the induction of ER stress after proteasome inhibition
ultrastructural changes in trastuzumab-induced cardiotoxicity. [4, 24, 25].
In this way downregulation of BCL-XL and activation of BCL- bevacizumab. The incidence of HF related to bevacizumab-
XS would lead to ATP depletion and contractile dysfunction containing treatment is 2.2% in phase III studies. Uncontrolled
[16, 17]. hypertension and inhibition of VEGF signalling that impairs the
ABL inhibitors: imatinib, dasatinib, nilotinib. Imatinib is a adaptive response of the heart to pressure overload have been
tyrosine kinase inhibitor of ABL, ABL-related gene (ARG), raised as potential mechanisms to explain this toxicity [26, 27].
platelet-derived growth factor receptor (PDGFR) a and b and Other kinases have been shown to present a role in survival
KIT. It binds to the unphosphorylated form of ABL preventing or adaptation of cardiomyocytes to different injuries, and for
In a multicentre study in acute promyelocytic leukaemia cases of cardiac ischaemia. In another phase I trial evaluating
40% of patients developed QT prolongation that returned to the combination of combrestatin with radiotherapy the QTc
baseline 8 weeks after the end of treatment [31]. was increased by a mean of 13 ms, in all patients without
histone deacetylase inhibitors. Histone deacetylase (HDAC) related symptoms [36].
inhibitors target an epigenetic mechanism of acetylation that
modulates transcription. Depsipeptide (FK228) is a cyclic protein kinase C inhibitors
peptide HDAC inhibitor. In a retrospective analysis of 500 Enzastaurin is a protein kinase C inhibitor that suppresses the
patients who had received depsipeptide, five sudden deaths PI3K/Akt pathway, leading to anti-angiogenic effects,
were reported. All these patients had risk factors for sudden impairment of tumour growth and induction of tumour death.
death such as electrolyte abnormalities, concomitant therapy In a phase I trial 3 out of 47 enrolled patients developed
with a QTc-prolonging agent or hypertrophic cardiomyopathy. asymptomatic grade 3 QTc prolongation. In the combination
Documentation of increased QT was not obtained before phase I trial of enzastaurin with gemcitabine one patient had
sudden death. In other patients ST and T wave changes and QT a grade 2 QTc interval prolongation [29]. Finally, a phase II
prolongation were described as a class effect due to HDAC trial of enzastaurin 500 mg daily as second- or third-line
inhibition [29, 30, 32]. The incidence of QT prolongation treatment in patients with non-small-cell lung cancer showed 1
associated with vorinostat, a phenylbutyrate-derived HDAC asymptomatic grade 1 QTc prolongation out of 55 patients
inhibitor, is 3.5%–6% [29]. [37].
Scr/Abl kinase inhibitors. QT prolongation was reported to Hdm2 inhibitors. Serdemetan, a novel inhibitor of the
occur in 2%–3% of patients treated with dasatinib, but <1% Hdm2–p53 complex, has shown prolongation of the QTc
experienced a QTc interval of >500 ms [4]. interval in the preliminary presentation of a phase I study [38].
multitargeted tyrosine kinase inhibitors. Sunitinib has been
shown to prolong the QT interval, the PR interval, causing
bradycardia, and to induce ST and T wave changes [22]. ischaemia
Torsade de pointes has been observed in <0.1% of patients
receiving sunitinib. The effect of sunitinib on QT interval is Several cancer treatments, including not only cytotoxic drugs
dose dependent [23]. but also radiotherapy and targeted therapies, are associated
Vandetanib is an oral selective inhibitor of VEGFR, epidermal with an increased risk of coronary artery disease with or
growth factor receptor (EGFR) and RET tyrosine kinases. without coronary syndrome.
Preclinical studies showed an interaction of vandetanib with
cardiac ion channels leading to repolarization abnormalities. In fluoropyrimidines
a phase I trial of vandetanib in solid tumours 9% of patients fluorouracil. The incidence of cardiotoxicity with 5-fluorouracil
developed asymptomatic QTc prolongation, while in a Japanese (5FU) in the literature varies from 1% to 68% [4] with a mean
phase I trial the incidence rose to 61% [29]. A study carried out onset of 72 h after the initiation of administration. The most
in healthy volunteers showed that the effect on QT interval was common cardiac symptom is angina-like chest pain, but some
increased by the combination of vandetanib with ondansetron. cases developed myocardial infarction (MI), arrhythmias, HF,
In phase II trials the incidence of asymptomatic QT prolongation cardiogenic shock and sudden death [4, 11]. Risk factors are
was 15%, all events grade 1–2 [30]. not well defined but one of the most associated risk factors is
Nilotinib, a multitargeted tyrosine kinase inhibitor against a previous history of coronary artery disease. Other described
the Bcr–Abl fusion protein, KIT and PDGFR, has shown risk factors are previous mediastinal radiotherapy and
a mean QT prolongation in healthy volunteers of 18 ms [30]. In concomitant cisplatin therapy. The toxicity is described to be
phase II trials the incidence of grade 2 QT prolongation has dose and rate dependent, as high doses (>800 mg/m2) and
been 1%–4% [4]. In a phase I/II study five sudden deaths continuous infusion administration have been associated with
(0.6%) were reported considered probably or potentially higher rates of toxicity [4, 39].
related to nilotinib [34]. capecitabine. The incidence of cardiotoxicity related to
farnesyl protein transferase inhibitors. Farnesyl protein capecitabine ranges from 3% to 9%. The onset of angina
transferase inhibitors selectively inhibit post-translational symptoms ranges from 3 h to 4 days after treatment has been
farnesylation of Ras and other proteins from other signalling initiated [4]. The main risk factor described is previous
pathways. In a phase II trial of lonafarnib (SCH66336) in coronary artery disease.
patients with refractory head and neck squamous cell pathophysiology. The aetiology of fluoropyrimidine-related
carcinoma 1 out of 15 patients developed grade 3 QTc cardiotoxicity is still unknown. Several hypotheses have been
prolongation and another patient presented syncope without raised including vasospasm PKC mediated, direct toxicity on
previously documented QTc prolongation [29]. Asymptomatic the myocardium, coronary artery thrombus, activation of the
QT prolongation was observed in two phase I trials with coagulation system or autoimmune response [4, 11, 39]. The
L-778123 and also in the phase I trial of the combination of induction of vasospasm is the most likely mechanism as some
L-778123 with paclitaxel [29]. studies have shown a contraction in the brachial artery or an
vascular disruption agents. Combrestatin A4 phosphate (CA4P) increased level of endothelin-1 after 5FU infusion. In contrast,
is a targeted natural tubulin inhibitor with a vascular the co-administration of ergotamine to elicit vasospasm or the
disruption effect. In a phase I trial Dowlati et al. [35] described administration of vasodilating drugs as a preventive treatment
7 cases of QTc prolongation out of 25 enrolled patients and two has shown lack of benefit.
In the case of LVEF dysfunction, early interventional 13. Baliga RR, Pimental DR, Zhao YY et al. NRG-1 induced cardiomyocyte
treatment with ACE inhibitors and b-adrenergic blocking drugs hypertrophy. Role of PI-3-kinase, p70S6K, and MEK-MAPK-RSK. Am J Physiol
Heart Circ Physiol 1999; 277: 2026–2037.
has shown a clear benefit in adult patients with decreased LVEF
14. Perez EA, Koehler M, Byrne J et al. Cardiac safety of lapatinib: pooled analysis of 3689
not specifically caused by anthracycline therapy [4]. The benefit
patients enrolled in clinical trials. Mayo Clin Proc 2008; 83: 679–686.
of ACE inhibitors in cancer patients has been studied in a single-
15. Sliwkowski MX, Lofgren JA, Lewis GD et al. Nonclinical studies addressing the
centre study that proves that this treatment leads to LVEF mechanism of action of trastuzumab. Semin Oncol 1999; 26: 60–70.
recovery and also contributes to a reduction in cardiac events. 16. Grazette LP, Boecker W, Matsui T et al. Inhibition of ErbB2 causes mitochondrial
dysfunction in cardiomyocytes: implications for herceptin-induced
cardiomyopathy. J Am Coll Cardiol 2004; 44: 2231–2238.
conclusion
17. Potts MB, Vaughn AE, McDonough H et al. Reduced Apaf-1 levels in
Given the improvement in survival among cancer patients, cardiomyocytes engage a strict regulation of apoptosis by endogenous XIAP.
prevention and treatment of cardiovascular diseases in this J Cell Biol 2005; 171: 925–930.
population of patients have gained relevance. A better 18. Hatfield A, Owen S, Pilot PR. In reply to ‘Cardiotoxicity of the cancer therapeutic
agent imatinib mesylate’. Nat Med 2007; 13: 15–16.
knowledge about potential cardiac side-effects of antineoplastic
19. Wolf A, Couttet P, Dong M et al. Imatinib does not induce cardiotoxicity at
drugs and the identification of patients at higher risk is a key
clinically relevant concentrations in preclinical studies. Leuk Res 2010; Epub
strategy in reducing cardiotoxicity with these agents. Close ahead of print.
monitoring is essential for an early detection and treatment of 20. Motzer RJ, Hutson TE, Tomczak P et al. Sunitinib versus interferon alfa in
cardiovascular disease induced by antineoplastic treatment, and metastatic renal-cell carcinoma. N Engl J Med 2007; 356: 115–124.
this policy minimizes serious acute and chronic cardiac 21. Demetri GD, Oosterom AT, Garrett CR et al. Efficacy and safety of sunitinib in
consequences. The involvement of cardiologists during early patients with advanced gastrointestinal stromal tumour after failure of imatinib:
clinical drug development will be instrumental in order to a randomised controlled trial. Lancet 2006; 368: 1329–1338.
identify potential cardiac toxic effects in these patients and the 22. Chu TP, Rupnick MA, Kerkela R et al. Cardiotoxicity associated with tyrosine
best way to treat them from the onset. kinase inhibitor sunitinib. Lancet 2007; 370: 2011–2019.
23. 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:
disclosures 5204–5212.
24. Richardson PG, Barlogie B, Berenson J et al. A phase 2 study of bortezomib in
The authors have declared no conflict of interest. relapsed, refractory myeloma. N Engl J Med 2003; 348: 2609–2617.
25. Fu HY, Minamino T, Tsukamoto O et al. Overexpression of endoplasmic
references reticulum-resident chaperone attenuates cardiomyocyte death induced by
proteasome inhibition. Cardiovasc Res 2008; 79: 600–610.
1. Albini A, Penéis G, Donatelli F et al. Cardiotoxicity of anticancer drugs: the need 26. Miller K, Wang M, Gralow J et al. Paclitaxel plus bevacizumab versus paclitaxel
for cardio-oncology and cardio-oncological prevention. J Natl Cancer Inst 2010; alone for metastatic breast cancer. N Engl J Med 2007; 357: 2666–2676.
102: 14–25.
27. Chen MH, Kerkela R, Force T et al. Mechanisms of cardiac dysfunction
2. Oeffinger KC, Mertens AC, Sklar CA et al. Chronic health condition in adult associated with tyrosine kinase inhibitor cancer therapeutics. Circulation 2008;
survivors of childhood cancer. N Engl J Med 2006; 355: 1572–1582. 118: 84–95.
3. Kerkela R, Grazette L, Yacobi R et al. Cardiotoxicity of the cancer therapeutic 28. Hsieh PC, MacGillivray C, Gannon J et al. Local controlled intramyocardial
agent imatinib mesylate. Nat Med 2006; 12: 908–916. delivery of platelet-derived growth factor improves postinfarction ventricular
4. Yeh ET, Bickford CL. Cardiovascular complications of cancer therapy: incidence, function without pulmonary toxicity. Circulation 2006; 114: 637–644.
pathogenesis, diagnosis and management. J Am Coll Cardiol 2009; 53: 29. Strevel EL, Ing DJ, Siu LL. Molecularly targeted oncology therapeutics and
2231–2247. prolongation of the QT interval. J Clin Oncol 2007; 25: 3362–3371.
5. Jones RL, Swanton C, Ewer MS. Anthracycline cardiotoxicity. Expert Opin Drug 30. Ederhy S, Cohen A, Dufaitre G et al. QT interval prolongation among patients
Saf 2006; 5: 791–809. treated with angiogenesis inhibitors. Target Oncol 2009; 4: 89–97.
6. Piccart-Gebhart MJ, Procter M, Leyland-Jones B et al. Trastuzumab after 31. Soignet SL, Frankel SR, Douer D et al. United States multicenter study of arsenic
adjuvant chemotherapy in HER2-positive breast cancer. N Engl J Med 2005; trioxide in relapsed acute promyelocytic leukemia. J Clin Oncol 2001; 19:
353: 1659–1672. 3852–3860.
7. Seidman A, Hudis C, Pierri MK et al. Cardiac dysfunction in the trastuzumab 32. Piekarz RL, Frye AR, Wright JJ et al. Cardiac studies in patients treated with
clinical trials experience. J Clin Oncol 2002; 20: 1215–1221. depsipeptide, FK228, in a phase II trial for T-cell lymphoma. Clin Cancer Res
8. Tan-Chiu E, Yothers G, Romond E et al. Assessment of cardiac dysfunction in 2006; 12: 3762–3773.
a randomized trial comparing doxorubicin and cyclophosphamide followed by 33. Bello CL, Mulay M, Huang X et al. Electrocardiographic characterization of the
paclitaxel, with or without trastuzumab as adjuvant therapy in node-positive, QTc interval in patients with advanced solid tumors: pharmacokinetic–
human epidermal growth factor receptor 2-overexpressing breast cancer: NSABP pharmacodynamic evaluation of sunitinib. Clin Cancer Res 2009; 15: 7045–7052.
B-31. J Clin Oncol 2005; 23: 7811–7819. 34. Tam CS, Kantarjian H, Garcia-Manero G et al. Failure to achieve a major
9. Serrano C, Cortes J, Russillo M et al. Trastuzumab-related cardiotoxicity in the cytogenetic response by 12 months defines inadequate response in patients
elderly: which role for cardiovascular risk factors? Eur J Cancer 2009; 7 (Suppl): receiving nilotinib or dasatinib as second or subsequent line therapy for chronic
216 (Abstr 4005). myeloid leukemia. Blood 2008; 112: 516–518.
10. Force T, Krause DS, Van Etten RA. Molecular mechanisms of cardiotoxicity of 35. Dowlati A, Robertson K, Cooney M et al. A phase I pharmacokinetic and
tyrosine kinase inhibition. Nat Rev Cancer 2007; 7: 332–344. translational study of the novel vascular targeting agent combretastatin a-4
11. Jones RL, Ewer MS. Cardiac and cardiovascular toxicity of nonanthracycline phosphate on a single-dose intravenous schedule in patients with advanced
anticancer drugs. Expert Rev Anticancer Ther 2006; 6: 1249–1269. cancer. Cancer Res 2002; 62: 3408–3416.
12. Kuramochi Y, Guo X, Sawyer DB. Neuregulin activates ERBB2-dependent Src/ 36. Cooney MM, Radivoyevitch T, Dowlati A et al. Cardiovascular safety profile of
FAK signalling and cytoskeletal remodelling in isolated adult rat cardiac combretastatin a4 phosphate in a single-dose phase I study in patients with
myocytes. J Mol Cell Cardiol 2006; 41: 228–235. advanced cancer. Clin Cancer Res 2004; 10: 96–100.