Next Article in Journal
Structures and Anti-Inflammatory Evaluation of Phenylpropanoid Derivatives from the Aerial Parts of Dioscorea polystachya
Previous Article in Journal
Semi-Empirical Calculation of Bodipy Aggregate Spectroscopic Properties through Direct Sampling of Configurational Ensembles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cardiac Toxicity Associated with Immune Checkpoint Inhibitors: A Systematic Review

by
Angela Cozma
1,
Nicolae Dan Sporis
2,
Andrada Luciana Lazar
3,
Andrei Buruiana
2,*,
Andreea Maria Ganea
1,
Toma Vlad Malinescu
1,
Bianca Mihaela Berechet
1,
Adriana Fodor
4,
Adela Viviana Sitar-Taut
1,
Vasile Calin Vlad
1,
Vasile Negrean
1 and
Olga Hilda Orasan
1
1
Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
2
Department of Medical Oncology, Prof. Dr. I. Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
3
Department of Dermatology, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
4
Clinical Centre of Diabetes, Nutrition and Metabolic Disease, Iuliu Hatieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(18), 10948; https://doi.org/10.3390/ijms231810948
Submission received: 15 August 2022 / Revised: 9 September 2022 / Accepted: 16 September 2022 / Published: 19 September 2022
(This article belongs to the Section Biochemistry)

Abstract

:
Immune checkpoint inhibitors (ICIs) are an important advancement in the field of cancer treatment, significantly improving the survival of patients with a series of advanced malignancies, like melanoma, non-small cell lung cancer (NSCLC), hepatocellular carcinoma (HCC), renal cell carcinoma (RCC), and Hodgkin lymphoma. ICIs act upon T lymphocytes and antigen-presenting cells, targeting programmed cell death protein 1 (PD1), programmed cell death protein ligand 1 (PD-L1), and cytotoxic T-lymphocyte antigen 4 (CTLA-4), breaking the immune tolerance of the T cells against malignant cells and enhancing the body’s own immune response. A variety of cardiac-adverse effects are associated with ICI-based treatment, including pericarditis, arrhythmias, cardiomyopathy, and acute coronary syndrome, with myocarditis being the most studied due to its often-unexpected onset and severity. Overall, Myocarditis is rare but presents an immune-related adverse event (irAE) that has a high fatality rate. Considering the rising number of oncological patients treated with ICIs and the severity of their potential adverse effects, a good understanding and continuous investigation of cardiac irAEs is of the utmost importance. This systematic review aimed to revise recent publications (between 2016–2022) on ICI-induced cardiac toxicities and highlight the therapeutical approach and evolution in the selected cases.

1. Introduction

Immune checkpoint inhibitors (ICIs) are an important advancement in the field of cancer treatment, significantly improving the survival of patients with a series of advanced malignancies. The most common types of malignancies that benefited from the introduction of ICI-based therapies are melanoma, non-small cell lung cancer (NSCLC), hepatocellular carcinoma, renal cell carcinoma (RCC), and Hodgkin lymphoma [1,2].
Normally, antigens are processed by antigen-presenting cells (APCs) and presented as peptide-major histocompatibility complexes (MHC) to naive lymphocytes in secondary lymphoid organs. This interaction is facilitated by the additional ligand-receptor binding of CD28 (expressed on T cells) and CD80 or CD86 (expressed on APCs). As a consequence, activated lymphocytes clonally proliferate and differentiate towards CD4+ helper or CD8+ cytotoxic phenotypes. These effector cells will further migrate and recognize the antigens in their original location via T cell receptors (TCRs). In physiologic conditions, several mechanisms dampen the activation of T-cells in order to prevent autoimmunity. Therefore, CTLA-4 is upregulated in activated T cells and negatively competes with CD28 in this process. Besides this, CTLA-4 is crucial for the maintenance of self-tolerance by the FoxP3+ regulatory T lymphocytes (Treg). Similarly, PD-1 expressed by activated T cells interacts with its ligand, PD-L1, which is expressed by APCs, lymphocytes, and epithelial cells in order to prevent aberrant immune reactions. In cancer, PD-L1 expression by tumors cells is a well-known mechanism of immune evasion. Blocking these pathways by use of ICIs restores the activity of the T cells but, as mentioned before, diminishes the immune tolerance and consequently promotes autoimmunity.
As ICI-based treatment is emerging in a wide spectrum of cancers with different primary sites such as gastro-intestinal (GI), head and neck, triple-negative breast cancer, and bladder cancers, it is of significant importance to identify and manage the adverse effects that are arising [2]. The lowered immune tolerance induced by ICIs brought to light a series of immune-related adverse events (irAEs) involving various organ systems.
IrAEs include dermatologic, GI, hepatic, endocrine, and other less common inflammatory events [1]. While most common irAEs can be managed with temporary immunosuppression using corticosteroids and/or other agents, some pose a vital risk for the patient and require prompt management and the complete discontinuation of ICI administration. IrAEs are classified according to the Common Terminology Criteria for Adverse Events from grades 1 to 5. Between 60 to 80% of patients treated with ICIs experience irAEs, and around 13–23% require permanent discontinuation as a result of grade 2, 3, or 4 AEs [1,3]. The risk and severity of irAEs is increased in patients treated with a combination of CTLA-4 and anti-PD1/PD-L1 [4]. A variety of cardiac adverse effects are associated with ICI-based treatment, including pericarditis, arrhythmias, cardiomyopathy, and acute coronary syndrome, with myocarditis being the most studied due to its often-unexpected onset and severity.
In this review, we have mainly focused on the clinical aspects of the ICI-related cardiac autoimmunity; a more detailed picture of these mechanisms is presented elsewhere [5,6]. Briefly, CD8+ lymphocytes recognize myocardial antigens, kill cardiomyocytes, and secrete IFN-γ. Further on, IFN-γ can induce PD-L1 expression in various cells, including cardiomyocytes and endothelial cells (ECs), as a protection mechanism. On the other hand, CD4+ lymphocytes are recruited and favor macrophage activation (Figure 1). It appears that the inhibition of the PD-1/PD-L1 and CTLA4 axes leads to cardiotoxicity in slightly different manners. For instance, in PD-1−/− mice, cardiac troponin I autoantibodies are present and contribute to the development of autoimmune dilative cardiomyopathy [7], and PD-1-deficient MRL mice acquire lymphocytic myocarditis [8]. The immunization of PD-1−/−, but not PD-L1−/−, mice, with a peptide of the mouse α-myosin H chain, results in a primarily neutrophilic infiltration, also with an important lymphocytic component responsible for the secretion of IL-17A and IFN-γ [9]. Additionally, the transfer of PD-1−/− CD8+ T cells in cMy-mOVA mice results in the higher proliferative activity of cytotoxic lymphocytes and elevated levels of cytokines and chemokines (IFN-γ, TNF-α, CXCL10, CCL3, CCL5) compared to their PD-1+/+ counterparts [9]. Compared to PD-1−/− mice, CTLA-4−/− mice do not exhibit autoantibodies, and a non-specific activation of CD4+ T cells may be responsible for the inflammatory response. Johnson et al. showed, by next generation sequencing, that myocardial and tumoral T cells resemble the same clonal origin [10], suggesting the existence of a shared epitope between the tumor and the myocardial cells. Thus, besides necrosis and oedema, histological samples of ICI-induced myocarditis consist of mononuclear cell infiltration: mixed CD4+ and CD8+ lymphocytic populations (usually with the predominance of cytotoxic T cells), macrophages (CD68+), and occasionally CD20+ B cells. Multinuclear giant cells were often encountered in severe, fulminant cases. Moreover, strong PD-L1 expression and the positive expression of HLA-DR (marker for IFN-γ production) are usually described. Notably, the absence of FoxP3+ Treg is also suggestive of ICI exposure [11,12]. Myocarditis is overall seen as a rare but frequently fatal irAE, with a fatality rate of up to 46% [13].
Regarding acute coronary syndromes, checkpoint blockade seems to be involved in the pathogenesis of atherosclerotic lesions, although the results are contradictory. Therefore, even if PD-1−/− LDLR−/− mice present an elevated formation of atherosclerosis plaques [14], a retrospective study showed that PD-1 inhibitor, nivolumab, was associated with a favorable course for atherosclerotic lesions [15].
Several novel checkpoint pathways are also currently under investigation. Lymphocyte activation gene-3 (LAG-3) binds to MHC-II and blocks antigen presentation, while T cell immunoglobulin-3 (TIM-3) and T cell immunoreceptor, with Ig and ITIM domains (TIGIT), are intrinsic inhibitors of T cells. In clinical settings, LAG-3 deficiency was shown to be associated with a proinflammatory state and a higher risk of coronary artery disease [16]. Similarly, TIGIT-positive Tregs possess a protective role regarding atherosclerotic lesions and are severely reduced in acute coronary syndrome [17]. However, TIM-3 expression was correlated with the severity of CHD, suggesting a different effect on lipid metabolism and atherosclerosis formation [18].
Considering the rising number of oncological patients treated with ICIs and the severity of the potential adverse effects, a good understanding and continuous investigation of cardiac irAEs is of the utmost importance. This systematic review aimed to revise recent publications published between January 2016–September 2022 on ICI-induced cardiac toxicities. Furthermore, we highlighted the treatment approach and evolution in selected cases.

2. Results

2.1. ICI-Induced Myocarditis

The most common cardiovascular side effect in ICI-treated oncologic patients is myocarditis. The inflammation of the heart muscle is often associated with arrhythmias, myositis, or myasthenia gravis (MG) (Table 1, the complete data regarding ICI-induced myocarditis is presented in the Supplementary material Table S1). Thus, of the total studies included, 88 out of 128 (68.75%) were of case reports of ICI-associated myocarditis.
The majority of ICI-induced myocarditis cases were in male patients. At the same time, most of the cases were reported in subjects between 70 and 79 years, the youngest being 25 yo and the oldest 86 yo [19]. Regarding the underlying oncological disease, most of the cases were reported for patients diagnosed with melanoma [11,12,20,21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39], renal cancer [40,41,42,43,44,45,46,47,48,49], and lung neoplasms [33,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69]
Most patients with ICI-induced myocarditis complain of shortness of breath, tachycardia or palpitations, fatigue, chest pain, cough, and episodes of syncope (See Table 1); cardiogenic shock was encountered in some of the cases [46,59,66]. It is noteworthy that a minority of patients had subclinical myocarditis [29,44,49,69]. However, even if they were asymptomatic from a cardiovascular standpoint, the presence of other irAEs, especially neuromuscular, guided further investigations. Notably, signs and symptoms of myositis/MG, such as diplopia, ptosis, blurred vision, dysphagia, muscle weakness, posterior neck pain, and neck drop, were reported in several studies (See Table 1). In such patients, dyspnea may be the result of both myocarditis-induced heart failure and myositis/MG.
In approximatively 25% of cases, ICI-induced myocarditis is a part of an overlap syndrome with MG and/or myositis [10,13]. A plausible explanation of this association is the presence of cross-antigens between the tumor and striated muscles [70,71]. Cancers with high-tumor burden (e.g., melanoma, NSCLC) display a higher incidence of irAEs, probably due to antigen spreading [72]. Although, in idiopathic MG, the association with myositis and/or myocarditis is relatively rare, the presence of anti-striational antibodies is correlated with a higher incidence of cardiac involvement [73,74]. Anti-AChR antibodies do not cross-react with heart muscles [75]. In severe myositis, troponin T could reflect the active regeneration of skeletal muscles [76].
Hepatitis, renal disturbances, thyroid dysfunction [28,30,57,77], Stevens–Johnson syndrome/toxic epidermal necrolysis [78], and DRESS syndrome [79] were also described in patients with ICI-induced myocarditis. Skin rashes and fever (See Table 1) were encountered in several patients.
Regarding the diagnosis, although a few patients presented a normal electrocardiogram (ECG) in spite of the cardiac symptoms (See Table 1), most of them presented electrocardiographic changes such as sinus tachycardia [30,80], bradycardia [24], atrial flutter [81] and atrial fibrillation [82], atrioventricular block [43,47,78], complete heart block [24,50,55], left bundle branch block [49], right bundle branch block (See Table 1), ST-segment depression [25,83], elevation [30,46,58,78] or non-specific ST-T changes [25,28], T wave inversion [77] ventricular tachycardia [26,59,84], and other changes, such as small QRS complexes [25,55].
Laboratory analyses, such as hs-TnI, CK, CK-MB, BNP, and NT-proBNP, were determined in the majority of cases (See Table 1). It is noteworthy that, when myositis/MG was suspected, complementary laboratory tests, such as anti-acetylcholine receptor antibodies (AChR), myohemoglobin, RyR-Ab, anti-NOR-90, anti-Ro-52 antibodies, anti-muscle specific kinase (MuSK) [85], anti-striated muscle, Asialo-GM1 [40], anti-GM1 [20], anti-SM [86], anti-TG, anti-TPO, anti-mitochondria, anti-SRP, and anti-PM/Scl100 [65] were carried out (See Table 1). In some patients, the EMG studies revealed a myogenic disorder [47] with a short duration and a reduced amplitude of muscle potential [80].
Normal wall motion and preserved EF were found in some of the cases (See Table 1); however, most patients had an abnormal echocardiogram. Thus, TTE revealed a depreciated EF (See Table 1), wall motion abnormalities, or global hypokinesis (See Table 1). Serial ultrasonographic examinations should be taken into consideration since, initially, normal kinetics or regional abnormalities can rapidly progress to global hypokinesis/akinesis [36,67]. Long-term screening is also needed in order to identify a possible evolution towards apical ballooning (Takotsubo cardiomyopathy) [55]. In terms of structural abnormalities, the echocardiogram depicted dilated atrial and ventricular cavities (See Table 1) and a thickening of the interventricular septum [38,49,83,87] and left ventricular wall, respectively [49,55,85,88]. Pericardial effusion [11,37,51,55], apical thrombi [55], and valvular regurgitation [38,83] were also found. Additionally, GLS measurements were performed in five patients, showing abnormal values in four of those patients (−14.4%; −15.7%) [11,20,39,49].
Chest X-rays uncovered an abnormal cardiothoracic index with pulmonary congestion [55,83], and pleural effusion [89]. The aforementioned abnormalities were also found with chest computer tomography (CT), which showed pleural and pericardial effusions, pulmonary oedema, and atelectasis [25]. Coronary angiographies ruled out acute coronary syndrome (See Table 1) in those patients with ICI-induced myocarditis.
Cardiac magnetic resonance imaging (cardiac MRI, CMR) was performed in the majority of cases. The most frequent findings associated with ICI-induced myocarditis are represented by oedema of the myocardium (T2-weighted imaging revealed high signal intensities consistent with acute myocarditis) and areas of late gadolinium enhancement (LGE), together with pericardial effusion (See Table 1). Extensive ventricular fibrosis was identified by MRI in some cases [46].
Cardiac catheterization and myocardial biopsies were performed on 23 patients. Endomyocardial biopsy (EMB) represents the gold standard procedure for myocarditis diagnosis [90]. The histopathology, together with the immunochemistry examinations, revealed an admixed interstitial inflammatory infiltration, consisting of T lymphocytes CD3+, CD8+, and macrophages (CD68+) (See Table 1). Furthermore, T lymphocytes expressed PD-1, while the CD68+ macrophages expressed PD-L1 checkpoint proteins [55,87]. The histopathological findings suggest that T lymphocytes play a crucial role in the pathogenesis of ICI-associated myocarditis. Additionally, several animal studies proved that PD-1 [8] and CTLA-4 [91] deficiencies predispose the animal to autoimmune myocarditis with a fatal outcome [8]. Interestingly, a recent study by Xia et al. showed that PD-L1-inhibitor treatment is responsible for the upregulation of M1 macrophage populations. An increased population of M1 macrophages has been documented in cardiac inflammation. Additionally, miR-34a expression was found to be higher in the macrophages of PD-L1 inhibitor-treated mice compared to the controls. The M1/M2 polarization of macrophages is regulated by miR-34a via KLF4 [92]. It is noteworthy that miR-34a exacerbates myocardial inflammation via the TGF-β/Smad-signaling pathway [93]. Collagen was also found admixed in with the inflammatory cells in a case reported by Norwood et al. Furthermore, the predominance of CD8+ with few CD4+ lymphocytes in the inflammatory infiltrate was once again established [28]. A case of chronic smoldering myocarditis was diagnosed when the EBM revealed myocardial infiltration by CD4+ and CD8+ T lymphocytes together with a considerable number of histiocytes and macrophages [46]. Neutrophils and eosinophils, together with myocardial necrosis [58,89], were also found in anatomopathological examinations. To better understand the pathophysiology and improve the diagnosis, several authors made use of immunohistochemistry (IHC) or cytokine analysis [55,87,94]. These could provide a better stratification of these entities and identify cell populations and cytokines in order to elect specific immunomodulatory therapies [11]. It is noteworthy that, in several patients, viral serology was positive for influenza [84], Coxsackie B [47], parvovirus B19, and HHV6 [64]. In these cases, it is not clear if the etiology of myocarditis was viral, autoimmune, or a combination of both.

2.1.1. Treatment of ICI-Induced Myocarditis

The first therapeutic intervention in ICI myocarditis cases was the cessation of the incriminated drug. Steroid therapy is the main treatment for ICI-induced myocarditis. The efficacy of high doses of systemic corticosteroids, such as intravenous methylprednisolone, was documented in several studies (See Table 1). However, steroids could promote a transient myasthenic crisis in half of MG patients [49,95], and for that reason, it is recommended to associate it with intravenous immunoglobulin (IVIG) or plasmapheresis in these patients [96]. IVIG and plasmapheresis reduce autoantibodies in neuromuscular AEs [97]. Even if IVIG presents a relatively safer profile compared to other immunomodulatory strategies [94], it should be restricted in patients with high thromboembolic risk [85]. Additionally, plasma exchange (PLEX) is considered more suitable for MG patients with positive anti-MuSK antibodies [98].
In cases with an unfavorable evolution, the association of IVIG to the therapeutic regimen represents a reasonable option (See Table 1). The oral corticosteroid taper, following intravenous methylprednisolone administration, is mandatory (See Table 1). Other therapeutic interventions are represented by infliximab (monoclonal anti-tumor necrosis alpha antibody) [27,66,89,99], mycophenolate mofetil (MMF), together with oral prednisone [25], abatacept [47], or tacrolimus [30]. Furthermore, several patients do not respond to corticotherapy; in these patients, plasmapheresis, MMF, antithymocyte globulin, infliximab, alemtuzumab, tacrolimus, abatacept, tofacitinib, or tocilizumab, may be taken into consideration (See Table 1). In myasthenia-gravis, several alternatives include cyclophosphamide, azathioprine, methotrexate, or rituximab [63].
When a complete heart block is proved via ECG, a pacemaker (temporary or permanent) is required (See Table 1). In one case of hemophagocytic syndrome, cyclosporine was added to the therapeutic regimen [55]. PLEX [26] plasmapheresis [31,78], haemodialysis [87], non-invasive positive pressure ventilation [58], ECMO [30,66] intra-aortic balloon pump (IABP), and a defibrillator with cardiac resynchronization [66] were also needed in critically ill patients. Most of the patients required a complex therapeutic regimen consisting of a combination of the aforementioned interventions [29,31,78,89].

2.1.2. Evolution of ICI-Induced Myocarditis

ICI myocarditis was lethal in 23 of the cases included in the present review. An anti-PD-1 antibody was administered in most of the fatal cases, namely pembrolizumab [55,89,99] or nivolumab [45,59,87]. Fatal outcomes saw myocarditis patients treated with an association of ICI (anti-PD-1 + anti-CTLA-4 inhibitors) [27,31]. In the present review, myocarditis onset presented large temporal variations in relation to ICI administration. Thus, it developed as early as 1 week after the first ipilimumab + nivolumab infusion [29], but it was also diagnosed after long-lasting treatment with pembrolizumab (8 cycles, 1 year) [57].
It is noteworthy that there is a lack of medical history documentation in the majority of ICI-associated myocarditis patients. Special consideration should have been attributed to the previously diagnosed cardiovascular pathologies and autoimmune diseases. Another critical aspect that should be cautiously taken into consideration is represented by the infectious ethology of myocarditis. Additionally, several patients had a history of thymoma (See Table 1). The association between thymoma, myocarditis [100], MG, and myositis [101,102] is well known. Thus, thymoma-induced autoimmunity is also plausible in patients with thymic tumors treated with ICI [35].

2.2. ICI-Induced Pericarditis

ICIs have been associated with the development of other various cardiovascular toxicities, including pericarditis. Regarding pericardial diseases associated with ICI-based therapy, we analyzed several case reports and the time to onset of pericarditis was 5 to 340 days; most of the cases were reported in male subjects with an average age of 60 years. Moreover, at particular intervals of time, the patients developed clinical manifestations, like dyspnea, thoracic pain, fatigue, and general malaise (See Table 2). Furthermore, a patient presented dyspnea with tachycardia and a low-grade fever [103], limb oedema, and increased body weight [104], while others developed severe hypotension due to cardiogenic shock [105]. With regard to the associated comorbidities with ICI-related pericarditis, only asthma and hypertension were mentioned [106]. It is also noteworthy that previous immune toxicity, characterized by thyroiditis [103,107], autoimmune hepatitis [104,107], generalized pruriginous rash, [106,107], colitis, and MG [106], may show a predisposition to autoimmune toxicity with these drugs.
After the initial clinical evaluation, for a complete diagnosis, the management of ICI-related pericarditis included an ECG, echocardiogram, CT, and MRI. The ECG findings ranged from normal ECGs [103,104,108,109] to new T-wave inversions in anterolateral leads and prolonged QT intervals [107,110], sinus tachycardia and decreased QRS voltage [105,107], atrial fibrillation with a high heart rate and an ST segment elevation [96,111], and in some cases, non-specific repolarisation anomalies [106].
Most of the patients with ICI-induced pericarditis experienced preserved LVEF and pericardial effusion without signs of hemodynamic impairment [104,105,106,107,108,109,111], but there were two cases described in which the echocardiogram emphasized an important restrictive pericardial effusion with “swimming heart”, suggestive of cardiac tamponade [103,110].
A single chest X-ray showed moderate bilateral pleural effusions and an enlarged heard silhouette [107]; in most of the cases, for better accuracy, CT scans were performed, which confirmed the pericardial effusion [103,104,107,109]. For a better investigation, the cardiac MRI showed a marked pericardial hyperintensity in T2-weighted sequences as a sign of active inflammation [109]. Cardiac MRI proved the presence of pericardial thickening, although without evidence of myocardial oedema or marked late godalinium enhancement in the pericardium [104].
What is more, the cardiac enzymes, such as troponin levels and BNP, remained within the normal range in patients with ICI-related pericarditis [105,107,110,111], except for one case where the serum troponin-T level was high, but there was no evidence of active myocarditis from the endomyocardial biopsy. Elevated serum troponin indicated myocardial inflammation that was associated with severe inflammation of the pericardium [104]. In some cases of ICI-pericarditis, the histopathologic evaluation showed only non-specific chronic inflammation with extensive fibrosis and lymphocyte infiltration, with no evidence of malignancy or microorganisms [103,105,107].
Regarding the treatment of ICI, pericardiocentesis remains the preferred procedure for the acute management of massive pericardial effusions by either mechanism [103,104,105,107,108,110]. Corticosteroids are a prefered treatment, needing only a prompt initiation. [96] In our reviewed cases, corticosteroids were administered to all the patients with ICI-related pericardial effusions (See Table 2). The treatment starts with high doses of prednisone (1–2 mg/kg) with a tapering off of at least 4 weeks in selected cases. Transplant rejection doses must be administered in unresponsive cases, which means 1 g methylprednisolone given daily in association with mycophenolate mofetil, infliximab [104], or anti thymocyte globulin [110].
In the present review, the majority of ICI-induced pericarditis has not developed any other immune-related adverse events (See Table 2). These case reports highlight the importance of prompt diagnosis and high-dose corticosteroid treatment for immune-related pericarditis. Nevertheless, in one case (of a 54-year-old patient diagnosed with lung adenocarcinoma who was treated with ICI), it was discovered that the patient had moderate paraneoplastic pericarditis; a tapering scheme of prednisone was administered (10 mg/day, every 2 weeks) in combination with colchicine for 6 months to prevent another pericardial effusion. His cardiac condition was evaluated again by echocardiogram, which was substantially stable compared to the previous one. Thus, a second cycle of chemotherapy was initiated without administering pembrolizumab (given the high doses of prednisone ongoing). Unfortunately, the patient experienced acute dyspnea and succumbed to sudden death after less than two months after the first cycle of chemotherapy. No autopsy was conducted as per the family’s wish [111].
To stratify the patient risk before starting ICI treatment, it is recommended that complete blood tests be performed, including troponin and brain natriuretic peptide (BNP), ECG, and echocardiogram, besides the usual clinical follow-up. The echocardiogram, cardiac biomarkers, and/or cardiac MRI are useful investigations for diagnosing late-onset cardiotoxicity, both in the initial and later phases of ICI therapy [96].

2.3. ICI-Induced Arrythmias

New-onset or pre-existing arrhythmias with ICI-use (Table 3) ranged from tachyarrhythmias, such as atrial fibrillation [112] and sinus tachycardia [113] to bradyarrhythmias, such as atrioventricular block [114]. These adverse effects were observed mostly in male patients with metastatic melanoma [112,115,116], metastatic sarcoma [113], metastatic NSCLC [115,117], and end-stage liver cancer [118].
After a period (running from two weeks to five months) following the initiation of ICI therapy, the patients met the definition of ICI-induced arrhythmias. They tended to be older and more likely to have a history of hypertension, diabetes mellitus [112,115,117], and hyperlipidemia [117]. It is noteworthy that a single patient with pre-existing atrial fibrillation underwent three prior ablations in the past [112]. Regarding the anamnesis and clinical examination in patients with arrhythmias, some patients remained asymptomatic [112,116]. Most of them developed manifestations, like increased fatigue, generalized malaise, dizziness, or syncope [112,113,118,119], while others experienced palpitations [119] or worsening dyspnea [115].
A combination of ECGs, echocardiograms, cardiac catheterizations, and biomarkers was used to identify cases of ICI-induced arrhythmias after the initiation of immune checkpoint inhibitor therapy. Twelve-lead ECGs were recorded with the presence of atrial fibrillation with rapid ventricular response [112], sinus tachycardia associated with first-degree atrioventricular block, right bundle branch block, and left anterior fascicular block, then accelerated junctional rhythm with complete heart block [113], symptomatic sinus bradycardia of 40 bpm [112,118], a new first-degree AV block [116], Mobitz type 2 s-degree AV block of 30 bpm followed by complete heart block with a ventricular rate of 22 bpm after 3 h [117], a third-degree AV block with a bradycardia of 44 bpm [115], and complete AV block with wide QRS complexes [119].
Furthermore, QT dispersion (QTd) is measured as the difference between the longest and the shortest QT intervals within a 12-lead ECG. Prolongation of corrected QT interval (QTc) is an important indicator of arrhythmogenesis [116]. In this review, there was one study that mentioned a significant increase in QTd after treatment initiation with a combination of ICI, while no significant difference was observed in patients treated with PD-1i monotherapy [116].
In 90% of the cases with ICI-induced arrhythmias, the transthoracic echocardiogram did not reveal abnormalities in wall motion, with the preservation of the ejection fraction of the left ventricle. Nevertheless, an unusual case of progressive conduction abnormalities after the initiation of checkpoint inhibitors, anti-PD-L1 and anti-CTL4, showed a moderate decrease in the left ventricle ejection fraction (30–35%), associated with kinetic disorders (anteroseptal hypokinesis and apical akinesis with septal motion consistent with conduction abnormality) in the setting of a normally sized left ventricle. The systolic function and contractility of the right ventricle were reduced in spite of no significant valvular disease or pericardial effusion [113].
Laboratory results were negative for elevations in troponin or BNP in half of the cases [116,117,118], but increased myoglobin, creatin-kinase, and troponin were detected in the rest of them [113,115,119]. Cardiac catheterization was also performed in three cases of ICI-induced arrhythmias, which did not reveal any significant stenosis [115,119] or there were minor luminal irregularities in the coronary arteries [113].
The evaluation for the immune-related effects, like hyperthyroidism, was notable in only one case, which was felt to possibly be drug-induced toxicity from the ICIs, treated with methimazole and steroids [112]. A suggested approach to the management of a patient with suspected ICI-induced arrhythmias is outlined following the guidelines for the management of patients presenting different arrhythmias. Self-limited AF resolved spontaneously after about 48 h [112], persistent AF managed with an echocardiogram (TEE)-guided cardioversion, anticoagulation and beta blockers and antiarrhythmic drugs [112], implantation of a temporary pacemaker, then permanent pacemaker for the heart conduction disorders [113,115,119] and association of cortisone in immune-mediated sinoatrial node dysfunction with immediate improvement of symptoms and bradycardia without relapsing-symptoms [118]
None of the ICI-induced arrhythmias cases required stopping treatment with the ICIs, and in most instances, the arrhythmias could be managed in a relatively straightforward manner with positive evolution, except for one case where pembrolizumab was discontinued due to the progressive status of his HCC. Unfortunately, one patient 26 days after beginning anti-PD-1treatment. Considering the metastatic malignant underlying disease, and the worsening hemodynamic and respiratory situation, the relatives made the decision to reduce medical care with an unfavorable prognosis.

2.4. Takotsubo Cardiomyopathy and Acute Heart Failure

ICIs have also been associated with the development of other various cardiovascular toxicities, such as acute heart failure, cardiomyopathies, Takotsubo syndrome, and acute coronary syndromes. Concerning cardiomyopathies, including Takotsubo syndrome, we analyzed several case reports and the time to onset of cardiomyopathy is 4 to 112 days; most of the cases were reported in male subjects with an average age of 64 yo, the youngest being 42 yo and the oldest 79 yo.
Regarding the underlying oncological disease, cases have been reported in patients diagnosed with melanoma [120,121,122,123,124], lung neoplasms [125,126], hepatocellular carcinoma [127], breast cancer [128], and squamous cell carcinoma of the lip [129]. Most of the cases were described as Takotsubo cardiomyopathies [120,121,122,123,127,129], while two cases of acute heart failure were also described [125,126]. It is noteworthy that, in two cases, the ICI-induced cardiomyopathies were associated with acute renal failure [129] and with myopericarditis, respectively [127]. The majority of patients with ICI-induced cardiomyopathies complained of chest pain and shortness of breath. Diaphoresis, nausea, vomiting, and lower extremity oedema were encountered in some of the cases (Table 4).
In regard to the diagnosis, most of the patients presented ECG changes, such as sinus tachycardia [123], atrial flutter [126], ST elevation [121,126], or T wave changes [120,122]. A TTE was performed in all the cases and was always abnormal. In patients diagnosed with Takotsubo syndrome, TTE revealed apical akinesis with apical ballooning, hyperdynamic basal LV segments, and a reduced cardiac ejection fraction [120,121,122,127,129]. One case of atypical Takotsubo cardiomyopathy was also described whereby the TTE revealed hypokinesia of the mid inferior and mid anterior septum, mid inferolateral, mid anterolateral, mid inferior and mid anterior wall, sparing the apical and basal segments [123]. In acute heart failure cases, the wall motion abnormalities were not systematized [125,126].
Chest X-rays uncovered pulmonary congestion, pulmonary effusion, and cardiac dilation, but these were performed only in a few cases [123,126]. A CMR was obtained in the majority of cases, mainly to rule out the diagnosis of ICI-induced myocarditis. It is noteworthy that one patient had concomitant myopericarditis and Takotsubo syndrome following ICI therapy. In this case, the CMR revealed mid-myocardial to subepicardial delayed enhancement (MDE) in the inferior lateral walls, as well as in the apex [127].
The laboratory analyses (TnI, CK, CK-MB, BNP, and NT-proBNP) were determined in the majority of cases and were almost always elevated, which raised the question of an acute coronary syndrome [120,121,122,123,125,127,129]. Subsequently, a coronary angiography was performed in the majority of the cases and an acute myocardial infarction was ruled out [121,122,123,125,126,127,129]. Cardiac catheterization and myocardial biopsies were performed in two patients, which excluded a possible myocarditis [120,126].
The first therapeutic intervention (when ICI-induced side effects were suspected) was the cessation of the incriminated drug. Immunosuppressive therapy with intravenous corticosteroids was initiated and continued with oral prednisone taper when myocarditis was diagnosed [127] or suspected [122,125,127,129]. In all cases, guideline-directed heart failure treatment was started with betablockers, ACE inhibitors, and diuretics, as well as IV dopamine and tolvaptan in one severe acute heart failure case [126]. After the ICI treatment was discontinued and heart failure treatment was initiated, the evolution was favorable in all the cases, with an improvement in the symptoms and the left ventricular ejection fraction [120,121,122,123,125,126,127,129].

2.5. Acute Coronary Syndrome

We analyzed the case reports of acute coronary syndromes (ACS) related to ICI administration, and the time to onset of ACS is 2–365 days, and most of the cases were reported in male subjects with an average age of 67 years, the youngest being 52 yo and the oldest 87 yo. Concerning underlying oncological disease, the cases were reported in patients diagnosed with lung cancer [130,131,132,133,134], melanoma [135], and renal cell carcinoma [136], as we have presented in Table 5.
More than 70% of the cases were described as non-ST elevation myocardial infarction (NSTEMI), but ST-elevation myocardial infarction (STEMI) [133] and vasospastic angina were also described [136,137,138]. The main clinical manifestations of ICI-associated ACS were chest pain and dyspnea; only one case reported no symptoms, and the diagnosis was based on the elevated cardiac biomarkers [131]. Concerning concomitant autoimmune diseases, one patient developed grade five colitis while being hospitalized for ICI-related myocardial infarction [134]. Another patient presented related concomitant autoimmune manifestations, namely erythema multiforme, thyroid dysfunction, and interstitial pneumonia [131], while another case with associated ICI-induced hepatitis was also reported [131]. The concomitant autoimmune diseases were treated with high-dose intravenous corticosteroids, followed by oral corticosteroids taper.
Most of the patients had an abnormal ECG with ST-depression in various territories [130,135,136], T-wave inversions [132], Q-waves [116,134] and ST-elevation [132]. One case report revealed no ECG abnormalities [131]. The laboratory analyses (TnI, CK, CK-MB, BNP and NT-proBNP) were determined in the majority of cases and revealed abnormal values, particularly the troponins [130,131,132,133,134,135,136]. In every case, emergency coronary angiography was performed and revealed significant stenosis or occlusion of the coronary arteries [130,131,132,133,134,135], except for the case of vasospastic angina [136].
Most of the patients underwent a percutaneous coronary intervention with implantation of drug-eluting stents [112,113,114,115,116]. However, one patient with underlying severe CAD was surgically treated with coronary artery bypass grafts (CABG) for the affected arteries [135]. The patient diagnosed with vasospastic angina was treated with calcium channel blockers and nitrates, and the symptoms gradually disappeared [136]. All the patients were discharged with chronic myocardial infarction treatment, namely dual antiplatelet therapy, ACE inhibitors, beta-blockers, and statins, except for the patient diagnosed with vasospastic angina [136]. The evolution of the patients was almost always favorable [130,131,132,133,134,135,136], except for one patient who died 16 days after stenting due to refractory shock without ACS recurrence [134].

2.6. IrAEs Reported in Clinical Trials

Between 2017 and January 2022, there were published trials that reported cardiac irAEs following the administration of ICIs, the most common AE being myocarditis. Maio et al. [139] conducted a double-blind placebo-controlled trial, evaluating the use of tremelimumab as second- or third-line treatment in patients with relapsed malignant melanoma, reporting a multitude of cardiac AE. In the treatment group, one case of myocardial infarction (vs. none in the placebo group) was identified, along with one case of cardiac failure (vs. one in the placebo group) and twelve cases of pericardial effusion (vs. six in the placebo group). A dose-finding and dose-expansion phase 1b trial conducted by Choueiri et al. [140], regarding the use of avelumab, a PD-L1 inhibitor, in association with axitinib, a tyrosine kinase inhibitor (TKi), in patients with clear-cell renal-cell carcinoma reported one case of myocarditis diagnosed before the first evaluation of the patients, resulting in the death of the patient. Among the six deaths reported in this study, this was the only one caused by irAEs, with the other five being a result of disease progression. Juergens et al. [141] assessed the efficacy of durvalumab with or without tremelimumab in combination with platinum-doublet based chemotherapy in several types of malignancies, the most common being NSCLC. There was one reported case of myocarditis, which was confirmed using post-mortem. The ESCORT trial assessed the efficacy of camrelizumab vs. the investigator’s choice chemotherapy regime in advanced or metastatic esophageal squamous cell carcinoma. A total of 10 treatment-related deaths were reported, 7 of them for camrelizumab (one with myocarditis) [142].
Hasson et al. [143] assessed the safety of reintroducing immunotherapy in patients that survived ICI-induced myocarditis. Three patients were chosen from a total of seven and ICIs were reintroduced under close surveillance and in combination with low dose PO prednisone and cardio-protective therapy (angiotensin-converting-enzyme inhibitors, beta-blockers, or mineralocorticoid receptor antagonists). One of the three patients exhibited heart failure symptoms which required the cessation of the ICI regiment, while the other two continued treatment with durvalumab and pembrolizumab without irAEs at the time of data cut-off.

Limitations of the Study

For the moment, a lot of information is emerging from daily clinical practice but must be treated with caution. Considering the lack of full data about patients’ profiles (regarding their cardiovascular and non-cardiovascular history and associated risk factors), further research is needed to fully assess the relationship between ICI and cardiotoxicity.

3. Material and Methods

We reviewed all publications from PubMed, Embase, and Medline databases using the terms: (“immunotherapy”[tw] OR “CTLA4”[tw] OR “PD1”[tw] OR “PDL1”[tw] OR “PD-1”[tw] OR “PD-L1”[tw] OR “Immune checkpoint inhibitors”[tw]) AND (“cardiotoxicity”[tw] OR “myocarditis”[tw] OR “Myocardial Infarction”[tw] OR “Heart Failure”[tw] OR “Acute Coronary Syndrome”[tw] OR “Arrhythmias, Cardiac”[Mesh] OR “Takotsubo Cardiomyopathy”[tw] OR “Pericarditis”[tw] OR “Coronary Vasospasm”[tw] OR “Shock, Cardiogenic”[tw] OR “Heart Conduction System”[tw]) NOT (“review”[tiab] OR “meta-analysis”[tiab]). Only human studies from the past five years were selected for screening, including case reports/case series. Duplicates were removed and only articles written in English with the presence of an abstract were reviewed. Reviews, systematic reviews, animal, and in vitro studies were excluded (Figure 2).

4. Conclusions

ICI-induced cardiac toxicity comprises a large field of morphological and functional abnormalities, including myocarditis, pericarditis, Takotsubo cardiomyopathy, and acute coronary syndrome. Despite only a few reports from the first clinical trials involving ICI, many cases were documented in later years. For this review, we identified and collected relevant data from the last five years in the literature in order to highlight the clinical and paraclinical patterns, as well as any therapeutic approaches. Although rare, these adverse events can lead to fatal outcomes, especially in the case of myocarditis. The precise mechanisms are not yet fully elucidated, and consequently, the treatment lacks standardization. Further research is needed for the better stratification of these pathological entities and the proper design for early diagnosis and treatment strategies.

Supplementary Materials

The supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms231810948/s1.

Author Contributions

Conceptualization, A.C. and O.H.O.; methodology, A.C.; validation, A.C., A.B. and O.H.O.; formal analysis, A.C.; investigation, A.L.L. and A.B.; resources, A.B.; data curation, A.B., A.L.L., T.V.M., A.M.G., A.F., B.M.B., A.V.S.-T., V.C.V., V.N. and N.D.S.; writing—A.B., A.L.L., T.V.M., A.M.G. and N.D.S.; writing—review and editing, A.C., O.H.O. and A.B.; visualization A.B.; supervision, O.H.O.; project administration A.C. All authors have read and agreed to the published version of the manuscript.

Funding

The APC was funded by authors and “Iuliu Hatieganu” University of Medicine and Pharmacy Cluj-Napoca.

Institutional Review Board Statement

Ethical review and approval were waived for this study because we performed an analysis of the existing published literature.

Informed Consent Statement

Not applicable.

Acknowledgments

This work was granted by project PDI-PFE-CDI 2021, entitled Increasing the Performance of Scientific Research, Supporting Excellence in Medical Research and Innovation, PROGRES, no. 40PFE/30.12.2021.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Postow, M.A.; Callahan, M.K.; Wolchok, J.D. Immune Checkpoint Blockade in Cancer Therapy. J. Clin. Oncol. 2015, 33, 1974–1982. [Google Scholar] [CrossRef] [PubMed]
  2. Nardi Agmon, I.; Itzhaki Ben Zadok, O.; Kornowski, R. The Potential Cardiotoxicity of Immune Checkpoint Inhibitors. J. Clin. Med. 2022, 11, 865. [Google Scholar] [CrossRef]
  3. Puzanov, I.; Diab, A.; Abdallah, K.; Bingham, C.O.; Brogdon, C.; Dadu, R.; Hamad, L.; Kim, S.; Lacouture, M.E.; LeBoeuf, N.R.; et al. Managing toxicities associated with immune checkpoint inhibitors: Consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J. Immunother. Cancer 2017, 5, 95. [Google Scholar] [CrossRef] [PubMed]
  4. Larkin, J.; Hodi, F.; Wolchok, J. Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma. N. Engl. J. Med. 2015, 373, 1270–1271. [Google Scholar] [CrossRef] [PubMed]
  5. Grabie, N.; Lichtman, A.H.; Padera, R. T cell checkpoint regulators in the heart. Cardiovasc. Res. 2019, 115, 869–877. [Google Scholar] [CrossRef] [PubMed]
  6. Arangalage, D.; Degrauwe, N.; Michielin, O.; Monney, P.; Özdemir, B.C. Pathophysiology, diagnosis and management of cardiac toxicity induced by immune checkpoint inhibitors and BRAF and MEK inhibitors. Cancer Treat. Rev. 2021, 100, 102282. [Google Scholar] [CrossRef]
  7. Okazaki, T.; Tanaka, Y.; Nishio, R.; Mitsuiye, T.; Mizoguchi, A.; Wang, J.; Ishida, M.; Hiai, H.; Matsumori, A.; Minato, N.; et al. Autoantibodies against cardiac troponin I are responsible for dilated cardiomyopathy in PD-1-deficient mice. Nat. Med. 2003, 9, 1477–1483. [Google Scholar] [CrossRef]
  8. Wang, J.; Okazaki, I.; Yoshida, T.; Chikuma, S.; Kato, Y.; Nakaki, F.; Hiai, H.; Honjo, T.; Okazaki, T. PD-1 deficiency results in the development of fatal myocarditis in MRL mice. Int. Immunol. 2010, 22, 443–452. [Google Scholar] [CrossRef]
  9. Tarrio, M.L.; Grabie, N.; Bu, D.; Sharpe, A.H.; Lichtman, A.H. PD-1 Protects against Inflammation and Myocyte Damage in T Cell-Mediated Myocarditis. J. Immunol. 2012, 188, 4876–4884. [Google Scholar] [CrossRef]
  10. Johnson, D.B.; Balko, J.M.; Compton, M.L.; Chalkias, S.; Gorham, J.; Xu, Y.; Hicks, M.; Puzanov, I.; Alexander, M.R.; Bloom-er, T.L.; et al. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N. Engl. J. Med. 2016, 375, 1749–1755. [Google Scholar] [CrossRef]
  11. Balanescu, D.V.; Donisan, T.; Palaskas, N.; Lopez-Mattei, J.; Kim, P.Y.; Buja, L.M.; McNamara, D.M.; Kobashigawa, J.A.; Durand, J.-B.; Iliescu, C.A. Immunomodulatory treatment of immune checkpoint inhibitor-induced myocarditis: Pathway toward precision-based therapy. Cardiovasc. Pathol. 2020, 47, 107211. [Google Scholar] [CrossRef] [PubMed]
  12. Tajmir-Riahi, A.; Bergmann, T.; Schmid, M.; Agaimy, A.; Schuler, G.; Heinzerling, L. Life-threatening Autoimmune Cardio-myopathy Reproducibly Induced in a Patient by Checkpoint Inhibitor Therapy. J. Immunother. 2018, 41, 35–38. [Google Scholar] [CrossRef] [PubMed]
  13. Moslehi, J.J.; Salem, J.-E.; Sosman, J.A.; Lebrun-Vignes, B.; Johnson, D.B. Increased reporting of fatal immune checkpoint in-hibitor-associated myocarditis. Lancet 2018, 391, 933. [Google Scholar] [CrossRef]
  14. Bu, D.; Tarrio, M.; Maganto-Garcia, E.; Stavrakis, G.; Tajima, G.; Lederer, J.; Jarolim, P.; Freeman, G.J.; Sharpe, A.H.; Licht-man, A.H. Impairment of the Programmed Cell Death-1 Pathway Increases Atherosclerotic Lesion Development and In-flammation. Arterioscler. Thromb. Vasc. Biol. 2011, 31, 1100–1107. [Google Scholar] [CrossRef] [PubMed]
  15. Gelsomino, F.; Fiorentino, M.; Zompatori, M.; Poerio, A.; Melotti, B.; Sperandi, F.; Gargiulo, M.; Borghi, C.; Ardizzoni, A. Pro-grammed death-1 inhibition and atherosclerosis: Can nivolumab vanish complicated atheromatous plaques? Ann. Oncol. 2018, 29, 284–286. [Google Scholar] [CrossRef]
  16. Chocarro, L.; Blanco, E.; Zuazo, M.; Arasanz, H.; Bocanegra, A.; Fernández-Rubio, L.; Morente, P.; Fernández-Hinojal, G.; Echaide, M.; Garnica, M.; et al. Understanding LAG-3 Signaling. Int. J. Mol. Sci. 2021, 22, 5282. [Google Scholar] [CrossRef]
  17. Xiong, X.; Luo, Z.; Zhou, H.; Duan, Z.; Niu, L.; Zhang, K.; Huang, G.; Li, W. Downregulation of TIGIT Expression in FOXP3+Regulatory T Cells in Acute Coronary Syndrome. J. Inflamm. Res. 2022, 15, 1195–1207. [Google Scholar] [CrossRef]
  18. Zhang, J.; Zhan, F.; Liu, H. Expression Level and Significance of Tim-3 in CD4+ T Lymphocytes in Peripheral Blood of Pa-tients with Coronary Heart Disease. Braz. J. Cardiovasc. Surg. 2022, 37, 350–355. [Google Scholar] [CrossRef]
  19. Nguyen, L.S.; Bretagne, M.; Arrondeau, J.; Zahr, N.; Ederhy, S.; Abbar, B.; Pinna, B.; Allenbach, Y.; Mira, J.P.; Moslehi, J.; et al. Reversal of immune-checkpoint inhibitor fulminant myocarditis using personalized-dose-adjusted abatacept and rux-olitinib: Proof of concept. J. Immunother. Cancer 2022, 10, e004699. [Google Scholar] [CrossRef]
  20. Diamantopoulos, P.T.; Tsatsou, K.; Benopoulou, O.; Bonou, M.; Anastasopoulou, A.; Mastrogianni, E.; Gogas, H. Concomi-tant development of neurologic and cardiac immune-related adverse effects in patients treated with immune checkpoint in-hibitors for melanoma. Melanoma Res. 2020, 30, 484–491. [Google Scholar] [CrossRef]
  21. Fazal, M.; Prentice, D.A.; Kho, L.K.; Fysh, E. Nivolumab-associated myositis myocarditis and myasthenia and anti-striated muscle antibodies. Intern. Med. J. 2020, 50, 1003–1006. [Google Scholar] [CrossRef] [PubMed]
  22. Samara, Y.; Yu, C.L.; Dasanu, C.A. Acute autoimmune myocarditis and hepatitis due to ipilimumab monotherapy for malig-nant melanoma. J. Oncol. Pharm. Pract. 2019, 25, 966–968. [Google Scholar] [CrossRef] [PubMed]
  23. Liu, S.; Chan, J.; Brinc, D.; Gandhi, S.; Izenberg, A.; Delgado, D.; Abdel-Qadir, H.; Wintersperger, B.J.; Thavendiranathan, P. Immune Checkpoint Inhibitor-Associated Myocarditis With Persistent Troponin Elevation Despite Abatacept and Prolonged Immunosuppression. JACC Cardio Oncol. 2020, 2, 800–804. [Google Scholar] [CrossRef] [PubMed]
  24. Barham, W.; Guo, R.; Park, S.S.; Herrmann, J.; Dong, H.; Yan, Y. Case Report: Simultaneous Hyperprogression and Fulmi-nant Myocarditis in a Patient with Advanced Melanoma Following Treatment With Immune Checkpoint Inhibitor Therapy. Front. Immunol. 2021, 11, 561083. [Google Scholar] [CrossRef] [PubMed]
  25. Wintersperger, B.J.; Calvillo-Argüelles, O.; Lheureux, S.; Houbois, C.P.; Spreafico, A.; Bedard, P.L.; Neilan, T.G.; Thavendiranathan, P. Immune checkpoint inhibitor-related myocarditis: An illustrative case series of applying the updated Cardiovascular Magnetic Resonance Lake Louise Criteria. Eur. Heart J.-Case Rep. 2022, 6, ytab478. [Google Scholar] [CrossRef]
  26. Saishu, Y.; Yoshida, T.; Seino, Y.; Nomura, T. Nivolumab-related myasthenia gravis with myositis requiring prolonged me-chanical ventilation: A case report. J. Med. Case Rep. 2022, 16, 61. [Google Scholar] [CrossRef]
  27. Saibil, S.D.; Bonilla, L.; Majeed, H.; Sotov, V.; Hogg, D.; Chappell, M.A.; Cybulsky, M.; Butler, M.O. Fatal Myocarditis and Rhabdomyositis in a Patient with Stage Iv Melanoma Treated with Combined Ipilimumab and Nivolumab. Curr. Oncol. 2019, 26, 418–421. [Google Scholar] [CrossRef]
  28. Norwood, T.G.; Westbrook, B.C.; Johnson, D.B.; Litovsky, S.H.; Terry, N.L.; McKee, S.B.; Gertler, A.S.; Moslehi, J.J.; Conry, R.M. Smoldering myocarditis following immune checkpoint blockade. J. Immunother. Cancer 2017, 5, 91. [Google Scholar] [CrossRef]
  29. Giblin, G.T.; Dennehy, C.; Featherstone, H.; Clarke, R.; Murphy, L.; Timlin, D.; O’Keane, C.; Mulligan, N.; Kelly, C.M.; Joyce, E. Subclinical Myocarditis After Combination Immune Checkpoint Inhibitor Therapy. Circ. Heart Fail. 2021, 14, e007524. [Google Scholar] [CrossRef] [PubMed]
  30. Arangalage, D.; Delyon, J.; Lermuzeaux, M.; Ekpe, K.; Ederhy, S.; Pages, C.; Lebbé, C. Survival After Fulminant Myocarditis Induced by Immune-Checkpoint Inhibitors. Ann. Intern. Med. 2017, 167, 683. [Google Scholar] [CrossRef]
  31. Fazel, M.; Jedlowski, P.M. Severe Myositis, Myocarditis, and Myasthenia Gravis with Elevated Anti-Striated Muscle Antibody following Single Dose of Ipilimumab-Nivolumab Therapy in a Patient with Metastatic Melanoma. Case Rep. Immunol. 2019, 2019, 1–3. [Google Scholar] [CrossRef] [PubMed]
  32. Ida, M.; Nakamori, S.; Yamamoto, S.; Watanabe, S.; Imanaka-Yoshida, K.; Ishida, M.; Sakuma, H.; Yamanaka, K.; Dohi, K. Subtle-but-smouldering myocardial injury after immune checkpoint inhibitor treatment accompanied by amyloid deposits. ESC Heart Fail. 2022, 9, 2027–2031. [Google Scholar] [CrossRef] [PubMed]
  33. Arora, P.; Talamo, L.; Dillon, P.; Gentzler, R.D.; Millard, T.; Salerno, M.; Slingluff, C.L.; Gaughan, E.M. Severe combined car-diac and neuromuscular toxicity from immune checkpoint blockade: An institutional case series. Cardio Oncol. 2020, 6, 21. [Google Scholar] [CrossRef] [PubMed]
  34. So, H.; Ikeguchi, R.; Kobayashi, M.; Suzuki, M.; Shimizu, Y.; Kitagawa, K. PD-1 inhibitor-associated severe myasthenia gravis with necrotizing myopathy and myocarditis. J. Neurol. Sci. 2019, 399, 97–100. [Google Scholar] [CrossRef]
  35. Esfahani, K.; Buhlaiga, N.; Thébault, P.; Lapointe, R.; Johnson, N.A.; Miller, W.H. Alemtuzumab for Immune-Related Myo-carditis Due to PD-1 Therapy. N. Engl. J. Med. 2019, 380, 2375–2376. [Google Scholar] [CrossRef]
  36. Yamaguchi, S.; Morimoto, R.; Okumura, T.; Yamashita, Y.; Haga, T.; Kuwayama, T.; Yokoi, T.; Hiraiwa, H.; Kondo, T.; Sugiura, Y.; et al. Late-Onset Fulminant Myocarditis with Immune Checkpoint Inhibitor Nivolumab. Can. J. Cardiol. 2018, 34, 812.e1–812.e3. [Google Scholar] [CrossRef]
  37. Ansari-Gilani, K.; Tirumani, S.H.; Smith, D.A.; Nelson, A.; Alahmadi, A.; Hoimes, C.J.; Ramaiya, N.H. Myocarditis associat-ed with immune checkpoint inhibitor therapy: A case report of three patients. Emerg. Radiol. 2020, 27, 455–460. [Google Scholar] [CrossRef]
  38. Shalata, W.; Peled, N.; Gabizon, I.; Abu Saleh, O.; Kian, W.; Yakobson, A. Associated Myocarditis: A Predictive Factor for Response? Case Rep. Oncol. 2020, 13, 550–557. [Google Scholar] [CrossRef]
  39. Leaver, P.J.; Jang, H.S.-I.; Vernon, S.T.; Fernando, S.L. Immune checkpoint inhibitor-mediated myasthenia gravis with focal subclinical myocarditis progressing to symptomatic cardiac disease. BMJ Case Rep. 2020, 13, e232920. [Google Scholar] [CrossRef]
  40. Hardy, T.; Yin, M.; Chavez, J.A.; Ivanov, I.; Chen, W.; Nadasdy, T.; Brodsky, S.V. Acute fatal myocarditis after a single dose of anti-PD-1 immunotherapy, autopsy findings: A case report. Cardiovasc. Pathol. 2020, 46, 107202. [Google Scholar] [CrossRef]
  41. Sato, T.; Nakamori, S.; Watanabe, S.; Nishikawa, K.; Inoue, T.; Imanaka-Yoshida, K.; Ishida, M.; Sakuma, H.; Ito, M.; Dohi, K. Monitoring of the Evolution of Immune Checkpoint Inhibitor Myocarditis With Cardiovascular Magnetic Resonance. Circ. Cardiovasc. Imaging 2020, 13, e010633. [Google Scholar] [CrossRef] [PubMed]
  42. Mirabel, M.; Callon, D.; Bruneval, P.; Lebreil, A.-L.; Mousseaux, E.; Oudard, S.; Hulot, J.-S.; Andreoletti, L. Late-Onset Giant Cell Myocarditis Due to Enterovirus During Treatment With Immune Checkpoint Inhibitors. JACC Cardio Oncol. 2020, 2, 511–514. [Google Scholar] [CrossRef] [PubMed]
  43. 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. 2021, 34, 101508. [Google Scholar] [CrossRef] [PubMed]
  44. Thibault, C.; Vano, Y.; Soulat, G.; Mirabel, M. Immune checkpoint inhibitors myocarditis: Not all cases are clinically patent. Eur. Heart J. 2018, 39, 3553. [Google Scholar] [CrossRef]
  45. Rota, E.; Varese, P.; Agosti, S.; Celli, L.; Ghiglione, E.; Pappalardo, I.; Zaccone, G.; Paglia, A.; Morelli, N. Concomitant myas-thenia gravis, myositis, myocarditis and polyneuropathy, induced by immune-checkpoint inhibitors: A life–threatening con-tinuum of neuromuscular and cardiac toxicity. eNeurologicalSci 2019, 14, 4–5. [Google Scholar] [CrossRef]
  46. Miyauchi, Y.; Naito, H.; Tsunemori, H.; Tani, R.; Hasui, Y.; Miyake, Y.; Minamino, T.; Ishikawa, R.; Kushida, Y.; Haba, R.; et al. Myocarditis as an immune-related adverse event following treatment with ipilimumab and nivolumab combination thera-py for metastatic renal cell carcinoma: A case report. J. Med. Case Rep. 2021, 15, 508. [Google Scholar] [CrossRef]
  47. Jespersen, M.S.; Fanø, S.; Stenør, C.; Møller, A.K. A case report of immune checkpoint inhibitor-related steroid-refractory my-ocarditis and myasthenia gravis-like myositis treated with abatacept and mycophenolate mofetil. Eur. Heart J.-Case Rep. 2021, 5, ytab342. [Google Scholar] [CrossRef]
  48. Lorente-Ros, Á.; Rajjoub-Al-Mahdi, E.A.; Monteagudo Ruiz, J.M.; Rivas García, S.; Ortega Pérez, R.; Fernández Golfín, C.; Álvarez-García, J.; Zamorano Gómez, J.L. Checkpoint Immunotherapy-Induced Myocarditis and Encephalitis Complicated With Complete AV Block: Not All Hope Is Lost. JACC Case Rep. 2022, 4, 1032–1036. [Google Scholar] [CrossRef]
  49. Tanabe, J.; Watanabe, N.; Endo, A.; Nagami, T.; Inagaki, S.; Tanabe, K. Asymptomatic Immune Checkpoint Inhibitor-associated Myocarditis. Intern. Med. 2021, 60, 569–573. [Google Scholar] [CrossRef]
  50. Xie, X.; Wang, F.; Qin, Y.; Lin, X.; Xie, Z.; Liu, M.; Ouyang, M.; Luo, B.; Gu, Y.; Li, S.; et al. Case Report: Fatal Multiorgan Failure and Heterochronous Pneumonitis Following Pembrolizumab Treatment in a Patient with Large-Cell Neuroendocrine Carcinoma of Lung. Front. Pharmacol. 2021, 11, 569466. [Google Scholar] [CrossRef]
  51. Tomoaia, R.; Beyer, R.Ș.; Pop, D.; Minciună, I.A.; Dădârlat-Pop, A. Fatal association of fulminant myocarditis and rhabdo-myolysis after immune checkpoint blockade. Eur. J. Cancer 2020, 132, 224–227. [Google Scholar] [CrossRef] [PubMed]
  52. Sakai, T.; Yahagi, K.; Hoshino, T.; Yokota, T.; Tanabe, K.; Mori, M.; Ikeda, S. Nivolumab-induced myocardial necrosis in a patient with lung cancer: A case report. Respir. Med. Case Rep. 2019, 27, 100839. [Google Scholar] [CrossRef]
  53. von Itzstein, M.S.; Khan, S.; Popat, V.; Lu, R.; Khan, S.A.; Fattah, F.J.; Park, J.Y.; Bermas, B.L.; Karp, D.R.; Ahmed, M.; et al. Statin Intolerance, Anti-HMGCR Antibodies, and Immune Checkpoint Inhibitor-Associated Myositis: A “Two-Hit” Autoim-mune Toxicity or Clinical Predisposition? Oncologist 2020, 25, e1242–e1245. [Google Scholar] [CrossRef] [PubMed]
  54. Salido Iniesta, M.; López López, L.; Carreras Costa, F.; Sionis, A. A different type of acute myocarditis: A case report of acute autoimmune myocarditis mediated by anti-PD-1 T lymphocyte receptor (pembrolizumab). Eur. Heart J.-Case Rep. 2020, 4, 1–6. [Google Scholar] [CrossRef] [PubMed]
  55. Tsuruda, T.; Yoshikawa, N.; Kai, M.; Yamaguchi, M.; Toida, R.; Kodama, T.; Kajihara, K.; Kawabata, T.; Nakamura, T.; Sa-kata, K.; et al. The Cytokine Expression in Patients with Cardiac Complication after Immune Checkpoint Inhibitor Therapy. Intern. Med. 2021, 60, 423–429. [Google Scholar] [CrossRef]
  56. Doms, J.; Prior, J.O.; Peters, S.; Obeid, M. Tocilizumab for refractory severe immune checkpoint inhibitor-associated myocar-ditis. Ann. Oncol. 2020, 31, 1273–1275. [Google Scholar] [CrossRef] [PubMed]
  57. Li, Y.; Hu, Y.; Yang, B.; Jin, C.; Ren, H.; Wu, J.; Wang, Z.; Wei, Y.; Yang, L.; Hu, Y. Immunotherapy-Related Cardiotoxicity Re-Emergence in Non-Small Cell Lung Cancer—A Case Report. Onco. Targets. Ther. 2021, 14, 5309–5314. [Google Scholar] [CrossRef]
  58. Fukasawa, Y.; Sasaki, K.; Natsume, M.; Nakashima, M.; Ota, S.; Watanabe, K.; Takahashi, Y.; Kondo, F.; Kozuma, K.; Seki, N. Nivolumab-Induced Myocarditis Concomitant with Myasthenia Gravis. Case Rep. Oncol. 2017, 10, 809–812. [Google Scholar] [CrossRef]
  59. Matson, D.R.; Accola, M.A.; Rehrauer, W.M.; Corliss, R.F. Fatal Myocarditis Following Treatment with the PD-1 Inhibitor Nivolumab. J. Forensic. Sci. 2018, 63, 954–957. [Google Scholar] [CrossRef]
  60. Okauchi, S.; Sasatani, Y.; Yamada, H.; Satoh, H. Late-onset pulmonary and cardiac toxicities in a patient treted with immune checkpoint inhibitor monotherapy. Klin. Onkol. 2022, 35, 150–154. [Google Scholar] [CrossRef]
  61. Ederhy, S.; Fenioux, C.; Cholet, C.; Rouvier, P.; Redheuil, A.; Cohen, A.; Salem, J.-E. Immune Checkpoint Inhibitor Myocardi-tis with Normal Cardiac Magnetic Resonance Imaging: Importance of Cardiac Biopsy and Early Diagnosis. Can. J. Cardiol. 2021, 37, 1654–1656. [Google Scholar] [CrossRef] [PubMed]
  62. Zhou, B.; Li, M.; Chen, T.; She, J. Case Report: Acute Myocarditis Due to PD-L1 Inhibitor Durvalumab Monotherapy in a Patient with Lung Squamous Cell Carcinoma. Front. Med. 2022, 9, 7–10. [Google Scholar] [CrossRef] [PubMed]
  63. Xing, Q.; Zhang, Z.-W.; Lin, Q.-H.; Shen, L.-H.; Wang, P.-M.; Zhang, S.; Fan, M.; Zhu, B. Myositis-myasthenia gravis overlap syndrome complicated with myasthenia crisis and myocarditis associated with anti-programmed cell death-1 (sintilimab) therapy for lung adenocarcinoma. Ann. Transl. Med. 2020, 8, 250. [Google Scholar] [CrossRef] [PubMed]
  64. Chatzantonis, G.; Evers, G.; Meier, C.; Bietenbeck, M.; Florian, A.; Klingel, K.; Bleckmann, A.; Yilmaz, A. Immune Check-point Inhibitor-Associated Myocarditis. JACC Case Rep. 2020, 2, 630–635. [Google Scholar] [CrossRef]
  65. Fuentes-Antrás, J.; Peinado, P.; Guevara-Hoyer, K.; del Arco, C.D.; Sánchez-Ramón, S.; Aguado, C. Fatal autoimmune storm after a single cycle of anti-PD-1 therapy: A case of lethal toxicity but pathological complete response in metastatic lung ade-nocarcinoma. Hematol. Oncol. Stem Cell Ther. 2020, in press. [Google Scholar] [CrossRef]
  66. Frigeri, M.; Meyer, P.; Banfi, C.; Giraud, R.; Hachulla, A.-L.; Spoerl, D.; Friedlaender, A.; Pugliesi-Rinaldi, A.; Dietrich, P.-Y. Immune Checkpoint Inhibitor-Associated Myocarditis: A New Challenge for Cardiologists. Can. J. Cardiol. 2018, 34, 92.e1–92.e3. [Google Scholar] [CrossRef]
  67. Nierstedt, R.T.; Yeahia, R.; Barnett, K.M. Unanticipated Myocarditis in a Surgical Patient Treated with Pembrolizumab: A Case Report. AA Pract. 2020, 14, e01177. [Google Scholar] [CrossRef]
  68. Valenti-Azcarate, R.; Esparragosa Vazquez, I.; Toledano Illan, C.; Idoate Gastearena, M.A.; Gállego Pérez-Larraya, J. Nivolumab and Ipilimumab-induced myositis and myocarditis mimicking a myasthenia gravis presentation. Neuromuscul. Disord. 2020, 30, 67–69. [Google Scholar] [CrossRef]
  69. Tu, L.; Liu, J.; Li, Z.; Liu, Y.; Luo, F. Early detection and management of immune-related myocarditis: Experience from a case with advanced squamous cell lung carcinoma. Eur. J. Cancer 2020, 131, 5–8. [Google Scholar] [CrossRef]
  70. Möhn, N.; Beutel, G.; Gutzmer, R.; Ivanyi, P.; Satzger, I.; Skripu, T. Neurological Immune Related Adverse Events Associated with Nivolumab, Ipilimumab, and Pembrolizumab Therapy—Review of the Literature and Future Outlook. J. Clin. Med. 2019, 8, 1777. [Google Scholar] [CrossRef] [Green Version]
  71. Tajiri, K.; Aonuma, K.; Sekine, I. Immune checkpoint inhibitor-related myocarditis. Jpn. J. Clin. Oncol. 2018, 48, 7–12. [Google Scholar] [CrossRef] [PubMed]
  72. Bomze, D.; Hasan Ali, O.; Bate, A.; Flatz, L. Association Between Immune-Related Adverse Events During Anti–PD-1 Ther-apy and Tumor Mutational Burden. JAMA Oncol. 2019, 5, 1633. [Google Scholar] [CrossRef] [PubMed]
  73. Suzuki, S. Autoimmune Targets of Heart and Skeletal Muscles in Myasthenia Gravis. Arch. Neurol. 2009, 66, 1334. [Google Scholar] [CrossRef] [PubMed]
  74. Romi, F.; Skeie, G.O.; Gilhus, N.E.; Aarli, J.A. Striational Antibodies in Myasthenia Gravis. Arch. Neurol. 2005, 62, 442. [Google Scholar] [CrossRef] [PubMed]
  75. Shivamurthy, P.; Parker, M.W. Cardiac manifestations of myasthenia gravis: A systematic review. IJC Metab. Endocr. 2014, 5, 3–6. [Google Scholar] [CrossRef]
  76. Hughes, M.; Lilleker, J.B.; Herrick, A.L.; Chinoy, H. Cardiac troponin testing in idiopathic inflammatory myopathies and systemic sclerosis-spectrum disorders: Biomarkers to distinguish between primary cardiac involvement and low-grade skeletal muscle disease activity. Ann. Rheum. Dis. 2015, 74, 795–798. [Google Scholar] [CrossRef]
  77. Shen, L.; Chen, H.; Wei, Q. Immune-Therapy-Related Toxicity Events and Dramatic Remission After a Single Dose of Pem-brolizumab Treatment in Metastatic Thymoma: A Case Report. Front. Immunol. 2021, 12, e621858. [Google Scholar] [CrossRef]
  78. Cao, J.; Li, Q.; Zhi, X.; Yang, F.; Zhu, W.; Zhou, T.; Hou, X.; Chen, D. Pembrolizumab-induced autoimmune Stevens-Johnson syndrome/toxic epidermal necrolysis with myositis and myocarditis in a patient with esophagogastric junction carcinoma: A case report. Transl. Cancer Res. 2021, 10, 3870–3876. [Google Scholar] [CrossRef]
  79. Ai, L.; Gao, J.; Zhao, S.; Li, Q.; Cui, Y.-H.; Liu, Q.; Wu, D.; Wang, Y.; Jin, X.; Ji, Y.; et al. Nivolumab-associated DRESS in a genetic susceptible individual. J. Immunother. Cancer 2021, 9, e002879. [Google Scholar] [CrossRef]
  80. Luo, Y.-B.; Tang, W.; Zeng, Q.; Duan, W.; Li, S.; Yang, X.; Bi, F. Case Report: The Neuromusclar Triad of Immune Checkpoint Inhibitors: A Case Report of Myositis, Myocarditis, and Myasthenia Gravis Overlap Following Toripalimab Treatment. Front. Cardiovasc. Med. 2021, 8, 714460. [Google Scholar] [CrossRef]
  81. Zhao, L.-Z.; Liu, G.; Li, Q.-F.; Chen, G.; Jin, G.-W. A case of carrelizumab-associated immune myocarditis. Asian J. Surg. 2022, 45, 496–497. [Google Scholar] [CrossRef] [PubMed]
  82. Monge, C.; Maeng, H.; Brofferio, A.; Apolo, A.B.; Sathya, B.; Arai, A.E.; Gulley, J.L.; Bilusic, M. Myocarditis in a patient treat-ed with Nivolumab and PROSTVAC: A case report. J. Immunother. Cancer 2018, 6, 150. [Google Scholar] [CrossRef]
  83. Iwasaki, S.; Hidaka, H.; Uojima, H.; Hashimura, M.; Nabeta, T.; Sanoyama, I.; Wada, N.; Kubota, K.; Nakazawa, T.; Shibuya, A.; et al. A case of immune checkpoint inhibitor-associated myocarditis after initiation of atezolizumab plus bevaci-zumab therapy for advanced hepatocellular carcinoma. Clin. J. Gastroenterol. 2021, 14, 1233–1239. [Google Scholar] [CrossRef]
  84. Stein-Merlob, A.F.; Hsu, J.J.; Colton, B.; Berg, C.J.; Ferreira, A.; Price, M.M.; Wainberg, Z.; Baas, A.S.; Deng, M.C.; Parikh, R.V.; et al. Keeping immune checkpoint inhibitor myocarditis in check: Advanced circulatory mechanical support as a bridge to recovery. ESC Heart Fail. 2021, 8, 4301–4306. [Google Scholar] [CrossRef] [PubMed]
  85. Szuchan, C.; Elson, L.; Alley, E.; Leung, K.; Camargo, A.L.; Elimimian, E.; Nahleh, Z.; Sadler, D. Checkpoint inhibitor-induced myocarditis and myasthenia gravis in a recurrent/metastatic thymic carcinoma patient: A case report. Eur. Heart J.-Case Rep. 2020, 4, 1–8. [Google Scholar] [CrossRef] [PubMed]
  86. Jeyakumar, N.; Etchegaray, M.; Henry, J.; Lelenwa, L.; Zhao, B.; Segura, A.; Buja, L.M. The Terrible Triad of Checkpoint In-hibition: A Case Report of Myasthenia Gravis, Myocarditis, and Myositis Induced by Cemiplimab in a Patient with Metastatic Cutaneous Squamous Cell Carcinoma. Case Rep. Immunol. 2020, 2020, 1–4. [Google Scholar] [CrossRef] [PubMed]
  87. Chen, Q.; Huang, D.-S.; Zhang, L.-W.; Li, Y.-Q.; Wang, H.-W.; Liu, H. Fatal myocarditis and rhabdomyolysis induced by nivolumab during the treatment of type B3 thymoma. Clin. Toxicol. 2018, 56, 667–671. [Google Scholar] [CrossRef]
  88. Wang, Q.; Hu, B. Successful therapy for autoimmune myocarditis with pembrolizumab treatment for nasopharyngeal carci-noma. Ann. Transl. Med. 2019, 7, 247. [Google Scholar] [CrossRef]
  89. Portolés Hernández, A.; Blanco Clemente, M.; Escribano García, D.; Velasco Calvo, R.; Núñez García, B.; Oteo Domínguez, J.F.; Salas Antón, C.; Méndez García, M.; Segovia Cubero, J.; Domínguez, F. Checkpoint inhibitor-induced fulminant myo-carditis, complete atrioventricular block and myasthenia gravis—A case report. Cardiovasc. Diagn. Ther. 2021, 11, 1013–1019. [Google Scholar] [CrossRef]
  90. Thiene, G.; Bruneval, P.; Veinot, J.; Leone, O. Diagnostic use of the endomyocardial biopsy: A consensus statement. Virchows Arch. 2013, 463, 1–5. [Google Scholar] [CrossRef]
  91. Love, V.A.; 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, 248–257. [Google Scholar] [CrossRef] [PubMed]
  92. Xia, W.; Zou, C.; Chen, H.; Xie, C.; Hou, M. Immune checkpoint inhibitor induces cardiac injury through polarizing macro-phages via modulating microRNA-34a/Kruppel-like factor 4 signaling. Cell Death Dis. 2020, 11, 575. [Google Scholar] [CrossRef] [PubMed]
  93. Hua, C.-C.; Liu, X.-M.; Liang, L.-R.; Wang, L.-F.; Zhong, J.-C. Targeting the microRNA-34a as a Novel Therapeutic Strategy for Cardiovascular Diseases. Front. Cardiovasc. Med. 2022, 8, 784044. [Google Scholar] [CrossRef]
  94. Liu, Y.; Jiang, L. Tofacitinib for treatment in immune-mediated myocarditis: The first reported cases. J. Oncol. Pharm. Pract. 2021, 27, 739–746. [Google Scholar] [CrossRef] [PubMed]
  95. Safa, H.; Johnson, D.H.; Trinh, V.A.; Rodgers, T.E.; Lin, H.; Suarez-Almazor, M.E.; Fa’ak, F.; Saberian, C.; Yee, C.; Davies, M.A.; et al. Immune checkpoint inhibitor related myasthenia gravis: Single center experience and systematic review of the lit-erature. J. Immunother. Cancer 2019, 7, 319. [Google Scholar] [CrossRef] [PubMed]
  96. Chang, E.; Sabichi, A.L.; Sada, Y.H. Myasthenia Gravis After Nivolumab Therapy for Squamous Cell Carcinoma of the Blad-der. J. Immunother. 2017, 40, 114–116. [Google Scholar] [CrossRef]
  97. Brahmer, J.R.; Lacchetti, C.; Schneider, B.J.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; Ernstoff, M.S.; Gardner, J.M.; Ginex, P.; et al. 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, 1714–1768. [Google Scholar] [CrossRef]
  98. Dhawan, P.S.; Goodman, B.P.; Harper, C.M.; Bosch, P.E.; Hoffman-Snyder, C.R.; Wellik, K.E.; Wingerchuk, D.M.; Demaerschalk, B.M. IVIG Versus PLEX in the Treatment of Worsening Myasthenia Gravis. Neurologist 2015, 19, 145–148. [Google Scholar] [CrossRef]
  99. Martinez-Calle, N.; Rodriguez-Otero, P.; Villar, S.; Mejías, L.; Melero, I.; Prosper, F.; Marinello, P.; Paiva, B.; Idoate, M.; San-Miguel, J. Anti-PD1 associated fulminant myocarditis after a single pembrolizumab dose: The role of occult pre-existing auto-immunity. Haematologica 2018, 103, e318–e321. [Google Scholar] [CrossRef]
  100. Koul, D.; Kanwar, M.; Jefic, D.; Kolluru, A.; Singh, T.; Dhar, S.; Kumar, P.; Cohen, G. Fulminant Giant Cell Myocarditis and Cardiogenic Shock: An Unusual Presentation of Malignant Thymoma. Cardiol. Res. Pract. 2010, 2010, 1–4. [Google Scholar] [CrossRef] [Green Version]
  101. Furuta, C.; Yano, M.; Numanami, H.; Yamaji, M.; Taguchi, R.; Haniuda, M. A case of thymoma-associated multiorgan auto-immunity including polymyositis and myocarditis. Surg. Case Rep. 2021, 7, 226. [Google Scholar] [CrossRef] [PubMed]
  102. Mygland, Å.; Vincent, A.; Newsom-Davis, J.; Kaminski, H.; Zorzato, F.; Agius, M.; Gilhus, N.E.; Aarli, J.A. Autoantibodies in Thymoma-Associated Myasthenia Gravis With Myositis or Neuromyotonia. Arch. Neurol. 2000, 57, 527. [Google Scholar] [CrossRef] [PubMed]
  103. de Almeida, D.V.P.; Gomes, J.R.; Haddad, F.J.; Buzaid, A.C. Immune-mediated Pericarditis With Pericardial Tamponade During Nivolumab Therapy. J. Immunother. 2018, 41, 329–331. [Google Scholar] [CrossRef] [PubMed]
  104. Moriyama, S.; Fukata, M.; Tatsumoto, R.; Kono, M. Refractory constrictive pericarditis caused by an immune checkpoint in-hibitor properly managed with infliximab: A case report. Eur. Heart J.-Case Rep. 2021, 5, ytab002. [Google Scholar] [CrossRef] [PubMed]
  105. Oristrell, G.; Bañeras, J.; Ros, J.; Muñoz, E. Cardiac tamponade and adrenal insufficiency due to pembrolizumab: A case report. Eur. Heart J.-Case Rep. 2018, 2, yty038. [Google Scholar] [CrossRef]
  106. Dhenin, A.; Samartzi, V.; Lejeune, S.; Seront, E. Cascade of immunologic adverse events related to pembrolizumab treatment. BMJ Case Rep. 2019, 12, e229149. [Google Scholar] [CrossRef]
  107. Dasanu, C.A.; Jen, T.; Skulski, R. Late-onset pericardial tamponade, bilateral pleural effusions and recurrent immune mono-arthritis induced by ipilimumab use for metastatic melanoma. J. Oncol. Pharm. Pract. 2017, 23, 231–234. [Google Scholar] [CrossRef]
  108. Zarogoulidis, P.; Chinelis, P.; Athanasiadou, A.; Tsiouda, T.; Trakada, G.; Kallianos, A.; Veletza, L.; Hatzibougias, D.; Mi-halopoulou, E.; Goupou, E.; et al. Possible adverse effects of immunotherapy in non-small cell lung cancer; treatment and fol-low-up of three cases. Respir. Med. Case Rep. 2017, 22, 101–105. [Google Scholar] [CrossRef]
  109. Öztürk, C.; Luetkens, J.A. Cardiac MRI in Immune Checkpoint Inhibitor Associated Pericarditis. Clin. Case Rep. 2021, 9, e04926. [Google Scholar] [CrossRef]
  110. Khan, A.M.; Munir, A.; Thalody, V.; Munshi, M.K.; Mehdi, S. Cardiac tamponade in a patient with stage IV lung adenocar-cinoma treated with pembrolizumab. Immunotherapy 2019, 11, 1533–1540. [Google Scholar] [CrossRef]
  111. Jacobs, F.; Carnio, S.; De Stefanis, P.; Luciano, A.; Novello, S. Pericarditis during chemoimmunotherapy for non–small cell lung cancer: An adverse event to prevent and recognise. Eur. J. Cancer 2021, 149, 114–116. [Google Scholar] [CrossRef] [PubMed]
  112. Joseph, L.; Nickel, A.; Patel, A.; Saba, N.; Leon, A.; El-Chami, M.; Merchant, F. Incidence of Cancer Treatment Induced Ar-rhythmia Associated with Immune Checkpoint Inhibitors. J. Atr. Fibrillation 2021, 13, 2461. [Google Scholar] [PubMed]
  113. Reddy, N.; Moudgil, R.; Lopez-Mattei, J.C.; Karimzad, K.; Mouhayar, E.N.; Somaiah, N.; Conley, A.P.; Patel, S.; Giza, D.E.; Iliescu, C. Progressive and Reversible Conduction Disease with Checkpoint Inhibitors. Can. J. Cardiol. 2017, 33, 1335.e13–1335.e15. [Google Scholar] [CrossRef]
  114. Giancaterino, S.; Abushamat, F.; Duran, J.; Lupercio, F.; DeMaria, A.; Hsu, J.C. Complete heart block and subsequent sudden cardiac death from immune checkpoint inhibitor–associated myocarditis. Heart Case Rep. 2020, 6, 761–764. [Google Scholar] [CrossRef] [PubMed]
  115. 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, 155–158. [Google Scholar] [CrossRef] [PubMed]
  116. Pohl, J.; Mincu, R.-I.; Mrotzek, S.M.; Hinrichs, L.; Michel, L.; Livingstone, E.; Zimmer, L.; Wakili, R.; Schadendorf, D.; Rassaf, T.; et al. ECG Changes in Melanoma Patients Undergoing Cancer Therapy—Data from the ECoR Registry. J. Clin. Med. 2020, 9, 2060. [Google Scholar] [CrossRef] [PubMed]
  117. Khan, A.; Riaz, S.; Carhart, R. Pembrolizumab-Induced Mobitz Type 2 Second-Degree Atrioventricular Block. Case Rep. Cardiol. 2020, 2020, 1–5. [Google Scholar] [CrossRef]
  118. Hsu, C.-Y.; Su, Y.-W.; Chen, S.-C. Sick sinus syndrome associated with anti-programmed cell death-1. J. Immunother. Cancer 2018, 6, 72. [Google Scholar] [CrossRef]
  119. Katsume, Y.; Isawa, T.; Toi, Y.; Fukuda, R.; Kondo, Y.; Sugawara, S.; Ootomo, T. Complete Atrioventricular Block Associated with Pembrolizumab-induced Acute Myocarditis: The Need for Close Cardiac Monitoring. Intern. Med. 2018, 57, 3157–3162. [Google Scholar] [CrossRef]
  120. Serzan, M.; Rapisuwon, S.; Krishnan, J.; Chang, I.C.; Barac, A. Takotsubo Cardiomyopathy Associated with Checkpoint In-hibitor Therapy. JACC Cardio Oncol. 2021, 3, 330–334. [Google Scholar] [CrossRef]
  121. Oldfield, K.; Jayasinghe, R.; Niranjan, S.; Chadha, S. Immune checkpoint inhibitor-induced takotsubo syndrome and diabetic ketoacidosis: Rare reactions. BMJ Case Rep. 2021, 14, e237217. [Google Scholar] [CrossRef] [PubMed]
  122. Ederhy, S.; Cautela, J.; Ancedy, Y.; Escudier, M.; Thuny, F.; Cohen, A. Takotsubo-like Syndrome in Cancer Patients Treated with Immune Checkpoint Inhibitors. JACC Cardiovasc. Imaging 2018, 11, 1187–1190. [Google Scholar] [CrossRef] [PubMed]
  123. Khan, N.A.J.; Pacioles, T.; Alsharedi, M. Atypical Takotsubo Cardiomyopathy Secondary to Combination of Chemo-Immunotherapy in a Patient with Non-Small Cell Lung Cancer. Cureus 2020, 12, e9429. [Google Scholar] [CrossRef] [PubMed]
  124. Roth, M.E.; Muluneh, B.; Jensen, B.C.; Madamanchi, C.; Lee, C.B. Left ventricular dysfunction after treatment with ipili-mumab for metastatic melanoma. Am. J. Ther. 2016, 23, e1925–e1928. [Google Scholar] [CrossRef] [PubMed]
  125. Al-Obaidi, A.; Parker, N.; Choucair, K.; Alderson, J.; Deutsch, J.M. A Case of Acute Heart Failure Following Immunotherapy for Metastatic Lung Cancer. Cureus 2020, 12, e8093. [Google Scholar] [CrossRef]
  126. Samejima, Y.; Iuchi, A.; Kanai, T.; Noda, Y.; Nasu, S.; Tanaka, A.; Morishita, N.; Suzuki, H.; Okamoto, N.; Harada, H.; et al. Development of Severe Heart Failure in a Patient with Squamous Non-small-cell Lung Cancer During Nivolumab Treatment. Intern. Med. 2020, 59, 2003–2008. [Google Scholar] [CrossRef]
  127. Tan, N.Y.L.; Anavekar, N.S.; Wiley, B.M. Concomitant myopericarditis and takotsubo syndrome following immune check-point inhibitor therapy. BMJ Case Rep. 2020, 13, e235265. [Google Scholar] [CrossRef]
  128. Anderson, R.D.; Brooks, M. Apical takotsubo syndrome in a patient with metastatic breast carcinoma on novel immunother-apy. Int. J. Cardiol. 2016, 222, 760–761. [Google Scholar] [CrossRef]
  129. Schwab, K.S.; Kristiansen, G.; Isaak, A.; Held, S.E.A.; Heine, A.; Brossart, P. Long Term Remission and Cardiac Toxicity of a Combination of Ipilimumab and Nivolumab in a Patient With Metastatic Head and Neck Carcinoma After Progression Fol-lowing Nivolumab Monotherapy. Front. Oncol. 2019, 9, 00403. [Google Scholar] [CrossRef]
  130. Cheng, Y.; Nie, L.; Ma, W.; Zheng, B. Early Onset Acute Coronary Artery Occlusion After Pembrolizumab in Advanced Non-Small Cell Lung Cancer: A Case Report. Cardiovasc. Toxicol. 2021, 21, 683–686. [Google Scholar] [CrossRef]
  131. Tomita, Y.; Sueta, D.; Kakiuchi, Y.; Saeki, S.; Saruwatari, K.; Sakata, S.; Jodai, T.; Migiyama, Y.; Akaike, K.; Hirosako, S.; et al. Acute coronary syndrome as a possible immune-related adverse event in a lung cancer patient achieving a complete response to anti-PD-1 immune checkpoint antibody. Ann. Oncol. 2017, 28, 2893–2895. [Google Scholar] [CrossRef] [PubMed]
  132. Kwan, J.M.; Cheng, R.; Feldman, L.E. Hepatotoxicity and Recurrent NSTEMI While on Pembrolizumab for Metastatic Giant Cell Bone Tumor. Am. J. Med. Sci. 2019, 357, 343–347. [Google Scholar] [CrossRef] [PubMed]
  133. Cancela-Díez, B.; Gómez-De Rueda, F.; Antolinos Pérez, M.J.; Jiménez-Morales, A.; López-Hidalgo, J.L. Acute coronary syn-drome and recurrent colitis as immune-related adverse events in a lung cancer patient. J. Oncol. Pharm. Pract. 2020, 26, 252–255. [Google Scholar] [CrossRef] [PubMed]
  134. Cautela, J.; Rouby, F.; Salem, J.-E.; Alexandre, J.; Scemama, U.; Dolladille, C.; Cohen, A.; Paganelli, F.; Ederhy, S.; Thuny, F. Acute Coronary Syndrome with Immune Checkpoint Inhibitors: A Proof-of-Concept Case and Pharmacovigilance Analysis of a Life-Threatening Adverse Event. Can. J. Cardiol. 2020, 36, 476–481. [Google Scholar] [CrossRef] [PubMed]
  135. Masson, R.; Manthripragada, G.; Liu, R.; Tavakoli, J.; Mok, K. Possible Precipitation of Acute Coronary Syndrome with Im-mune Checkpoint Blockade: A Case Report. Perm. J. 2020, 24, 1. [Google Scholar] [CrossRef]
  136. Otsu, K.; Tajiri, K.; Sakai, S.; Ieda, M. Vasospastic angina following immune checkpoint blockade. Eur. Heart J. 2020, 41, 1702. [Google Scholar] [CrossRef]
  137. Kumamoto, T.; Kawano, H.; Kurobe, M.; Akashi, R.; Yonekura, T.; Ikeda, S.; Maemura, K. Vasospastic Angina: An Immune-related Adverse Event. Intern. Med. 2022, 61, 1983–1986. [Google Scholar] [CrossRef]
  138. Guo, K.; Chen, M.; Li, J. PD-1 Inhibitor-Induced Thyrotoxicosis Associated with Coronary Artery Spasm and Ventricular Tachycardia. Cardiovasc. Toxicol. 2022. [Google Scholar] [CrossRef]
  139. Maio, M.; Scherpereel, A.; Calabrò, L.; Aerts, J.; Perez, S.C.; Bearz, A.; Nackaerts, K.; Fennell, D.A.; Kowalski, D.; Tsao, A.S.; et al. Tremelimumab as second-line or third-line treatment in relapsed malignant mesothelioma (DETERMINE): A multicen-tre, international, randomised, double-blind, placebo-controlled phase 2b trial. Lancet Oncol. 2017, 18, 1261–1273. [Google Scholar] [CrossRef]
  140. Choueiri, T.K.; Larkin, J.; Oya, M.; Thistlethwaite, F.; Martignoni, M.; Nathan, P.; Powles, T.; McDermott, D.; Robbins, P.B.; Chism, D.D.; et al. Preliminary results for avelumab plus axitinib as first-line therapy in patients with advanced clear-cell re-nal-cell carcinoma (JAVELIN Renal 100): An open-label, dose-finding and dose-expansion, phase 1b trial. Lancet Oncol. 2018, 19, 451–460. [Google Scholar] [CrossRef]
  141. Juergens, R.A.; Hao, D.; Ellis, P.M.; Tu, D.; Mates, M.; Kollmannsberger, C.; Bradbury, P.A.; Tehfe, M.; Wheatley-Price, P.; Robinson, A.; et al. A phase IB study of durvalumab with or without tremelimumab and platinum-doublet chemotherapy in advanced solid tumours: Canadian Cancer Trials Group Study IND226. Lung Cancer 2020, 143, 1–11. [Google Scholar] [CrossRef] [PubMed]
  142. Huang, J.; Xu, J.; Chen, Y.; Zhuang, W.; Zhang, Y.; Chen, Z.; Chen, J.; Zhang, H.; Niu, Z.; Fan, Q.; et al. Camrelizumab ver-sus investigator’s choice of chemotherapy as second-line therapy for advanced or metastatic oesophageal squamous cell car-cinoma (ESCORT): A multicentre, randomised, open-label, phase 3 study. Lancet Oncol. 2020, 21, 832–842. [Google Scholar] [CrossRef]
  143. Peleg Hasson, S.; Salwen, B.; Sivan, A.; Shamai, S.; Geva, R.; Merimsky, O.; Raphael, A.; Shmilovich, H.; Moshkovits, Y.; Kapusta, L.; et al. Re-introducing immunotherapy in patients surviving immune checkpoint inhibitors-mediated myocarditis. Clin. Res. Cardiol. 2021, 110, 50–60. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The CD8+, CD4+ T cells, and macrophages interplay within the pathogenesis of ICI-related cardiotoxicity.
Figure 1. The CD8+, CD4+ T cells, and macrophages interplay within the pathogenesis of ICI-related cardiotoxicity.
Ijms 23 10948 g001
Figure 2. Flow diagram of the paper selection processes.
Figure 2. Flow diagram of the paper selection processes.
Ijms 23 10948 g002
Table 1. Case reports published from 2021–2022—ICI-related myocarditis.
Table 1. Case reports published from 2021–2022—ICI-related myocarditis.
StudyPatient CharacteristicsMedical HistorySymptomsDiagnosisCV Side EffectICIType of CancerMyocarditis Onset Myocarditis TreatmentEvolution
Ida, 2022♀, 81HBP
dyslipidemia
high-grade fever
whole-body rash
altered consciousness
↑CK; ↑CKMB, troponin I, C-reactive protein, ECG, TTE
CMR
endomyocardial biopsy
MyocarditisPD-L1i + PD-1iadvanced melanoma,1 weekMethylprednisolone
followed by prednisolone
IGIV
favorable
Nguyen, 2022♂, 25N/Achest pain, subtle myalgiaCoronary angiography
cardiac MRI
endomyocardial biopsy
Troponin-T
creatine-kinase
Myocarditis
myositis
PD-1ihymoma2 weeksMethylprednisolone
mycophenolate-mofetil
loading dose of intravenous abatacept
oral ruxolitinib
cardiogenic shock
VT
extracorporeal life support
On day 40, the patient fully recovered clinically
Okauchi, 2022♂, 60Smoking history Cough, dyspnea Chest RX
BNP
TTE
MyocarditisPD-1iSquamous cell carcinoma 130 weeks from initiation Diuretics, beta-blocker Favorable
Zhou, 2022♂, 67N/Afever, chest pain and dyspneaChest computed tomography, TTE, ECG, BNP, troponin T, CK myocarditisPD-L1ilung squamous
cell carcinoma stage IV
days after lastcyclemethylprednisolonefavorable
Zhao, 2022♂, 60N/Afever, tachycardia, hypotension, fatigue, dyspneaECG,
TTE
Myocarditis + hypothyroidismPD-1isoft tissue sarcoma8 weeksIV mPSLfavorable
Lorente-Ros, 2022♂, 70nephrectomy2 episodes in the previous 12 h of severe dizziness, dyspnea, and profuse sweating.ECG
troponin I,C- reactive protein, TTE ECG- 3rd AVB, coronary angiography, brain CT autoimmunity lab tests, brain, MRI, lumbar puncture, EEG
Myocarditis + EncephalitisPD-1i + CTLA-4irenal cell carcinoma19 days Temporary pacemaker
high-dose iv corticosteroids
intravenous immunoglobulins
delirium deterioration in his level of consciousness,
intubation.
Extubate -> reintubation
discharged
Saishu 2022♀, 55N/Aquadrantanopia, ocular motility disorder, diplopia, dysphagia, ocular motility
disorder,
muscle weakness of the extremities,
bilateral ptosis muscle weakness.
↑CK;
ECG,
TTE; anti-AchR ab.
Myocarditis, myositis/MGPD-1imela-
noma
2 weeksIGIV, prednisolone
Intubation for MV,
mPSL
plasma exchange
Tracheostomy
favorable
Yang, 2022♀, 51contrast agent allergyhigh fever,
mild dyspnea, and systemic rash.
Liver function indexes
cardiac markers
CT examination
myocarditis
hepatitis
PD-1ibreast cancer (TNBC) 3 daysIv methylprednisolone
Antibiotics
hepatic protectors
favorable
Ederhy, 2021♀, 60–70 approx.N/Adiplopia↑TnI;
ECG;
CMR, coronary angiography, EMB
Myocarditis (subclinical)PD-1iLung cancer (unmentioned type, metastatic)3 infusions + 10 dayssteroids, plasmapheresisfavorable
Tsuruda, 2021♂, 75N/Aasymptomatic↑cTnT, CK, CK-MB; ECG, Echocardiogram, CMR, EMBMyocarditis (subclinical), TTSPD-1iNSCLS (squamous, recurrent)3 weeksmPSLfatal
♂, 47N/Aasymptomatic↑cTnT, CK, CK-MB; CMRMyocarditis (subclinical)PD-1iEthmoid sinus cancer3 infusions + 16 daysmPSL; IVIG (progressive thrombocytopenia); cyclosporine (hemophagocytic syndrome)favorable
♂, 63N/Ahypotension (84/42 mmHg), tachycardia (132 bpm), tachypnea (22 rpm); high fever, decreased appetite,↑cTnT, CK, AST, ALT, CRP, Cr;
↓WBC, Hb, PLT;
ECG, Echocardiogram
cardiac complication of cytokine-releasing syndromePD-1iHypopharyngeal cancer5 infusions + 32 dayscardioversion, extracorporeal hemoperfusion with polymyxin B + continuous hemodiafiltration, catechocardiographylamines, broad-spectrum antibiotics, recombinant thrombomodulin, IVIG, high-dose corticosteroidsfavorable
Tanabe, 2021♂, 75N/Aposterior neck pain, neck drop↑TnI, CK, CK-MB; ↑Eo (834/μL), ↓eRFG; DLST (+); Echocardiogram, coronary angiography, CMR, Myocarditis (subclinical)PD-1i + CTLA-4iRCC (clear cell, metastatic)53 daysprednisolonefavorable
Barham, 2021♀, 79N/Adizziness, abdominal bloating, hypoxic↑LDH;
ECG;
EMB
Myocarditis (grade 4); hyperprogressionPD-1i + CTLA-4iMelanoma (vaginal, metastatic)23 dayssteroids; carboplatin + paclitaxel (salvage therapy), atropine, pacemaker (for AVB III)fatal
Xie, 2021♂, 67N/Aexertional dyspnea, ptosis, blurred vision, quadriparesis↑TnI, CK, CK-MB, AST, ALT, BNP, Mb;
ECG;
Echocardiogram;
coronary angiography
Myocarditis (fulminant), MG crisis, hepatic dysfunction; delayed ir pneumonitisPD-1i + pemetrexed + carboplatinLCNEC (metastatic)2 weeksmPSL; pacemaker (temporary permanent); ganciclovir/cefmetazolefavorable
Hu, 2021♂, 63N/Achest tightness,
limb weakness, dorsal myasthenia, diplopia, dysphagia
↑Hs-TnI,
CK-MB,
NT-proBNP,
CK;
Echocardiogram, CMR; anti-β1AR ab,
CC ab, anti-myosin heavy
chain ab,
ribonucleoprotein ab
Myocarditis + MGPD-1iureteral urothelial cancer IV3 weeksmPSL
IVIG
favorable
Wintersperger, 2021♂, 52N/Afatigue dyspnea↑hsTnI,
CK, BNP;
ECG, Chest CT, Echocardiogram, CMR,
EMB
MyocarditisPD-L1i + investigational ICImelanoma3 weeksmPSL
prednisone infliximab IV MMF
favorable
♀, 60N/Ageneral-
ized weakness muscle pain
fatigue fever
↑CK, hsTnI;
ECG, Coronary angiography, CMR, EMB
MyocarditisPD-L1igynecological cancer2 weeksmPSL
prednisone
favorable
♀, 49N/Afever cough↑hsTnI,
BNP;
chest CT,
ECG, CMR
MyocarditisPD-L1itriple-negative breast cance2 weeksMMF
prednisone
favorable
♀, 74N/Ageneral pain, progressive muscle weakness diplopia↑hsTnI,
BNP, ECG,
Coronary angiography, CMR
MyocarditisPD-L1igynecological cancer2 weeksmPSL
prednisone
MMF
favorable
Stein-Merlob, 2021♀, 60N/Apalpitations
reduced exercise tolerance, cool extremities
altered
mental status
↑Tn,
BNP;
ECG, Echocardiogram,
Coronary angiography,
CMR
Myocarditis
Ocular myasthenia, Colitis
hepatitis
PD-1iColon cancer Metoprolol succinate, lisinopril
Continued immunosuppression spironolactone,
Oral amiodarone, wearable defibrillator.dopamine Nitroprusside
milrinone, VA-ECMO
favorable
Shen, 2021♀, 53N/Acough
chest
congestion,
muscle weakness fatigability drooping eyelids,
↑CK, CK-MB;
ECG
Myocarditis, hepatitis, renal dysfunction, hypothyroidismPD-1i +
paclitaxel + platinum
type B3 thymoma3 weeksMagnesium
isoglycyrrhizinate reduced glutathione injections, prednisone
mPSL
euthyrox
pyridostigmine
favorable
Miyauchi, 2021♂, 71hypertension, DM2, hyperuricemiaAsymptomatic,
chest tightness, shortness of breath,
cardiogenic shock
↑CK, CM-MB, TnI,
NT-proBNP; ECG,
catheterization, EMB, CMR
MyocarditisCTLA-4i + PD-1iRCC8 weeksdopamine, dobutamine, noradrenaline
intra-aortic balloon pump was inserted,
adaptive servo ventilation
mPSL
prednisolone
favorable
Luo, 2021♀, 47N/Adiplopia, myalgia, limb weakness, dysphagia, dyspnea↑TnI, CK,
ECG, EMG;
RyR-ab, AChR-ab,
anti-fibrillarin ab, anti-NOR-90 ab
anti-Ro-52 ab
Myocarditis, myositis, MGPD-1ithymoma3 weeksneostigmine IVIG
mPSL
prednisolone
pacemaker
favorable
Li, 2021♂, 62hypertension, coronary heart diseasefever lethargy, cognitive dysfunction tachypnea hypoxia hypotension
oliguria
↑Mb, Tn, CK-MB; ECGcardiotoxicity kidney toxicity.PD-1ilung adenocarcinoma48 weeksmPSL
continuous renal replacement therapy
favorable
Jespersen, 2021♂, 57N/Aheadache myalgia, palpitations
binocular diplopia, ptosis,
muscle weakness
↑TnI, CK-MB,
Mb, CK;
ECG, EMG,
Echocardiogram, CMR, AChR-ab,
Myocarditis + myositisCTLA-4i + PD-1iRCC2 weekstemporary pacemaker. mPSL abatacept MMF
implantable cardio-defibrilator
favorable
Iwasaki, 2021♀, 70hypertension, aortic stenosis,
chronic renal failure
shortness of breath fatigue↑CK, CK-MB,
TnT, NT-proBNP;
ECG, Echocardiogram, CMR, Cardiac catheterization, EBMCoronary angiography
Myocarditis + myositisPD-L1iHCC<1 weekcariperitide,
furosemide,
mPSL
prednisolone
favorable
Hernández, 2021♀, 48N/Ashortness of breath,
dyspnea,
bilateral ptosis blurred vision
↑Hs-TnI, NT-proBNP, CRP,
CK;
ECG,
Echocardiogram,
Coronary angiography,
EMB;
AChR-ab
Myocarditis + myositis (MG)PD-1ithymoma<2 weeksIV isoproterenol drip mPSL
Infliximab
temporary pacemaker, dual-chamber pacemaker. intravenous amiodarone, noradrenaline dobutamine, intravenous anti-thymocyte globuline, pyridostigmine, ECMO
fatal
Giblin, 2021♀, 47N/Adermatitisdiarrhea, palpitations↑hS-TnI,
BNP,
Echocardiogram, CMR, Coronary angiography,
EMB
Myocarditis (subclinical)CTLA-4i + PD-1imelanoma1 weekmPSL
prednisolon, IVIG
favorable
Cao, 2021♂, 69N/Aptosis, diplopia, shortness of breath,↑CK, CK-MB,
Mb,
hs-TnT,
NT-proBNP,
LDH, ECG;
Echocardiogram,
EMG
Myositis,
Myocarditis
SJS/TEN
PD-1iesophagogastric junction
carcinoma
2 weeksmPSL
IVIG
plasmapheresis
favorable
Ai, 2021♂, 72N/AasymptomaticECG; CMRMyocarditis (DRESS)PD-1igastric adenocarcinoma3 weeksSCS (for DRESS)
Abbreviations: ↑, increase; ↓, decrease; Ab, antibody; ACEi, angiotensin-converting-enzyme inhibitors, AchR, acetylcholine receptor, AF, atrial fibrillation; ALT, alanine transaminase, ANA, antinuclear antibodies, ARB, angiotensin receptor blockers; AST, aspartate aminotransferase, AVB, atrioventricular block; β1AR, beta-1-adrenergic receptor; BB, beta-blocker; BiPAP, bilevel positive airway pressure; BNP, brain natriuretic peptide; CAD, coronary artery disease; CC, calcium channel; CK, creatine kinase; CK-MB, creatine kinase—muscle/brain; CMR, cardiovascular magnetic resonance imaging; cN1A, cytosolic 5’-nucleotidase 1A; Cr, serum creatinine; CRP, C-reactive protein; CT, computer tomography; CTLA-4i, cytotoxic T-lymphocyte-associated protein 4 inhibitor; DLST, drug lymphocyte stimulation test; DM2, diabetes mellitus type 2; ECG, electrocardiography; eGFR, estimated glomerular filtration rate; EMB, endomyocardial biopsy; Eo, eosinophils; ESC, esophageal squamous cell carcinoma; GFAP, glial fibrillary acidic protein, Hb, hemoglobin; HCC, hepatocellular carcinoma; HCV, hepatitis C virus; HCQ, hydroxychloroquine; HMGCR, HMG-CoA reductase; HF, heart failure; HfpEF, heart failure with preserved ejection fraction; IABP, intra-aortic balloon pump; ICD, implantable cardioverter-defibrillator; IV, intravenous; IVIG, intravenous immune globulin; LCNEC, large cell neuroendocrine carcinoma; LDH, lactate dehydrogenasem; LyT, T lymphocytes; Mb, myoglobin; MPM, malignant pleural mesothelioma; mPSL, methylprednisolone; MG, myasthenia gravis; MMF, mycophenolate mofetil; MuSK, muscle-specific kinase; MV, mechanical ventilation; NC, nasal cannula; NG, nasogastric; NIPPV, nasal intermittent positive pressure ventilation; NOR90, nucleolus organizer region; NSCLC, non-small cell lung cancer; NT-pro-BNP, N-terminal pro hormone BNP; PO, per oral; PD-1i, programmed cell death 1 inhibitor; PD-L1i, programmed cell death ligand 1 inhibitor; PLEX, plasmapheresis; PLT, platelet count; PM/SCL, polymyositis/scleroderma; RCC, renal cell carcinoma; RYR, ryanodine receptor; SCS, systemic corticosteroid therapy; SJS, Stevens-Johnson syndrome; SRP, signal recognition particle; TEN, toxic epidermal necrolysis; Tn, troponin; TTS, Takotsubo syndrome, VA ECMO, veno-arterial extracorporeal membrane oxygenation, VT, ventricular tachicardia, WBC, white blood cells count.
Table 2. Case reports with ICI-associated pericarditis.
Table 2. Case reports with ICI-associated pericarditis.
StudyPatient CharacteristicsCV Side Effect Symptoms Diagnosis Pre-Existent CVDICIType of CancerPericarditis Onset Concomitant Treatments Pericarditis Treatment Evolution Concomitant AID
Khan,
2019
♂, 62pericarditisdyspneaCT, echocardiogram, ECG, TnI, BNPnot mentionedPD-1itonsillar cancer (squamous)15 weeksnot mentionedpericardiocentesis, prednisoneresolutionnot mentioned
Arora,
2020
♂, 83pericarditismarked fatigue
weakness chest pain orthopnea
left eye ptosis
TnI, CK, TTE, brain MRI Hypertension,
HyperlipidemiaAtrial fibrillation
PD-1imelanoma1 month after first dosenot mentionedcolchicine and naproxen
IV methylprednisolonePlasmapheresis
Intubation
inability to reduce
ventilatory support–transitioned to
comfort measures
Hepatitis
MG
de Almeida, 2018♂, 69pericarditisdyspnea,
tachycardia, low-grade fever
CT, echocardiogramnot mentionedPD-1iNSCLC (adenocarcinoma)48 weeksnot mentionedpericardiocentesis, prednisonefavorablethyroiditis
Oristrell, 2018♀, 55pericarditispericardial chest painechocardiogram, ECG, TnInot mentionedPD-1iductal carcinoma of the left breast30 weeksnot mentionedPericardiocentesis followed by pericardiectomy steroidsfavorablenot mentioned
Zarogoulidis, 2017♂, 60pericarditisnot mentionednot mentionednot mentionedPD-1iNSCLC17 weeksnot mentionedpericardiocentesis mPSL favorablenot mentioned
Öztürk,
2021
♂, 61pericarditisnot mentionedCT, echocardiogram, MRInot mentionedPD-1iNSCLC17 weekspemetrexednot mentionednot mentionednot mentioned
Moriyama,
2021
♂, 58pericarditisfatigue, limb oedema, increased body weightechocardiogram, cardiac CT, cardiac catheterization, EMB, MRI, ECG, TnTnot mentioned PD-1iNSCLC77 weeksnot mentionedprednisolone, furosemide, mPSL, infliximab favorableautoimmune hepatitis
Jacobs, 2021♂, 54pericarditischest pain, general malaise, dyspneaechocardiogram, CT, MRI, ECG, TnInot mentionedPD-1iNSCLC (adenocarcinoma)5 weekscarboplatin + pemetrexed + mPSLfatal not mentioned
Dasanu, 2016♀, 65pericarditisprogressive dyspnea, chest discomfortX-ray, CT, echocardiogram, ECGnot mentionedPD-1imelanoma (nodular type)37 weeksnot mentionedpericardiocentesis, mPSLfavorableabnormal thyroid function, hepatitis, rash
Dhenin, 2019♀, 79pericarditisintense thoracic pain, increasingwhen leaning, fatigue, general malaiseechocardiogram, ECGhypertensionPD-1iNSCLC (adenocarcinoma)3 weeksnot mentionedmPSLfavorablerash, colitis, MG
Abbreviations: CT, computer tomography; ECG, electrocardiography; EMB, endomyocardial biopsy; IV, intravenous; LAFB, left anterior fascicular block; mPSL, methylprednisolone; MG, myasthenia gravis; MRI, magnetic resonance imaging; NSCLC, non-small cell lung cancer; PD-1i, programmed cell death 1 inhibitor; Tn, troponin.
Table 3. Case reports with ICI-associated arrhythmia.
Table 3. Case reports with ICI-associated arrhythmia.
StudyPatient CharacteristicsCV Side EffectSymptomsDiagnosisPre-Existent CVDICIType of CancerArrhythmias Onset Concomitant Treatments Arrhythmias Treatment Evolution Concomitant AID
Joseph, 2021♂, 78AFnot mentionedECGhypertensionPD-1imetastatic melanoma35 weeksnot mentionedTEE-guided cardioversionfavorablenot mentioned
♂, 68AFnot mentionedECGhypertensionPD-1imetastatic melanoma14 weeksnot mentionedself-limited AF (48 h)favorablenot mentioned
♂, 66AFnot mentionedECGhypertensionPD-1imetastatic melanoma2 weeksnot mentionedbeta-blockers favorablehyperthyroidism
♂, 74sinus bradycardia,
AF with RVR
fatigue and
dizziness
ECG3 ablations for AFPD-1imetastatic melanoma21 weeksnot mentionedbeta-blockersfavorablenot mentioned
Reddy, 2017♂, 68sinus tachycardia, 1st degree AVB, RBBB, LAFB followed bycomplete AVB) fatigue, generalized malaise, weakness with ambulation NYHA IIB symptomsECG, Tn, CK-MB, echocardiogram, cardiac catheterizationnot mentionedPD-1i + CTLA-4imetastatic sarcoma 2 weeksnot mentionedhigh-dose IV steroids, temporary transvenous pacemaker, MMFfavorablenot mentioned
Giancaterino, 2020♂, 88ECGs–progression normal SR + with
PAC- 3rd AVB (hospital day 5)
generalized weaknessECG, Tn, CK-MB, TTEnot mentioned invasive melanomafirst
- 22 days prior
prednisone 40 mg daily
nivolumab infusions were held
IV methylprednisolone
Infliximab
dual-chamber pacemaker—day 10
decline clinically- VF- fatal myositis
Behling, 2017♂, 63complete AVB (44 bpm)worsening of a pre-existing dyspneaECG, echocardiogram, cardiac catheterization, myoglobin, TnhypertensionPD-1imetastatic melanoma3 weeksnot mentionedtemporary pacemaker, corticosteroids, oxygen therapyfatalnot mentioned
Katsume, 2018♂, 73complete AVB (wide QRS complexes)fatigue, faintness, syncope, palpitationsECG, echocardiogram, cardiac catheterization, TnT, CKnot mentionedPD-1imetastatic NSCLC2 weeksnot mentionedIV steroids, pacemaker (temporary permanent)favorablenot mentioned
Hsu, 2018♂, 42sinus bradycardia (40 bpm)fatigue, dizziness, anorexia, hypotensionECG, TnInot mentionedPD-1imetastatic liver cancernot mentionednot mentionedPO steroidsfavorablenot mentioned
Pohl, 2020♀, 61new 1st degree AVB, QTc prolongationnot mentionedECG, echocardiogramnot mentionedPD-1imetastatic melanoma4–12 weeksnot mentionednot mentionednot mentionednot mentioned
Khan, 2020♀, 67Mobitz type 2 2nd degree AVB (30 bpm) complete AVB (22 bpm) after 3 h asymptomaticECG, echocardiogram, TnThypertension, hyperlipidemiaPD-1imetastatic NSCLC3 weeksnot mentioneddobutamine, pacemaker (temporary permanent)favorablenot mentioned
Abbreviations: AF, atrial fibrillation; AVB, atrioventricular block; CK, creatine kinase; CK-MB, creatine kinase–muscle/brain; CTLA-4i, cytotoxic T-lymphocyte-associated protein 4 inhibitor; ECG, electrocardiography; IV, intravenous; LAFB, left anterior fascicular block; MMF, mycophenolate mofetil; NSCLC, non-small cell lung cancer; NYHA, New York Heart Association; PO, per oral; PD-1i, programmed cell death 1 inhibitor; RBBB, right bundle branch block; RVR, rapid ventricular response; TEE, transesophageal echocardiogram; Tn, troponin.
Table 4. Case reports with ICI-related cardiomyopathies.
Table 4. Case reports with ICI-related cardiomyopathies.
StudyPatientCV Side EffectSymptomsDiagnosisPre-Existent CVDICIType of CancerCVD Onset CVD TreatmentEvolution
Serzan, (2021)♀, 66Takotsubo cardiomyopathyExertional dyspnea, generalized painTNI, ECG, TTE, CT angiography, RV catheterization, EMB, CMRnot mentionedCTLA-4i + PD-1iChoroidal melanoma16 weeksMetoprololFavorable
Oldfield, (2021)♂, 76Takotsubo cardiomyopathy, diabetic ketoacidosisChest pain, diaphoresisECG, TNI, Coronary angiography, TTE, CMRT2DM, dyslipidemia, hypertensionCTLA-4i + PD-1iMelanoma4 daysAspirin, bisoprolol, ramiprilFavorable
Schwab, (2018)♂, 69Takotsubo cardiomyopathyChest pain, shortness of breathCoronary angiography, TTE, CMRnot mentionedPD-1i and CTLA-4i + PD-1iSquamous cell cancer of the lower lip7 cyclesHeart failure treatment, prednisolone Favorable
Ederhy, (2018)♂, 41Takotsubo-like syndromenot mentionedECG, TNI, TTE, Coronary angiography, CMRnot mentionedCTLA-4i + PD-1imelanoma5 daysIV mPSL Favorable
♂, 77Takotsubo-like syndromenot mentionedECG, CMR, TNI, Coronary angiographynot mentionedCTLA-4i + PD-1iesophageal melanoma2 cycles of ipilimumab + nivolumab and 1 cycle of nivolumabmPSL, ACEi, beta blockersFavorable
Tan, (2020)♂, 62Takotsubo cardiomyopathyChest
pain, nausea, vomiting
ECG, Coronary angiography, TNI, NT-proBNP, TTE, CMRnot mentionedPD-1iHCC3 weeksIV mPSL, PrednisoneFavorable
Al-Obaidi, (2020)♀, 52Acute HFDyspnea on exertion, angina-like chest pain, lower extremity oedemaTTE, chest X-ray, CT angiography, ECGnot mentionedCTLA-4i + PD-1iNSCLC 1 yearIV mPSL, oral prednisoneFavorable
Khan, (2020)♀, 57Atypical Takotsubo cardiomyopathyChest pain, palpitations, tachypnoea, tachycardiaChest X-ray, ECG, TNI, TTE, Coronary angiographynot mentionedPD-L-1iNSCLC (adenocarcinoma)2 weeks following 4th cycle Guideline-directed HF treatmentFavorable
Samejima, (2020)♂, 79Acute HFDyspneaChest X-ray, CT, ECG, CRP, CK, CK-MB, TnI, BNP, TTE, Coronary angiography, EMBnot mentionedPD-1iNSCLC 20 daysFurosemide, dopamine, tolvaptan, bisoprolol, spironolactone, enalaprilFavorable
Roth, (2016)♂,60Left Ventricular Dysfunctionheart palpitationsECG, TTE, pharmacologic
stress test
hypertension, anxiety, and Raynaud
syndrome
CTLA-4iBRAFwild-type stage IIIA (T2, N1a) melanoma 4 cycles + another 4 cycles (liver metastasis)–after 4 months beta blockers, ACEI Favorable
Andersen, (2016)♀, 56Apical takotsubo syndromechest pain after
severe episode
of abdominal cramping with diarrhea
ECG, highly sensitive troponin
Chest X-ray, TTE, Coronary angiography
no cardiac
risk factors. No CVD
PD-1ibreast carcinoma3 weeks ACEi, beta blockersFavorable
Abbreviations: ACEi, angiotensin-converting enzyme inhibitor; ACS, acute coronary syndrome; BNP, brain natriuretic peptide; CCB, calcium channel blocker; CTLA-4i, cytotoxic T-lymphocyte-associated protein 4 inhibitor; cTnI, cardiac troponin I; CK, creatine kinase; CK-MB, creatine kinase-MB; CMR, Cardiac MRI; CRP, C-reactive protein; ECG, electrocardiogram; HF, heart failure; HCC, hepatocellular carcinoma; mPSL, methylprednisolone; NSCLC, non-small cell lung cancer; NT-proBNP, N-terminal pro-brain natriuretic peptide; PD-1i, programmed cell death protein 1 inhibitor; PD-L1i, programmed death-ligand 1 inhibitor; RV, right ventricle; T2DM, Type 2 diabetes mellitus; TTE, transthoracic echocardiogram.
Table 5. Case reports with ICI-related acute coronary syndrome.
Table 5. Case reports with ICI-related acute coronary syndrome.
StudyPatientCV Side EffectSymptomsDiagnosisPreexistent CVDICIType of CancerCVD Onset CVD TreatmentEvolution
Arora, 2020♂,69ACS (NSTEMI)diffuse body pain
weakness
ECG, CK-MB, cTNI, TTE
Coronary angiography
CKD, Hypertension, Hyperlipidemia
Type 2 DM
CAD
PD-1imetastatic
urothelial carcinoma
cycle
day 2
IV
Steroids, MMF
transition to
comfort measures
Cheng, (2021)♀, 87ACS (NSTEMI)Chest pain and dyspneaECG, CK-MB, cTNI, CRP, Coronary angiographyHypertension, 3-vessel CADPD-1iNSCLC (adenocarcinoma)2 daysPCI + DESFavorable
Tomita, (2017)♂, 61ACS (NSTEMI)not mentionedCK, CK-MB, cTNI, Coronary angiography, OCTDyslipidemiaPD-1iNSCLC (adenocarcinoma)11th cyclePCI + DES; thrombus aspirationFavorable
Kwan, (2019)♀, 71ACS (NSTEMI)Chest paincTNI, ECG, Coronary angiographyHypertension, T2DM, peripheral
artery disease
PD-1iGiant cell tumor of the bone2 YearsFirst ACS: atherectomy of the LAD with 3 x DES, aspirin + clopidogrel, atorvastatin;
Second ACS: DES, DAPT
Favorable
Cancela-Díez, (2019)♂, 79ACS (STEMI)Chest
pain, oppression and dyspnea.
TNI, TTE, ECG, Coronary angiographyInfrarenal abdominal aortic aneurysmPD-1iNSCLC (epidermoid)10 days after the last cycle (10th) PCI + DES, aspirin + clopidogrel, nitro-glycerine, beta blockers, enalaprilFavorable
Masson, (2020)♂, 62ACS (NSTEMI)Chest painECG, TNI, BNP, TTE, Coronary angiographyT2DM, multi-vessel CAD, STEMIPD-1iMelanoma1 week after cycle 4 of Nivolumab therapyCABGFavorable
Cautela, (2020)♀, 52ACS (NSTEMI)Chest painECG, TNT, NT-proBNP, TTE, Coronary angiography, CMRnot mentionedPD-1iNSCLC (?)5 daysMethylprednisolone prednisolone; aspirin + clopidogrel, statinsFatal (due to refractory shock)
Otsu, (2020)♂, 57Vasospastic anginaRest anginaECG, Coronary
angiography
not mentionedPD-1iRenal cell carcinoma4 weeksCCB, nitratesFavorable
Kumamato, (2022)♀, 54Vasospastic anginachest pain at rest for 2 monthsECG, Chest radiography, TTE, Gadolinium enhanced
cardiac MRI
cardiac catheterization–coronary vasospasm provoked by ergonovine
not mentionedPD-1iHypopharyngeal cancer21 months Benidipine 8 mgFavorable
Guo, (2022)♂, 6oCoronary
Artery Spasm
ventricular tachycardia
1-week history of chest tightness and
palpitation
ECG
Thyroid function
Holter ECG
TTE
acute coronary syndrome
8 months ago–complete revascularization
with stents
PD-1iMetastatic liver cancerPre-evaluation of 3rd dose isosorbide mononitrate
and diltiazem
sedative drug
aspirin, clopidogrel, and atorvastatin
1
week later–discharged
Abbreviations: ACS, acute coronary syndrome; BNP, brain natriuretic peptide; CABG, coronary artery bypass grafting; CAD, coronary artery disease; CCB, calcium channel blockers; cTnI, cardiac troponin I; CK, creatine kinase; CK-MB, creatine kinase-MB; CMR, Cardiac MRI; CRP, C-reactive protein; DAPT, dual antiplatelet therapy; DES, drug-eluting stent; ECG, electrocardiogram; LAD, left anterior descending artery; NSCLC, non-small cell lung cancer; NSTEMI, non-ST segment elevation myocardial infarction; NT-proBNP, N-terminal pro-brain natriuretic peptide; OCT, Optical coherence tomography; PCI, percutaneous coronary intervention; PD-1i, programmed cell death protein 1 inhibitor; STEMI, ST segment elevation myocardial infarction; T2DM, type 2 diabetes mellitus; TTE, transthoracic echocardiogram.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Cozma, A.; Sporis, N.D.; Lazar, A.L.; Buruiana, A.; Ganea, A.M.; Malinescu, T.V.; Berechet, B.M.; Fodor, A.; Sitar-Taut, A.V.; Vlad, V.C.; et al. Cardiac Toxicity Associated with Immune Checkpoint Inhibitors: A Systematic Review. Int. J. Mol. Sci. 2022, 23, 10948. https://doi.org/10.3390/ijms231810948

AMA Style

Cozma A, Sporis ND, Lazar AL, Buruiana A, Ganea AM, Malinescu TV, Berechet BM, Fodor A, Sitar-Taut AV, Vlad VC, et al. Cardiac Toxicity Associated with Immune Checkpoint Inhibitors: A Systematic Review. International Journal of Molecular Sciences. 2022; 23(18):10948. https://doi.org/10.3390/ijms231810948

Chicago/Turabian Style

Cozma, Angela, Nicolae Dan Sporis, Andrada Luciana Lazar, Andrei Buruiana, Andreea Maria Ganea, Toma Vlad Malinescu, Bianca Mihaela Berechet, Adriana Fodor, Adela Viviana Sitar-Taut, Vasile Calin Vlad, and et al. 2022. "Cardiac Toxicity Associated with Immune Checkpoint Inhibitors: A Systematic Review" International Journal of Molecular Sciences 23, no. 18: 10948. https://doi.org/10.3390/ijms231810948

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop