Review Article

Drug-induced AF: Arrhythmogenic Mechanisms and Management Strategies

Register or Login to View PDF Permissions
Permissions× For commercial reprint enquiries please contact Springer Healthcare: ReprintsWarehouse@springernature.com.

For permissions and non-commercial reprint enquiries, please visit Copyright.com to start a request.

For author reprints, please email rob.barclay@radcliffe-group.com.
Information image
Average (ratings)
No ratings
Your rating

Abstract

AF is a prevalent condition that is associated with various modifiable and unmodifiable risk factors. Drug-induced AF, despite being commonly under-recognised, can be relatively easy to manage. Numerous cardiovascular and non-cardiovascular agents, including catecholaminergic agents, adenosine, anti-tumour agents and others, have been reported to induce AF. However, the mechanisms underlying drug-induced AF are diverse and not fully understood. The complexity of clinical scenarios and insufficient knowledge regarding drug-induced AF have rendered the management of this condition complicated, and current treatment guidelines follow those for other types of AF. Here, we present a review of the epidemiology of drug-induced AF and highlight a range of drugs that can induce or exacerbate AF, along with their molecular and electrophysiological mechanisms. Given the inadequate evidence and lack of attention, further research is crucial to underscore the clinical significance of drug-induced AF, clarify the underlying mechanisms and develop effective treatment strategies for the condition.

Disclosure:The authors have no conflicts of interest to declare.

Received:

Accepted:

Published online:

Funding:

This study was funded by the National Natural Science Foundation of China (Grant no: 81930105).

Correspondence Details:Lin Wu, Department of Cardiology, Peking University First Hospital, #8, Xishiku St, West District, Beijing 100034, China. E: wuepgroup@126.com

Open Access:

This work is open access under the CC-BY-NC 4.0 License which allows users to copy, redistribute and make derivative works for non-commercial purposes, provided the original work is cited correctly.

AF is the most common sustained cardiac arrhythmia, affecting millions of people globally.1 It is associated with an increased risk of various adverse health outcomes, such as heart failure, stroke, dementia and other conditions, placing a substantial burden on healthcare systems worldwide.2

Despite significant advances in the development of drugs and interventional procedures for the treatment of AF, the therapeutic effects of these treatments remain suboptimal. The current management and prevention strategies for AF through upstream treatments highlight the importance of addressing risk factors. However, there is still much to be developed in terms of effective prevention strategies for AF.3

Risk factors for AF include advanced age, alcohol consumption, family history of AF, hypertension, thyroid dysfunction, sleep apnoea, structural heart diseases and gut microbiota dysbiosis (Table 1).4,5 Notably, drugs represent a significant risk factor for AF not mentioned in the latest AF guidelines.6 Drug-induced AF (DIAF) is a type of AF that is induced or precipitated by drug therapy and determining the underlying cause can be challenging. Despite the difficulty in diagnosis, DIAF can cause significant symptoms, which can have an impact on the management of relevant diseases. However, compared with other types of AF, DIAF is often more preventable and manageable once the causal drug is discontinued.

Drugs that might induce AF have been collectively reviewed in previous literature.7–10 The aim of this review is to comprehensively discuss DIAF, including potential molecular and electrophysiological mechanisms and suggested management strategies. By enhancing the understanding of DIAF, this review aims to raise awareness among healthcare providers of its possible occurrence and highlight the need for prompt and appropriate management.

Epidemiology

Assessing the prevalence of DIAF precisely and comprehensively is a formidable undertaking, as the available data pertaining to it are relatively scant. The incidence of DIAF is estimated to be relatively low and variable among different drugs. For example, the incidence has been reported as 0–18% for dobutamine, 4.6–5% for milrinone, 6–10.3% for anthracycline agents, 3–16% for ibrutinib, 1–12% for adenosine, 1.2% for trastuzumab, 1.3% for ivabradine, 0.1–17.1% for intracoronary acetylcholine (ACh), 1.1–30% for immune checkpoint inhibitors (ICIs), 2.07% for chimeric antigen receptor T-cell (CAR-T) therapy, 0.8% for acute alcohol consumption, 25–47% for persistent and heavy alcohol consumption and 1.17% for substance use disorder.10–29

It is worth noting that DIAF may exhibit a higher frequency among patients with coexisting severe cardiovascular disorders. For instance, in patients diagnosed with anthracycline-associated left ventricular dysfunction, the likelihood of developing AF has been reported to reach as high as 56.6%.30

Figure 1: Mechanisms Underlying AF

Article image

Table 1: Common Conditions Underlying AF and Corresponding Therapeutics Associated with AF-inducing Effects

Article image

Numerous accounts of DIAF rely on isolated case reports and substantial, all-encompassing clinical investigations are currently lacking. Consequently, the estimates of DIAF described above may not be entirely precise and are prone to underestimation. Additionally, the exact incidence of DIAF caused by other drugs is difficult to assess. Reasons for this may include – but are not limited to – the following:

  • The prescription of many medications by non-cardiologists who may not be fully aware of the possible adverse effect of DIAF, leading to under-recognition of some cases.
  • The symptomless nature of AF, which can be paroxysmal and asymptomatic (i.e., silent AF), unlike drug-induced QT prolongation, which may result in fatal ventricular arrhythmias with severe symptoms such as torsade de pointes; additionally, the incidence of AF is directly proportional to the frequency of screening, making it challenging to precisely evaluate its actual prevalence.
  • Establishing a causal relationship between drugs and AF can be complicated because of the intricacies of pathophysiology, such as in patients with polypharmacy or in those with both diseases and relevant therapies that are associated with AF (for example, both cancer and anticancer treatment may induce AF), and because of the unavailability of a comprehensive medical history.31

As a result, despite the estimated low incidence of DIAF, pinpointing the precise prevalence remains a challenging task.

Mechanisms of AF and Drug-induced AF

AF pathogenesis is complex and multifactorial, involving a multitude of triggers and substrates (Figure 1). Ectopic/triggered activity can be attributed to early afterdepolarisation (EAD), delayed afterdepolarisation (DAD) or abnormal automaticity. EAD often happens at the action potential (AP) phase 2 or 3, which is associated with AP prolongation that allows L-type Ca2+ channels to recover and activate again. In contrast, DAD is often associated with intracellular Ca2+ overload.32

There is a lack of evidence about the role of EAD in AF patients without delayed repolarisation; therefore, DAD is regarded as the major type of ectopic/triggered activity in AF arrhythmogenesis.33 Reentry refers to a phenomenon whereby the impulse travels around a specific obstacle (either anatomic or functional) and excites tissue that regains activation ability from previous excitation without extinguishing.34 Short effective repolarisation duration (ERP), slow conduction velocities as well as heterogenous electrophysiological properties are prominent factors in the formation of reentry.35,36 Often, these two factors interact and enhance each other in the development of AF. The mechanisms underlying DIAF are also diverse and may involve direct or indirect effects on atrial electrophysiology, ion channels and signalling pathways, leading to the development of initiators and/or substrates for AF. Identification of the substrates and triggers of DIAF is the first step in its management.

Drugs Affecting the Autonomic Nervous System

The pathophysiology of AF involves the critical contribution of both the vagal and sympathetic nervous systems, each with unique mechanisms.37 However, in most cases, disharmony of autonomic nervous system activation, rather than increased activity of each side respectively alone, is of greater importance for AF onset.

Sympathetic Activating Agents

Upon binding to cardiac β1-adrenergic receptors, the sympathetic neurotransmitters, catecholamines, activate adenylyl cyclase and initiate cyclic adenosine monophosphate (cAMP) synthesis. This results in the elevation of cAMP levels, which in turn activates protein kinase A (PKA). PKA activation subsequently triggers the phosphorylation of several crucial Ca2+-handling proteins. As a result of sympathetic activation, intracellular Ca2+ levels increase, which may result in Ca2+ overload and DAD.38 Catecholamines and other sympathomimetic agents are widely used as inotropes or bronchodilators, many of which can trigger AF, and dobutamine-induced AF is common in dobutamine stress echocardiography testing.39,40 Given the crucial role of the sympathetic nervous system in AF and the increased prevalence of catecholamine-induced AF, β-blockers have been widely used in the management of AF, particularly in individuals with heightened sympathetic tone.

Cholinergic Activators

ACh is the neurotransmitter released by parasympathetic efferent fibres. Vagal stimulation induces the release of ACh, which primarily binds to cardiac M2 receptors. This activation, in turn, stimulates the inward rectifier potassium current channel, IKAch, causing an outward potassium current and shortening of atrial ERP, which facilitates the formation of reentry. In addition, the non-uniform distribution of vagal nerve endings can lead to increased atrial heterogeneity, further contributing to the formation of reentry.37 Furthermore, ACh can also cause vasoconstriction in the intracoronary ACh provocation test used in the diagnosis of vasospastic angina, triggering paroxysmal AF in patients with ischaemia.20

Cardiovascular Drugs

Diuretics

In addition to their intended effects, diuretics, such as loop diuretics and thiazides, also inhibit the reabsorption of potassium ions, leading to hypokalaemia. The clinical consequences of hypokalaemia can vary, ranging from asymptomatic to potentially fatal arrhythmias.41 Current estimates suggest that up to 80% of patients receiving diuretics are at risk of developing hypokalaemia, which may lead to AF.42,43

A study by Lin et al. found that the loop diuretic furosemide was associated with an increased risk of developing AF after pacemaker implantation in elderly patients, while the thiazide diuretic hydrochlorothiazide was protective against AF in these patients.44 The researchers attributed the proarrhythmic effects of furosemide to the activation of the renin–angiotensin–aldosterone system and sympathetic nervous system, as well as hypomagnesaemia.44 Anti-aldosterone diuretics do not exhibit a significant impact on potassium levels and certain studies suggest that they may reduce the risk of AF.45

Cardiac Calcitropes

Traditional inotropes, also known as cardiac calcitropes, augment myocardial contractility by elevating Ca2+ and are commonly administered to preserve cardiac output in patients with heart failure and those undergoing post-surgical procedures. Nevertheless, heightened energy demand, as well as higher Ca2+, can lead to arrhythmias and exacerbate clinical conditions.46 Catecholamines and some sympathomimetic drugs are also important inotropes able to induce AF, as discussed earlier.

Milrinone, a phosphodiesterase 3 inhibitor, elevates cytosolic Ca2+ by preventing the hydrolysis and degradation of cyclic nucleotides cAMP/cyclic guanosine monophosphate through non-catecholamine pathways, which can promote the generation of ectopic impulses and potentially facilitate AF.11,12,47 Digitalis inhibits Na+–K+–ATPase and leads to an increase in intracellular Na+, which in turn activates the Na+/Ca2+ exchanger to promote Ca2+ influx. While atrioventricular (AV) nodal block and/or premature contractions (both atrial and ventricular) are the most frequently observed types of digitalis-induced arrhythmias, it is theoretically possible for digitalis to result in any form of arrhythmia, including AF.48,49

Vasodilators

Nicorandil, an ATP-sensitive potassium channel activator, exhibits pronounced vasodilatory effects on both coronary and peripheral vessels, rendering it a frequently used therapeutic agent in individuals with coronary artery disease. However, the activation of potassium channels also shortens atrial ERP, facilitating the formation of reentry, which plays a pivotal role in the pathophysiology of AF. Lee et al. revealed that the new application of nicorandil is linked to an augmented risk of AF/atrial flutter (OR 2.34; 95% CI [1.07–5.13]) compared with nitrate use.50 However, because of a lack of associated research, the exact incidence of nicorandil-induced AF remains unknown.

Antiarrhythmic Agents

Proarrhythmic effects remain an important issue for antiarrhythmic agents; for example, Vaughan Williams class III drugs prolong QT interval and can even cause fatal torsades de pointes.51 However, with much attention paid to ventricular arrhythmia, many antiarrhythmic drugs, such as adenosine, amiodarone and ivabradine, may also lead to or exaggerate AF.

Adenosine

Adenosine activates A1 receptors, the predominant cardiac subtype receptor, and stimulates IKAdo, an inward potassium current also activated by M2 ACh receptors.52 Consequently, adenosine-induced IKAdo current shortens AP duration (APD) and abbreviates atrial ERP in a heterogeneous manner, similar to ACh-induced atrial electrophysiological alterations that promote AF.53,54 Notably, adenosine is frequently used for the diagnosis or termination of paroxysmal supraventricular tachycardia involving the AV node, primarily because of its ability to impede AV node conduction. Nonetheless, a well-known adverse effect of adenosine is the induction of AF.17,18 While its impact is brief because of its short half-life (lasting only 20–30 seconds), this poses a significant risk, especially in patients with Wolff-Parkinson-White (WPW) syndrome, as it can potentially progress to VF and result in unstable haemodynamics.55 In such cases, the emergency department should be prepared for continuous monitoring and the availability of resuscitation equipment for the rare cases of adenosine-induced fast ventricular preexcitation in patients with WPW syndrome.55

Amiodarone

Amiodarone – used for both rate control and pharmacological cardioversion for AF patients – may also induce AF.56,57 Amiodarone, with a chemical structure resembling that of thyroid hormones, contains approximately 37% iodine. Because of its lipophilicity, slow absorption rate, moderate to low bioavailability (approximately 40–50%) and long half-life, amiodarone can accumulate in various organs and tissues, particularly with prolonged use.58 Accumulated amiodarone results in elevated iodine content, excessive hormone synthesis and release and release of stored hormone because of autoimmune destructive thyroiditis.58 As a result, chronic amiodarone administration may increase thyroid function, leading to AF. Dronedarone, developed as a non-iodinated analogue of amiodarone with similar antiarrhythmic properties, is associated with a higher rate of AF recurrence compared with amiodarone, but with fewer extracardiac adverse effects, such as thyroid dysfunction.59,60 Therefore, in selected patients (such as those with paroxysmal AF without structural heart diseases), dronedarone, but not amiodarone, may be considered the preferred choice.

Ivabradine

Ivabradine selectively inhibits the sinoatrial node (SAN) If current to decrease resting heart rate; primary clinical applications include the treatment of stable angina pectoris, left ventricular systolic dysfunction and chronic heart failure.61 However, debate remains about the relationship between ivabradine and AF.62 Ivabradine might be used to control ventricular rate in AF patients and to treat atrial tachycardia.63,64 Additionally, it shows benefits in the prevention of postoperative AF.65 There have also been various preclinical experiments supporting the anti-AF properties of ivabradine.66–69 On the contrary, other studies demonstrate evidence of elevated risks of AF by unselective ivabradine treatment in patients with chronic heart failure and coronary heart diseases, although ivabradine-induced AF did not appear to affect the incidence of non-fatal stroke compared with placebo.65,70 Laboratory studies on the underlying mechanisms are insufficient to accurately capture this complexity. It was supposed that AF might be secondary to hyperpolarization-activated cyclic nucleotide-gated potassium channel 4 downregulation and SAN dysfunction.62 Ivabradine also leads to bradycardia, hypotension and resultant sympathetic nervous system excitation. Simultaneous vagal (i.e., heart rate-lowering) and sympathetic activation may lead to Ca2+ instability and ectopic firing.62 In a canine model, AF inducibility remained unchanged despite obvious ivabradine-induced bradycardia and vagal activity alterations.71 Overall, the inconsistent findings between studies call for further research to explore how the application of ivabradine influences AF in patients.

Anti-tumour Drugs

The use of anti-cancer drugs has been demonstrated to be an independent risk factor for AF in a dose-dependent manner.31 The underlying mechanisms of this association are still being investigated and include – but are not limited to – direct myocardial damage, modification of molecules associated with electrophysiological alterations and immune cell infiltration.

Traditional Chemotherapy

Traditional chemotherapies are typical cytotoxic drugs that target tumour cells by disrupting their metabolism or interfering with DNA replication. However, these drugs also exhibit significant cardiotoxicity and are closely associated with the development of AF.

Anthracyclines, a class of traditional antineoplastic agents, exert their anti-tumour effects by damaging DNA, generating reactive oxidative species (ROS) and inhibiting molecular synthesis. Their cardiotoxicity is cumulative, dose-dependent and augmented by risk factors such as age, pre-existing cardiovascular disease and concomitant administration of drugs with potential cardiovascular effects. While much attention is given to anthracycline-related left ventricular dysfunction, it is important to note that anthracyclines can also lead to atrial abnormalities and AF.13,14,72

In a study by Tan et al., post-surgery breast cancer patients receiving anthracycline-containing regimens showed unchanged ventricular parameters and functions before and after chemotherapy, but left atrial reservoir and conduit function were decreased post-chemotherapy.72 The authors also showed that mice treated with doxorubicin showed significant atrial remodelling and increased susceptibility to AF, which could be alleviated by dexrazoxane (a Food and Drug Administration-approved drug for management of doxorubicin cardiotoxicity) or antioxidants.72

Although the mechanisms underlying anthracycline-induced cardiotoxicity have not been fully elucidated, several pathways have been implicated, including generation of ROS, lipid peroxidation, iron-mediated damage and disruption of calcium homeostasis.73 The main mechanism of anthracycline cardiotoxicity is now thought to be through the inhibition of topoisomerase 2β, leading to the activation of the cell death pathway and concurrent inhibition of mitochondrial biogenesis.73

Novel Molecular Anti-tumour Therapies

Traditional chemotherapies – while used for many years – lack specificity toward cancer cells, causing unexpected adverse effects in normal tissues. To address this, targeted therapies have been developed to focus on molecules specific to malignant cells, aiming for higher efficacy and fewer adverse effects. However, it is important to note that even targeted therapies can lead to cardiotoxicity and heart dysfunction.

Erb-b2 Receptor Tyrosine Kinase 2 Inhibitors

Trastuzumab is a humanised monoclonal antibody that specifically targets and blocks the erb-b2 receptor tyrosine kinase 2 (ErbB2) pathway; it has become an integral part of the standard treatment of ErbB2-positive breast cancer. However, it also causes cardiac dysfunction, such as heart failure and AF, especially when used in combination with other cytotoxic drugs.19

The cardiac adverse effects of trastuzumab are attributed to blockade of the neuregulin 1 (NRG-1)/ErbB2 axis. NRG-1, released from cardiac endothelial cells in a paracrine manner, is critical in foetal cardiac tissue development and protective for adult hearts under various stimuli, e.g. stress. In cardiac tissues, there are four types of ErbB receptor, with ErbB2 being of particular importance.74 The combination of NRG-1 with ErbB receptors activates downstream effectors, such as ERK1/2 and phosphoinositide 3-kinase (PI3K)–Akt.75 ErbB signals protect the heart from various stimuli, like anthracycline treatment, possibly via anti-oxidative action.76 It has been observed that trastuzumab induced blockade of NRG-1/ErbB pathway, increased ROS production and removed its cardio-protective effect.77 ErbB2 inhibition also caused cardiac mitochondrial-associated cell apoptosis.78 All these changes eventually led to cardiac injury and predispose the heart to arrhythmias.

Ibrutinib

Ibrutinib, a first-generation Bruton’s tyrosine kinase (BTK) inhibitor, has demonstrated remarkable clinical efficacy in treating various B-cell malignancies.79 While generally well-tolerated, ibrutinib is associated with several adverse effects, with AF being one of the most common.80 Notably, among all anti-tumour agents, ibrutinib stands out as the most prominent inducer of AF.

Although the exact mechanisms underlying ibrutinib-induced AF remain unclear, it is generally considered that the blockade of cardiac off-targets, such as PI3K–Akt signalling, ErbB2 signalling and C-terminal Src kinase-Src family tyrosine kinase signalling, is responsible for its proarrhythmic effects.81–84 BTK–PI3K–Akt signalling is cardiac-protective under stress conditions and plays an important role in preventing stress-induced cardiomyopathy. Experimental evidence shows that genetically modified mice with reduced cardiac PI3K–Akt activity are more susceptible to AF, indicating a potential link between reduced PI3K–Akt activity and AF susceptibility.85 Furthermore, clinical studies have reported that patients with AF tend to exhibit lower cardiac PI3K–Akt activity levels, supporting the involvement of the pathway in the development and progression of AF.86 The proarrhythmic effect of ibrutinib was also considered to be associated with augmented late sodium current, although APD prolongation was not identified in atrial cardiomyocytes isolated from ibrutinib-treated mice.84,87 In comparison, second-generation BTK inhibitors with better selectivity for BTK have been found not to increase the risk of AF and bleeding compared with ibrutinib.88

Immunotherapies

Immune escape is an important mechanism by which cancer cells avoid being detected and eliminated; immunotherapy, which modulates the immune system to regain the ability to kill cancer cells, has been developed recently. Cancer immunotherapies – predominantly CAR-T and ICIs – elevate the host immune response targeted to neoplastic cells, which has been effectively applied clinically.89

Immune Checkpoint Inhibitors

ICIs are monoclonal antibodies designed for immune checkpoint blockade that can upregulate T-cell-related immune responses to kill cancer cells. However, systematic intensification of T-cell immune response also leads to autoimmune reaction and damage. Although those adverse effects are mild and often reversible, cardiac adverse effects can be fatal.90 The incidence of ICI-induced AF varies vastly among different studies, from 1.1% to as high as 30%.21–25 The high diversity of incidence might be because of heterogeneity among patients enrolled in studies, and because some studies were not intended to investigate the incidence of cardiovascular complications such as AF.

ICI-related cardiotoxicity and myocarditis might predispose the heart to AF.91 Expression of immune checkpoints in the heart, while low under physiological conditions, is upregulated under stimuli such as inflammation, which can protect the heart from T-cell damage.92 The use of ICIs cancels this protective effect, therefore aggravating the impact of stimuli on the heart. It has also been suggested that neoplastic cells and the myocardium might share the same antigen as the target of T-cells. ICIs cause non-specific activation of the immune system and inflammatory cell infiltration in both neoplastic and cardiac tissue, which predisposes to AF.93

Chimeric Antigen Receptor T-cell Therapy

CAR-T therapy is a novel antineoplastic treatment in which patient’s own T cells are extracted, artificially engineered in vitro to enable them to recognise tumours specifically, multiplied and finally infused back into the patient to target cancer cells.94 Clinical research has demonstrated that CAR-T therapy can induce AF.95–97 CAR-T-induced cardiotoxicity is attributed to cytokine release syndrome, i.e., general activation of the immune system and resultant hypercytokinaemia.98 Marked supraphysiological circulating inflammatory molecules such as interleukin (IL)-6 and interferon-γ can lead to fever, vasodilation, hypotension, tachycardia and multiorgan dysfunction.99 It is postulated that IL-6 is critical in CAR-T-induced cardiotoxicity.100 Because IL-6 acts as a key biomarker of AF and elevated IL-6 contributes to generation and persistence of AF, it may link systematic inflammation with AF onset, accounting for CAR-T-induced AF.101

Drugs of Abuse

Alcohol

Alcohol abuse is a global health burden associated with many diseases.102 Among these, arrhythmias – particularly AF – have a strong correlation with alcohol consumption. The term ‘holiday heart syndrome’ was first introduced in the 1970s to characterise the emergence of dysrhythmias, particularly AF, in otherwise healthy individuals following acute alcohol consumption.103 Nevertheless, the incidence of AF subsequent to binge drinking in individuals without pre-existing AF is relatively low at approximately 0.8%.27 Persistent and heavy alcohol consumption is associated with higher AF burden; Han et al. reported a 25% elevated risk of incident AF associated with a higher cumulative alcohol burden over a 4-year period and up to 47% with sustained heavy drinking during the same timeframe.28 However, there is ongoing debate regarding the association between low to moderate alcohol consumption and AF.104–106

Mechanisms underlying AF – whether induced acutely or through chronic consumption – share certain common features but also exhibit distinctions. Acute alcohol ingestion precipitates reversible and self-terminating arrhythmias, induces autonomic imbalance and diminishes atrial ERP, particularly in the pulmonary veins.107 In contrast, chronic alcohol intake frequently results in enduring atrial remodelling, structural alterations, such as left-atrial enlargement, increased left-atrial volume and reduced left-atrial mechanical performance.108–110

Tobacco and Illicit Substances

Tobacco is one of the most detrimental drug substances abused globally. A large meta-analysis reported that tobacco smoking increased the risk of developing AF by 33% compared with never smokers.111 In addition, exposure to environmental tobacco smoke has also been linked to an increased prevalence of AF, even among non-smokers.112,113 Nicotine, the primary active ingredient in tobacco, has both addictive and sympathomimetic properties, which can trigger AF by stimulating sympathetic neurotransmission and increasing sympathetic activity.114 Nicotine is also profibrotic and the fibrosis provides the arrhythmogenic substrate.115

It has been suggested that marijuana use may also contribute to the development of AF, particularly in young individuals without significant cardiac comorbidities.116,117 The exact incidence of illicit substance-induced AF is difficult to determine for several reasons. First, it can be challenging to accurately determine the consumption of illicit substances in the real world, as there may be under-reporting or non-disclosure of drug use by individuals. Second, individuals with substance use disorders may be reluctant to seek medical attention or disclose their drug use to medical providers, further complicating the identification and diagnosis of illicit substance-induced AF. Finally, individuals who use illicit substances may often use multiple drugs of abuse simultaneously, making it difficult to attribute the onset of AF to a specific drug. Marijuana smoking resulted in immediate heart rate elevation and serum norepinephrine level increase, indicating sympathetic overactivity.118 The principal active metabolite of marijuana, δ-9-tetrahydrocannabinol, inhibits cAMP formation and decreases Ca2+ influx that, in turn, results in negative cardiac inotropy, vasodilation and hypotension by binding to the cannabinoid 1 receptor.119,120 Collectively, elevated sympathetic tone by marijuana smoking may be attributed to indirect action via cannabinoid 1 receptors.

Treatment of Drug-induced AF

It is essential to conduct more extensive studies to obtain a more precise estimate of the prevalence of DIAF and to identify the underlying mechanisms and risk factors associated with this condition. Such investigations are crucial for improving our understanding of DIAF and developing effective prevention and management strategies. There is no specific expert consensus for the treatment of DIAF, and its management is primary according to the current AF guideline.6 Because DIAF often occurs during acute hospitalisation, management strategies for DIAF can also refer to the recent scientific statement put forward by the American Heart Association.121 The principal goals of managing DIAF are to optimise haemodynamics, alleviate patient symptoms and reduce the short- and long-term risks of thromboembolism. Achieving these goals requires a comprehensive approach that includes identifying and discontinuing the causative drug, assessing and managing underlying risk factors and implementing appropriate pharmacological and non-pharmacological interventions. Individualised management strategies that consider comorbidities and drug therapy are recommended for optimal management of DIAF.

Table 2: Summary of Mechanisms Underlying Drug-induced AF Discussed in this Review

Article image

Figure 2: Mechanisms of Drug-induced AF

Article image

The first step is to find patients with DIAF and to establish a causative relationship between the suspected drug and AF, which can be challenging. Distinguishing between DIAF and other forms of AF can be difficult, especially in patients with multiple risk factors and comorbidities. Accurately ascribing the causality of DIAF requires a thorough evaluation of the patient’s medical history, medication usage and potential underlying mechanisms of the AF. Therefore, healthcare providers need to maintain a high level of awareness of DIAF and be knowledgeable about the various drugs that can cause or exacerbate AF.

Treatment strategies may include drug dose modification or withdrawal with adequate ECG monitoring for AF recurrence. In some cases, DIAF is transient, haemodynamically stable and spontaneously reverts to sinus rhythm (e.g. adenosine), which can be considered a benign condition with a favourable prognosis. For patients who fail to spontaneously convert to sinus rhythm after drug withdrawal, additional clinical interventions to control rate and rhythm are recommended according to current guidelines for haemodynamic optimisation as well as symptom relief. Patients can switch to other drugs with similar therapeutic effects but lower AF inducibility. However, drug discontinuation must be weighed against the risk of disease progression, particularly in cancer patients in whom use of the drug may be essential because of the seriousness of the disease being treated.31 For instance, despite a relatively high incidence of AF in patients treated with ibrutinib, patients tend to be generally manageable without discontinuation of ibrutinib and attempting to mitigate the risk of AF by reducing the dose of ibrutinib may have limited effectiveness.122,123

While there are currently no reports of DIAF leading to thrombogenesis and stroke, it is theoretically possible – particularly in cases of long-lasting DIAF. As a result, it may be appropriate to use the CHA2DS2-VASc scoring system to determine if anticoagulation therapy is necessary. However, determining the need for anticoagulation in cancer patients with DIAF can be challenging, as they are at a higher risk of both thrombosis and bleeding.31 While the CHA2DS2-VASc scoring system has not been validated in patients with malignancies, given their high risk of thrombosis, anticoagulation therapy may still be recommended. Additionally, a study of ivabradine-induced AF found that it did not affect the incidence of non-fatal stroke compared with placebo, suggesting that the CHA2DS2-VASc scoring system may not be sufficient in all situations to guide clinical practice.70 Therefore, a comprehensive assessment of stroke risk in patients with DIAF should incorporate multiple factors and be tailored to individual patient characteristics.

Close monitoring and follow-up are crucial in patients with DIAF in light of the concern of AF recurrence. This involves regular clinical assessments, ECG monitoring and medication adjustments as needed. Additionally, patients with DIAF should receive education on lifestyle modifications and avoidance of drugs that may trigger AF. As the underlying mechanisms of DIAF are not fully understood, ongoing research is necessary to improve our understanding of this condition and to develop more effective prevention and management strategies. By implementing close monitoring and follow-up, healthcare providers can optimise the management of DIAF and improve patient outcomes.

Conclusion

DIAF, a condition that can stem from a wide range of both cardiac and non-cardiac medications, is a prevailing clinical issue. The intricate mechanisms of DIAF include effects on ion channels, Ca2+ handling, ROS generation, inflammation and autonomic regulation of the heart (Table 2 and Figure 2). Unfortunately, DIAF is frequently overlooked and under-treated. To address this, a comprehensive understanding of the specific drug mechanisms and the individual patient’s needs and conditions are crucial for optimal management. As such, individualised approaches are recommended, with a focus on developing effective prevention and management measures. It is imperative to conduct future research investigating the underlying mechanisms of DIAF.

Clinical Perspective

  • Drug-induced AF (DIAF) is a prevalent condition that is often underestimated and overlooked and that can cause significant symptoms and outcomes.
  • The management of DIAF aims to optimise haemodynamics, alleviate patient symptoms and reduce the risks of thromboembolism. Close monitoring and follow-up are crucial because of the high risks of AF recurrence. Additionally, the CHA2DS2-VASc scoring system may not be sufficient in all situations to guide clinical practice.
  • Further research is needed to highlight the clinical significance of DIAF, clarify the underlying mechanisms and improve treatment strategies.

References

  1. Bai Y, Wang YL, Shantsila A, Lip GYH. The global burden of atrial fibrillation and stroke: a systematic review of the clinical epidemiology of atrial fibrillation in Asia. Chest 2017;152:810–20. 
    Crossref | Pubmed
  2. Staerk L, Sherer JA, Ko D, et al. Atrial fibrillation: epidemiology, pathophysiology, and clinical outcomes. Circ Res 2017;120:1501–17. 
    Crossref | Pubmed
  3. Lau DH, Nattel S, Kalman JM, Sanders P. Modifiable risk factors and atrial fibrillation. Circulation 2017;136:583–96. 
    Crossref | Pubmed
  4. Elliott AD, Middeldorp ME, Van Gelder IC, et al. Epidemiology and modifiable risk factors for atrial fibrillation. Nat Rev Cardiol 2023;20:404–17. 
    Crossref | Pubmed
  5. Al-Kaisey AM, Figgett W, Hawson J, et al. Gut microbiota and atrial fibrillation: pathogenesis, mechanisms and therapies. Arrhythm Electrophysiol Rev 2023;12:e14. 
    Crossref | Pubmed
  6. Joglar JA, Chung MK, Armbruster AL, et al. 2023 ACC/AHA/ACCP/HRS guideline for the diagnosis and management of atrial fibrillation: a report of the American College of Cardiology/American Heart Association joint committee on clinical practice guidelines. Circulation 2024;149:e1–156. 
    Crossref | Pubmed
  7. Tamargo J, Caballero R, Delpón E. Drug-induced atrial fibrillation. Expert Opin Drug Saf 2012;11:615–34. 
    Crossref | Pubmed
  8. van der Hooft CS, Heeringa J, van Herpen G, et al. Drug-induced atrial fibrillation. J Am Coll Cardiol 2004;44:2117–24. 
    Crossref | Pubmed
  9. Kaakeh Y, Overholser BR, Lopshire JC, Tisdale JE. Drug-induced atrial fibrillation. Drugs 2012;72:1617–30. 
    Crossref | Pubmed
  10. Tisdale JE, Chung MK, Campbell KB, et al. Drug-induced arrhythmias: a scientific statement from the American Heart Association. Circulation 2020;142:e214–33. 
    Crossref | Pubmed
  11. Feneck RO, Sherry KM, Withington PS, et al. Comparison of the hemodynamic effects of milrinone with dobutamine in patients after cardiac surgery. J Cardiothorac Vasc Anesth 2001;15:306–15. 
    Crossref | Pubmed
  12. Cuffe MS, Califf RM, Adams KF, et al. Short-term intravenous milrinone for acute exacerbation of chronic heart failurea randomized controlled trial. JAMA 2002;287:1541–7. 
    Crossref | Pubmed
  13. Kilickap S, Barista I, Akgul E, et al. Early and late arrhythmogenic effects of doxorubicin. South Med J 2007;100:262–5. 
    Crossref | Pubmed
  14. Amioka M, Sairaku A, Ochi T, et al. Prognostic significance of new-onset atrial fibrillation in patients with non-Hodgkin’s lymphoma treated with anthracyclines. Am J Cardiol 2016;118:1386–9. 
    Crossref | Pubmed
  15. Yun S, Vincelette ND, Acharya U, Abraham I. Risk of atrial fibrillation and bleeding diathesis associated with ibrutinib treatment: a systematic review and pooled analysis of four randomized controlled trials. Clin Lymphoma Myeloma Leuk 2017;17:31–37.e13. 
    Crossref | Pubmed
  16. Leong DP, Caron F, Hillis C, et al. The risk of atrial fibrillation with ibrutinib use: a systematic review and meta-analysis. Blood 2016;128:138–40. 
    Crossref | Pubmed
  17. Camaiti A, Pieralli F, Olivotto I, et al. Prospective evaluation of adenosine-induced proarrhythmia in the emergency room. Eur J Emerg Med 2001;8:99–105. 
    Crossref | Pubmed
  18. Strickberger SA, Man KC, Daoud EG, et al. Adenosine-induced atrial arrhythmia: a prospective analysis. Ann Intern Med 1997;127:417–22. 
    Crossref | Pubmed
  19. Yuan M, Tse G, Zhang Z, et al. The incidence of atrial fibrillation with trastuzumab treatment: a systematic review and meta-analysis. Cardiovasc Ther 2018;36:e12475. 
    Crossref | Pubmed
  20. Saito Y, Kitahara H, Shoji T, et al. Paroxysmal atrial fibrillation during intracoronary acetylcholine provocation test. Heart Vessels 2017;32:902–8. 
    Crossref | Pubmed
  21. Brumberger ZL, Branch ME, Klein MW, et al. Cardiotoxicity risk factors with immune checkpoint inhibitors. Cardiooncology 2022;8:3. 
    Crossref | Pubmed
  22. Mascolo A, Scavone C, Ferrajolo C, et al. Immune checkpoint inhibitors and cardiotoxicity: an analysis of spontaneous reports in eudravigilance. Drug Saf 2021;44:957–71. 
    Crossref | Pubmed
  23. Nso N, Antwi-Amoabeng D, Beutler BD, et al. Cardiac adverse events of immune checkpoint inhibitors in oncology patients: a systematic review and meta-analysis. World J Cardiol 2020;12:584–98. 
    Crossref | Pubmed
  24. Escudier M, Cautela J, Malissen N, et al. Clinical features, management, and outcomes of immune checkpoint inhibitor-related cardiotoxicity. Circulation 2017;136:2085–7. 
    Crossref | Pubmed
  25. Joseph L, Nickel AC, Patel A, et al. Incidence of cancer treatment induced arrhythmia associated with immune checkpoint inhibitors. J Atr Fibriliation 2021;13:2461. 
    Crossref | Pubmed
  26. Goldman A, Maor E, Bomze D, et al. Adverse cardiovascular and pulmonary events associated with chimeric antigen receptor T-cell therapy. J Am Coll Cardiol 2021;78:1800–13. 
    Crossref | Pubmed
  27. Brunner S, Herbel R, Drobesch C, et al. Alcohol consumption, sinus tachycardia, and cardiac arrhythmias at the Munich Octoberfest: results from the Munich beer Related Electrocardiogram Workup study (MunichBREW). Eur Heart J 2017;38:2100–6. 
    Crossref | Pubmed
  28. Han M, Lee SR, Choi EK, et al. Habitual alcohol intake and risk of atrial fibrillation in young adults in Korea. JAMA Netw Open 2022;5:e2229799. 
    Crossref | Pubmed
  29. Doshi R, Dave M, Majmundar M, et al. National rates and trends of tobacco and substance use disorders among atrial fibrillation hospitalizations. Heart Lung 2021;50:244–51. 
    Crossref | Pubmed
  30. Mazur M, Wang F, Hodge DO, et al. Burden of cardiac arrhythmias in patients with anthracycline-related cardiomyopathy. JACC Clin Electrophysiol 2017;3:139–50. 
    Crossref | Pubmed
  31. Alexandre J, Salem JE, Moslehi J, et al. Identification of anticancer drugs associated with atrial fibrillation: analysis of the who pharmacovigilance database. Eur Heart J Cardiovasc Pharmacother 2021;7:312–20. 
    Crossref | Pubmed
  32. Wit AL, Boyden PA. Triggered activity and atrial fibrillation. Heart Rhythm 2007;4(Suppl):S17–23. 
    Crossref | Pubmed
  33. Nattel S, Heijman J, Zhou L, Dobrev D. Molecular basis of atrial fibrillation pathophysiology and therapy: a translational perspective. Circ Res 2020;127:51–72. 
    Crossref | Pubmed
  34. Comtois P, Kneller J, Nattel S. Of circles and spirals: bridging the gap between the leading circle and spiral wave concepts of cardiac reentry. Europace 2005;7(Suppl 2):10–20. 
    Crossref | Pubmed
  35. Aguilar M, Nattel S. The pioneering work of George Mines on cardiac arrhythmias: groundbreaking ideas that remain influential in contemporary cardiac electrophysiology. J Physiol 2016;594:2377–86. 
    Crossref | Pubmed
  36. Diker E, Ozdemir M, Aydoğdu S, et al. Dispersion of repolarization in paroxysmal atrial fibrillation. Int J Cardiol 1998;63:281–6. 
    Crossref | Pubmed
  37. Linz D, Elliott AD, Hohl M, et al. Role of autonomic nervous system in atrial fibrillation. Int J Cardiol 2019;287:181–8. 
    Crossref | Pubmed
  38. Bers DM. Cardiac excitation–contraction coupling. Nature 2002;415:198–205. 
    Crossref | Pubmed
  39. Huerta C, Lanes SF, García Rodríguez LA. Respiratory medications and the risk of cardiac arrhythmias. Epidemiol (Camb Mass) 2005;16:360–6. 
    Crossref | Pubmed
  40. Mansencal N, Mustafic H, Hauguel-Moreau M, et al. Occurrence of atrial fibrillation during dobutamine stress echocardiography. Am J Cardiol 2019;123:1277–82. 
    Crossref | Pubmed
  41. Lin Z, Wong LYF, Cheung BMY. Diuretic-induced hypokalaemia: an updated review. Postgrad Med J 2022;98:477–82. 
    Crossref | Pubmed
  42. Kardalas E, Paschou SA, Anagnostis P, et al. Hypokalemia: a clinical update. Endocr Connect 2018;7:R135–46. 
    Crossref | Pubmed
  43. Emara MK, Saadet AM. Transient atrial fibrillation in hypertensive patients with thiazide induced hypokalaemia. Postgrad Med J 1986;62:1125–7. 
    Crossref | Pubmed
  44. Lin DW, Jiang F, Wu C, et al. Association of furosemide or hydrochlorothiazide use with risk of atrial fibrillation post pacemaker implantation among elderly patients. Ann Transl Med 2021;9:855. 
    Crossref | Pubmed
  45. Filippatos G, Bakris GL, Pitt B, et al. Finerenone reduces new-onset atrial fibrillation in patients with chronic kidney disease and type 2 diabetes. J Am Coll Cardiol 2021;78:142–52. 
    Crossref | Pubmed
  46. Psotka MA, Gottlieb SS, Francis GS, et al. Cardiac calcitropes, myotropes, and mitotropes: JACC review topic of the week. J Am Coll Cardiol 2019;73:2345–53. 
    Crossref | Pubmed
  47. Fleming GA, Murray KT, Yu C, et al. Milrinone use is associated with postoperative atrial fibrillation after cardiac surgery. Circulation 2008;118:1619–25. 
    Crossref | Pubmed
  48. Ma G, Brady WJ, Pollack M, Chan TC. Electrocardiographic manifestations: digitalis toxicity. J Emerg Med 2001;20:145–52. 
    Crossref | Pubmed
  49. Fisch C, Knoebel SB. Digitalis cardiotoxicity. J Am Coll Cardiol 1985;5(Suppl A):91A–8A. 
    Crossref | Pubmed
  50. Lee CC, Chang SN, Tehrani B, et al. Use of nicorandil is associated with increased risk of incident atrial fibrillation. Aging 2022;14:6975–92. 
    Crossref | Pubmed
  51. Frommeyer G, Eckardt L. Drug-induced proarrhythmia: risk factors and electrophysiological mechanisms. Nat Rev Cardiol 2016;13:36–47. 
    Crossref | Pubmed
  52. Krapivinsky G, Gordon EA, Wickman K, et al. The G-protein-gated atrial K+ channel IKACH is a heteromultimer of two inwardly rectifying K(+)-channel proteins. Nature 1995;374:135–41. 
    Crossref | Pubmed
  53. Kabell G, Buchanan LV, Gibson JK, Belardinelli L. Effects of adenosine on atrial refractoriness and arrhythmias. Cardiovasc Res 1994;28:1385–9. 
    Crossref | Pubmed
  54. Li N, Csepe TA, Hansen BJ, et al. Adenosine-induced atrial fibrillation: localized reentrant drivers in lateral right atria due to heterogeneous expression of adenosine A1 receptors and GIRK4 subunits in the human heart. Circulation 2016;134:486–98. 
    Crossref | Pubmed
  55. Turley AJ, Murray S, Thambyrajah J. Pre-excited atrial fibrillation triggered by intravenous adenosine: a commonly used drug with potentially life-threatening adverse effects. Emerg Med J 2008;25:46–8. 
    Crossref | Pubmed
  56. Kurt IH, Yigit T, Karademir BM. Atrial fibrillation due to late amiodarone-induced thyrotoxicosis. Clin Drug Investig 2008;28:527–31. 
    Crossref | Pubmed
  57. Schreiber DH, DeFreest MS. Paroxysmal atrial fibrillation precipitated by amiodarone-induced thyrotoxicosis five months after cessation of therapy. J Emerg Med 2006;31:61–4. 
    Crossref | Pubmed
  58. Trohman RG, Sharma PS, McAninch EA, Bianco AC. Amiodarone and thyroid physiology, pathophysiology, diagnosis and management. Trends Cardiovasc Med 2019;29:285–95. 
    Crossref | Pubmed
  59. Freemantle N, Lafuente-Lafuente C, Mitchell S, et al. Mixed treatment comparison of dronedarone, amiodarone, sotalol, flecainide, and propafenone, for the management of atrial fibrillation. Europace 2011;13:329–45. 
    Crossref | Pubmed
  60. Le Heuzey JY, De Ferrari GM, Radzik D, et al. A short-term, randomized, double-blind, parallel-group study to evaluate the efficacy and safety of dronedarone versus amiodarone in patients with persistent atrial fibrillation: the Dionysos study. J Cardiovasc Electrophysiol 2010;21:597–605. 
    Crossref | Pubmed
  61. Ide T, Ohtani K, Higo T, et al. Ivabradine for the treatment of cardiovascular diseases. Circ J 2019;83:252–60. 
    Crossref | Pubmed
  62. Marciszek M, Paterek A, Oknińska M, et al. Effect of ivabradine on cardiac arrhythmias: antiarrhythmic or proarrhythmic? Heart Rhythm 2021;18:1230–8. 
    Crossref | Pubmed
  63. Fontenla A, Tamargo J, Salgado R, et al. Ivabradine for controlling heart rate in permanent atrial fibrillation: a translational clinical trial. Heart Rhythm 2023;20:822–30. 
    Crossref | Pubmed
  64. Xu X, Guo Y, Gao W, et al. Ivabradine monotherapy in pediatric patients with focal atrial tachycardia: a single-center study. Eur J Pediatr 2023;182:2265–71. 
    Crossref | Pubmed
  65. Wang Z, Wang W, Li H, et al. Ivabradine and atrial fibrillation: a meta-analysis of randomized controlled trials. J Cardiovasc Pharmacol 2022;79:549–57. 
    Crossref | Pubmed
  66. Suenari K, Cheng CC, Chen YC, et al. Effects of ivabradine on the pulmonary vein electrical activity and modulation of pacemaker currents and calcium homeostasis. J Cardiovasc Electrophysiol 2012;23:200–6. 
    Crossref | Pubmed
  67. Li YD, Ji YT, Zhou XH, et al. Effects of ivabradine on cardiac electrophysiology in dogs with age-related atrial fibrillation. Med Sci Monit 2015;21:1414–20. 
    Crossref | Pubmed
  68. Frommeyer G, Sterneberg M, Dechering DG, et al. Effective suppression of atrial fibrillation by ivabradine: novel target for an established drug? Int J Cardiol 2017;236:237–43. 
    Crossref | Pubmed
  69. Wang J, Yang YM, Li Y, et al. Long-term treatment with ivabradine in transgenic atrial fibrillation mice counteracts hyperpolarization-activated cyclic nucleotide gated channel overexpression. J Cardiovasc Electrophysiol 2019;30:242–52. 
    Crossref | Pubmed
  70. Fox K, Ford I, Steg PG, et al. Bradycardia and atrial fibrillation in patients with stable coronary artery disease treated with ivabradine: an analysis from the signify study. Eur Heart J 2015;36:3291–6. 
    Crossref | Pubmed
  71. Uemura K, Inagaki M, Zheng C, et al. Acute ivabradine treatment reduces heart rate without increasing atrial fibrillation inducibility irrespective of underlying vagal activity in dogs. Heart Vessels 2017;32:484–94. 
    Crossref | Pubmed
  72. Tan R, Cong T, Xu G, et al. Anthracycline-induced atrial structural and electrical remodeling characterizes early cardiotoxicity and contributes to atrial conductive instability and dysfunction. Antioxid Redox Signal 2022;37:19–39. 
    Crossref | Pubmed
  73. Zhang S, Liu X, Bawa-Khalfe T, et al. Identification of the molecular basis of doxorubicin-induced cardiotoxicity. Nat Med 2012;18:1639–42. 
    Crossref | Pubmed
  74. Odiete O, Hill MF, Sawyer DB. Neuregulin in cardiovascular development and disease. Circ Res 2012;111:1376-1385.
    Pubmed
    https://10.1161/circresaha.112.267286
  75. Leemasawat K, Phrommintikul A, Chattipakorn SC, Chattipakorn N. Mechanisms and potential interventions associated with the cardiotoxicity of ErbB2-targeted drugs: insights from in vitro, in vivo, and clinical studies in breast cancer patients. Cell Mol Life Sci 2020;77:1571–89. 
    Crossref | Pubmed
  76. Timolati F, Ott D, Pentassuglia L, et al. Neuregulin-1 beta attenuates doxorubicin-induced alterations of excitation-contraction coupling and reduces oxidative stress in adult rat cardiomyocytes. J Mol Cell Cardiol 2006;41:845–54. 
    Crossref | Pubmed
  77. Gordon LI, Burke MA, Singh AT, et al. Blockade of the ErbB2 receptor induces cardiomyocyte death through mitochondrial and reactive oxygen species-dependent pathways. J Biol Chem 2009;284:2080–7. 
    Crossref | Pubmed
  78. 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;44:2231–8. 
    Crossref | Pubmed
  79. Burger JA, Tedeschi A, Barr PM, et al. Ibrutinib as initial therapy for patients with chronic lymphocytic leukemia. N Engl J Med 2015;373:2425–37. 
    Crossref | Pubmed
  80. Paydas S. Management of adverse effects/toxicity of ibrutinib. Crit Rev Oncol Hematol 2019;136:56-63. 
    Crossref | Pubmed
  81. Honigberg LA, Smith AM, Sirisawad M, et al. The Bruton tyrosine kinase inhibitor PCI-32765 blocks B-cell activation and is efficacious in models of autoimmune disease and B-cell malignancy. Proc Natl Acad Sci U S A 2010;107:13075–80. 
    Crossref | Pubmed
  82. McMullen JR, Boey EJ, Ooi JY, et al. Ibrutinib increases the risk of atrial fibrillation, potentially through inhibition of cardiac PI3K-Akt signaling. Blood 2014;124:3829–30. 
    Crossref | Pubmed
  83. Chen J, Kinoshita T, Sukbuntherng J, et al. Ibrutinib inhibits ERBB receptor tyrosine kinases and HER2-amplified breast cancer cell growth. Mol Cancer Ther 2016;15:2835–44. 
    Crossref | Pubmed
  84. Xiao L, Salem JE, Clauss S, et al. Ibrutinib-mediated atrial fibrillation attributable to inhibition of C-terminal Src kinase. Circulation 2020;142:2443–55. 
    Crossref | Pubmed
  85. Pretorius L, Du XJ, Woodcock EA, et al. Reduced phosphoinositide 3-kinase (p110alpha) activation increases the susceptibility to atrial fibrillation. Am J Pathol 2009;175:998–1009. 
    Crossref | Pubmed
  86. Weeks KL, Gao X, Du XJ, et al. Phosphoinositide 3-kinase p110α is a master regulator of exercise-induced cardioprotection and PI3K gene therapy rescues cardiac dysfunction. Circ Heart Fail 2012;5:523–34. 
    Crossref | Pubmed
  87. Tao Yang, Javid J Moslehi, Dan M Roden. Proarrhythmic Effects of ibrutinib, a clinically approved inhibitor of Bruton’s tyrosine kinase (BTK) used in cancer therapy. Presented at: American Heart Association Scientific Sessions, Orlando, FL, US, 6 November 2015. Abstract number 14587.
  88. Patel V, Balakrishnan K, Bibikova E, et al. Comparison of acalabrutinib, a selective Bruton tyrosine kinase inhibitor, with ibrutinib in chronic lymphocytic leukemia cells. Clin Cancer Res 2017;23:3734–43. 
    Crossref | Pubmed
  89. Waldman AD, Fritz JM, Lenardo MJ. A guide to cancer immunotherapy: from T cell basic science to clinical practice. Nat Rev Immunol 2020;20:651–68. 
    Crossref | Pubmed
  90. Lyon AR, Yousaf N, Battisti NML, et al. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncol 2018;19:e447–58. 
    Crossref | Pubmed
  91. Fradley MG, Beckie TM, Brown SA, et al. Recognition, prevention, and management of arrhythmias and autonomic disorders in cardio-oncology: a scientific statement from the American Heart Association. Circulation 2021;144:e41–55. 
    Crossref | Pubmed
  92. Grabie N, Lichtman AH, Padera R. T cell checkpoint regulators in the heart. Cardiovasc Res 2019;115:869–77. 
    Crossref | Pubmed
  93. Palaskas N, Lopez-Mattei J, Durand JB, et al. Immune checkpoint inhibitor myocarditis: pathophysiological characteristics, diagnosis, and treatment. J Am Heart Assoc 2020;9:e013757. 
    Crossref | Pubmed
  94. Holstein SA, Lunning MA. Car T-cell therapy in hematologic malignancies: a voyage in progress. Clin Pharmacol Ther 2020;107:112–22. 
    Crossref | Pubmed
  95. Alvi RM, Frigault MJ, Fradley MG, et al. Cardiovascular events among adults treated with chimeric antigen receptor T-cells (CAR-T). J Am Coll Cardiol 2019;74:3099–108. 
    Crossref | Pubmed
  96. Davila ML, Riviere I, Wang X, et al. Efficacy and toxicity management of 19–28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci Transl Med 2014;6:224ra25. 
    Crossref | Pubmed
  97. Lefebvre B, Kang Y, Smith AM, et al. Cardiovascular effects of CAR T cell therapy: a retrospective study. JACC CardioOncol 2020;2:193–203. 
    Crossref | Pubmed
  98. Oved JH, Barrett DM, Teachey DT. Cellular therapy: immune-related complications. Immunol Rev 2019;290:114–26. 
    Crossref | Pubmed
  99. Kennedy LB, Salama AKS. A review of cancer immunotherapy toxicity. CA Cancer J Clin 2020;70:86–104. 
    Crossref | Pubmed
  100. Ganatra S, Carver JR, Hayek SS, et al. Chimeric antigen receptor T-cell therapy for cancer and heart: JACC council perspectives. J Am Coll Cardiol 2019;74:3153–63. 
    Crossref | Pubmed
  101. Issac TT, Dokainish H, Lakkis NM. Role of inflammation in initiation and perpetuation of atrial fibrillation: a systematic review of the published data. J Am Coll Cardiol 2007;50:2021–8. 
    Crossref | Pubmed
  102. GBD 2016 Alcohol Collaborators. Alcohol use and burden for 195 countries and territories, 1990–2016: a systematic analysis for the Global Burden of Disease study 2016. Lancet 2018;392:1015–35. 
    Crossref | Pubmed
  103. Ettinger PO, Wu CF, De La Cruz C, Jr, et al. Arrhythmias and the “holiday heart”: alcohol-associated cardiac rhythm disorders. Am Heart J 1978;95:555–62. 
    Crossref | pubmed
  104. Gémes K, Malmo V, Laugsand LE, et al. Does moderate drinking increase the risk of atrial fibrillation? The Norwegian HUNT (Nord-Trøndelag Health) study. J Am Heart Assoc 2017;6:e007094. 
    Crossref | Pubmed
  105. Larsson SC, Drca N, Wolk A. Alcohol consumption and risk of atrial fibrillation: a prospective study and dose-response meta-analysis. J Am Coll Cardiol 2014;64:281–9. 
    Crossref | Pubmed
  106. Di Castelnuovo A, Costanzo S, Bonaccio M, et al. Moderate alcohol consumption is associated with lower risk for heart failure but not atrial fibrillation. JACC. Heart Fail 2017;5:837–44. 
    Crossref | Pubmed
  107. Marcus GM, Dukes JW, Vittinghoff E, et al. A randomized, double-blind, placebo-controlled trial of intravenous alcohol to assess changes in atrial electrophysiology. JACC Clin Electrophysiol 2021;7:662–70. 
    Crossref | Pubmed
  108. McManus DD, Yin X, Gladstone R, et al. Alcohol consumption, left atrial diameter, and atrial fibrillation. J Am Heart Assoc 2016;5:e004060. 
    Crossref | Pubmed