CC BY-NC-ND 4.0 · South Asian J Cancer 2021; 10(03): 195-210
DOI: 10.1055/s-0041-1731907
Cardio-Oncology

Bradyarrhythmias in Cardio-Oncology

Marta Fonseca
1   Division of Cardiology, Cardiac-Oncology Service, Bart’s Heart Centre, St Bartholomew’s Hospital West Smithfield, London, United Kingdom
2   Hatter Cardiovascular Institute, Institute of Cardiovascular Science UCL, University College London Hospital, London, United Kingdom
,
3   UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, California, United States
,
Duc Do
3   UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, California, United States
,
Shouvik Haldar
4   Division of Cardiology, Heart Rhythm Centre, The Royal Brompton and Harefield Hospitals, Guys & St Thomas’ NHS Foundation Trust, London, United Kingdom
5   National Heart and Lung Institute, Imperial College, London, United Kingdom
,
Shelby Kutty
6   The Helen B. Taussig Heart Center, The Johns Hopkins Hospital and Johns Hopkins University, Baltimore, Maryland, United States
,
Eric H. Yang
3   UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, California, United States
,
Arjun K. Ghosh#
1   Division of Cardiology, Cardiac-Oncology Service, Bart’s Heart Centre, St Bartholomew’s Hospital West Smithfield, London, United Kingdom
2   Hatter Cardiovascular Institute, Institute of Cardiovascular Science UCL, University College London Hospital, London, United Kingdom
,
Avirup Guha#
7   Harrington Heart and Vascular Institute, Case Western Reserve University, Cleveland, Ohio, United States
8   Division of Cardiology, Department of Medicine, Augusta University, Augusta, Georgia, United States
9   Division of Cardiology-Oncology Program, The Ohio State University, Columbus, Ohio, United States
› Institutsangaben
Funding None.
 

Abstract

The relationship between bradyarrhythmias and cancer therapies has not been well described but is increasingly recognized. There have been extensive advances in oncological pharmacotherapy, with several new classes of drugs available including targeted agents, immune checkpoint inhibitors and CAR T cell therapy. This increasing repertoire of available drugs has revolutionized overall prognosis and survival of cancer patients but the true extent of their cardiovascular toxicity is only beginning to be understood. Previous studies and published reviews have traditionally focused on conventional chemotherapies and in arrhythmias in general, particularly tachyarrhythmias. The number of patients with both cancer and cardiovascular problems is increasing globally and oncologists and cardiologists need to be adept at managing arrythmia based scenarios. Greater collaboration between the two specialties including studies with prospective data collection in Cardio-Oncology are much needed to fill in knowledge gaps in this arena. This case-based review summarizes current available evidence of cancer treatment-related bradyarrhythmia incidence (including its different subtypes), possible mechanisms and outcomes. Furthermore, we propose a stepwise surveillance and management protocol for patients with suspected bradyarrhythmia related to cancer treatment.


#
Zoom Image
Avirup Guha

Case 1: Bradycardia with Antithymocyte Globulin

A 30-year-old male with aplastic anemia started treatment with antithymocyte globulin (ATG). He had no significant cardiovascular risk factors or cardiovascular disease. On the first ATG cycle, his heart rate dropped to 40 bpm (no electrocardiogram recorded at this point). He remained asymptomatic from the cardiac perspective. The Oncology team referred the patient to the Cardio-Oncology clinic. The echocardiogram and the 24-hour Holter monitoring performed 7 days after the bradycardic episode were normal. Bradycardia is an uncommon cardiotoxic sequelae of ATG, with current data limited to some case reports and one series.[1] We suggested admission with cardiac monitoring during the next ATG cycle.

Bradycardia and Bradyarrhythmias with Conventional Chemotherapies and Targeted Agents

Bradyarrhythmias have been reported in patients treated with alkylating agents. Four out of 39 patients with breast cancer, previously treated with anthracyclines and receiving high-dose chemotherapy with cyclophosphamide before stem cell transplantation, developed some degree of atrioventricular block (AVB).[2] Two of them developed complete heart block (CHB) managed with isoproterenol infusion, but in all cases, the conduction abnormality was transient, and the patients were able to complete the scheduled regimen. Nevertheless, other reports of CHB required discontinuation of cyclophosphamide and/or temporary as well as permanent pacemaker implantation.[3] [4] Arrhythmias related to alkylating agents are thought to arise from direct cardiotoxicity to myocytes and ischemic damage caused by coronary vasospasm and vascular endothelial injury with subsequent intracapillary microthrombi. An increased vagal tone has also been proposed as a mechanism given that some patients have uncontrolled emesis associated with CHB.[5] [6]

Anthracyclines are traditionally associated with cardiomyopathy/left ventricular dysfunction, with arrhythmias resulting from the former. In a prospective study of patients receiving doxorubicin, 3.4% developed bradycardia detected by 24-hour Holter monitoring with one patient developing CHB requiring permanent pacing.[7] There have been no specific studies addressing the mechanisms of bradyarrhythmias due to anthracyclines. Anthracyclines can have a direct electrophysiological effect by interfering with cardiac Ca2+ channels. They also generate, lead to the accumulation of and increase cardiomyocyte’s susceptibility to reactive oxygen species. Abnormal Ca2+ homeostasis due to abnormal functioning of sarcoplasmic reticulum calcium–ATPase can cause intracellular Ca2+ overload and subsequent myocardial necrosis. Reduction in adenosine monophosphate-activated protein kinase expression can lead to myocardial apoptosis. Cardiac accumulation of doxorubicinol, a toxic metabolite of doxorubicin, may be involved in some of these mechanisms.[5] [6]

Cisplatin infusion is associated with sinus bradycardia (SB) which can start immediately during infusion, can be marked, and tends to recur with each cycle.[8] [9] [10] [11] Coronary vasospasm, endothelial damage, and electrolyte disturbances are postulated mechanisms for cisplatin cardiotoxicity.[5] [6] Cisplatin deposition in the sinoatrial (SA) node and the vasculature supplying it are likely to be the cause of the bradycardia.[8]

There have been isolated case reports of SB related to 5-fluorouracil (5-FU) treatment with a published case series of six patients that developed asymptomatic SB with a heart rate below 50 bpm. Two of them required repeated interruptions of the infusion, and two of them required discontinuation of the drug due to sustained bradycardia in the second cycle of drug administration.[12] In a second case series of six patients, one of them developed Mobitz type I and another one SB with first-degree AVB. Interestingly there were no ST-segment changes, and troponin levels were normal.[13] A relatively large retrospective study showed that 5-FU induced SB occurred in 12% of the patients (36 out of 301 patients), and it was the most common cardiotoxicity.[14] In a prospective study including patients treated with either 5-FU or oral capecitabine, 26 out of 644 (4.03%) had cardiotoxicity, and three of them had conduction abnormalities that included either Mobitz type II or CHB, with one death occurring because of high-grade AVB and subsequent cardiac arrest.[15] The classic mechanism of 5-FU mediated cardiotoxicity is coronary vasospasm either through a direct toxic effect involving endothelial nitric oxide synthase or through endothelium-independent protein kinase C vasoconstriction. Another postulated mechanism is ischemia due to arteritis and/or thrombosis.[5] [6] There is a paucity of data regarding bradycardia and other drugs of the antimetabolites class with only one case report of symptomatic SB with cytarabine.[16]

The most common paclitaxel-related arrhythmia is SB, which can occur in as much as 29% of the patients (13 out of 45 patients in one prospective study) but is usually transient and asymptomatic.[17] More advanced AVB is infrequent. In the National Cancer Institute database, including more than 4,000 patients, only four had second- or third-degree AVB. Paclitaxel-induced conduction abnormalities might be explained by reduced coronary blood flow due to vasoconstriction. Histamine-induced release can also cause bradycardia and atrioventricular (AV) conduction delays.[5] [6]

CHB requiring pacing to continue arsenic trioxide treatment has been described, but it is infrequent, and the most common described cardiotoxicity of this drug still is QT interval changes.[18] Arsenic trioxide increases Ca2+ currents, inhibits K+ channels, and reduces surface expression of the human ether-a-go-go-related K+ channels, although these actions are more clearly related to the QTc changes previously mentioned.[5] [6]

Isolated cases of varying degrees of AVB have been described with interleukin-2, and it has been speculated that lymphoid infiltration of the AV node or the conduction system of the heart might be at its origin.[19]

In a phase II multicenter study, three out of 131 patients with mantle-cell lymphoma developed bradycardia related with rituximab, requiring some form of treatment.[20] Two case reports of CHB have been published, but one of the patients had previous conduction disease, and the other was an aged patient; hence the causality is difficult to ascertain.[21] [22] It was postulated that rituximab might affect the cardiac conduction system by inhibiting Ca2+ channel properties of the CD20 antigen on cardiac myocytes.[22]

In an extension of a phase II study, including 63 relapsed multiple myeloma patients treated with bortezomib, one case of nonspecified and CHB was reported.[23] In another study, two out of 69 patients with multiple myeloma treated with bortezomib developed nonspecified and CHB, leading to pacemaker implantation.[24] Bortezomib upregulates the adrenergic G-protein-coupled receptor kinase 2 (GRK2), which can predispose to bradyarrhythmia and AV conduction abnormalities.[5] [6]

SB is a common cardiotoxicity of thalidomide and occurred in as many as 53% of the patients included in one study. Although it usually resolved with discontinuation of the drug, some patients required pacemaker implantation.[25] [26] AVB is limited to rare case reports.[27] It has been hypothesized that thalidomide inhibits dorsal motor neurons of the nucleus of the vagus nerve by reducing TNF-alfa levels, with subsequent parasympathetic over-reactivity. Bradycardia has also been associated with thalidomide-induced hypothyroidism.[5] [6]

Tyrosine kinase inhibitors (TKIs) are implicated in the modulation of growth factor signaling and, as such, play an essential role in targeted cancer therapy. However, they are not entirely selective for protein kinases in cancer cells. SB is therefore observed with various TKIs but is particularly common with ALK-TKIs. In a retrospective analysis of two multinational trials, SB occurred in 42% of the patients treated with crizotinib, with an average decrease in heart rate of 25 bpm. The likelihood of experiencing SB was higher among patients with a pretreatment heart rate of less than 70 bpm.[28] Alectinib treatment resulted in a mean decrease of heart rate of 11 to 13 beats per minute. In approximately 5% of the patients, bradycardia or SB’s adverse events were reported, but when analyzing electrocardiograms (ECG), 20% of the patients had heart rates below 50 bpm. Bradycardia is usually transient and asymptomatic and does not require discontinuation of the drug.[29] The occurrence of AVB needing further management is low. In a phase II trial of 295 patients who received lorlatinib, 1% experienced AVB, and 0.3% experienced Grade 3 AVB and underwent pacemaker placement.[30] Symptomatic bradyarrhythmias that led to pacemaker implantation occurred in 1% (three out of 449) of the patients treated with ponatinib.[31] Arrhythmias including AVB are described as common with the use of nilotinib, but the specific percentage of each arrhythmia type is not specified.[32]

[Table 1] summarizes the type and incidence of bradycardia/bradyarrhythmia with each class of agents, as well as the mechanism behind it.

Table 1

Bradycardia/bradyarrhythmia type, incidence, and mechanism with conventional chemotherapy and targeted agents5,686

Agent

Adverse event

Incidence of adverse event

Mechanism

Sinus bradycardia

Atrioventricular block

Abbreviations: AVB, atrioventricular block; FU, fluorouracil; GRK2, G-protein-coupled receptor kinase 2; ND, not defined; SB, sinus bradycardia.

Alkylating agents

Cyclophosphamide

+

+

ND

Endothelial damage, myocardial ischemia, direct cardiotoxicity to myocytes, increased vagal tone.

Ifosfamide

+

ND

Busulfan

+

ND

Anthracyclines

+

+

Bradycardia 3.4%

Abnormal Ca2+ homeostasis, cardiac apoptosis, mitochondrial injury, oxidative stress.

Cisplatin

+

+

<1%

Coronary artery vasospasm, electrolyte disturbances, autonomic nervous system imbalance.

Antimetabolites

5-FU

+

+

SB up to 12%, AVB ND

Coronary ischemia (vasospasm, thrombosis due to endothelial damage).

Capecitabine

+

+

SB in 4.03% (5-FU or Capecitabine), AVB ND

Clofarabine

+

ND

Cytarabine

+

ND

Fludarabine

+

ND

Pentostatin

+

ND

Paclitaxel

+

+

SB up to 29%, AVB 0.11%

Reduced coronary blood flow due to vasoconstriction, histamine- induced release.

Arsenic trioxide

+

ND

Hman ether-a-go-go-related gene (hERG) channel blockade, abnormal Ca2+ homeostasis.

Interleukin-2

+

+

ND

Lymphoid infiltration of the atrioventricular node or the conduction system.

Monoclonal antibodies

Rituximab

+

+

<1%

Inhibition of calcium channel properties of the CD20 antigen on cardiac myocytes.

Alemtuzumab

+

SB 1–10%

Cetuximab

+

SB <1%

Pertuzumab

+

SB <1%

Trastuzumab

+

SB <1%

Proteasome inhibitors

Bortezomib

+

+

ND

Upregulation of GRK2

Carfilzomib

+

+

ND

Immunomodulatory drugs

Thalidomide

+

+

SB >10%, AVB ND

Parasympathetic over- reactivity, thalidomide-induced hypothyroidism

Lenalidomide

+

ND

Multi-target kinase inhibitors (TKIs)

Alectinib

+

SB 5.1–20%

Ceritinib

+

SB 3%

Crizotinib

+

SB 5–69%

Brigatinib

+

Bradycardia 5.7–8.1%

Lorlatinib

+

ND

Nilotinib

+

+

Bradycardia 1–10%

Ponatinib

+

+

Symptomatic bradycardia 1%

Bosutinib

+

ND

Pazopanib

+

Bradycardia 19%

Sorafenib

+

+

<1%

Sunitinib

+

<1%

Vemurafenib

+

<1%

Trametinib

+

1–19%

Ruxolitinib

+

<1%

Histone deacetylase inhibitors

Panobinostat

+

ND

Romidepsin

+

ND

Vorinostat

+

ND

Amsacrine

+

ND


#
#

Case 2: CHB with Pembrolizumab

A 65-year-old male with a background of hypertension, diabetes, obesity, and smoking history was diagnosed with metastatic gastric adenocarcinoma and was receiving palliative treatment with pembrolizumab. He then presented to the emergency department with a 2-day history of progressive worsening exertional dyspnea leading to shortness of breath at rest, lightheadedness, and dizziness. The patient received two cycles of pembrolizumab beginning 6 weeks prior. On admission, vital signs were stable, and a physical exam was notable for irregular heart rhythm and trace bilateral lower extremity pitting edema. The ECG showed CHB with a heart rate of approximately 50 bpm and multiple premature ventricular contractions ( [Fig. 1] ). Cardiac troponin I, creatinine kinase, liver enzymes, and C-reactive protein were elevated. A bedside echocardiogram revealed mildly impaired systolic function (left ventricular ejection fraction [LVEF] approximately 50% with anteroseptal hypokinesis with an LVEF of 65% from a prior echocardiogram). Given concerns for acute coronary syndrome (ACS) with electrical instability, the catheterization laboratory was activated. After input from both the cardiology and oncology teams, the patient received only 1 mg/kg prednisone before the procedure for empiric ICI myocarditis to avoid the risks of pulse dose steroids if the final diagnosis was, in fact, an ACS. The patient was found to have nonobstructive coronary artery disease. A temporary–permanent pacemaker was placed in conjunction with immediate pulse IV methylprednisolone treatment in the catheterization laboratory. After additional immunosuppressive therapy (mycophenolate mofetil) and intravenous immunoglobulin therapy, the patient’s AV conduction returned to normal after 1 week, and the pacemaker was removed.

Zoom Image
Fig. 1 Immune checkpoint inhibitor-induced bradyarrhythmia ECG (case 2). ECG, electrocardiogram.

Bradycardia and Bradyarrhythmias with Immune Checkpoint Inhibitors

Immune checkpoint inhibitors (ICIs) are part of a relatively novel drug category which has demonstrated efficacy in prolonging overall survival in numerous cancers with previous poor prognosis. Conceptually, cancer cells can evade the patient’s immune system. ICIs aim to restore antitumor immune response by blocking immune checkpoints, i.e., intrinsic downregulation of immunity, such as programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), or cytotoxic T lymphocyte-associated protein 4 (CTLA-4). By suppressing immune regulation and tolerance, ICIs are traditionally associated with immune-related adverse events (irAEs). There are concerns that cardiovascular events were not consistently reported in randomized control trials of ICIs.[33] Moreover, there are other cardiotoxic effects related to ICIs besides myocarditis that were likely not identified in registration trials, owing to their limited power.[34] Cardiovascular toxicity is infrequent when compared with other irAEs, but now it is increasingly reported in the literature, and with the rapidly expanding indications of ICIs, we should expect to see these more often.

There are no retrospective studies regarding bradyarrhythmias and the use of ICIs. Most of the published data in relation to cardiovascular toxicities have focused on myocarditis, with some of them reporting concomitant conduction disease but not in a systematic fashion, so the type of bradyarrhythmia, risk factors, treatment, and the outcome are not always clear.

Escudier et al published in 2017 a review of 30 cases of ICIs cardiotoxicity from their cardio-oncology clinic combined with selected cases from a literature review. Of those, 17% (n = 5) had conduction abnormalities. Cardiotoxicity was diagnosed 65 (2–454) days after the initiation of ICIs. Cardiovascular mortality was significantly associated with conduction abnormalities (80 vs. 16%; p = 0.003).[35]

In a retrospective study conducted using VigiBase, which is the World Health Organization’s global database of individual case safety reports, Salem et al. reported an incidence of ICI-related cardiac conductive disorders of 0.12%, similar to the incidence in the entire database.[36]

In a multicenter observational study that included 35 patients with ICI-associated myocarditis, three had hemodynamically significant CHB. Myocarditis was diagnosed at a median of 34 days after initiation of ICIs. Diabetes, sleep apnea, obesity, and combination checkpoint blockade were all identified as risk factors for myocarditis.

In a 2018 systematic review of published case series and case reports, Mir et al concluded that myocarditis was the most commonly observed cardiovascular irAE, corresponding to 45% (n = 45) of the cases. Conduction disease was present in 12% (n = 12) of the patients with cardiovascular toxicities. Of those, three had isolated rhythm disturbances, and nine had another toxicity. The overall mortality rate was 35%, and 50% in patients presenting with CHB or other conduction abnormalities.[34]

In a 2019 review by Atallah-Yunes et al, of 42 myocarditis cases related to pembrolizumab, nivolumab, or ipilimumab use, CHB occurred in 36% of them. In agreement with previously published data, 33% of the myocarditis cases occurred after a single ICI dose and 29% after two doses. Notably, CHB occurred in 64% of the patients that died, and 60% of those who developed CHB died.[37]

A recent study by Zlotoff et al. found that QRS duration is increased in ICI myocarditis and is associated with an increased risk of major adverse cardiac events, including CHB (HR 3.28, 95% CI 1.98–5.62, p < 0.001).[38]

The large majority of bradycardia cases related to ICIs use are due to CHB, with most of the patients having concomitant myocarditis. [Table 2] describes the characteristics and outcomes of ICIs-related bradycardia case reports and case series. Most strikingly, half of the patients with bradyarrhythmia died; however, it is not clear if this association only denotes the severity of the myocarditis behind it. Definitive pacemakers were inserted in almost two-thirds of the patients, but there are three reported cases of recovery of intrinsic sinus rhythm after steroid therapy.[39] [40] [41]

Table 2

Case reports and case series of bradycardia and bradyarrhythmia related to immune checkpoint inhibitors

Age

Sex

Cancer

ICI

Number of cycles

Bradyarrhythmia

Pacemaker

Cardiovascular risk factors/cardiovascular disease/past medical history

Laboratory markers

Imaging studies

Histopathology

Treatment

Death

Abbreviations: AV, atrioventricular; CK, creatine kinase; CMR, cardiac magnetic resonance; LVEF, left ventricular ejection fraction; MG, myasthenia gravis; MMF, mycophenolate mofetil; TTE, transthoracic echocardiography.

Tshuma et al[87]

74

Female

Urothelial (bladder) cancer

Atezolizumab

12

Varying degree of AV block

Yes

Sleep apnea, hypopituitarism

Mild LV impairment on CMR

Steroids

Yes

Bukamur et al[88]

88

Female

Squamous cell lung carcinoma

Nivolumab

2

Complete AV block

Yes

Hypertension, hyperlipidemia

High CK and troponin

Increased wall thickness on TTE

Steroids

No

Prevel et al[89]

80

Male

Squamous cell lung carcinoma

Nivolumab

4

Complete AV block

No

Smoker, hypertension, diabetes mellitus type 2

High CK

No

Yes

Tan et al[90]

74

Male

Nonsmall cell lung cancer

Nivolumab

2

Complete AV block

Yes

Smoker, chronic obstructive pulmonary disease

High troponin and BNP

LVEF 31% on TTE, increased T2 on CMR

Steroids

No

Behling et al[91]

63

Male

Melanoma

Nivolumab

2

Complete AV block

Yes

Chronic obstructive pulmonary disease, hypertension, hyperlipidemia, diabetes mellitus type 2

High CK and troponin

Increased wall thickness on TTE

Steroids

No

Samara et al[92]

77

Male

Melanoma

Ipilimumab

4

Junctional bradycardia

No

Hypertension

High troponin and BNP

LVEF 45% on TTE

Steroids

Yes

Johnson et al[48]

65

Female

Melanoma

Nivolumab + ipilimumab

1

Complete AV block

No

Hypertension

High CK and troponin

LVEF 73% on TTE

Patchy lymphocytic infiltrate within the myocardium, cardiac sinus, and atrioventricular nodes.

Steroids

Yes

Johnson et al[48]

63

Male

Melanoma

Nivolumab + Ipilimumab

1

Complete AV block

Yes

Hypertension

High troponin

LVEF 73% on TTE

Patchy lymphocytic infiltrate within the myocardium, cardiac sinus, and atrioventricular nodes.

Steroids and Infliximab

Yes

Hardy et al[93]

81

Male

Renal cell carcinoma

Nivolumab + Ipilimumab

1

Complete AV block

Yes

High CK and troponin

LVEF 60–65% on TTE

CD3-positive T cells, occasional CD20-positive B cells, and CD68-positive macrophages. More CD4-positive cells as compared with CD8-positive cells. Staining with antibodies to PD-1 and PD-L1 in the areas of inflammation.

Steroids and plasmapheresis

Yes

Yanase et al[41]

59

Male

Renal cell carcinoma

Nivolumab + ipilimumab

1

Transient complete AV block

No

High CK and troponin

Steroids, plasmapheresis, and IV immunoglobulin (concomitant MG)

No

Szuchan et al[94]

70

Female

Thymic carcinoma

Pembrolizumab

1

Complete AV block

Yes

High CK and troponin

LVEF 63% on TTE

Lymphocyte infiltration of the myocardium

Steroids and plasmapheresis (concomitant MG)

No

Katsume et al[40]

73

Male

Nonsmall cell lung cancer

Pembrolizumab

1

Complete AV block

No

High CK and troponin

LVEF 70%

Steroids

No

Lee et al[95]

37

Female

Alveolar soft part sarcoma

Pembrolizumab

13

Sinus bradycardia

No

Normal troponin

LVEF 60–65% on TTE; High T2 on CMR

Steroids

No

Agrawal et al[96]

73

Male

Mesothelioma

Pembrolizumab

1

Complete AV block

Yes

High CK and troponin

LVEF 50–60%

Steroids, plasmapheresis and IV immunoglobulin (concomitant MG)

Yes

Agrawal et al[96]

89

Male

Melanoma

Pembrolizumab

1

Complete AV block

Yes

Hypertension, hyperlipidemia, diabetes mellitus type 2

High CK, troponin, and BNP

Normal TTE

Steroids and IV immunoglobulin

Yes

Khan et al[78]

67

Female

Nonsmall cell lung cancer

Pembrolizumab

1

Complete AV block

Yes

Hypertension and hyperlipidemia

Normal CK, troponin, and BNP

LVEF 60–65% on TTE

No

Mahmood et al[39]

75

Female

Endometrial cancer

Durvalumab + tremelimumab

1

Transient complete AV block

No

High CK and troponin

LVEF 54%

Steroids and MMF

No

The exact mechanism(s) of ICI-related conduction disease is not fully understood. It is postulated that myocarditis, pre-existing cardiovascular diseases, past cardiotoxic chemotherapy, myocardial metastases, old age, and systemic inflammatory conditions could all contribute to arrhythmias in cancer patients treated with PD-1/PDL-1 inhibitors.[42] Lyon et al hypothesized that ventricular myocarditis, inflammation of the His-Purkinje conduction system, either via direct T-cell-Purkinje interactions or via activation of local macrophages resident in the Purkinje system, increased systemic inflammatory state (without myocarditis), left ventricular impairment due to noninflammatory functional cardiotoxicity, or inflammation of myocardial metastases (if present) could all be at the origin of arrhythmias in the context of ICIs use.[43] In murine models, the deletion of PD-1 leads to autoimmune dilated cardiomyopathy.[44] Later studies detected circulating autoantibodies against cardiac troponin I in PD-1 knockout mice.[45] CTLA-4 knockout mice also develop autoimmune myocarditis with CD4+ and CD8+ T lymphocyte infiltration of the myocardium.[46] In mice models of T cell-mediated myocarditis, there is upregulation of myocardial PD-L1. ICIs can interfere with this likely cytokine-induced cardioprotective mechanism by blocking PD-L1.[47] Johnson et al. described a robust T-cell infiltration, activation, and clonal expansion across the myocardium, striated muscle, and tumor tissue, with shared high-frequency T-cell receptors, in their two reported cases of myocarditis with ICIs use. They then postulated that T cells could either be targeting an antigen shared by those tissue types or that the same T-cell receptor was targeting a tumor antigen and a different but homologous muscle antigen. Backing the first possibility was the fact they observed high levels of muscle-specific antigens (desmin and troponin) in tumors from both patients. However, they still considered the possibility that their finding of clonal, high-frequency, T-cell receptor sequences across tumor and muscle samples could be misleading and that distinct T-cell receptors were targeting dissimilar antigens.[48]


#
#

Case 3: Mobitz Type II AVB with Radiation Therapy

A 51-year-old female with a history of stage IIIA nodular sclerosing Hodgkin’s lymphoma 32 years ago, stage IIA estrogen-receptor-negative right-sided breast cancer 12 years ago, and radiation-induced mitral stenosis presented after an episode of syncope. Her Hodgkin’s lymphoma was previously treated with MOPP (mechlorethamine, oncovin, procarbazine, and prednisone) chemotherapy and mediastinal and mantle field radiation therapy. Her breast cancer was previously treated with AC-T (doxorubicin, cyclophosphamide, and paclitaxel) chemotherapy with subsequent bilateral mastectomy. After this syncopal episode, a 14-day ECG patch was ordered and showed six episodes of Mobitz type II AVB. At that time, the patient was on metoprolol succinate (50 mg daily) for inappropriate sinus tachycardia, which was discontinued. Four months later, the patient was admitted after a recurrent episode of syncope. ECG showed sinus tachycardia at a ventricular rate of 117 bpm with Mobitz type II, underlying right-bundle branch block, and right atrial enlargement ( [Fig. 2] ). Inpatient continuous telemetry revealed additional periods of 2:1 AVB with suspected infra-Hisian disease. She subsequently underwent successful implantation of a dual-chamber pacemaker with a left bundle branch pacing lead. The patient has not had any recurrent syncopal episodes since pacemaker implantation at the time of publication.

Zoom Image
Fig. 2 Radiation-induced bradyarrhythmia ECG (case 3). ECG, electrocardiogram.

#

Case 4: Mobitz 2:1 AVB with Radiation Therapy

A 54-year-old female with a history of Hodgkin’s lymphoma 27 years ago, radiation-induced aortic stenosis, type 2 diabetes mellitus, hyperlipidemia, and hypothyroidism presented with progressive dyspnea on exertion. Her Hodgkin’s lymphoma was previously treated with ABVD (doxorubicin, bleomycin, vinblastine, and dacarbazine) chemotherapy and mediastinal and neck radiation therapy. Subsequently, the patient developed radiation-induced moderate to severe aortic stenosis and aortic insufficiency with associated symptomatic heart failure with preserved ejection fraction. After persistent dyspnea refractory to medical management, the patient was admitted to evaluate the suitability of transcatheter aortic valve replacement (TAVR). During this admission, ECG showed normal sinus rhythm with 2:1 AVB and right-bundle branch block with a ventricular rate of 45 bpm, concerning for infra-Hisian disease ( [Fig. 3] ). Inpatient continuous telemetry showed persistency of the rhythm disturbance. She subsequently underwent successful implantation of a dual-chamber pacemaker with left bundle branch pacing lead before TAVR.

Zoom Image
Fig. 3 Radiation-induced bradyarrhythmia ECG (case 4). ECG, electrocardiogram.

Bradycardia and Bradyarrhythmias with Radiation Therapy

Radiation therapy for thoracic malignancies such as Hodgkin’s lymphoma, left-sided breast cancer, lung cancer, and other mediastinal tumors can lead to radiation-associated cardiac disease due to unwanted cardiac exposure.[49] SB and bradyarrhythmia can develop from the direct effects of irradiation on the conduction system and myocardium, often manifesting years to decades after the radiation treatment.[50] In 48 patients with Hodgkin’s lymphoma treated with mediastinal irradiation, 75% had conduction defects on ECG with a mean follow-up of 14 years.[51] Case series for other patients with irradiation for Hodgkin’s lymphoma 10 to 20 years prior described the development of CHB, predominantly due to infranodal conduction block.[52] [53] [54] Other types of bradyarrhythmia after mediastinal irradiation include first-degree and Mobitz type II AVB.[54] Right bundle branch block was also commonly observed due to its anterior location.[52] [55] Second- and third-degree AVB have also been reported after irradiation for left-sided breast cancer 18 to 23 years prior.[56] Valvular disease is also a latent consequence of radiation therapy. Radiation-associated valvular thickening and calcification can lead to stenosis or regurgitation, more commonly affecting left-sided valves. The mechanism for radiation-associated bradyarrhythmia is primarily driven by damage to the conduction system from chronic, progressive fibrosis.[49] Reactive oxygen species are formed from radiation exposure, creating an inflammatory response.[57] Subsequently, progressive fibrosis and endothelial dysfunction are enhanced by the induction of transforming growth factor-β expression.[58] Experiments of irradiation in animal models have shown increased deposition of collagen in the myocardium, which exacerbates over time.[59] Similar findings are observed in patients with CHB who underwent autopsy. Microscopic examination of the conduction system of patients with CHB after mediastinal irradiation showed sclerosis of the SA node arterioles and arteries, moderate fibrosis near the SA and AV nodes, and substantial fibrosis of both right and left bundle branches.[53] [54]


#

Interaction between Anticancer Agents and Nodal Blocking Drugs

A drug–drug interaction occurs when the combined administration of drugs leads to an adverse pharmacological or clinical response in patients.[60] Patients with cancer have an increased risk of potential drug–drug interactions due to polypharmacy among anticancer therapies and medications for comorbidities as well as age-related physiologic changes that alter drug absorption or excretion.[60] [61] In particular, drug–drug interactions have been reported in 14 to 16% of patients receiving anticancer therapy.[62] [63] Pharmacodynamic interactions occur when two or more drugs have similar physiological outcomes, leading to excessive response or toxicity. Pharmacokinetic interactions occur when one drug alters the absorption, distribution, metabolism, or elimination of another drug. Commonly, metabolism by cytochrome P450 enzymes in the liver, such as the CYP34A isozyme, can be affected by drugs that act as inhibitors or inducers.[60] Calcium-channel blockers are substrates for CYP34A isozymes, and several β-blockers are substrates for CYP2D6 isozymes.[64] [65] Another common mechanism of interaction involves the P-glycoprotein (P-gp), an efflux pump that enables the transport of drugs across cell membranes for excretion.[60] Substrates for P-gp include many anticancer therapies, digoxin, and calcium-channel blockers.[65]

Drug databases are frequently used to identify potentially harmful drug interactions, with reported sensitivities and specificities over 80%.[66] [67] [68] [Fig. 4] shows the risk of bradycardia and bradyarrhythmia with drug–drug interactions between nodal blocking agents and anticancer therapies, including data from Lexi-Interact (Wolters Kluwer) and Micromedex (IBM Corporation).[69] [70] Among conventional chemotherapies, cases of additive bradycardia have been reported from pharmacodynamic drug–drug interactions between thalidomide and β-blockers, particularly in elderly patients with cardiac comorbidities.[71] [72] Further, many targeted agents such as TKIs are known to interact with medications that affect CYP3A4 metabolism, leading to the risk of pharmacokinetic drug–drug interactions with calcium-channel blockers in particular.[73] [74] For example, there are moderate to severe interactions between calcium-channel blockers and the TKIs imatinib, nilotinib, or vemurafenib from the inhibition of CYP3A4-mediated metabolism leading to increased calcium-channel blocker concentration. Ibrutinib and vemurafenib, in particular, also have pharmacokinetic drug–drug interactions with digoxin due to P-gp-mediated digoxin efflux transport inhibition. Among histone deacetylase inhibitors, panobinostat has a minor pharmacokinetic interaction with metoprolol due to inhibition of CYP2D6-mediated metabolism. On the other hand, no formal pharmacokinetic drug–drug interactions have been conducted for nivolumab, pembrolizumab, and ipilimumab.[75] [76] [77] Further evaluation is needed to better define drug–drug interactions with nodal blocking agents and ICIs, especially given case reports of high-grade AVB in ICI-associated myocarditis.[40] [48] [78] [79] [80]

Zoom Image
Fig. 4 Drug–Drug interactions between anticancer and nodal blocking agents (created by biorender.com).

In reverse, nodal blocking agents can also potentiate anticancer therapies’ concentration due to similar pharmacokinetic mechanisms. For example, doxorubicin, paclitaxel, vincristine, and vinblastine are P-gp substrates.[65] Thus, their drug concentrations can be increased in patients who are taking carvedilol, a moderate P-gp inhibitor. Similarly, diltiazem is a potent CYP3A4 inhibitor, and co-administration with CYP3A4 substrates such as doxorubicin, ibrutinib, busulfan, imatinib, vincristine, and vinblastine can increase the systemic concentration of such anticancer therapies.[60] [64] For example, venetoclax exposure is increased with the concurrent use of both CYP3A4 inhibitors or P-gp inhibitors, such as carvedilol and diltiazem, that can result in increased neutropenia and risk of tumor lysis syndrome. Alternative nodal blocking agents should be considered to avoid such chemotherapy toxicities.[81]


#

Diagnosis and Management

The American College of Cardiology/American Heart Association/Heart Rhythm Society (ACC/AHA/HRS) bradycardia clinical practice guidelines define a sinus rate <50 bpm and/or a sinus pause >3 seconds as potential components of sinus node dysfunction (SND). For the acute management of bradycardia related to moderate or severely symptomatic SND, symptomatic second- or third-degree AVB believed to be at the AV nodal level or any of the above resulting in hemodynamic compromise, atropine can be helpful to transiently increase sinus rate, and is a reasonable first step. If symptoms persist despite this and given atropine’s short duration of action, β-adrenergic agonists such as isoproterenol, dopamine, dobutamine, or epinephrine may be considered to increase heart rate and/or improve AV conduction and/or symptoms. In the presence of symptoms or hemodynamic compromise refractory to medical therapy, temporary transvenous pacing is reasonable, and temporary transcutaneous pacing may be considered until a temporary transvenous, or permanent pacemaker is placed.[82] The need for temporary pacing is more likely when AV block is infra-Hisian.

There are no specific bradycardia-management guidelines produced by the European Society of Cardiology (ESC). Instead, there are guidelines related to cardiac pacing indications. They state that temporary transvenous pacing shall only be used in bradycardia refractory to positive chronotropic drugs. Temporary transvenous pacing should also be limited to cases of high-degree AVB without escape rhythm or life-threatening bradyarrhythmias.[83]

As per the ACC/AHA/HRS guidelines, for the management of bradycardia attributable to AVB in the chronic setting, such as the one related to radiation therapy, class I indications for permanent pacemaker implantation include Mobitz type II AVB, high-grade AVB, or CHB that is not attributable to reversible causes. For bradycardia attributable to SND, class I indication for permanent pacemaker implantation includes symptoms directly attributable to SND.[82] The ESC pacing guidelines recommend permanent pacemaker implantation (Class I recommendation) in patients whose symptoms are attributable to SND and patients with third- or second-degree type 2 AVB.[83]

[Fig. 5] represents a proposed algorithm for managing patients with cancer treatment-related bradycardia/bradyarrhythmia, with a special focus on ICI- and radiation-induced conduction abnormalities.

Zoom Image
Fig. 5 Proposed algorithm for the management of bradycardia/bradyarrhythmia, with a special focus on ICI- and radiation-induced conduction abnormalities [49] [97] [98] [99]w[100] (created by biorender.com). CK, creatine kinase; CMR, cardiac magnetic resonance; CRP, C-reactive protein; CXR, chest X-ray; ECG, electrocardiogram; EMB, endomyocardial biopsy; ESR, erythrocyte sedimentation rate; NT proBNP, N-terminal pro-brain natriuretic peptide; TTE, transthoracic echocardiography; WBC, white blood cell count.

#

Future Directions

There is a limited body of literature regarding the true incidence of bradycardia and bradyarrhythmia related to chemotherapeutic agents, targeted molecular therapies, immunotherapies, and radiation. This is partially explained by the lack of systematic cardiac monitoring and reporting of arrhythmic events. In our view, a structured cardiac surveillance strategy with a homogenous classification of the arrhythmic events to be reported should be applied to clinical trials investigating the safety and efficacy of new cancer drugs. Databases/registries could also be established to obtain real-world quantitative data. Research of the possible mechanisms behind this type of cardiotoxicity is as well much needed. The already published evidence has mainly focused on arrhythmias in general, and there is a lack of data regarding the pathophysiology behind cancer drug-related bradycardia/bradyarrhythmia. Such knowledge could help develop evidence-based models to predict the patients who are more at risk of developing bradycardia/bradyarrhythmia and define the ones who may benefit from tighter monitoring strategies. Longer-term follow-up data are essential to determine the role of bradycardia on cancer patients’ overall outcomes. In particular, in ICI-myocarditis patients presenting with CHB who frequently undergo pacemaker implantation, long-term data regarding the percentage of pacing would be helpful to evaluate the degree of temporal recovery of these conduction abnormalities if at all. The rapid advances in pharmacotherapy in oncology show no signs of abating and if we are to understand the nuances of the adverse effects of these drugs on the cardiovascular system then the future mandates much closer collaboration between cardiologists and oncologists to optimize patient outcomes.


#
#
#

Conflict of Interest

None declared.

# Joint senior authors.


  • References

  • 1 Elazhary S, Alawyat HA. Bradycardia associated with antithymocyte globulin treatment of a pediatric patient with sickle cell disease: a case report and literature review. Hematol Transfus Cell Ther 2020; (e-pub ahead of print). doi: DOI: 10.1016/j.htct.2020.08.018.
  • 2 Ando M, Yokozawa T, Sawada J. et al Cardiac conduction abnormalities in patients with breast cancer undergoing high-dose chemotherapy and stem cell transplantation. Bone Marrow Transplant 2000; 25 (02) 185-189
  • 3 Ramireddy K, Kane KM, Adhar GC. Acquired episodic complete heart block after high-dose chemotherapy with cyclophosphamide and thiotepa. Am Heart J 1994; 127 (03) 701-704
  • 4 Agarwal N, Burkart TA. Transient, high-grade atrioventricular block from high-dose cyclophosphamide. Tex Heart Inst J 2013; 40 (05) 626-627
  • 5 Haddad D, Guha A, Awan F, Daoud EG, Baliga R. Arrhythmias in cancer and cancer treatment: a review. In: Encyclopedia of Cardiovascular Research and Medicine Vol 1. Elsevier 2018: 162-181
  • 6 Tamargo J, Caballero R, Delpón E. Cancer chemotherapy and cardiac arrhythmias: a review. Drug Saf 2015; 38 (02) 129-152
  • 7 Kilickap S, Barista I, Akgul E, Aytemir K, Aksoy S, Tekuzman G. Early and late arrhythmogenic effects of doxorubicin. South Med J 2007; 100 (03) 262-265
  • 8 Darling HS. Cisplatin induced bradycardia. Int J Cardiol 2015; 182: 304-306
  • 9 Kucharz J, Michalowska-Kaczmarczyk A, Zygulska AL. et al Bradycardia as a rare symptom of cisplatin cardiotoxicity: a case report. Oncol Lett 2016; 11 (03) 2297-2299
  • 10 Schlumbrecht MP, Hehr K. Cisplatin-induced bradycardia and the importance of the QT interval. J Oncol Pharm Pract 2015; 21 (02) 157-160
  • 11 Altundağ O, Çelik I, Kars A. Recurrent asymptomatic bradycardia episodes after cisplatin infusion. Ann Pharmacother 2001; 35 (05) 641-642
  • 12 Talapatra K, Rajesh I, Rajesh B, Selvamani B, Subhashini J. Transient asymptomatic bradycardia in patients on infusional 5-fluorouracil. J Cancer Res Ther 2007; 3 (03) 169-171
  • 13 Stewart T, Pavlakis N, Ward M. Cardiotoxicity with 5-fluorouracil and capecitabine: more than just vasospastic angina. Intern Med J 2010; 40 (04) 303-307
  • 14 Khan MA, Masood N, Husain N, Ahmad B, Aziz T, Naeem A. A retrospective study of cardiotoxicities induced by 5-fluouracil (5-FU) and 5-FU based chemotherapy regimens in Pakistani adult cancer patients at Shaukat Khanum Memorial Cancer Hospital & Research Center. J Pak Med Assoc 2012; 62 (05) 430-434
  • 15 Kosmas C, Kallistratos MS, Kopterides P. et al Cardiotoxicity of fluoropyrimidines in different schedules of administration: a prospective study. J Cancer Res Clin Oncol 2008; 134 (01) 75-82
  • 16 Cil T, Kaplan MA, Altintas A, Pasa S, Isikdogan A. Cytosine-arabinoside induced bradycardia in patient with non-Hodgkin lymphoma: a case report. Leuk Lymphoma 2007; 48 (06) 1247-1249
  • 17 McGuire W, Rowinsky E, Rosenhein N. et al Taxol: a unique antineoplastic agent with significant activity in advanced ovarian epithelial neoplasms. Int J Gynaecol Obstet 1990; 31 (03) 298
  • 18 Huang CH, Chen WJ, Wu CC, Chen YC, Lee YT. Complete atrioventricular block after arsenic trioxide treatment in an acute promyelocytic leukemic patient. Pacing Clin Electrophysiol 1999; 22 (6 Pt 1) 965-967
  • 19 Shridhar PL. Interleukin-2 induced atrioventricular dissociation. Int J Collaborative Res Internal Med Public Health 2016; 8 (08) 510-511
  • 20 Foran JM, Rohatiner AZS, Cunningham D. et al European phase II study of rituximab (chimeric anti-CD20 monoclonal antibody) for patients with newly diagnosed mantle-cell lymphoma and previously treated mantle-cell lymphoma, immunocytoma, and small B-cell lymphocytic lymphoma. J Clin Oncol 2000; 18 (02) 317-324
  • 21 Ko Ko NL, Minaskeian N, El Masry HZ. A case of irreversible bradycardia after rituximab therapy for diffuse large B-cell lymphoma. Cardiooncology 2020; 6 (01) 22
  • 22 Cervera Grau JM, Esquerdo Galiana G, Belso Candela A, Llorca Ferrándiz C, Juárez Marroquí A, Maciá Escalante S. Complete atrioventricular block induced by rituximab in monotherapy in an aged patient with non-Hodgkin’s diffuse large B-cell lymphoma. Clin Transl Oncol 2008; 10 (05) 298-299
  • 23 Berenson JR, Jagannath S, Barlogie B. et al Safety of prolonged therapy with bortezomib in relapsed or refractory multiple myeloma. Cancer 2005; 104 (10) 2141-2148
  • 24 Enrico O, Gabriele B, Nadia C. et al Unexpected cardiotoxicity in haematological bortezomib treated patients. Br J Haematol 2007; 138 (03) 396-397
  • 25 Rajkumar SV, Gertz MA, Lacy MQ. et al Thalidomide as initial therapy for early-stage myeloma. Leukemia 2003; 17 (04) 775-779
  • 26 Fahdi IE, Gaddam V, Saucedo JF. et al Bradycardia during therapy for multiple myeloma with thalidomide. Am J Cardiol 2004; 93 (08) 1052-1055
  • 27 Hinterseer M, Becker A, Kääb S, Lang N, Näbauer M, Steinbeck G. Thalidomide-induced symptomatic third-degree atrioventricular block. Clin Res Cardiol 2006; 95 (09) 474-476
  • 28 Ou S-HI, Tang Y, Polli A, Wilner KD, Schnell P. Factors associated with sinus bradycardia during crizotinib treatment: a retrospective analysis of two large-scale multinational trials (PROFILE 1005 and 1007). Cancer Med 2016; 5 (04) 617-622
  • 29 Morcos PN, Bogman K, Hubeaux S. et al Effect of alectinib on cardiac electrophysiology: results from intensive electrocardiogram monitoring from the pivotal phase II NP28761 and NP28673 studies. Cancer Chemother Pharmacol 2017; 79 (03) 559-568
  • 30 Lorlatinib. FDA package insert. Published 2018. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/ 210868s000lbl.pdf. Accessed February 15, 2021
  • 31 Ponatinib. FDA package insert. Published 2012. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/ 203469s009lbl.pdf. Accessed February 15, 2021
  • 32 Nilotinib. FDA package insert. Published 2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/ 022068s026lbl.pdf. Accessed February 15, 2021
  • 33 Ball S, Ghosh RK, Wongsaengsak S. et al Cardiovascular toxicities of immune checkpoint inhibitors: JACC Review Topic of the Week. J Am Coll Cardiol 2019; 74 (13) 1714-1727
  • 34 Mir H, Alhussein M, Alrashidi S. et al Cardiac complications associated with checkpoint inhibition: a systematic review of the literature in an important emerging area. Can J Cardiol 2018; 34 (08) 1059-1068
  • 35 Escudier M, Cautela J, Malissen N. et al Clinical features, management, and outcomes of immune checkpoint inhibitor-related cardiotoxicity. Circulation 2017; 136 (21) 2085-2087
  • 36 Salem J-E, Manouchehri A, Moey M. et al Cardiovascular toxicities associated with immune checkpoint inhibitors: an observational, retrospective, pharmacovigilance study. Lancet Oncol 2018; 19 (12) 1579-1589
  • 37 Atallah-Yunes SA, Kadado AJ, Kaufman GP, Hernandez-Montfort J. Immune checkpoint inhibitor therapy and myocarditis: a systematic review of reported cases. J Cancer Res Clin Oncol 2019; 145 (06) 1527-1557
  • 38 Zlotoff DA, Hassan MZO, Zafar A. et al Electrocardiographic features of immune checkpoint inhibitor associated myocarditis. J Immunother Cancer 2021; 9 (03) e002007
  • 39 Mahmood SS, Chen CL, Shapnik N, Krishnan U, Singh HS, Makker V. Myocarditis with tremelimumab plus durvalumab combination therapy for endometrial cancer: a case report. Gynecol Oncol Rep 2018; 25: 74-77
  • 40 Katsume Y, Isawa T, Toi Y. et al Complete atrioventricular block associated with pembrolizumab-induced acute myocarditis: the need for close cardiac monitoring. Intern Med 2018; 57 (21) 3157-3162
  • 41 Yanase T, Moritoki Y, Kondo H, Ueyama D, Akita H, Yasui T. Myocarditis and myasthenia gravis by combined nivolumab and ipilimumab immunotherapy for renal cell carcinoma: a case report of successful management. Urol Case Rep 2020; 34: 101508
  • 42 Safi M, Ahmed H, Al-Azab M. et al PD-1/PDL-1 inhibitors and cardiotoxicity; molecular, etiological and management outlines. J Adv Res 2020; 29: 45-54
  • 43 Lyon AR, Yousaf N, Battisti NML, Moslehi J, Larkin J. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncol 2018; 19 (09) e447-e458
  • 44 Nishimura H, Okazaki T, Tanaka Y. et al Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001; 291 (5502) 319-322
  • 45 Okazaki T, Tanaka Y, Nishio R. et al Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med 2003; 9 (12) 1477-1483
  • 46 Love VA, Grabie N, Duramad P, Stavrakis G, Sharpe A, Lichtman A. CTLA-4 ablation and interleukin-12 driven differentiation synergistically augment cardiac pathogenicity of cytotoxic T lymphocytes. Circ Res 2007; 101 (03) 248-257
  • 47 Grabie N, Gotsman I, DaCosta R. et al Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart. Circulation 2007; 116 (18) 2062-2071
  • 48 Johnson DB, Balko JM, Compton ML. et al Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med 2016; 375 (18) 1749-1755
  • 49 Desai MY, Windecker S, Lancellotti P. et al Prevention, diagnosis, and management of radiation-associated cardiac disease: JACC Scientific Expert Panel. J Am Coll Cardiol 2019; 74 (07) 905-927
  • 50 Desai MY, Jellis CL, Kotecha R, Johnston DR, Griffin BP. Radiation-associated cardiac disease: a practical approach to diagnosis and management. JACC Cardiovasc Imaging 2018; 11 (08) 1132-1149
  • 51 Adams MJ, Lipsitz SR, Colan SD. et al Cardiovascular status in long-term survivors of Hodgkin’s disease treated with chest radiotherapy. J Clin Oncol 2004; 22 (15) 3139-3148
  • 52 Orzan F, Brusca A, Gaita F, Giustetto C, Figliomeni MC, Libero L. Associated cardiac lesions in patients with radiation-induced complete heart block. Int J Cardiol 1993; 39 (02) 151-156
  • 53 Cohen SI, Bharati S, Glass J, Lev M. Radiotherapy as a cause of complete atrioventricular block in Hodgkin’s disease. An electrophysiological-pathological correlation. Arch Intern Med 1981; 141 (05) 676-679
  • 54 Slama MS, Le Guludec D, Sebag C. et al Complete atrioventricular block following mediastinal irradiation: a report of six cases. Pacing Clin Electrophysiol 1991; 14 (07) 1112-1118
  • 55 Heidenreich PA, Kapoor JR. Radiation induced heart disease: systemic disorders in heart disease. Heart 2009; 95 (03) 252-258
  • 56 Tzivoni D, Ratzkowski E, Biran S, Brook JG, Stern S. Complete heart block following therapeutic irradiation of the left side of the chest. Chest 1977; 71 (02) 231-234
  • 57 Donnellan E, Jellis CL, Griffin BP. Radiation-associated cardiac disease: from molecular mechanisms to clinical management. Curr Treat Options Cardiovasc Med 2019; 21 (05) 22
  • 58 Tapio S. Pathology and biology of radiation-induced cardiac disease. J Radiat Res (Tokyo) 2016; 57 (05) 439-448
  • 59 Boerma M, Sridharan V, Mao XW. et al Effects of ionizing radiation on the heart. Mutat Res 2016; 770 (Pt B) 319-327
  • 60 Scripture CD, Figg WD. Drug interactions in cancer therapy. Nat Rev Cancer 2006; 6 (07) 546-558
  • 61 Campen CJ, Vogel WH, Shah PJ. Managing drug interactions in cancer therapy: a guide for the advanced practitioner. J Adv Pract Oncol 2017; 8 (06) 609-620
  • 62 Stoll P, Kopittke L. Potential drug-drug interactions in hospitalized patients undergoing systemic chemotherapy: a prospective cohort study. Int J Clin Pharm 2015; 37 (03) 475-484
  • 63 van Leeuwen RWF, Brundel DHS, Neef C. et al Prevalence of potential drug-drug interactions in cancer patients treated with oral anticancer drugs. Br J Cancer 2013; 108 (05) 1071-1078
  • 64 Lam MSH, Ignoffo RJ. A guide to clinically relevant drug interactions in oncology. J Oncol Pharm Pract 2003; 9 (2-3) 45-85
  • 65 Kennedy C, Brewer L, Williams D. Drug interactions. Med (United Kingdom) 2016; 44 (07) 422-426
  • 66 Kheshti R, Aalipour M, Namazi S. A comparison of five common drug-drug interaction software programs regarding accuracy and comprehensiveness. J Res Pharm Pract 2016; 5 (04) 257-263
  • 67 Roblek T, Vaupotic T, Mrhar A, Lainscak M. Drug-drug interaction software in clinical practice: a systematic review. Eur J Clin Pharmacol 2015; 71 (02) 131-142
  • 68 Marcath LA, Xi J, Hoylman EK, Kidwell KM, Kraft SL, Hertz DL. Comparison of nine tools for screening drug-drug interactions of oral oncolytics. J Oncol Pract 2018; 14 (06) e368-e374
  • 69 Wolters Kluwer. Lexi-Interact Online. Published 2021. Available at: https/ www.uptodate.com/drug-interactions/ Accessed February 7, 2021
  • 70 Corporation IBM. Micromedex. Published 2021. Available at: https/ www.micromedexsolutions.com
  • 71 Kaur A, Yu SS, Lee AJ, Chiao TB. Thalidomide-induced sinus bradycardia. Ann Pharmacother 2003; 37 (7-8) 1040-1043
  • 72 Yamaguchi T. Syncope and sinus bradycardia from combined use of thalidomide and beta-blocker. Pharmacoepidemiol Drug Saf 2008; 17 (10) 1033-1035
  • 73 Rogala BG, Charpentier MM, Nguyen MK, Landolf KM, Hamad L, Gaertner KM. Oral anticancer therapy: management of drug interactions. J Oncol Pract 2019; 15 (02) 81-90
  • 74 Haouala A, Widmer N, Duchosal MA, Montemurro M, Buclin T, Decosterd LA. Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib. Blood 2011; 117 (08) e75-e87
  • 75 Pembrolizumab Label FDA. Published 2014. Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/ 125514lbl.pdf. Accessed February 15, 2021
  • 76 Ipilimumab Label FDA. Published 2011. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/ 125377s094lbl.pdf. Accessed February 15, 2021
  • 77 Nivolumab Label FDA. Published 2014. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/ 125554s070lbl.pdf. Accessed February 15, 2021
  • 78 Khan A, Riaz S, Carhart Jr R. Pembrolizumab-induced mobitz type 2 second-degree atrioventricular block. Case Rep Cardiol 2020; 2020: 8428210
  • 79 Reddy N, Moudgil R, Lopez-Mattei JC. et al Progressive and reversible conduction disease with checkpoint inhibitors. Can J Cardiol 2017; 33 (10) 1335.e13-1335.e15
  • 80 Giancaterino S, Abushamat F, Duran J, Lupercio F, DeMaria A, Hsu JC. Complete heart block and subsequent sudden cardiac death from immune checkpoint inhibitor-associated myocarditis. HeartRhythm Case Rep 2020; 6 (10) 761-764
  • 81 AbbVie Inc. and Genentech USA I. Management of potential VENCLEXTA drug interactions. Published 2021. Available at: https://www.venclextahcp.com/cll/dosing/drug-interactions.html. Accessed April 4, 2021
  • 82 Kusumoto FM, Schoenfeld MH, Barrett C. et al 2018 ACC/AHA/HRS guideline on the evaluation and management of patients with bradycardia and cardiac conduction delay: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. Circulation 2019; 140 (08) e382-e482
  • 83 Brignole M, Auricchio A, Baron-Esquivias G. et al ESC Committee for Practice Guidelines (CPG), Document Reviewers. 2013 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy: the Task Force on cardiac pacing and resynchronization therapy of the European Society of Cardiology (ESC). Developed in collaboration with the European Heart Rhythm Association (EHRA). Eur Heart J 2013; 34 (29) 2281-2329
  • 84 Herrmann J. Adverse cardiac effects of cancer therapies: cardiotoxicity and arrhythmia. Nat Rev Cardiol 2020; 17 (08) 474-502
  • 85 Guglin M, Aljayeh M, Saiyad S, Ali R, Curtis AB. Introducing a new entity: chemotherapy-induced arrhythmia. Europace 2009; 11 (12) 1579-1586
  • 86 Buza V, Rajagopalan B, Curtis AB. Cancer treatment-induced arrhythmias: focus on chemotherapy and targeted therapies. Circ Arrhythm Electrophysiol 2017; 10 (08) 1-12
  • 87 Tshuma N, Glynn N, Evanson J, Powles T, Drake WM. Hypothalamitis and severe hypothalamic dysfunction associated with anti-programmed cell death ligand 1 antibody treatment. Eur J Cancer 2018; 104: 247-249
  • 88 Bukamur HS, Mezughi H, Karem E, Shahoub I, Shweihat Y. Nivolumab-induced third degree atrioventricular block in a patient with stage IV squamous cell lung carcinoma. Cureus 2019; 11 (06) e4869
  • 89 Prevel R, Colin G, Calès V, Renault PA, Mazieres J. Bloc atrio-ventriculaire de grade III compliquant une myocardite au décours d’un traitement par anti-PD1: observation d’un cas et revue de la littérature. Rev Med Interne 2020; 41 (04) 284-288
  • 90 Tan JL, Mugwagwa AN, Cieslik L, Joshi R. Nivolumab-induced myocarditis complicated by complete atrioventricular block in a patient with metastatic non-small cell lung cancer. BMJ Case Rep 2019; 12 (07) e229963
  • 91 Behling J, Kaes J, Münzel T, Grabbe S, Loquai C. New-onset third-degree atrioventricular block because of autoimmune-induced myositis under treatment with anti-programmed cell death-1 (nivolumab) for metastatic melanoma. Melanoma Res 2017; 27 (02) 155-158
  • 92 Samara Y, Yu CL, Dasanu CA. Acute autoimmune myocarditis and hepatitis due to ipilimumab monotherapy for malignant melanoma. J Oncol Pharm Pract 2019; 25 (04) 966-968
  • 93 Hardy T, Yin M, Chavez JA. et al Acute fatal myocarditis after a single dose of anti-PD-1 immunotherapy, autopsy findings: a case report. Cardiovasc Pathol 2020; 46: 107202
  • 94 Szuchan C, Elson L, Alley E. et al Checkpoint inhibitor-induced myocarditis and myasthenia gravis in a recurrent/metastatic thymic carcinoma patient: a case report. Eur Heart J Case Rep 2020; 4 (03) 1-8
  • 95 Lee DH, Armanious M, Huang J, Jeong D, Druta M, Fradley MG. Case of pembrolizumab-induced myocarditis presenting as torsades de pointes with safe re-challenge. J Oncol Pharm Pract 2020; 26 (06) 1544-1548
  • 96 Agrawal N, Khunger A, Vachhani P. et al Cardiac toxicity associated with immune checkpoint inhibitors: case series and review of the literature. Case Rep Oncol 2019; 12 (01) 260-276
  • 97 Brahmer JR, Lacchetti C, Schneider BJ. et al National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2018; 36 (17) 1714-1768
  • 98 Thompson JA, Schneider BJ, Brahmer J. et al NCCN guidelines insights: management of immunotherapy-related toxicities, version 1.2020. J Natl Compr Canc Netw 2020; 18 (03) 230-241
  • 99 Adams MJ, Lipshultz SE, Schwartz C, Fajardo LF, Coen V, Constine LS. Radiation-associated cardiovascular disease: manifestations and management. Semin Radiat Oncol 2003; 13 (03) 346-356
  • 100 Shen W-K, Sheldon RS, Benditt DG. et al 2017 ACC/AHA/HRS guideline for the evaluation and management of patients with syncope: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol 2017; 70 (05) e39-e110

Address for correspondence

Avirup Guha, MD
Harrington Heart and Vascular Institute, Case Western Reserve University
11100 Euclid Ave, Cleveland, OH 44106
United States   

Publikationsverlauf

Artikel online veröffentlicht:
15. Oktober 2021

© 2021. MedIntel Services Pvt Ltd. This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial-License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/).

Thieme Medical and Scientific Publishers Private Ltd
A-12, Second Floor, Sector -2, NOIDA -201301, India

  • References

  • 1 Elazhary S, Alawyat HA. Bradycardia associated with antithymocyte globulin treatment of a pediatric patient with sickle cell disease: a case report and literature review. Hematol Transfus Cell Ther 2020; (e-pub ahead of print). doi: DOI: 10.1016/j.htct.2020.08.018.
  • 2 Ando M, Yokozawa T, Sawada J. et al Cardiac conduction abnormalities in patients with breast cancer undergoing high-dose chemotherapy and stem cell transplantation. Bone Marrow Transplant 2000; 25 (02) 185-189
  • 3 Ramireddy K, Kane KM, Adhar GC. Acquired episodic complete heart block after high-dose chemotherapy with cyclophosphamide and thiotepa. Am Heart J 1994; 127 (03) 701-704
  • 4 Agarwal N, Burkart TA. Transient, high-grade atrioventricular block from high-dose cyclophosphamide. Tex Heart Inst J 2013; 40 (05) 626-627
  • 5 Haddad D, Guha A, Awan F, Daoud EG, Baliga R. Arrhythmias in cancer and cancer treatment: a review. In: Encyclopedia of Cardiovascular Research and Medicine Vol 1. Elsevier 2018: 162-181
  • 6 Tamargo J, Caballero R, Delpón E. Cancer chemotherapy and cardiac arrhythmias: a review. Drug Saf 2015; 38 (02) 129-152
  • 7 Kilickap S, Barista I, Akgul E, Aytemir K, Aksoy S, Tekuzman G. Early and late arrhythmogenic effects of doxorubicin. South Med J 2007; 100 (03) 262-265
  • 8 Darling HS. Cisplatin induced bradycardia. Int J Cardiol 2015; 182: 304-306
  • 9 Kucharz J, Michalowska-Kaczmarczyk A, Zygulska AL. et al Bradycardia as a rare symptom of cisplatin cardiotoxicity: a case report. Oncol Lett 2016; 11 (03) 2297-2299
  • 10 Schlumbrecht MP, Hehr K. Cisplatin-induced bradycardia and the importance of the QT interval. J Oncol Pharm Pract 2015; 21 (02) 157-160
  • 11 Altundağ O, Çelik I, Kars A. Recurrent asymptomatic bradycardia episodes after cisplatin infusion. Ann Pharmacother 2001; 35 (05) 641-642
  • 12 Talapatra K, Rajesh I, Rajesh B, Selvamani B, Subhashini J. Transient asymptomatic bradycardia in patients on infusional 5-fluorouracil. J Cancer Res Ther 2007; 3 (03) 169-171
  • 13 Stewart T, Pavlakis N, Ward M. Cardiotoxicity with 5-fluorouracil and capecitabine: more than just vasospastic angina. Intern Med J 2010; 40 (04) 303-307
  • 14 Khan MA, Masood N, Husain N, Ahmad B, Aziz T, Naeem A. A retrospective study of cardiotoxicities induced by 5-fluouracil (5-FU) and 5-FU based chemotherapy regimens in Pakistani adult cancer patients at Shaukat Khanum Memorial Cancer Hospital & Research Center. J Pak Med Assoc 2012; 62 (05) 430-434
  • 15 Kosmas C, Kallistratos MS, Kopterides P. et al Cardiotoxicity of fluoropyrimidines in different schedules of administration: a prospective study. J Cancer Res Clin Oncol 2008; 134 (01) 75-82
  • 16 Cil T, Kaplan MA, Altintas A, Pasa S, Isikdogan A. Cytosine-arabinoside induced bradycardia in patient with non-Hodgkin lymphoma: a case report. Leuk Lymphoma 2007; 48 (06) 1247-1249
  • 17 McGuire W, Rowinsky E, Rosenhein N. et al Taxol: a unique antineoplastic agent with significant activity in advanced ovarian epithelial neoplasms. Int J Gynaecol Obstet 1990; 31 (03) 298
  • 18 Huang CH, Chen WJ, Wu CC, Chen YC, Lee YT. Complete atrioventricular block after arsenic trioxide treatment in an acute promyelocytic leukemic patient. Pacing Clin Electrophysiol 1999; 22 (6 Pt 1) 965-967
  • 19 Shridhar PL. Interleukin-2 induced atrioventricular dissociation. Int J Collaborative Res Internal Med Public Health 2016; 8 (08) 510-511
  • 20 Foran JM, Rohatiner AZS, Cunningham D. et al European phase II study of rituximab (chimeric anti-CD20 monoclonal antibody) for patients with newly diagnosed mantle-cell lymphoma and previously treated mantle-cell lymphoma, immunocytoma, and small B-cell lymphocytic lymphoma. J Clin Oncol 2000; 18 (02) 317-324
  • 21 Ko Ko NL, Minaskeian N, El Masry HZ. A case of irreversible bradycardia after rituximab therapy for diffuse large B-cell lymphoma. Cardiooncology 2020; 6 (01) 22
  • 22 Cervera Grau JM, Esquerdo Galiana G, Belso Candela A, Llorca Ferrándiz C, Juárez Marroquí A, Maciá Escalante S. Complete atrioventricular block induced by rituximab in monotherapy in an aged patient with non-Hodgkin’s diffuse large B-cell lymphoma. Clin Transl Oncol 2008; 10 (05) 298-299
  • 23 Berenson JR, Jagannath S, Barlogie B. et al Safety of prolonged therapy with bortezomib in relapsed or refractory multiple myeloma. Cancer 2005; 104 (10) 2141-2148
  • 24 Enrico O, Gabriele B, Nadia C. et al Unexpected cardiotoxicity in haematological bortezomib treated patients. Br J Haematol 2007; 138 (03) 396-397
  • 25 Rajkumar SV, Gertz MA, Lacy MQ. et al Thalidomide as initial therapy for early-stage myeloma. Leukemia 2003; 17 (04) 775-779
  • 26 Fahdi IE, Gaddam V, Saucedo JF. et al Bradycardia during therapy for multiple myeloma with thalidomide. Am J Cardiol 2004; 93 (08) 1052-1055
  • 27 Hinterseer M, Becker A, Kääb S, Lang N, Näbauer M, Steinbeck G. Thalidomide-induced symptomatic third-degree atrioventricular block. Clin Res Cardiol 2006; 95 (09) 474-476
  • 28 Ou S-HI, Tang Y, Polli A, Wilner KD, Schnell P. Factors associated with sinus bradycardia during crizotinib treatment: a retrospective analysis of two large-scale multinational trials (PROFILE 1005 and 1007). Cancer Med 2016; 5 (04) 617-622
  • 29 Morcos PN, Bogman K, Hubeaux S. et al Effect of alectinib on cardiac electrophysiology: results from intensive electrocardiogram monitoring from the pivotal phase II NP28761 and NP28673 studies. Cancer Chemother Pharmacol 2017; 79 (03) 559-568
  • 30 Lorlatinib. FDA package insert. Published 2018. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/ 210868s000lbl.pdf. Accessed February 15, 2021
  • 31 Ponatinib. FDA package insert. Published 2012. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2014/ 203469s009lbl.pdf. Accessed February 15, 2021
  • 32 Nilotinib. FDA package insert. Published 2017. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2017/ 022068s026lbl.pdf. Accessed February 15, 2021
  • 33 Ball S, Ghosh RK, Wongsaengsak S. et al Cardiovascular toxicities of immune checkpoint inhibitors: JACC Review Topic of the Week. J Am Coll Cardiol 2019; 74 (13) 1714-1727
  • 34 Mir H, Alhussein M, Alrashidi S. et al Cardiac complications associated with checkpoint inhibition: a systematic review of the literature in an important emerging area. Can J Cardiol 2018; 34 (08) 1059-1068
  • 35 Escudier M, Cautela J, Malissen N. et al Clinical features, management, and outcomes of immune checkpoint inhibitor-related cardiotoxicity. Circulation 2017; 136 (21) 2085-2087
  • 36 Salem J-E, Manouchehri A, Moey M. et al Cardiovascular toxicities associated with immune checkpoint inhibitors: an observational, retrospective, pharmacovigilance study. Lancet Oncol 2018; 19 (12) 1579-1589
  • 37 Atallah-Yunes SA, Kadado AJ, Kaufman GP, Hernandez-Montfort J. Immune checkpoint inhibitor therapy and myocarditis: a systematic review of reported cases. J Cancer Res Clin Oncol 2019; 145 (06) 1527-1557
  • 38 Zlotoff DA, Hassan MZO, Zafar A. et al Electrocardiographic features of immune checkpoint inhibitor associated myocarditis. J Immunother Cancer 2021; 9 (03) e002007
  • 39 Mahmood SS, Chen CL, Shapnik N, Krishnan U, Singh HS, Makker V. Myocarditis with tremelimumab plus durvalumab combination therapy for endometrial cancer: a case report. Gynecol Oncol Rep 2018; 25: 74-77
  • 40 Katsume Y, Isawa T, Toi Y. et al Complete atrioventricular block associated with pembrolizumab-induced acute myocarditis: the need for close cardiac monitoring. Intern Med 2018; 57 (21) 3157-3162
  • 41 Yanase T, Moritoki Y, Kondo H, Ueyama D, Akita H, Yasui T. Myocarditis and myasthenia gravis by combined nivolumab and ipilimumab immunotherapy for renal cell carcinoma: a case report of successful management. Urol Case Rep 2020; 34: 101508
  • 42 Safi M, Ahmed H, Al-Azab M. et al PD-1/PDL-1 inhibitors and cardiotoxicity; molecular, etiological and management outlines. J Adv Res 2020; 29: 45-54
  • 43 Lyon AR, Yousaf N, Battisti NML, Moslehi J, Larkin J. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncol 2018; 19 (09) e447-e458
  • 44 Nishimura H, Okazaki T, Tanaka Y. et al Autoimmune dilated cardiomyopathy in PD-1 receptor-deficient mice. Science 2001; 291 (5502) 319-322
  • 45 Okazaki T, Tanaka Y, Nishio R. et al Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat Med 2003; 9 (12) 1477-1483
  • 46 Love VA, Grabie N, Duramad P, Stavrakis G, Sharpe A, Lichtman A. CTLA-4 ablation and interleukin-12 driven differentiation synergistically augment cardiac pathogenicity of cytotoxic T lymphocytes. Circ Res 2007; 101 (03) 248-257
  • 47 Grabie N, Gotsman I, DaCosta R. et al Endothelial programmed death-1 ligand 1 (PD-L1) regulates CD8+ T-cell mediated injury in the heart. Circulation 2007; 116 (18) 2062-2071
  • 48 Johnson DB, Balko JM, Compton ML. et al Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med 2016; 375 (18) 1749-1755
  • 49 Desai MY, Windecker S, Lancellotti P. et al Prevention, diagnosis, and management of radiation-associated cardiac disease: JACC Scientific Expert Panel. J Am Coll Cardiol 2019; 74 (07) 905-927
  • 50 Desai MY, Jellis CL, Kotecha R, Johnston DR, Griffin BP. Radiation-associated cardiac disease: a practical approach to diagnosis and management. JACC Cardiovasc Imaging 2018; 11 (08) 1132-1149
  • 51 Adams MJ, Lipsitz SR, Colan SD. et al Cardiovascular status in long-term survivors of Hodgkin’s disease treated with chest radiotherapy. J Clin Oncol 2004; 22 (15) 3139-3148
  • 52 Orzan F, Brusca A, Gaita F, Giustetto C, Figliomeni MC, Libero L. Associated cardiac lesions in patients with radiation-induced complete heart block. Int J Cardiol 1993; 39 (02) 151-156
  • 53 Cohen SI, Bharati S, Glass J, Lev M. Radiotherapy as a cause of complete atrioventricular block in Hodgkin’s disease. An electrophysiological-pathological correlation. Arch Intern Med 1981; 141 (05) 676-679
  • 54 Slama MS, Le Guludec D, Sebag C. et al Complete atrioventricular block following mediastinal irradiation: a report of six cases. Pacing Clin Electrophysiol 1991; 14 (07) 1112-1118
  • 55 Heidenreich PA, Kapoor JR. Radiation induced heart disease: systemic disorders in heart disease. Heart 2009; 95 (03) 252-258
  • 56 Tzivoni D, Ratzkowski E, Biran S, Brook JG, Stern S. Complete heart block following therapeutic irradiation of the left side of the chest. Chest 1977; 71 (02) 231-234
  • 57 Donnellan E, Jellis CL, Griffin BP. Radiation-associated cardiac disease: from molecular mechanisms to clinical management. Curr Treat Options Cardiovasc Med 2019; 21 (05) 22
  • 58 Tapio S. Pathology and biology of radiation-induced cardiac disease. J Radiat Res (Tokyo) 2016; 57 (05) 439-448
  • 59 Boerma M, Sridharan V, Mao XW. et al Effects of ionizing radiation on the heart. Mutat Res 2016; 770 (Pt B) 319-327
  • 60 Scripture CD, Figg WD. Drug interactions in cancer therapy. Nat Rev Cancer 2006; 6 (07) 546-558
  • 61 Campen CJ, Vogel WH, Shah PJ. Managing drug interactions in cancer therapy: a guide for the advanced practitioner. J Adv Pract Oncol 2017; 8 (06) 609-620
  • 62 Stoll P, Kopittke L. Potential drug-drug interactions in hospitalized patients undergoing systemic chemotherapy: a prospective cohort study. Int J Clin Pharm 2015; 37 (03) 475-484
  • 63 van Leeuwen RWF, Brundel DHS, Neef C. et al Prevalence of potential drug-drug interactions in cancer patients treated with oral anticancer drugs. Br J Cancer 2013; 108 (05) 1071-1078
  • 64 Lam MSH, Ignoffo RJ. A guide to clinically relevant drug interactions in oncology. J Oncol Pharm Pract 2003; 9 (2-3) 45-85
  • 65 Kennedy C, Brewer L, Williams D. Drug interactions. Med (United Kingdom) 2016; 44 (07) 422-426
  • 66 Kheshti R, Aalipour M, Namazi S. A comparison of five common drug-drug interaction software programs regarding accuracy and comprehensiveness. J Res Pharm Pract 2016; 5 (04) 257-263
  • 67 Roblek T, Vaupotic T, Mrhar A, Lainscak M. Drug-drug interaction software in clinical practice: a systematic review. Eur J Clin Pharmacol 2015; 71 (02) 131-142
  • 68 Marcath LA, Xi J, Hoylman EK, Kidwell KM, Kraft SL, Hertz DL. Comparison of nine tools for screening drug-drug interactions of oral oncolytics. J Oncol Pract 2018; 14 (06) e368-e374
  • 69 Wolters Kluwer. Lexi-Interact Online. Published 2021. Available at: https/ www.uptodate.com/drug-interactions/ Accessed February 7, 2021
  • 70 Corporation IBM. Micromedex. Published 2021. Available at: https/ www.micromedexsolutions.com
  • 71 Kaur A, Yu SS, Lee AJ, Chiao TB. Thalidomide-induced sinus bradycardia. Ann Pharmacother 2003; 37 (7-8) 1040-1043
  • 72 Yamaguchi T. Syncope and sinus bradycardia from combined use of thalidomide and beta-blocker. Pharmacoepidemiol Drug Saf 2008; 17 (10) 1033-1035
  • 73 Rogala BG, Charpentier MM, Nguyen MK, Landolf KM, Hamad L, Gaertner KM. Oral anticancer therapy: management of drug interactions. J Oncol Pract 2019; 15 (02) 81-90
  • 74 Haouala A, Widmer N, Duchosal MA, Montemurro M, Buclin T, Decosterd LA. Drug interactions with the tyrosine kinase inhibitors imatinib, dasatinib, and nilotinib. Blood 2011; 117 (08) e75-e87
  • 75 Pembrolizumab Label FDA. Published 2014. Available at: http://www.accessdata.fda.gov/drugsatfda_docs/label/2014/ 125514lbl.pdf. Accessed February 15, 2021
  • 76 Ipilimumab Label FDA. Published 2011. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2018/ 125377s094lbl.pdf. Accessed February 15, 2021
  • 77 Nivolumab Label FDA. Published 2014. Available at: https://www.accessdata.fda.gov/drugsatfda_docs/label/2019/ 125554s070lbl.pdf. Accessed February 15, 2021
  • 78 Khan A, Riaz S, Carhart Jr R. Pembrolizumab-induced mobitz type 2 second-degree atrioventricular block. Case Rep Cardiol 2020; 2020: 8428210
  • 79 Reddy N, Moudgil R, Lopez-Mattei JC. et al Progressive and reversible conduction disease with checkpoint inhibitors. Can J Cardiol 2017; 33 (10) 1335.e13-1335.e15
  • 80 Giancaterino S, Abushamat F, Duran J, Lupercio F, DeMaria A, Hsu JC. Complete heart block and subsequent sudden cardiac death from immune checkpoint inhibitor-associated myocarditis. HeartRhythm Case Rep 2020; 6 (10) 761-764
  • 81 AbbVie Inc. and Genentech USA I. Management of potential VENCLEXTA drug interactions. Published 2021. Available at: https://www.venclextahcp.com/cll/dosing/drug-interactions.html. Accessed April 4, 2021
  • 82 Kusumoto FM, Schoenfeld MH, Barrett C. et al 2018 ACC/AHA/HRS guideline on the evaluation and management of patients with bradycardia and cardiac conduction delay: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. Circulation 2019; 140 (08) e382-e482
  • 83 Brignole M, Auricchio A, Baron-Esquivias G. et al ESC Committee for Practice Guidelines (CPG), Document Reviewers. 2013 ESC Guidelines on cardiac pacing and cardiac resynchronization therapy: the Task Force on cardiac pacing and resynchronization therapy of the European Society of Cardiology (ESC). Developed in collaboration with the European Heart Rhythm Association (EHRA). Eur Heart J 2013; 34 (29) 2281-2329
  • 84 Herrmann J. Adverse cardiac effects of cancer therapies: cardiotoxicity and arrhythmia. Nat Rev Cardiol 2020; 17 (08) 474-502
  • 85 Guglin M, Aljayeh M, Saiyad S, Ali R, Curtis AB. Introducing a new entity: chemotherapy-induced arrhythmia. Europace 2009; 11 (12) 1579-1586
  • 86 Buza V, Rajagopalan B, Curtis AB. Cancer treatment-induced arrhythmias: focus on chemotherapy and targeted therapies. Circ Arrhythm Electrophysiol 2017; 10 (08) 1-12
  • 87 Tshuma N, Glynn N, Evanson J, Powles T, Drake WM. Hypothalamitis and severe hypothalamic dysfunction associated with anti-programmed cell death ligand 1 antibody treatment. Eur J Cancer 2018; 104: 247-249
  • 88 Bukamur HS, Mezughi H, Karem E, Shahoub I, Shweihat Y. Nivolumab-induced third degree atrioventricular block in a patient with stage IV squamous cell lung carcinoma. Cureus 2019; 11 (06) e4869
  • 89 Prevel R, Colin G, Calès V, Renault PA, Mazieres J. Bloc atrio-ventriculaire de grade III compliquant une myocardite au décours d’un traitement par anti-PD1: observation d’un cas et revue de la littérature. Rev Med Interne 2020; 41 (04) 284-288
  • 90 Tan JL, Mugwagwa AN, Cieslik L, Joshi R. Nivolumab-induced myocarditis complicated by complete atrioventricular block in a patient with metastatic non-small cell lung cancer. BMJ Case Rep 2019; 12 (07) e229963
  • 91 Behling J, Kaes J, Münzel T, Grabbe S, Loquai C. New-onset third-degree atrioventricular block because of autoimmune-induced myositis under treatment with anti-programmed cell death-1 (nivolumab) for metastatic melanoma. Melanoma Res 2017; 27 (02) 155-158
  • 92 Samara Y, Yu CL, Dasanu CA. Acute autoimmune myocarditis and hepatitis due to ipilimumab monotherapy for malignant melanoma. J Oncol Pharm Pract 2019; 25 (04) 966-968
  • 93 Hardy T, Yin M, Chavez JA. et al Acute fatal myocarditis after a single dose of anti-PD-1 immunotherapy, autopsy findings: a case report. Cardiovasc Pathol 2020; 46: 107202
  • 94 Szuchan C, Elson L, Alley E. et al Checkpoint inhibitor-induced myocarditis and myasthenia gravis in a recurrent/metastatic thymic carcinoma patient: a case report. Eur Heart J Case Rep 2020; 4 (03) 1-8
  • 95 Lee DH, Armanious M, Huang J, Jeong D, Druta M, Fradley MG. Case of pembrolizumab-induced myocarditis presenting as torsades de pointes with safe re-challenge. J Oncol Pharm Pract 2020; 26 (06) 1544-1548
  • 96 Agrawal N, Khunger A, Vachhani P. et al Cardiac toxicity associated with immune checkpoint inhibitors: case series and review of the literature. Case Rep Oncol 2019; 12 (01) 260-276
  • 97 Brahmer JR, Lacchetti C, Schneider BJ. et al National Comprehensive Cancer Network. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2018; 36 (17) 1714-1768
  • 98 Thompson JA, Schneider BJ, Brahmer J. et al NCCN guidelines insights: management of immunotherapy-related toxicities, version 1.2020. J Natl Compr Canc Netw 2020; 18 (03) 230-241
  • 99 Adams MJ, Lipshultz SE, Schwartz C, Fajardo LF, Coen V, Constine LS. Radiation-associated cardiovascular disease: manifestations and management. Semin Radiat Oncol 2003; 13 (03) 346-356
  • 100 Shen W-K, Sheldon RS, Benditt DG. et al 2017 ACC/AHA/HRS guideline for the evaluation and management of patients with syncope: a report of the American College of Cardiology/American Heart Association Task Force on Clinical Practice Guidelines and the Heart Rhythm Society. J Am Coll Cardiol 2017; 70 (05) e39-e110

Zoom Image
Avirup Guha
Zoom Image
Fig. 1 Immune checkpoint inhibitor-induced bradyarrhythmia ECG (case 2). ECG, electrocardiogram.
Zoom Image
Fig. 2 Radiation-induced bradyarrhythmia ECG (case 3). ECG, electrocardiogram.
Zoom Image
Fig. 3 Radiation-induced bradyarrhythmia ECG (case 4). ECG, electrocardiogram.
Zoom Image
Fig. 4 Drug–Drug interactions between anticancer and nodal blocking agents (created by biorender.com).
Zoom Image
Fig. 5 Proposed algorithm for the management of bradycardia/bradyarrhythmia, with a special focus on ICI- and radiation-induced conduction abnormalities [49] [97] [98] [99]w[100] (created by biorender.com). CK, creatine kinase; CMR, cardiac magnetic resonance; CRP, C-reactive protein; CXR, chest X-ray; ECG, electrocardiogram; EMB, endomyocardial biopsy; ESR, erythrocyte sedimentation rate; NT proBNP, N-terminal pro-brain natriuretic peptide; TTE, transthoracic echocardiography; WBC, white blood cell count.