Skip to main content
Therapeutic Advances in Cardiovascular Disease logoLink to Therapeutic Advances in Cardiovascular Disease
editorial
. 2019 Apr 18;13:1753944719843435. doi: 10.1177/1753944719843435

Cardiotoxicity due to targeted anticancer agents: a growing challenge

Chintan P Shah 1,, Jan S Moreb 2
PMCID: PMC6475850  PMID: 30995890

Abstract

The emergence of various targeted anticancer agents has led us to uncharted territory secondary to their cardiotoxic potential with many burning questions, which in turn has led to the evolution of the cardio-oncology field. These targeted agents differ in their cardiovascular complication (CVC) potential even within the same class and it is very difficult to design screening tests that can predict CVCs. Moreover, there is a need for more research to answer many crucial questions, since these toxicities are unanticipated and can lead to poor overall survival of cancer patients. We still do not clearly understand the mechanism for such toxicity, risk factors, and natural history. A better understanding of the underlying risk factors and identification of biomarkers would help us develop protocols for appropriate monitoring strategies which in turn would help capture these toxicities at early stages. In this succinct review, we try to focus on CVC definition, summarize some published research, and point to areas of unmet need in this new field.

Keywords: cancer, cardiotoxicity, cardiovascular toxicity, targeted agents, therapy

Defining the problem

Emergence of various targeted agents has shifted the paradigm of cancer treatment, and as our understanding of molecular pathogenesis expands further many more molecular agents are likely to become important in the treatment of various cancers. Apart from cancer recurrence, cancer survivors today face many challenges related to the long-term side effects of anticancer therapies, including cardiovascular complications (CVCs) that can potentially be a big obstacle in their battle against cancer.1 In general, CVCs from cancer therapy include heart failure, coronary artery disease, arrhythmias, QT prolongation, valvular disease, arterial hypertension, thromboembolic disease, and peripheral vascular disease as per the European Society of Cardiology (ESC) guidelines.2 According to the ESC, cardiotoxicity leading to heart failure is defined as a decrease in the left ventricular ejection fraction (LVEF) >10% points to a value below the lower limit of normality on an echocardiograph, and a relative reduction in global longitudinal strain of >15% from baseline signifies a risk for cardiotoxicity.2 Improvement in the overall survival (OS) of patients with various cancers has led to a higher proportion of cancer patients living with concomitant cardiovascular diseases.3 The field of cardiotoxicity induced by targeted drugs is rapidly expanding leading to the evolution of a new subspecialty of cardio-oncology to deal with the new health burden. It is imperative for an oncologist to know the toxicity profile of newer agents so that the risk of CVCs can be weighed against the benefit of these agents. The list of anticancer targeted drugs that can potentially cause CVCs is growing, and the most recent class of drugs added to the list is the immune checkpoint inhibitors which are shown to cause immune-mediated myocarditis46 (Table 1). Different agents differ in their cardiotoxicity potential as well as types and this variability is seen not only among different classes but also within same class of drugs.710 For instance, within the proteasome inhibitors, carfilzomib was shown to cause significantly more CVCs compared with bortezomib which is less cardiotoxic but causes neuropathy more frequently.11 Similarly, among different tyrosine kinase inhibitors (TKIs), imatinib showed minimal CVCs,7 while dasatinib12 was associated with more cardiopulmonary issues, while ponatinib was associated with more vascular than cardiac events.13 Of note, although many of these new drugs are supposedly targeted towards one gene, apart from a few drugs, most of the time other targets or the same target in other normal organs are also affected.

Table 1.

Anticancer targeted drugs causing cardiovascular complications.

Agents Type of toxicity with approximate frequency when known
Small molecule TKIs
Imatinib Heart failure (<1%), arrhythmias (<1%)
Dasatinib Pulmonary hypertension (0.1–<1%), heart failure (8–9%)
Nilotinib Heart failure, myocardial infarction/ischemia (5–9.4%), QT prolongation (1–2%), PAD, pulmonary hypertension,
Ponatinib Arrhythmias (1–5%), heart failure (3–15%), myocardial infarction/ischemia (12%), PAD, hypertension (2–68%), thromboembolic events (3%)
Bosutinib Pericarditis (1%), pericardial effusion, QT prolongation (<1%)
Sunitinib Heart failure (1–27%), thromboembolic events (1–3%), hypertension (4–34%)
Sorafenib Heart failure (1.9–11%), thromboembolic events, hypertension (9–16%)
Lapatinib Heart failure (0.9–4.9%), QT prolongation (6.1%)
Pazopanib Heart failure (0.6–11%), QT prolongation (<2%)
Monoclonal antibody-based
Rituximab Arrhythmia, heart failure, myocardial infarction/ischemia
Trastuzumab Heart failure (2–28%), thromboembolic events (2–3%), hypertension (4%)
Bevacizumab Heart failure (1–10.9%), myocardial infarction/ischemia (0.6–8.5%), thromboembolic events (3–21%), hypertension (5–18%)
Pertuzumab Heart failure (0.9–16%)
Histone deacetylase inhibitors
Vorinostat QT prolongation (1–4%), thromboembolic events
Immunomodulators
Thalidomide Arrhythmia, thromboembolic events (8–22.5%)
Lenalidomide myocardial infarction/ischemia (0–1.9%), thromboembolic events (4–9%), hypertension (7–8%), hypotension (7%)
Pomalidomide Thromboembolic events (3%), atrial fibrillation (2%)
Proteasome inhibitors
Bortezomib Heart failure (2–5%), arrhythmia, myocardial infarction/ischemia
Carfilzomib Heart failure (4–28%), hypertension (5–27%), arrhythmia, myocardial infarction/ischemia, pulmonary hypertension (1%)
Ixazomib Heart failure (2–4%), myocardial infarction/ischemia, hypotension,
Immune checkpoint inhibitors
Ipilimumab
Pembrolizumab
Nivolumab
Atezolizumab
Durvalumab
Myocarditis (<1%), heart failure (<1%), pericarditis (<1%)
Miscellaneous
Temsirolimus Hypertension (7%), thromboembolic events (2%), heart failure (<1%)
Everolimus Hypertension (4%), heart failure (<1%), tachycardia

Abs, antibodies; PAD, peripheral artery disease; TKI, tyrosine kinase inhibitor; VEGF, vascular endothelial growth factor.

Natural history and outcome

The exact mechanism for the development of CVCs secondary to various targeted agents is poorly understood. Moreover, significant uncertainty prevails in understanding the predisposing factors and the natural history of CVCs. In our retrospective study of patients,14 who had both hematologic malignancy (multiple myeloma, leukemia, and lymphoma) and CVCs, we found that 3.5% (29 of 820) of patients experienced cardiotoxicity (study group) due to targeted agents (such as proteasome inhibitors, TKIs, anti CD20 rituximab, and immunomodulators) over the 10-year study period (2005–2014). The median time from the exposure to cardiac event was 132 days (range 1–1176 days). A total of 8 patients developed various arrhythmias, 27 patients developed reduced LVEF and were diagnosed with cardiomyopathy, while two had non-ST elevation myocardial infarction. Furthermore, we compared the study group with patients who did not develop cardiotoxicity (the reference group, n = 70) after exposure to similar class of targeted agents. Interestingly, we did not find any association between the development of cardiotoxicity and traditional cardiovascular risk factors such as age, sex, hypertension, diabetes mellitus, hyperlipidemia, obesity, and smoking. Recently, another retrospective study also reported that the higher atherosclerotic cardiovascular disease score >7.5% did not significantly increase the incidence of adverse cardiovascular events.15 This raises the possibility of genetic predisposition for the development of CVCs in some patients and not others, which needs to be further explored. Moreover, some data also suggest that there can be common genetic risk factors, such as mutations of DNMT3, TETE2 or ASXL1, which may predispose patients to various cancers as well as CVCs.16 Clearly, and in some instances, the mechanism of CVCs related to drugs such as trastuzumab, TKIs and proteasome inhibitors, is attributed to the off-target effects of the drugs.

Unlike the anthracycline cardiotoxicity, unanticipated CVCs are not usually dose dependent and can happen any time during the course of therapy. Thus, we have to rely only on clinical judgment when weighing the risk of life-threatening CVCs against the benefit of potentially life-saving therapy. In order to understand the magnitude of these complications, it is imperative to develop a clear set of criteria that defines CVCs in the setting of targeted anticancer therapy that can help differentiate it from cardiovascular events that are not related to these drugs. Initial trials of carfilzomib mentioned dyspnea as a pulmonary complication where it could have been secondary to pulmonary edema caused by early cardiotoxic effect and before low LVEF is detected by regular echocardiograms.17 Clinical evidence suggests that the cardiovascular toxicity of targeted agents seems to be reversible after discontinuation of therapy.18,19 In our study,14 as many as 79% of patients had stable to improved LVEF on follow up. However, our study still showed that the study group had a significantly worse OS compared with the reference group, with no difference in cancer progression-free survival between the two groups. This worse OS of the patients with cardiotoxicity could be secondary to the added morbidity, interruption in anticancer therapy and more cautious use of less effective chemotherapeutic agents as a result of excluding certain drugs that can potentially add to the cardiac toxicity. Clinical trials usually exclude patients who experienced a New York Heart Association class III or IV functional heart failure or myocardial infarction in the 6 months prior to the onset of therapy,20 which limits our understanding of the safety of using these targeted agents in such a patient population, leaving retrospective studies as the only tool for more information. Designing trials in a way so that enrolled patients reflecting the real-world patient population with cardiovascular disorders would help oncologists in creating better interventional treatment plans. Moreover, a trial design that allows long-term monitoring would be ideal to capture delayed toxicities.

Early detection of toxicity

Unfortunately, there are no known predictive risk factors for the development of CVCs, and it is usually an unanticipated complication. LVEF measurement is a relatively insensitive tool for detecting cardiotoxicity at an early stage. This is largely because considerable change in LVEF does not occur until a critical amount of myocardial damage has taken place and all the compensatory mechanisms are exhausted. One of the biggest challenges is to identify toxicity in early stages of development by proper monitoring strategies. So far studies have shown that various cardiac markers such as troponin, B-type natriuretic peptide (BNP), and echocardiography do not effectively correlate with the degree of cardiac dysfunction, although in some cases and depending on the mechanism by which CVCs occur, troponin and electrocardiogram (EKG) periodic surveillance may be useful in early detection.20,21 In a randomized controlled trial, troponin I was used as an early detection tool which led to initiation of enalapril.22 During the follow up, none of the enalapril-treated patients developed left ventricular dysfunction versus 43% in control arm.22 In a substudy of the ENDEAVOR phase III trial, serial echocardiogram assessments were of limited utility as risk mitigation tools in patients treated with carfilzomib.18 Recent data suggest strain echocardiography might be able to detect chemotherapy-induced cardiac damage at relatively early stages; however, it needs further validation.23

Protection against cardiotoxicity

Obviously, we are in dire need for further research to develop therapeutic strategies for the prevention of such CVCs and reducing their adverse effect on overall outcome in cancer patients. Since CVC occurrence is unpredictable, and the drugs causing it are effective and needed, finding ways to provide cardiac protection makes most sense. There is vast literature about the protection against anthracycline cardiotoxicity, but not much published data on the protection against targeted therapy-related CVCs with the exception of recent trials about the protection against trastuzumab-induced cardiac toxicity.24,25 The two drug classes, beta blockers and angiotensin converting enzyme inhibitors, which are used for the treatment of cardiomyopathy and heart failure, have been reported to be protective against cardiac toxicity.22,26 However, studies have produced inconsistent results regarding the protective effects of different agents that belong to these drug classes27,28 and more studies and new protective agents are needed.

Future directions

The awareness of this complication is rising and the emerging of cardio-oncology is important, but we urgently need to pick up the pace to answer many of these burning questions. There are a few areas of unmet need: (1) Studies into the possible genetic predisposition to developing CVCs from targeted therapy; (2) What biomarkers can predict early cardiac damage/toxicity while cancer patients receive targeted therapies? (3) What can be done to prevent cardiac toxicity? Are there universal cardioprotective drugs that can be used concomitantly with targeted therapies? (4) What can we learn from the extensive research into the prevention of anthracycline cardiotoxicity?

Animal models and basic science laboratory research can probably answer some of these questions.2931 Furthermore, the establishment of the cardio-oncology discipline will hopefully help in answering some of these questions, especially with more government and industry funding and multilateral collaboration.

Footnotes

Funding: This research received no specific grant from any funding agency in the public, commercial, or not-for-profit sectors.

Conflict of interest statement: The authors declare that there is no conflict of interest.

ORCID iD: Chintan P. Shah Inline graphic https://orcid.org/0000-0002-2441-908X

Contributor Information

Chintan P. Shah, Department of Medicine, Hematology Oncology Division, University of Florida, 1600 SW Archer Road, Gainesville, 32608, FL, USA.

Jan S. Moreb, Department of Medicine, Hematology Oncology Division, University of Florida, Gainesville, FL, USA

References

  • 1. Eschenhagen T, Force T, Ewer MS, et al. Cardiovascular side effects of cancer therapies: a position statement from the Heart Failure Association of the European Society of Cardiology. Eur J Heart Fail 2011; 13: 1–10. [DOI] [PubMed] [Google Scholar]
  • 2. Zamorano JL, Lancellotti P, Rodriguez Muñoz D, et al. 2016 ESC position paper on cancer treatments and cardiovascular toxicity developed under the auspices of the ESC Committee for Practice Guidelines: The Task Force for Cancer Treatments and Cardiovascular Toxicity of the European Society of Cardiology (ESC). Eur Heart J 2016; 37: 2768–2801. [DOI] [PubMed] [Google Scholar]
  • 3. Siegel RL, Miller KD, Jemal A. Cancer statistics, 2016. CA Cancer J Clin 2016; 66: 7–30. [DOI] [PubMed] [Google Scholar]
  • 4. Mahmood SS, Fradley MG, Cohen JV, et al. Myocarditis in patients treated with immune checkpoint inhibitors. J Am Coll Cardiol 2018; 71: 1755–1764. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5. Nida W, Shah C, Boyd C, et al. Cardiotoxicity Associated with immune checkpoint inhibitors: a single center experience. Circulation 2018; 138(Suppl. 1): abstract A14262. [Google Scholar]
  • 6. Varricchi G, Galdiero MR, Marone G, et al. Cardiotoxicity of immune checkpoint inhibitors. ESMO Open 2017; 2: e000247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Kerkelä R, Grazette L, Yacobi R, et al. Cardiotoxicity of the cancer therapeutic agent imatinib mesylate. Nat Med 2006; 12: 908–916. [DOI] [PubMed] [Google Scholar]
  • 8. Shah C, Bishnoi R, Jain A, et al. Cardiotoxicity associated with carfilzomib: systematic review and meta-analysis. Leuk Lymphoma 2018; 59: 2557–2569. [DOI] [PubMed] [Google Scholar]
  • 9. Chang HM, Okwuosa TM, Scarabelli T, et al. Cardiovascular complications of cancer therapy: best practices in diagnosis, prevention, and management: part 2. J Am Coll Cardiol 2017; 70: 2552–2565. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10. Santoni M, Guerra F, Conti A, et al. Incidence and risk of cardiotoxicity in cancer patients treated with targeted therapies. Cancer Treat Rev 2017; 59: 123–131. [DOI] [PubMed] [Google Scholar]
  • 11. Shah C, Bishnoi R, Wang Y, et al. Efficacy and safety of carfilzomib in relapsed and/or refractory multiple myeloma: systematic review and meta-analysis of 14 trials. Oncotarget 2018; 9: 23704–23717. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. ÖzgürYurttaş N, Eşkazan AE. Dasatinib-induced pulmonary arterial hypertension. Br J Clin Pharmacol 2018; 84: 835–845. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13. Suh KJ, Lee JY, Shin DY, et al. Analysis of adverse events associated with dasatinib and nilotinib treatments in chronic-phase chronic myeloid leukemia patients outside clinical trials. Int J Hematol 2017; 106: 229–239. [DOI] [PubMed] [Google Scholar]
  • 14. Shah C, Gong Y, Szady A, et al. Unanticipated cardiotoxicity associated with targeted anticancer therapy in patients with hematologic malignancies patients: natural history and risk factors. Cardiovasc Toxicol 2018; 18: 184–191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15. Parrondo R, Andrews T, Panchal RM, et al. Carfilzomib cardiovascular disease: use of the atherosclerotic cardiovascular disease score to predict cardiovascular events in multiple myeloma patients treated with carfilzomib. Blood 2018; 132(Suppl. 1): 3254. [Google Scholar]
  • 16. Jaiswal S, Fontanillas P, Flannick J, et al. Age-related clonal hematopoiesis associated with adverse outcomes. N Engl J Med 2014; 371: 2488–2498. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Watanabe T, Tobinai K, Matsumoto M, et al. A phase 1/2 study of carfilzomib in Japanese patients with relapsed and/or refractory multiple myeloma. Br J Haematol 2016; 172: 745–756. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18. Russell S, Lyon A, Lenihan D, et al. Serial echocardiographic assessment of patients (PTS) with relapsed multiple myeloma (RMM) receiving carfilzomib and dexamethasone (KD) vs bortezomib and dexamethasone (VD): a substudy of the phase 3 ENDEAVOR trial (NCT01568866). Blood 2015; 126: abstract 4250. [Google Scholar]
  • 19. Hawkes EA, Okines AF, Plummer C, et al. Cardiotoxicity in patients treated with bevacizumab is potentially reversible. J Clin Oncol 2011; 29: e560–e562. [DOI] [PubMed] [Google Scholar]
  • 20. Dimopoulos MA, Moreau P, Palumbo A, et al. Carfilzomib and dexamethasone (Kd) vs bortezomib and dexamethasone (Vd) in patients (pts) with relapsed multiple myeloma (RMM): results from the phase III study ENDEAVOR. J Clin Oncol 2015; 33: abstract 8509. [Google Scholar]
  • 21. Zardavas D, Suter TM, Van Veldhuisen DJ, et al. Role of troponins I and T and N-terminal prohormone of brain natriuretic peptide in monitoring cardiac safety of patients with early-stage human epidermal growth factor receptor 2-positive breast cancer receiving trastuzumab: a herceptin adjuvant study cardiac marker substudy. J Clin Oncol 2017; 35: 878–884. [DOI] [PubMed] [Google Scholar]
  • 22. 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] [PubMed] [Google Scholar]
  • 23. Smiseth OA, Torp H, Opdahl A, et al. Myocardial strain imaging: how useful is it in clinical decision making? Eur Heart J 2016; 37: 1196–1207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24. Boekhout AH, Gietema JA, MilojkovicKerklaan B, et al. Angiotensin II-receptor inhibition with candesartan to prevent trastuzumab-related cardiotoxic effects in patients with early breast cancer: a randomized clinical trial. JAMA Oncol 2016; 2: 1030–1037. [DOI] [PubMed] [Google Scholar]
  • 25. Pituskin E, Mackey JR, Koshman S, et al. Multidisciplinary approach to novel therapies in cardio-oncology research (MANTICORE 101-Breast): a randomized trial for the prevention of trastuzumab-associated cardiotoxicity. J Clin Oncol 2017; 35: 870–877. [DOI] [PubMed] [Google Scholar]
  • 26. Kalay N, Basar E, Ozdogru I, et al. Protective effects of carvedilol against anthracycline-induced cardiomyopathy. J Am Coll Cardiol 2006; 48: 2258–2262. [DOI] [PubMed] [Google Scholar]
  • 27. Avila MS, Ayub-Ferreira SM, de Barros Wanderley MR, et al. Carvedilol for prevention of chemotherapy-related cardiotoxicity: the CECCY trial. J Am Coll Cardiol 2018; 71: 2281–2290. [DOI] [PubMed] [Google Scholar]
  • 28. 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: 2355–2362. [DOI] [PubMed] [Google Scholar]
  • 29. Horie T, Ono K, Nishi H, et al. Acute doxorubicin cardiotoxicity is associated with miR-146a-induced inhibition of the neuregulin-ErbB pathway. Cardiovasc Res 2010; 87: 656–664. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30. Imam F, Al-Harbi NO, Al-Harbia MM, et al. Rutin Attenuates Carfilzomib-Induced Cardiotoxicity Through Inhibition of NF-κB, Hypertrophic Gene Expression and Oxidative Stress. Cardiovasc Toxicol 2017; 17: 58–66. [DOI] [PubMed] [Google Scholar]
  • 31. Shah C, Moreb J, Kannampuzha J, et al. Identification of viral gene signatures in cancer patients with reduced ejection fraction after chemotherapy. Circulation 2018; 138(Suppl. 1): abstract A15398. [Google Scholar]

Articles from Therapeutic Advances in Cardiovascular Disease are provided here courtesy of SAGE Publications

RESOURCES