Arrhythmia and device assessment during cancer treatments


KEY POINTS

  • Atrial fibrillation and other atrial arrhythmias are common complications of many different cancer therapeutics.

  • The CHA 2 DS 2 -VASc and the HAS-BLED scores may not be appropriate to determine thromboembolic and bleeding risk in patients with cancer receiving active therapy.

  • Ventricular arrhythmias are an infrequent, but potentially life-threatening, complication of various cancer therapeutics, most often related to QT prolongation or secondary to other toxicities, such as ischemia or left ventricular dysfunction.

  • Bradyarrhythmias are frequently asymptomatic and rarely require intervention.

  • Heart block may be the first manifestation of checkpoint inhibitor myocarditis.

  • Autonomic dysfunction is commonly observed in patients after head, neck, and chest radiation.

  • The majority of patients with cardiac implantable devices can safely receive radiation therapy.

  • Additional monitoring precautions should be offered to pacemaker-dependent patients and those with devices who are exposed to high beam energy and/or cumulative absorbed dose.

The landscape of cancer therapeutics has significantly changed over the last decade owing to an improved understanding of cancer biology. Patients are now living longer and, in many cases, surviving their disease. Unfortunately, cardiovascular (CV) toxicities are an increasingly important and often treatment-limiting problem. Whereas much of the focus has been on left ventricular dysfunction and heart failure, many cancer treatments are known to be arrhythmogenic and can have a significant impact on patient morbidity and mortality (see Chapter 10 , Table 10.1 , concise overview provided in Table 19.1 ). Although atrial fibrillation and other supraventricular arrhythmias are more commonly encountered, ventricular arrhythmias and QT prolongation can also occur during cancer treatment (see Chapter 10 , Tables 10.1 and 10.4 ). Management of electrophysiology issues poses unique challenges in patients with cancer and requires a collaborative effort between cardio-oncologists and the rest of the treatment team.

TABLE 19.1
Arrhythmias Associated With Anticancer Agents
DRUG CLASS EXAMPLE ASSOCIATED ARRHYTHMIAS POTENTIAL MECHANISM
Alkylating agents Melphalan Atrial arrhythmias Unknown
Anthracyclines Doxorubicin Atrial arrhythmias

Ventricular arrhythmia

Free radical/toxin accumulation; direct myocardial damage/cardiomyopathy
Direct myocardial damage/cardiomyopathy; increased ventricular repolarization indices
Arsenic QT prolongation Potassium channel inhibition
Cyclin-dependent kinase 4/6 inhibitors Ribociclib QT prolongation Unknown
Fluoropyrimidines 5-Fluorouracil Ventricular arrhythmias Secondary to coronary vasospasm/myocardial ischemia
Immunotherapies Checkpoint inhibitors (e.g., pembrolizumab) Atrial arrhythmias
Ventricular arrhythmias
Bradyarrhythmias
Myocarditis/inflammatory
Myocarditis/inflammatory
Myocarditis/inflammatory
Proteasome inhibitors Carfilzomib Atrial arrhythmias Unknown: possibly owing to accumulation of abnormal intracellular proteins
Taxanes Paclitaxel Bradyarrhythmais Effects on histamine receptor
Tyrosine kinase inhibitors Ibrutinib (frequent)
Ponatinib, Bosutinib, Sorafenib (infrequent)
Nilotinib, Sunitinib, Vemurafenib, Vandetanib
Ibrutinib
ALK inhibitors
Atrial arrhythmias

QT prolongation

Ventricular arrhythmias
Bradyarrhythmias

PI3K pathway inhibition; impaired sarcoplasmic reticulum calcium handling; left atrial fibrosis
Impaired intracellular signaling leading to enhanced late sodium and decreased potassium currents
Unknown
Decrease I f (funny channel) currents in sinoatrial nodal cells
ALK, Anaplastic lymphoma kinase.

Atrial fibrillation and other supraventricular arrhythmias

Atrial arrhythmias, especially atrial fibrillation (AF) are frequently observed in patients undergoing cancer therapy. The “REasons for Geographic and Racial Differences in Stroke” (REGARDS) cohort of more than 15,000 patients reported a 20% higher adjusted risk of AF in patients with cancer, especially within the first year of diagnosis. This can be for multiple reasons, ranging from the malignancy itself and the associated inflammatory state (breast, colorectal, and hematologic malignancies are all associated with higher rates of AF), to specific cancer treatments, including chemo- and targeted therapies. Additionally, patients with cancer and AF have worse outcomes with a two-fold higher risk for thromboembolic complications, a 6-fold higher adjusted risk for heart failure, and a 10-fold higher risk of adjusted 30-day mortality compared with patients not having cancer. It is essential to identify and manage aggressively those patients with cancer with AF, whereas providing appropriate treatment can be challenging and often requires a nuanced, multidisciplinary approach in order for patients to have the best oncologic and CV outcomes.

Anthracyclines

Although heart failure and left ventricular dysfunction are the most common cardiotoxicities of doxorubicin and other anthracyclines, arrythmias are also frequently encountered. For example, rates of AF with doxorubicin infusion are reported between 6% and 10% in the absence of other cardiovascular pathology. , Moreover, also found are increased rates of AF in the setting of anthracycline-mediated cardiomyopathy, with a prevalence of more than 50%, which is similar to other nonischemic cardiomyopathy etiologies. , The arrhythmic mechanism is not clear, but is likely caused by impaired intracellular signaling, damage from free radicals and other toxins, and/or direct myocardial injury.

Alkylating agents and stem cell transplantation

Alkylating agents, including melphalan and cyclophosphamide, are a class of cytotoxic chemotherapeutics that disrupt the DNA double helix and are used in the treatment of various solid and hematologic malignancies. Moreover, melphalan is a common agent used in stem cell transplant (SCT) preconditioning regimens. In one study, 11% of patients who received melphalan prior to SCT developed atrial arrhythmias, including AF, which was significantly higher than in patients who received other treatment regimens. Moreover, SCT itself increases the likelihood of developing AF, with rates as high as 27%. Risk factors for the development of AF after transplant include renal failure, hypertension, and exposure to melphalan. Moreover, the development of AF during transplant portends a worse prognosis, with elevated rates of intensive care unit admissions as well as 30-day and 1-year mortality compared with those patients without arrhythmias.

Tyrosine kinase inhibitors

Tyrosine kinase inhibitors (TKI) affect intracellular pathways that regulate cell growth. A variety of cardiotoxicities are observed with these agents, including AF and other arrhythmias. In one study, the incidence of AF with the TKI sorafenib was reported at 5%. Similarly, arrhythmias can be seen with TKIs used to treat chronic myeloid leukemia that target the BCR-ABL protein. Although vascular toxicities, raging from stroke to myocardial infarction, are most often associated with these TKIs, arrhythmias can also occur. For example, in the EPIC study, the incidence of AF associated with ponatinib use was reported at 3%, compared with 0% for the imatinib treated group. Similarly, rates of AF are around 2% with bosutinib. Whereas the mechanism of arrhythmia remains unclear, it may be related to off-target effects on the phosphoinositide 3-kinase (PI3K) pathway.

Ibrutinib is the TKI most associated with the development of AF. This is a small molecule inhibitor of the Bruton’s tyrosine kinase, used to treat various B-cell malignancies, including chronic lymphocytic leukemia (CLL), mantle cell lymphoma, and Waldenstrom’s macroglobulinemia, as well as chronic graft-versus-host disease. Rates of AF have been reported between 10% and 15%, with ibrutinib itself identified as an independent risk factor for the development of AF. Other risk factors include a prior history of AF, left atrial enlargement, and an elevated Framingham Heart Study AF risk score. As other Bruton’s tyrosine kinase inhibitors, such as acalabrutinib, are associated with a significantly lower incidence of AF, off-target effects of ibrutinib have been postulated, among these. Inhibition of the PI3K pathway, enhanced left atrial fibrosis, and/or impaired sarcoplasmic reticulum calcium handling.

Proteasome inhibitors

Proteasomes are essential in the elimination of the majority of intracellular proteins, including those modified by oxidative stress and tagged for degradation by the ubiquitin system. Proteasome inhibitors (PIs) block the proteolytic activity of the proteasome complex, leading to protein accumulation, even aggregation, and the activation of apoptotic pathways and cell death. PIs are used primarily in the treatment of multiple myeloma and AL amyloidosis. AF is quite common in patients with multiple myeloma, with a prevalence of 14.6%. This is caused, in part, to PI exposure, particularly to carfilzomib, in which the associated incidence of AF/atrial flutter (AFL) is between 3% and 4%.

You're Reading a Preview

Become a Clinical Tree membership for Full access and enjoy Unlimited articles

Become membership

If you are a member. Log in here