Next Article in Journal
Evaluation of the Expression Levels of miR-21-5p and miR-429 Genes in Biopsy Samples from Patients with Oral Squamous Cell Carcinoma
Previous Article in Journal
Comparison of Diagnostic Performance and Image Quality between Topup-Corrected and Standard Readout-Segmented Echo-Planar Diffusion-Weighted Imaging for Cholesteatoma Diagnostics
Previous Article in Special Issue
The Prevalence and Risk Factors of Chronic Heart Failure in the Mongolian Population
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Immune Checkpoint Inhibitors-Related Myocarditis: A Review of Reported Clinical Cases

Department of Hospital Therapy with a Course of Medical and Social Expertise, Federal State Budgetary Educational Institution of Higher Education “Ryazan State Medical University named after Academician I.P. Pavlova” of the Ministry of Health of the Russian Federation, Ryazan 390026, Russia
Diagnostics 2023, 13(7), 1243; https://doi.org/10.3390/diagnostics13071243
Submission received: 27 February 2023 / Revised: 13 March 2023 / Accepted: 24 March 2023 / Published: 25 March 2023

Abstract

:
Myocarditis associated with the use of immune checkpoint inhibitors (ICI) is a rare manifestation of their cardiotoxicity, but is characterized by a high mortality rate. A literature search was conducted using PubMed using keywords, which resulted in the selection of 679 scientific works, from which 160 articles that described 244 clinical cases were selected. The median age of the patients was 67 years (IQR, 60–74). The median time from the start of ICI therapy to the development of the first adverse symptoms was 21 days (IQR, 14–38.3). In 37% of cases, myocarditis developed after the first administration of ICI. Cardiac symptoms were present in 47.1% of cases, neuromuscular symptoms in 30.3%, and other symptoms in 12.6%, while myocarditis was asymptomatic in 10.1% of cases. New changes in the electrocardiograms were detected in 85.1% of patients compared to the initial data. A high incidence of complete atrioventricular block (25.4%), right bundle branch block (18.4%), ventricular tachycardia (13%), and sinus tachycardia (12%) were noted. In 97% of the cases, the patients received prednisolone or methylprednisolone therapy. When using ICI, special attention should be paid to the early detection of possible cardiotoxicity by analyzing the condition and function of the myocardium before treatment and its dynamics.

1. Introduction

Statistical data show that over the past few decades, the incidence of malignant neoplasms has been steadily increasing, which can be explained by various factors, including population aging, particularly in countries with a high level of economic development, improvements in diagnostic capabilities, and advances in the treatment of oncological diseases, which in turn is reflected in the reduction in mortality rates [1]. During this time, a number of new groups of drugs have been developed and implemented in clinical practice in oncology, and immunotherapy has been actively developed, including monoclonal antibodies, non-specific cytokines, anti-cancer vaccines, and immune checkpoint inhibitors (ICI), which have made a breakthrough in the treatment of a whole range of oncological diseases. The first ICI approved by the Food and Drug Administration (FDA) was the drug ipilimumab (cytotoxic T-lymphocyte associated protein 4 (CTLA-4) inhibitor), which was approved for the treatment of melanoma in 2011 [2]. The next drug in this group to be approved by the FDA was nivolumab (programmed cell death ligand 1 (PD-1) molecular inhibitor) in 2014. Over the next few years, new programmed cell death protein 1 (PD-1) or its ligand, PD-L1, receptor inhibitors were approved all over the world [3,4]. The spectrum of indications for the use of this group of immune drugs in oncology practice is very wide (Table 1).
The mechanism of action of these drugs involves blocking various checkpoint proteins, including the following: CTLA-4 (cytotoxic T-lymphocyte-associated protein 4), PD-1 (programmed cell death protein 1), PD-L1 (programmed cell death ligand 1), LAG-3 (lymphocyte-activation gene 3).
Both CTLA-4 and PD-1, along with its partner PD-L1, are checkpoint proteins that can be inhibited by drugs. Other immune checkpoint drugs target PD-1 and PD-L1, which are partners in the immune system. Certain tumors overproduce PD-L1, which can inhibit the T cell response. Using this process, tumor cells increase the expression of PD-L1 to avoid recognition and destruction by the immune system. ICI block these immune checkpoints, “turning on” the cellular immune response against tumor cells. The mechanisms of action of these antibodies complement each other, which is why they are often used in combination. Other immune checkpoints that are currently being studied for their therapeutic potential include T cell immunoglobulin and mucin-containing protein 3 (TIM-3) [11], lymphocyte-activated gene-3 (LAG-3) [12], T cell immunoreceptor with Ig and ITIM domains (TIGIT) [13], B and T lymphocyte attenuator (BTLA) [14], V-domain Ig suppressor of T cell activation (VISTA, also known as PD-1 homologue, or PD-1H) [15], and others.
Due to their direct effect on the immune system, this class of drugs has specific immune-related adverse events (irAEs). The widespread use of ICI has led to an increase in the frequency of occurrence of specific toxicities. According to the data of various authors, the frequency of irAEs is around 30% with the use of CTLA-4 inhibitors, 20% with anti-PD-1 inhibitors, and 55% in patients receiving a combination of drugs [16]. ICI can have different toxicity profiles, such as dermatological, renal, gastrointestinal, cardiovascular, endocrine, neurological, ocular, pulmonary, musculoskeletal and hematological profiles [17].
IrAEs occur in 70–90% of patients receiving ICI, with severe irAEs occurring in 10–15% of patients [18]; these reactions result in a fatal outcome in 1.3% of patients [19]. IrAEs often occur within 1 year after the treatment [20], but the risk of developing any irAE increases over time [21].
Accumulated data formed the basis for recommendations for the diagnosis and treatment of irAEs [22,23,24]. When assessing the severity of IrAEs, general terminology criteria for adverse events are used, as recommended by the European Society of Medical Oncology and the American Society of Clinical Oncology (Table 2) [25,26].
Cardiotoxicity associated with ICI is relatively rare. However, this type of IrAE often leads to a fatal outcome [27]. Various manifestations of cardiotoxicity are possible, including myocarditis, arrhythmia, asequence, acute coronary syndrome, heart failure, and pericarditis [28]. The most common type of cardiotoxicity associated with ICI is myocarditis. The frequency of ICI-associated myocarditis is known to be between 0.09% and 2.4% [29,30,31]. However, the mortality rate is very high, at approximately 27–60% [32,33]. In 46% of cases, major adverse cardiac events (MACE) occur, including stroke, myocardial infarction, cardiovascular death, cardiogenic shock, cardiac arrest and hospitalization for severe heart failure [30].
The mechanism of cardiotoxicity is not fully studied, but the main cause is attributed to CD4+ T-cell-mediated inflammation [34]. The most common cytokines produced are tumor necrosis factor-alpha (TNF-α), granzyme B, and interferon gamma [35]. In addition, the expression of inflammatory transcription factors NLR family pyrin domain containing 3 (NLRP3), myeloid differentiation primary response gene 88 (MyD88) and p65/NF-κB is increased in cardiomyocytes [36]. It has been established that ICI does not induce the apoptosis of cardiomyocytes, meaning it does not have a direct cytotoxic effect [37].
Wang et al. demonstrated that fatal myocarditis developed in mice genetically predisposed the mice to systemic autoimmunity due to PD-1 deficiency, whereas in mice with genetic predisposition to autoimmunity but without PD-1 deficiency, myocarditis did not develop, suggesting that myocarditis prevention is likely to be mediated by PD-1 [38]. In a similar study, Love et al. found that the deletion of CTLA-4 on T cells also caused severe myocarditis in mice, but the absence of IL-12 prevented the proliferation of CD8+ T cells, thereby reducing the risk of developing myocarditis [39]. PD-L1, expressed in human myocardium, is involved in protecting against immune-mediated heart damage and inflammation [37].
In this article, a review of previously described cases of myocarditis caused by ICI is presented. The clinical demographic characteristics were analyzed, as well as the changes in the electrocardiograms (ECG), patient complaints, changes in laboratory parameters, data from echocardiography (EchoCG), cardiac magnetic resonance imaging (MRI), endomyocardial biopsy and performed therapy and outcomes.

2. Materials and Methods

The PubMed database was used to search for articles. The search was conducted using the following terms: (Checkpoint myocarditis) OR (Ipilimumab myocarditis) OR (Nivolumab myocarditis) OR (Pembrolizumab myocarditis) OR (Cemiplimab myocarditis) OR (Atezolizumab myocarditis) OR (Avelumab myocarditis) OR (Durvalumab myocarditis) or (Tremelimumab myocarditis) or (Relatlimab myocarditis). A total of 679 records were identified in the PubMed database. Subsequently, articles that were not accessible for research, articles not in English, and articles that did not contain descriptions of clinical cases of ICI-associated myocarditis were excluded. Some of the excluded data were used for the review section of our article. Additionally, the list of literature identified in the initially identified articles was manually checked to search for additional clinical cases. After that, duplicated descriptions of clinical cases that were found in different articles from the same institution were excluded. In the end, 160 articles describing 244 cases were selected [16,29,31,40,41,42,43,44,45,46,47,48,49,50,51,52,53,54,55,56,57,58,59,60,61,62,63,64,65,66,67,68,69,70,71,72,73,74,75,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102,103,104,105,106,107,108,109,110,111,112,113,114,115,116,117,118,119,120,121,122,123,124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147,148,149,150,151,152,153,154,155,156,157,158,159,160,161,162,163,164,165,166,167,168,169,170,171,172,173,174,175,176,177,178,179,180,181,182,183,184,185,186,187,188,189,190,191,192,193,194,195,196,197]. The majority of the scientific works were dated 2022 and 2021.
The continuous variables are presented as medians with interquartile ranges (IQR, Q1–Q3). The categorical variables are presented as frequencies (N) and percentages (%).

3. Results

3.1. The Analyzed Patients

In total, 244 cases of ICI-associated myocarditis were identified. The median age of the patients was 67 years (IQR, 06–74) and the patients were predominantly males (143; 40.6%). The age at which ICI-associated myocarditis developed was 66 years (IQR, 56–71) for females and was lower than that for males aged 69 years (IQR, 63–75).
In the analyzed group, patients received ICI therapy for the following types of cancer: melanoma (54 cases, 22.1%), lung cancer (51 cases, 20.9%), renal cell carcinoma (26 cases, 10.7%), thymoma (16 cases, 6.6%), gastric cancer (13 cases, 5.3%), hepatocellular carcinoma (10 cases, 4.1%), urothelial carcinoma (9 cases, 3.7%), and other locations with 1–8 described cases (mesothelioma, urothelial carcinoma, breast cancer, esophageal squamous cell carcinoma, cutaneous squamous cell carcinoma, prostate adenocarcinoma, and others) and 61 cases (25%).
An analysis of the administered therapy showed that 73% of the ICI-associated myocarditis cases occurred during monotherapy with anti-PD-1 drugs; in 17.6% of cases, a combination of anti-CTLA4 and anti-PD-1 therapy was used, while cases of myocarditis during monotherapy with anti-CTLA-4 (0.8%), anti-PD-L1 (7.8%), and combined anti-PD-1/anti-PD-L1 therapy (0.8%) were much less frequent. ICI-associated myocarditis most often occurred during the use of pembrolizumab (30.7%), nivolumab (20.1%), and combined therapy with nivolumab plus ipilimumab (16.8%). Based on these data, it can be assumed that the drugs in the anti-PD-1 group, especially in combination with anti-CTLA4 drugs, have the highest cardiotoxicity among ICI. The prescription of drugs in this group may be one of the risk factors for the development of autoimmune myocarditis, although this hypothesis requires further investigation in prospective studies. The characteristics of the analyzed group of patients are presented in Table 3.
The median time from the start of immunotherapy to the development of the first adverse symptoms was 21 days (IQR, 14–38.3). In 37% of cases, myocarditis developed after the first administration of ICI. However, there were cases where the first symptoms appeared later than a year after the start of ICI (both cases in the pembrolizumab group); in addition, cases of onset after more than 6 months of use of ICI were described in the pembrolizumab, nivolumab, and nivolumab plus ipilimumab groups. The median number of days from the start of ICI varied in the following ways depending on the treatment option: pembrolizumab—26 days (IQR, 16–61), nivolumab—23 days (IQR, 14–36), nivolumab plus ipilimumab—19 days (IQR, 12–32), sintilimab—21 days (IQR, 21–37.5), camrelizumab—25 days (IQR, 14–39.5), durvalumab—34 days (IQR, 26–45.5), toripalimab—20 days (IQR, 14–29.5), atezolizumab—14 days (IQR, 4–30); tislelizumab—20 days (IQR, 11–20) (Table 4).
In six of the analyzed works, there was no description of symptoms. The manifestation of ICI-associated myocarditis is characterized by a significant heterogeneity of symptoms. Depending on the prevailing type of symptoms, patients were divided into cases with predominantly cardiac symptoms (shortness of breath, chest pain, palpitations, heart rhythm disturbances, etc.), neuromuscular symptoms (muscle weakness, double vision, drooping eyelids, etc.), and other manifestations (fever, rash, diarrhea, etc.). Cardiac symptoms bothered patients in 47.1% of cases, neuromuscular symptoms in 30.3%, and other symptoms in 12.6%. It is noteworthy that in 10.1% of cases, ICI-associated myocarditis began asymptomatic and was diagnosed based on laboratory and/or ECG and/or cardiac MRI signs.
We conducted an analysis of the ECG changes associated with the development of ICI-associated myocarditis (Table 5). In 43 cases, there was no description of ECG; therefore, the evaluation was performed based on the data of 201 cases.
In total, 85.1% of patients had new ECG changes compared to before starting ICI. Various arrhythmias, conduction disturbances, ST segment changes, T wave inversion, new Q waves, and low voltage in multiple leads were recorded. Conduction disturbances predominated, including atrioventricular block of varying degrees and bundle branch blocks. Atrioventricular block was registered in 35.4% of cases, with complete atrioventricular block being the most common type. Bundle branch block was represented by left bundle branch block (6%) and right bundle branch block (18.4%). Arrhythmias manifested as supraventricular and ventricular variants. Among the supraventricular variants, sinus tachycardia was the most common type (12%). Among the cases analyzed, 13% of patients developed ventricular tachycardia. In addition to the described changes, changes in the ST segment were registered in 12.4% of cases. The detection of such changes was the basis for differential diagnosis with acute coronary syndrome and coronary angiography was performed. In all cases, no obstructive changes were detected in the coronary arteries, and subsequent dynamics of symptoms and laboratory parameters, as well as additional investigations (MRI and endomyocardial biopsy), allowed the exclusion of acute forms of ischemic heart disease. Low voltages were observed in several leads in 4% of cases, which was associated with pericardial effusion. No pathological changes in the ECG compared to the initial changes were recorded in 14.9% of patients, and the diagnosis of myocarditis was confirmed based on the clinical picture and changes in the laboratory parameters and/or MRI and/or myocardial biopsy data.
The results of EchoCG were not described in 53 cases. Despite the severe course of ICT-associated myocarditis, only 50 patients (26.2%) among the presented clinical cases had a violation of the contractile ability of the left ventricular myocardium with a decrease in the ejection fraction of <40%. In 12 cases (6%), the ejection fraction was moderately reduced (40–49%). In addition, in 129 cases (64.2%), a preserved ejection fraction was diagnosed.
The assessment of myocardial damage markers (troponin and creatine kinase (CK), CK-MB), markers of heart failure development (brain natriuretic peptide (BNP) or N-terminal probrain natriuretic peptide (NT-proBNP)) was not presented in all of the articles. The troponin level was presented in 197 clinical cases, CK-MB and/or CK in 174, and BNP or NT-proBNP in only 80. Among them, an increased level of troponin was detected in 93.9% of cases (n = 185), an increase in CK and/or CK-MB was noted in 93.7% of patients (n = 163), and an increase in BNP or NT-proBNP in 77.5% (n = 185).
Data on cardiac MRI were presented in 75 clinical cases, but in many studies, there was no clear information that would reliably confirm the diagnosis of myocarditis. In four cases, there were indications that MRI did not reveal any signs of the current inflammatory processes in the myocardium, but the diagnosis of myocarditis was confirmed during morphological examination, taking into account the clinical picture, laboratory changes, and instrumental studies.
Morphological examination of the myocardium was performed in 61 patients, and in other scientific works, there is no information about the biopsy performed. In eight cases, the inflammatory process in the myocardium was confirmed by autopsy examination. In most cases, a lymphocytic infiltrate was described (which was positive for T-cell markers such as CD4 and CD8) with positive CD68 macrophages.
In 103 cases (42.2%), in addition to myocardial damage, patients were diagnosed with myositis and/or myasthenia. Mortality in the group of patients with ICI-associated myocarditis and myositis/myasthenia in our analysis was 29.4%, which exceeded the mortality rates in the group of patients without myositis and/or myasthenia (23.9%).
The first line of therapy typically includes glucocorticosteroids (GCs), which are widely used to treat autoimmune diseases, cytokine storms, and can be effective in the managing of relief complications caused by checkpoint inhibitors. In 10 articles, there was no detailed description of the therapy performed. In 97% of cases, the patients received therapy with prednisolone or methylprednisolone. As a second line of therapy, 22.6% of the patients received intravenous immunoglobulin (IVIG) infusions and 3.4% received antithymocyte immunoglobulin infusions. Immunosuppressive drugs, such as mycophenolate mofetil (11.5%), infliximab (6.8%), tofacitinib (5.1%), abatacept (2.6%), rituximab (1.4%), methotrexate (0.9%), cyclophosphamide (0.9%), tocilizumab (0.4%), alemtuzumab (0.4%), ruxolitinib (0.4%), and tacrolimus (0.4%), were less frequently prescribed. Plasmapheresis was performed in 9% of cases, ECMO (extracorporeal membrane oxygenation) in 1.7%, temporary or permanent cardio stimulator was mentioned in 19.7% of cases, and intra-aortic balloon counterpulsation was performed in 2.6% of cases for life-saving indications.

3.2. Deceased Patients

Despite undergoing therapy, the lethality rate from ICI-associated myocarditis was 26.2%. In the subgroup of patients who died as a result of ICI-associated myocarditis, 42 (65.6%) were male, with a median age of 69.5 (IQR, 63–77) years. The age at which the first symptoms of the disease developed in women was 66 (IQR, 57–70.5) years, which was lower than that of men at 73 (IQR, 66–79.75) years, as well as among all the patients included in the analysis.
In the subgroup of deceased patients, checkpoint inhibitors were prescribed to treat the following oncological diseases: melanoma in 17 cases (26.6%), lung cancer in 12 (18.7%), thymoma in 8 (12.5%), renal cell carcinoma in 5 (7.8%), urothelial carcinoma in 3 (4.7%), and other locations each in 1–2 described cases—total in 19 (29.7%).
Upon evaluating the therapy being administered, it was found that fatal outcomes occurred in 82.8% of ICI-associated myocarditis cases against the background of monotherapy with drugs from the anti-PD-1 group (primarily pembrolizumab (32.8%) and nivolumab (26.6%)). Other treatment options were significantly less common; combination therapy with anti-CTLA4 and anti-PD-1 was used in 1.6% of cases, monotherapy with anti-CTLA-4 in 3.1%, and anti-PD-L1 in 3.1%. Thus, it can be assumed that there is the highest risk of an unfavorable outcome in the group receiving anti-PD-1 therapy, especially with pembrolizumab and nivolumab. However, it cannot be ignored that the obtained results are due to the higher frequency of the use of these drugs in real practice.
The median time from the beginning of ICI therapy to the development of the first symptoms from the cardiovascular system was 15.5 days (IQR, 12–28.5), and the median day of death after the first use of ICI was 31 days (IQR, 19–54).
In one case, the patient’s complaints were not described. Among the remaining 63 cases, cardiac manifestations at the time of onset of the disease were registered in 50.8% of patients, neuromuscular in 30.2%, and other manifestations in 12.7%. In 6.3% of cases, the disease started asymptomatically.
In seven of the analyzed cases, the results of the ECG were not described, so the frequency of occurrence of various types of rhythm and conduction disorders was evaluated in 57 patients. In 96.5% of cases, new changes in the ECG were registered compared to the data before the start of ICI use. As with all the analyzed patients, conduction disturbances predominated, including atrioventricular block of different degrees and bundle branch blocks. In 40.4% of cases, a third-degree atrioventricular block was registered, but less frequently (first degree in 7% and second degree in 3.5% of cases). Left bundle branch block was detected in 8.8% of cases, and right bundle branch block in 28.1%. Among the supraventricular arrhythmia types, sinus tachycardia (12.3%), atrial fibrillation (7%), and atrial flutter (1.8%) were described. Ventricular tachycardia developed in 17.5% of patients. Premature atrial contractions (1.8%) and premature ventricular contractions (7%) were infrequently registered. ST segment elevation in 22.7% of cases, ST segment depression in 3.5%, and T wave inversion in 5.3% were also described. In one case (1.8%), the appearance of a Q wave was observed. Low voltages in several leads were described in 3.5% of cases. Pathological changes in the ECG compared to the initial data were not detected in 3.5% of patients.
According to EchoCG data, which was described in 51 cases, decreased ejection fraction was detected in 29.4% of cases, moderately decreased in 17.6%, and preserved in 52.9%. Laboratory markers of myocardial damage were not mentioned in any of the analyzed scientific works. The level of CK and/or CK-MB was evaluated in 50 cases and was elevated in 94%. The level of troponins was evaluated in 55 cases and was elevated in 92.7%. The evaluation of BNP or NT-proBNP was indicated in 20 articles, and its increase was registered in 85%.
Cardiac MRI was performed in 20.3% of cases and endomyocardial biopsy in 29.7% (of which autopsy was performed in 12.5%).
Myositis and/or myasthenia developed in 46.9% of patients.
Therapy for ICI-associated myocarditis was described in 60 patients. Glucocorticoid therapy was performed in almost all cases (98.3%). IVIG was used in 26.7% of cases, and antithymocyte immunoglobulin infusions in 5%. The following drugs were also mentioned for treatment: mycophenolate mofetil (5%), infliximab (18.3%), tofacitinib (6.3%), rituximab (1.7%) and cyclophosphamide (1.7%). Plasmapheresis was performed in 10% of cases, ECMO in 1.7%, and in 28.1% of cases, a cardio stimulator was required.

4. Discussion

Cardiotoxicity, although a rare side effect of ICI inhibitors, is concerning due to its high mortality rate and early development that often occurs soon after the start of treatment. While most cases occur soon after the start of ICI treatment—with a median of 21 days (IQR, 14–38.3)—there are several cases that manifest significantly later. Based on this information, clinicians should pay particular vigilance to cardiac toxicity in patients soon after the start of treatment. For example, according to our analysis, nearly one third of patients developed myocarditis after the first use of ICI. Myocarditis develops more quickly in the Atezolizumab group—14 days (IQR, 4–30)—which is also confirmed by Shalata W et al. [197], but with the smaller number of patients in this subgroup, this issue should be studied in prospective studies. However, it should be remembered that the risk of cardiotoxicity never completely disappears, and patients may need to be monitored for suspicious symptoms even years after the start of ICI treatment.
Myocarditis has the highest mortality rate among all types of ICI-induced cardiotoxicity. However, if they do not progress to a fatal outcome, these side effects are often quickly treated with steroid treatment [194]. According to our data, the mortality rate from ICI-associated myocarditis was 26.2%. This index is at the lower end of those mentioned in earlier scientific works. Perhaps this is due to the large number of studies from 2021 and 2022 included in our review, when doctors were already alert to the possibility of ICI-associated myocarditis and, in cases of asymptomatic increase in cardiac-specific markers, stopped using the drug, conducted further testing of the patients, and provided necessary therapy, primarily with high doses of GCs.
The clinical presentation of ICI-associated myocarditis can vary from asymptomatic elevation of cardiac biomarkers (which corresponds to grade 1 severity) to severe decompensation with the involvement of target organs. Fulminant myocarditis is the most concerning, and cases are often reported in the literature. In these patients, myocarditis is often accompanied by serious arrhythmias and conduction disturbances, such as complete atrioventricular block and ventricular tachycardia. It should be noted that if patients with cardiac symptoms also have symptoms of another irAE, the possibility of myocarditis associated with ICI increases [198]. Myocarditis is most often associated with myasthenia and myositis in such cases. This is likely due to the antigenic relationship between cardiomyocytes and skeletal muscles.
There are several scientific works that describe the overlap of ICI-associated myocarditis with myositis and/or myasthenia, mainly in the form of case reports or small case series; the frequency of concomitant myositis and myocarditis is 30–40% and myasthenia and myocarditis is 10% [32,199,200]. According to our data, myositis and/or myasthenia developed in 46.9% of patients, and in 30.3% of cases, complaints related to the nervous system or muscle involvement were the first manifestation of the disease that prompted patients to ask for medical help. Given the relatively high incidence, patients with immune-mediated myositis and/or myocarditis should be promptly screened for myasthenia, considering the risk of myasthenic crisis and adverse outcomes, such as respiratory paralysis and death [40], through the analysis of repetitive nerve stimulation and analysis of autoantibodies against the acetylcholine receptor. However, they are not detected in all patients (only approximately 60%) [40]. The mortality rate in the group of patients with ICI-associated myocarditis and myositis/myasthenia, according to our analysis, was 29.4%, which exceeded the mortality rate in the group of patients without myositis and/or myasthenia, which was 23.9%. These indexes are lower than those previously described; Pathak R et al. found significant in-hospital mortality in cases of cross-involvement, with mortality rates approaching 60% [199]. In this situation, we can also suggest that doctors were more vigilant and used treatment methods that have previously shown high effectiveness, such as the addition of steroids, IVIG and plasmapheresis of other immunosuppressants such as tacrolimus, infliximab, mycophenolate mofetil, or antithymocyte globulin.
The diagnostics of ICI-associated myocarditis involves the use of ECG, evaluation of troponin, BNP or NT-proBNP, and echocardiograms. Depending on the indications, stress tests, coronary angiography, and cardiac MRI may be performed [16].
Overall, the diagnostics strategy of ICI-associated myocarditis is potentially more complex than the diagnostics strategy for myocarditis of another etiology. Its impact on patient treatment outcomes is significant, considering its consequences. Under-diagnosis may lead to the absence or delay of corticosteroid therapy and major adverse cardiac events (MACE). Overdiagnosis may lead to the discontinuation of ICI treatment and progression of cancer. Bonaca et al. proposed a unified definition of cancer therapy-related myocarditis [201]. According to this definition, we can speak of definite myocarditis in three situations. The first variant is associated with signs of myocarditis that are found during histological examination (myocardial biopsy or autopsy). The second variant is associated with typical changes in cardiac MRI, along with an increase in the markers of myocardial necrosis in the blood or in combination with typical changes on an ECG. In the third variant, abnormal movement of the myocardial walls is established during echocardiography, which cannot be explained by other causes in combination with clinical symptoms and an increase in markers of myocardial necrosis and changes on an ECG in the absence of changes during angiography (or other techniques that confirm the absence of obstructive coronary artery disease).
Probable myocarditis is confirmed in the following scenarios. For first variant, typical changes in cardiac MRI are not accompanied by a typical clinical picture, an increase in the markers of myocardial necrosis, or changes on an ECG. Usually, such changes are detected accidentally or as part of clinical research procedures. For the second variant, non-specific but suspicious myocarditis changes are detected on cardiac MRI, in combination with a typical clinical picture and/or an increase in the markers of myocardial necrosis and/or changes on an ECG. In the third variant, abnormal movement of the myocardial walls is established during EchoCG in combination with clinical symptoms of myocarditis and an increase in the markers of myocardial necrosis in the blood and changes on an ECG. In the fourth variant, suspicious changes are detected on PET-CT.
Possible myocarditis is established in the following variants. For the first variant, non-specific but suspicious changes in myocarditis are detected on cardiac MRI without a typical clinical picture, in addition to an increase in the markers of necrosis, or changes on an ECG. The second variant is the detection of abnormal movement of the myocardial walls during echoCG, in combination with changes on an ECG or clinical symptoms of myocarditis. The third variant is an increase in the markers of necrosis compared to the baseline level in combination, with clinical symptoms of myocarditis or typical changes on an ECG.
However, these recommendations are primarily intended for use in clinical trials [202]. Another definition of immune checkpoint inhibitor myocarditis has recently been proposed by the International Cardio-Oncology Society [203]. According to this definition, the diagnosis of myocarditis can be established in the following situations. The first variant is histological confirmation based on myocardial biopsy or autopsy data. The second variant is a clinically confirmed diagnosis in combination with elevated cardiac troponins and typical changes in cardiac MRI. The third variant is a clinically confirmed diagnosis in combination with elevated cardiac troponins with two of the following listed features: typical clinical symptoms, lower extremity edema, ventricular arrhythmia or new conduction disturbance, decreased ejection fraction, other irAEs and suspicious myocarditis changes in cardiac MRI.
Criteria for complete recovery and ongoing recovery have also been developed [204].
Complete recovery is associated with patients with a complete resolution of acute symptoms, normalization of biomarkers, and the recovery of ejection fraction after the discontinuation of immunosuppression. Cardiac MRI may still show late gadolinium enhancement or elevated T1 due to fibrosis, but any suggestion of acute oedema should be absent.
MRI is the gold standard for the non-invasive diagnostics of myocarditis. According to our analysis, cardiac MRI was performed in only 75 cases (30.7%), but in many scientific works, specific data confirming or excluding myocarditis according to the existing criteria [205] are absent. Myocardial biopsy as part of the diagnostics of ICI-associated myocarditis cannot always be performed due to the severity of the patient’s condition, patient refusal, or, conversely, the absence of serious clinical symptoms. According to our data, it was performed in 61 cases (25%). In addition, it cannot be excluded that these diagnostic methods were not needed or were uninformative due to the positive effect of the conducted GC therapy.
Cardiac troponins are sensitive and are specific markers of myocardial damage. In patients with clinical manifestations that indicate acute coronary syndrome, coronary angiography may be required for the purpose of differential diagnosis. In a previous case–control study, troponin was elevated in 94% of myocarditis cases associated with ICI, while BNP or NT-proBNP was elevated in 66% of cases [30]. In our research, troponin levels were elevated in 93.9% of cases, and CK and/or CK-MB levels were elevated in 93.7% of cases. Most patients that were suspected of having ACS underwent coronary angiography. No obstructive changes in the coronary arteries were detected in any of the cases. However, it should be noted that in a small percentage of cases, the markers of myocardial damage may be normal. NT-proBNP was elevated in 77.5% of cases according to our data, but its elevation is also possible in many cancer patients due to inflammation associated with cancer; therefore, the result is non-specific.
EchoCG allows us to evaluate the structural and functional changes provoked by myocarditis. In our research, the majority of patients—64.2%—showed preserved ejection fraction, even in the group of deceased patients (52.9%). However, it is possible to suggest that death occurred before the development of cardiac dysfunction in these cases. Ejection fraction of less than 40% was detected in only 26.2% of all patients and 29.4% of deceased patients according to our analysis. However, EchoCG allows us to evaluate the dynamics of functional parameters against the background of the conducted therapy. In addition, EchoCG can show changes in the heart chambers, geometry, or regional wall motion abnormalities. At the same time, the preservation of normal heart size may indicate an acute process, while remodeling and dilation indicate a chronic process.
ICI-associated myocarditis in most cases is accompanied by changes on an ECG. According to our data, ECG changes were detected in 85.1% of patients, which were not present before the use of ICI. Conduction disorders predominated, with complete atrioventricular block being registered in 25.4% and complete right bundle branch block in 18.4% of cases and among arrhythmias, the most common types of disturbances were sinus tachycardia (12%) and ventricular tachycardia (13%). The presence of a high frequency of serious conduction disturbances may be associated with T-cell-mediated cytotoxicity, which affects the cardiac conduction system. Histological studies have shown that ICI-mediated cardiomyocyte necrosis is characterized by the infiltration of CD4+ and CD8+ T-cells and is similar to the development of acute cardiac rejection after transplantation [205,206]. Lymphocytic infiltration may involve the sinoatrial and atrioventricular nodes [207].
The main treatment for ICI-associated cardiotoxicity involves the use of high doses of corticosteroids (1–2 mg/kg) [23,24]. As a rule, the duration of glucocorticoid use after the resolution of symptoms is at least 4–6 weeks [208]. If the effectiveness is insufficient, other drugs may be used, such as mycophenolate mofetil, infliximab, IV immunoglobulin, or antithymocyte globulin, but their use is not associated with the recommendations for the diagnostics and treatment of ICI-associated myocarditis [23]. According to our data, the most common treatment options were as follows: 97% of patients received therapy with prednisolone or methylprednisolone, 22.6% received IVIG infusions, 11.5% received mycophenolate mofetil, and 6.8% received infliximab.
Regarding the further use of ICI after the development of ICI-associated myocarditis, some scientific works recommend temporarily discontinuing ICI use even after minor cardiotoxicity; if there is evidence of severe toxicity, it should be permanently discontinued [209]. Among the clinical cases analyzed by us, it was only mentioned in two cases that ICI use was resumed later. However, ICI has a long half-life in the body, so discontinuing treatment will not lead to an immediate change in the biological activity of the drug [210]; this is a justification for the prolonged use of GCs.
How can we detect ICI-associated myocarditis in its early stage? In order to detect possible cardiotoxicity early, several studies recommend routine cardiac examination (testing for BNP or NT-proBNP, cardiac troponin, and ECG) before starting ICI treatment and during the first 1–4 cycles or up to 12 weeks of treatment [210,211,212,213]. The latest recommendations for cardio-oncology suggest the following scenario for detecting ICI-associated cardiotoxicity [213,214]: initial evaluation of ECG, BNP or NT-proBNP and cardiac troponin, followed by a reanalysis of ECG and cardiac troponins before the 2nd, 3rd, and 4th dose of the medication. If these tests do not show any changes, repeated examination should be performed after every three doses of the medication.

5. Conclusions

Immune checkpoint inhibitors can cause various side effects related to pathological autoimmunity. One of the rare side effects is cardiotoxicity manifested as myocarditis. Although these side effects are rare, they have a high mortality rate. When using ICI, particular attention should be paid to the early detection of possible cardiotoxicity, including analyzing the state and functions of the myocardium before treatment and in dynamics. The early detection of ICI-associated myocarditis and timely discontinuation of ICI use may lead to a more favorable outcome in such cases. The main proven treatment method is the use of high doses of steroids.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The author declares no conflict of interest.

References

  1. Sung, H.; Ferlay, J.; Siegel, R.L.; Laversanne, M.; Soerjomataram, I.; Jemal, A.; Bray, F. Global Cancer Statistics 2020: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries. CA Cancer J. Clin. 2021, 71, 209–249. [Google Scholar] [CrossRef] [PubMed]
  2. Decker, W.K.; da Silva, R.F.; Sanabria, M.H.; Angelo, L.S.; Guimarães, F.; Burt, B.M.; Kheradmand, F.; Paust, S. Cancer Immunotherapy: Historical Perspective of a Clinical Revolution and Emerging Preclinical Animal Models. Front. Immunol. 2017, 8, 829. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Fares, C.M.; Van Allen, E.M.; Drake, C.G.; Allison, J.P.; Hu-Lieskovan, S. Mechanisms of Resistance to Immune Checkpoint Blockade: Why Does Checkpoint Inhibitor Immunotherapy not Work for all Patients? Am. Soc. Clin. Oncol. Educ. Book 2019, 39, 147–164. [Google Scholar] [CrossRef] [PubMed]
  4. Dobosz, P.; Dzieciątkowski, T. The Intriguing History of Cancer Immunotherapy. Front. Immunol. 2019, 10, 2965. [Google Scholar] [CrossRef] [Green Version]
  5. Keam, S.J. Cadonilimab: First Approval. Drugs 2022, 82, 1333–1339. [Google Scholar] [CrossRef] [PubMed]
  6. Song, H.; Liu, X.; Jiang, L.; Li, F.; Zhang, R.; Wang, P. Current Status and Prospects of Camrelizumab, A Humanized Antibody Against Programmed Cell Death Receptor 1. Recent Patents Anti-Cancer Drug Discov. 2021, 16, 312–332. [Google Scholar] [CrossRef]
  7. Wang, J.; Su, S.; Li, J.; Li, Y. Efficacy and Safety of Camrelizumab Monotherapy and Combination Therapy for Cancers: A Systematic Review and Meta-Analysis. Front. Oncol. 2021, 11, 695512. [Google Scholar] [CrossRef] [PubMed]
  8. Investor.lilly. Available online: https://investor.lilly.com/news-releases/news-release-details/innovent-and-lilly-announce-successful-expansion-sintilimab (accessed on 10 February 2023).
  9. BeiGene. Available online: https://ir.beigene.com/news/china-nmpa-approves-tislelizumab-for-patients-with-second-line-esophageal-squamous-cell-carcinoma/50b0ad83-47cf-46e7-a6fd-53ccc04058f2/ (accessed on 13 February 2023).
  10. Zhang, L.; Hao, B.; Geng, Z.; Geng, Q. Toripalimab: The First Domestic Anti-Tumor PD-1 Antibody in China. Front. Immunol. 2022, 12, 730666. [Google Scholar] [CrossRef] [PubMed]
  11. Yan, J.; Zhang, Y.; Zhang, J.-P.; Liang, J.; Li, L.; Zheng, L. Tim-3 Expression Defines Regulatory T Cells in Human Tumors. PLoS ONE 2013, 8, e58006. [Google Scholar] [CrossRef] [Green Version]
  12. Huang, C.-T.; Workman, C.J.; Flies, D.; Pan, X.; Marson, A.L.; Zhou, G.; Hipkiss, E.L.; Ravi, S.; Kowalski, J.; Levitsky, H.I.; et al. Role of LAG-3 in Regulatory T Cells. Immunity 2004, 21, 503–513. [Google Scholar] [CrossRef] [Green Version]
  13. Yu, X.; Harden, K.; Gonzalez, L.C.; Francesco, M.; Chiang, E.; A Irving, B.; Tom, I.; Ivelja, S.; Refino, C.J.; Clark, H.; et al. The surface protein TIGIT suppresses T cell activation by promoting the generation of mature immunoregulatory dendritic cells. Nat. Immunol. 2008, 10, 48–57. [Google Scholar] [CrossRef] [PubMed]
  14. Fanale, D.; Corsini, L.R.; Brando, C.; Cutaia, S.; Di Donna, M.C.; Filorizzo, C.; Lisanti, M.C.; Randazzo, U.; Magrin, L.; Romano, R.; et al. Can circulating PD-1, PD-L1, BTN3A1, pan-BTN3As, BTN2A1 and BTLA levels enhance prognostic power of CA125 in patients with advanced high-grade serous ovarian cancer? Front. Oncol. 2022, 12, 946319. [Google Scholar] [CrossRef] [PubMed]
  15. Lines, J.L.; Pantazi, E.; Mak, J.; Sempere, L.F.; Wang, L.; O’Connell, S.; Ceeraz, S.; Suriawinata, A.A.; Yan, S.; Ernstoff, M.S.; et al. VISTA Is an Immune Checkpoint Molecule for Human T Cells. Cancer Res. 2014, 74, 1924–1932. [Google Scholar] [CrossRef] [Green Version]
  16. 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]
  17. Darnell, E.P.; Mooradian, M.J.; Baruch, E.N.; Yilmaz, M.; Reynolds, K.L. Immune-Related Adverse Events (irAEs): Diagnosis, Management, and Clinical Pearls. Curr. Oncol. Rep. 2020, 22, 39. [Google Scholar] [CrossRef] [PubMed]
  18. Zhang, L.; Reynolds, K.L.; Lyon, A.R.; Palaskas, N.; Neilan, T.G. The Evolving Immunotherapy Landscape and the Epidemiology, Diagnosis, and Management of Cardiotoxicity. JACC Cardio-Oncology 2021, 3, 35–47. [Google Scholar] [CrossRef] [PubMed]
  19. Waliany, S.; Lee, D.; Witteles, R.M.; Neal, J.W.; Nguyen, P.; Davis, M.M.; Salem, J.-E.; Wu, S.M.; Moslehi, J.J.; Zhu, H. Immune Checkpoint Inhibitor Cardiotoxicity: Understanding Basic Mechanisms and Clinical Characteristics and Finding a Cure. Annu. Rev. Pharmacol. Toxicol. 2021, 61, 113–134. [Google Scholar] [CrossRef]
  20. Michot, J.; Bigenwald, C.; Champiat, S.; Collins, M.; Carbonnel, F.; Postel-Vinay, S.; Berdelou, A.; Varga, A.; Bahleda, R.; Hollebecque, A.; et al. Immune-related adverse events with immune checkpoint blockade: A comprehensive review. Eur. J. Cancer 2016, 54, 139–148. [Google Scholar] [CrossRef]
  21. Cathcart-Rake, E.J.; Sangaralingham, L.R.; Henk, H.J.; Shah, N.D.; Bin Riaz, I.; Mansfield, A.S. A Population-based Study of Immunotherapy-related Toxicities in Lung Cancer. Clin. Lung Cancer 2020, 21, 421–427.e2. [Google Scholar] [CrossRef]
  22. Yu, Y.; Ruddy, K.J.; Tsuji, S.; Hong, N.; Liu, H.; Shah, N.; Jiang, G. Coverage Evaluation of CTCAE for Capturing the Immune-related Adverse Events Leveraging Text Mining Technologies. AMIA Jt. Summits Transl. Sci. proceedings. AMIA Jt. Summits Transl. Sci. 2019, 2019, 771–778. [Google Scholar]
  23. 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]
  24. Haanen, J.; Carbonnel, F.; Robert, C.; Kerr, K.; Peters, S.; Larkin, J.; Jordan, K. Corrections to “Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann. Oncol. 2018, 29, iv264–iv266. [Google Scholar] [CrossRef]
  25. Schneider, B.J.; Naidoo, J.; Santomasso, B.D.; Lacchetti, C.; Adkins, S.; Anadkat, M.; Atkins, M.B.; Brassil, K.J.; Caterino, J.M.; Chau, I.; et al. Management of Immune-Related Adverse Events in Patients Treated With Immune Checkpoint Inhibitor Therapy: ASCO Guideline Update. J. Clin. Oncol. 2021, 39, 4073–4126. [Google Scholar] [CrossRef]
  26. National Cancer Institute. Common Terminology Criteria for Adverse Events (CTCAE) Common Terminology Criteria for Adverse Events (CTCAE) v5.0. 2017. Available online: https://www.meddra.org/ (accessed on 7 January 2023).
  27. Wang, D.Y.; Salem, J.-E.; Cohen, J.V.; Chandra, S.; Menzer, C.; Ye, F.; Zhao, S.; Das, S.; Beckermann, K.E.; Ha, L.; et al. Fatal Toxic Effects Associated With Immune Checkpoint Inhibitors. JAMA Oncol. 2018, 4, 1721–1728. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Varricchi, G.; Galdiero, M.R.; Marone, G.; Criscuolo, G.; Triassi, M.; Bonaduce, D.; Marone, G.; Tocchetti, C.G. Cardiotoxicity of immune checkpoint inhibitors. ESMO Open 2017, 2, e000247. [Google Scholar] [CrossRef] [Green Version]
  29. Johnson, D.B.; Balko, J.M.; Compton, M.L.; Chalkias, S.; Gorham, J.; Xu, Y.; Hicks, M.; Puzanov, I.; Alexander, M.R.; Bloomer, T.L.; et al. Fulminant Myocarditis with Combination Immune Checkpoint Blockade. N. Engl. J. Med. 2016, 375, 1749–1755. [Google Scholar] [CrossRef] [PubMed]
  30. Mahmood, S.S.; Fradley, M.G.; Cohen, J.V.; Nohria, A.; Reynolds, K.L.; Heinzerling, L.M.; Sullivan, R.J.; Damrongwatanasuk, R.; Chen, C.L.; Gupta, D.; et al. Myocarditis in Patients Treated With Immune Checkpoint Inhibitors. J. Am. Coll. Cardiol. 2018, 71, 1755–1764. [Google Scholar] [CrossRef]
  31. Ganatra, S.; Neilan, T.G. Immune Checkpoint Inhibitor-Associated Myocarditis. Oncologist 2018, 23, 879–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Moslehi, J.J.; Salem, J.-E.; A Sosman, J.; Lebrun-Vignes, B.; Johnson, D.B. Increased reporting of fatal immune checkpoint inhibitor-associated myocarditis. Lancet 2018, 391, 933. [Google Scholar] [CrossRef] [Green Version]
  33. Escudier, M.; Cautela, J.; Malissen, N.; Ancedy, Y.; Orabona, M.; Pinto, J.; Monestier, S.; Grob, J.-J.; Scemama, U.; Jacquier, A.; et al. Clinical Features, Management, and Outcomes of Immune Checkpoint Inhibitor–Related Cardiotoxicity. Circulation 2017, 136, 2085–2087. [Google Scholar] [CrossRef]
  34. Baik, A.H.; Oluwole, O.O.; Johnson, D.B.; Shah, N.; Salem, J.-E.; Tsai, K.K.; Moslehi, J.J. Mechanisms of Cardiovascular Toxicities Associated With Immunotherapies. Circ. Res. 2021, 128, 1780–1801. [Google Scholar] [CrossRef] [PubMed]
  35. Zhou, Y.-W.; Zhu, Y.-J.; Wang, M.-N.; Xie, Y.; Chen, C.-Y.; Zhang, T.; Xia, F.; Ding, Z.-Y.; Liu, J.-Y. Immune Checkpoint Inhibitor-Associated Cardiotoxicity: Current Understanding on Its Mechanism, Diagnosis and Management. Front. Pharmacol. 2019, 10, 1350. [Google Scholar] [CrossRef] [PubMed]
  36. Quagliariello, V.; Passariello, M.; Rea, D.; Barbieri, A.; Iovine, M.; Bonelli, A.; Caronna, A.; Botti, G.; De Lorenzo, C.; Maurea, N. Evidences of CTLA-4 and PD-1 Blocking Agents-Induced Cardiotoxicity in Cellular and Preclinical Models. J. Pers. Med. 2020, 10, 179. [Google Scholar] [CrossRef] [PubMed]
  37. Tay, W.T.; Fang, Y.-H.; Beh, S.T.; Liu, Y.-W.; Hsu, L.-W.; Yen, C.-J.; Liu, P.-Y. Programmed Cell Death-1: Programmed Cell Death-Ligand 1 Interaction Protects Human Cardiomyocytes Against T-Cell Mediated Inflammation and Apoptosis Response In Vitro. Int. J. Mol. Sci. 2020, 21, 2399. [Google Scholar] [CrossRef] [Green Version]
  38. Wang, J.; Okazaki, I.-M.; 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]
  39. 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] [Green Version]
  40. Huang, Y.-T.; Chen, Y.-P.; Lin, W.-C.; Su, W.-C.; Sun, Y.-T. Immune Checkpoint Inhibitor-Induced Myasthenia Gravis. Front. Neurol. 2020, 11, 634. [Google Scholar] [CrossRef]
  41. Schiopu, S.R.I.; Käsmann, L.; Schönermarck, U.; Fischereder, M.; Grabmaier, U.; Manapov, F.; Rauch, J.; Orban, M. Pembrolizumab-induced myocarditis in a patient with malignant mesothelioma: Plasma exchange as a successful emerging therapy—Case report. Transl. Lung Cancer Res. 2021, 10, 1039–1046. [Google Scholar] [CrossRef]
  42. Shirai, T.; Kiniwa, Y.; Sato, R.; Sano, T.; Nakamura, K.; Mikoshiba, Y.; Ohashi, N.; Sekijima, Y.; Okuyama, R. Presence of antibodies to striated muscle and acetylcholine receptor in association with occurrence of myasthenia gravis with myositis and myocarditis in a patient with melanoma treated with an anti–programmed death 1 antibody. Eur. J. Cancer 2019, 106, 193–195. [Google Scholar] [CrossRef]
  43. Nasr, F.; El Rassy, E.; Maalouf, G.; Azar, C.; Haddad, F.; Helou, J.; Robert, C. Severe ophthalmoplegia and myocarditis following the administration of pembrolizumab. Eur. J. Cancer 2017, 91, 171–173. [Google Scholar] [CrossRef]
  44. Lipe, D.N.; Galvis-Carvajal, E.; Rajha, E.; Wechsler, A.H.; Gaeta, S. Immune checkpoint inhibitor-associated myasthenia gravis, myositis, and myocarditis overlap syndrome. Am. J. Emerg. Med. 2021, 46, 51–55. [Google Scholar] [CrossRef]
  45. Konstantina, T.; Konstantinos, R.; Anastasios, K.; Anastasia, M.; Eleni, L.; Ioannis, S.; Sofia, A.; Dimitris, M. Fatal adverse events in two thymoma patients treated with anti-PD-1 immune check point inhibitor and literature review. Lung Cancer 2019, 135, 29–32. [Google Scholar] [CrossRef] [PubMed]
  46. Jeyakumar, N.; Etchegaray, M.; Henry, J.; Lelenwa, L.; Zhao, B.; Segura, A.; Buja, L.M. The Terrible Triad of Checkpoint Inhibition: 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, 5126717. [Google Scholar] [CrossRef]
  47. Hellman, J.B.; Traynis, I.; Lin, L.K. Pembrolizumab and epacadostat induced fatal myocarditis and myositis presenting as a case of ptosis and ophthalmoplegia. Orbit 2018, 38, 244–247. [Google Scholar] [CrossRef] [PubMed]
  48. 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]
  49. 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]
  50. 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 bevacizumab therapy for advanced hepatocellular carcinoma. Clin. J. Gastroenterol. 2021, 14, 1233–1239. [Google Scholar] [CrossRef]
  51. 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]
  52. Zhao, L.-Z.; Liu, G.; Li, Q.-F.; Chen, G.; Jin, G.-W. A case of carrelizumab-associated immune myocarditis. Asian J. Surg. 2021, 45, 496–497. [Google Scholar] [CrossRef]
  53. Mehta, A.; Gupta, A.; Hannallah, F.; Koshy, T.; Reimold, S. Myocarditis as an immune-related adverse event with ipilimumab/nivolumab combination therapy for metastatic melanoma. Melanoma Res. 2016, 26, 319–320. [Google Scholar] [CrossRef]
  54. Chen, Q.; Huang, D.-S.; Zhang, L.-W.; Li, Y.-Q.; Wang, H.-W.; Liu, H.-B. Fatal myocarditis and rhabdomyolysis induced by nivolumab during the treatment of type B3 thymoma. Clin. Toxicol. 2017, 56, 667–671. [Google Scholar] [CrossRef] [PubMed]
  55. 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] [PubMed]
  56. Salem, J.-E.; Allenbach, Y.; Vozy, A.; Brechot, N.; Johnson, D.B.; Moslehi, J.J.; Kerneis, M. Abatacept for Severe Immune Checkpoint Inhibitor–Associated Myocarditis. N. Engl. J. Med. 2019, 380, 2377–2379. [Google Scholar] [CrossRef]
  57. Huertas, R.M.; Serrano, C.S.; Perna, C.; Gómez, A.F.; Gordoa, T.A. Cardiac toxicity of immune-checkpoint inhibitors: A clinical case of nivolumab-induced myocarditis and review of the evidence and new challenges. Cancer Manag. Res. 2019, 11, 4541–4548. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Sakai, T.; Sasada, S.; Jyo, C.; Ishioka, K.; Takahashi, S.; Nakamura, M. Acute myocarditis and pericarditis after nivolumab treatment in patients with non-small cell lung cancer. Ann. Oncol. 2017, 28, ix90. [Google Scholar] [CrossRef]
  59. 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] [PubMed]
  60. Chauhan, A.; Burkeen, G.; Houranieh, J.; Arnold, S.; Anthony, L. Immune checkpoint-associated cardiotoxicity: Case report with systematic review of literature. Ann. Oncol. 2017, 28, 2034–2038. [Google Scholar] [CrossRef] [PubMed]
  61. Valenti-Azcarate, R.; Vazquez, I.E.; Illan, C.T.; Idoate-Gastearena, M.; Pérez-Larraya, J.G. Nivolumab and Ipilimumab-induced myositis and myocarditis mimicking a myasthenia gravis presentation. Neuromuscul. Disord. 2019, 30, 67–69. [Google Scholar] [CrossRef]
  62. 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] [Green Version]
  63. Semper, H.; Muehlberg, F.; Schulz-Menger, J.; Allewelt, M.; Grohé, C. Drug-induced myocarditis after nivolumab treatment in a patient with PDL1- negative squamous cell carcinoma of the lung. Lung Cancer 2016, 99, 117–119. [Google Scholar] [CrossRef]
  64. 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. 2017, 63, 954–957. [Google Scholar] [CrossRef]
  65. Duarte, T.; Costa, C.; Gonçalves, S.; Raposo, L.; Ferreira, A.; Albuquerque, C.; Vau, N.; Caria, R. A case of lymphocytic myocarditis in a patient treated with an immune checkpoint inhibitor, a recent class of chemotherapy agents. Rev. Port. de Cardiol. 2022, 41, 1047–1051. [Google Scholar] [CrossRef] [PubMed]
  66. Li, W.-L.; Zhang, Y.; Zhang, X.-L.; Du, Y.-Y.; Hu, W.-Q.; Zhao, J. Long-term survival after immunotherapy and targeted therapy without chemotherapy in an elderly patient with HER2-positive gastroesophageal junction cancer: A case report. Hum. Vaccines Immunother. 2022, 6, 2121109. [Google Scholar] [CrossRef]
  67. Kadowaki, H.; Akazawa, H. Sick Sinus Syndrome: More Than a Needle-in-a-haystack Manifestation of Immune Checkpoint Inhibitor-associated Myocarditis. Intern. Med. 2022, 61, 2101–2102. [Google Scholar] [CrossRef] [PubMed]
  68. Berg, D.D.; Vaduganathan, M.; Davids, M.S.; Alyea, E.P.; Torre, M.; Padera, R.F.; Nohria, A. Immune-related fulminant myocarditis in a patient receiving ipilimumab therapy for relapsed chronic myelomonocytic leukaemia. Eur. J. Heart Fail. 2017, 19, 682–685. [Google Scholar] [CrossRef] [Green Version]
  69. Bukamur, H.S.; Mezughi, H.; Karem, E.; Shahoub, I.; Shweihat, Y. Nivolumab-induced Third Degree Atrioventricular Block in a Patient with Stage IV Squamous Cell Lung Carcinoma. Cureus 2019, 11, e4869. [Google Scholar] [CrossRef] [Green Version]
  70. 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, 2539493. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Imai, R.; Ono, M.; Nishimura, N.; Suzuki, K.; Komiyama, N.; Tamura, T. Fulminant Myocarditis Caused by an Immune Checkpoint Inhibitor: A Case Report With Pathologic Findings. J. Thorac. Oncol. 2019, 14, e36–e38. [Google Scholar] [CrossRef] [Green Version]
  72. Thibault, C.; Vano, Y.; Soulat, G.; Mirabel, M. Immune checkpoint inhibitors myocarditis: Not all cases are clinically patent. Eur. Heart J. 2018, 38, 3553. [Google Scholar] [CrossRef]
  73. 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]
  74. Chang, A.; Nasti, T.H.; Khan, M.K.; Parashar, S.; Kaufman, J.L.; Boise, L.H.; Lonial, S.; Ahmed, R.; Nooka, A.K. Myocarditis with Radiotherapy and Immunotherapy in Multiple Myeloma. J. Oncol. Pract. 2018, 14, 561–564. [Google Scholar] [CrossRef]
  75. A Laine, G.; Allen, S.J. Left ventricular myocardial edema. Lymph flow, interstitial fibrosis, and cardiac function. Circ. Res. 1991, 68, 1713–1721. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Guo, C.W.; Alexander, M.; Dib, Y.; Lau, P.K.; Weppler, A.M.; Au-Yeung, G.; Lee, B.; Khoo, C.; Mooney, D.; Joshi, S.B.; et al. A closer look at immune-mediated myocarditis in the era of combined checkpoint blockade and targeted therapies. Eur. J. Cancer 2019, 124, 15–24. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  77. 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]
  78. Wang, F.; Sun, X.; Qin, S.; Hua, H.; Liu, X.; Yang, L.; Yang, M. A retrospective study of immune checkpoint inhibitor-associated myocarditis in a single center in China. Chin. Clin. Oncol. 2020, 9, 16. [Google Scholar] [CrossRef] [PubMed]
  79. 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]
  80. Sakai, S.; Tajiri, K.; Li, S.; Ieda, M. Fatal cerebral haemorrhagic infarction due to left ventricular thrombus after healing of immune checkpoint inhibitor-associated myocarditis. Eur. Heart J. Case Rep. 2020, 4, 1–2. [Google Scholar] [CrossRef]
  81. 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-Oncology 2020, 2, 511–514. [Google Scholar] [CrossRef] [PubMed]
  82. Yanase, T.; Moritoki, Y.; Kondo, H.; Ueyama, D.; Akita, H.; Yasui, T. Myocarditis and myasthenia gravis by combined nivolumab and ipilimumab immunotherapy for renal cell carcinoma: A case report of successful management. Urol. Case Rep. 2020, 34, 101508. [Google Scholar] [CrossRef]
  83. Yogasundaram, H.; Alhumaid, W.; Chen, J.W.; Church, M.; Alhulaimi, N.; Kimber, S.; Paterson, D.I.; Senaratne, J.M. Plasma Exchange for Immune Checkpoint Inhibitor–Induced Myocarditis. CJC Open 2020, 3, 379–382. [Google Scholar] [CrossRef] [PubMed]
  84. 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]
  85. 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] [PubMed]
  86. Waliany, S.; Neal, J.W.; Reddy, S.; Wakelee, H.; Shah, S.A.; Srinivas, S.; Padda, S.K.; Fan, A.C.; Colevas, A.D.; Wu, S.M.; et al. Myocarditis Surveillance With High-Sensitivity Troponin I During Cancer Treatment With Immune Checkpoint Inhibitors. JACC Cardio-Oncology 2021, 3, 137–139. [Google Scholar] [CrossRef] [PubMed]
  87. Baik, A.H.; Tsai, K.K.; Oh, D.Y.; Aras, M.A. Mechanisms and clinical manifestations of cardiovascular toxicities associated with immune checkpoint inhibitors. Clin. Sci. 2021, 135, 703–724. [Google Scholar] [CrossRef]
  88. Ida, M.; Nakamori, S.; Ishida, M.; Dohi, K. Management of immune checkpoint inhibitor myocarditis: A serial cardiovascular magnetic resonance T2 mapping approach. Eur. Heart J. 2021, 42, 2869. [Google Scholar] [CrossRef]
  89. Wang, F.; Liu, Y.; Xu, W.; Zhang, C.; Lv, J.; Ma, S. Fulminant myocarditis induced by immune checkpoint inhibitor nivolumab: A case report and review of the literature. J. Med. Case Rep. 2021, 15, 336. [Google Scholar] [CrossRef]
  90. Matzen, E.; Bartels, L.E.; Løgstrup, B.; Horskær, S.; Stilling, C.; Donskov, F. Immune checkpoint inhibitor-induced myocarditis in cancer patients: A case report and review of reported cases. Cardio-Oncology 2021, 7, 27. [Google Scholar] [CrossRef]
  91. Shen, L.; Chen, H.; Wei, Q. Immune-Therapy-Related Toxicity Events and Dramatic Remission After a Single Dose of Pembrolizumab Treatment in Metastatic Thymoma: A Case Report. Front. Immunol. 2021, 12, 621858. [Google Scholar] [CrossRef]
  92. 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] [PubMed]
  93. Shindo, A.; Yamasaki, M.; Uchino, K.; Yamasaki, M. Asymptomatic Myocarditis with Mild Cardiac Marker Elevation Following Nivolumab-Induced Myositis. Int. Heart J. 2022, 63, 180–183. [Google Scholar] [CrossRef]
  94. 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 adenocarcinoma. Hematol. Stem Cell Ther. 2022, 15, 63–67. [Google Scholar] [CrossRef] [PubMed]
  95. Yang, Y.; Wu, Q.; Chen, L.; Qian, K.; Xu, X. Severe immune-related hepatitis and myocarditis caused by PD-1 inhibitors in the treatment of triple-negative breast cancer: A case report. Ann. Transl. Med. 2022, 10, 424. [Google Scholar] [CrossRef]
  96. Maetani, T.; Hamaguchi, T.; Nishimura, T.; Marumo, S.; Fukui, M. Durvalumab-associated Late-onset Myocarditis Successfully Treated with Corticosteroid Therapy. Intern. Med. 2022, 61, 527–531. [Google Scholar] [CrossRef]
  97. Nishimura, T.; Ninomiya, K.; Nakashima, M.; Akagi, S.; Kuribayashi, T.; Higo, H.; Hotta, K.; Maeda, Y.; Ito, H.; Kiura, K. Fulminant Myocarditis for Non-small-cell Carcinoma of the Lung with Nivolumab and Ipilimumab Plus Chemotherapy: A Case Report. Intern. Med. 2022; 0505–0522, online ahead of print. [Google Scholar] [CrossRef]
  98. Liu, S.M.; Ma, G.M.; Wang, H.M.; Yu, G.M.; Chen, J.; Song, W.M. Severe cardiotoxicity in 2 patients with thymoma receiving immune checkpoint inhibitor therapy: A case report. Medicine 2022, 101, e31873. [Google Scholar] [CrossRef] [PubMed]
  99. Zhang, B.; Gyawali, L.; Liu, Z.; Du, H.; Yin, Y. Camrelizumab-Related Lethal Arrhythmias and Myasthenic Crisis in a Patient with Metastatic Thymoma. Case Rep. Cardiol. 2022, 2022, 4042909. [Google Scholar] [CrossRef] [PubMed]
  100. Arora, P.; Talamo, L.; Dillon, P.; Gentzler, R.D.; Millard, T.; Salerno, M.; Slingluff, C.L., Jr.; Gaughan, E.M. Severe combined cardiac and neuromuscular toxicity from immune checkpoint blockade: An institutional case series. Cardio-Oncology 2020, 6, 21. [Google Scholar] [CrossRef] [PubMed]
  101. Yin, B.; Xiao, J.; Wang, X.; Li, X.; Guan, Y.; Chen, J.; Han, P.; Li, K.; Wang, J. Myocarditis and myositis/myasthenia gravis overlap syndrome induced by immune checkpoint inhibitor followed by esophageal hiatal hernia: A case report and review of the literature. Front. Med. 2022, 9, 950801. [Google Scholar] [CrossRef]
  102. Bai, J.; Li, D.; Yang, P.; Xu, K.; Wang, Y.; Li, Q.; Liu, J.; Du, W.; Zhang, F.; Feng, R. Camrelizumab-Related Myocarditis and Myositis With Myasthenia Gravis: A Case Report and Literature Review. Front. Oncol. 2022, 11, 778185. [Google Scholar] [CrossRef]
  103. Wang, S.; Peng, D.; Zhu, H.; Min, W.; Xue, M.; Wu, R.; Shao, Y.; Pan, L.; Zhu, M. Acetylcholine receptor binding antibody–associated myasthenia gravis, myocarditis, and rhabdomyolysis induced by tislelizumab in a patient with colon cancer: A case report and literature review. Front. Oncol. 2022, 12, 1053370. [Google Scholar] [CrossRef]
  104. Lin, Y.; Yuan, X.; Chen, L. Immune myocarditis related to sintilimab treatment in a patient with advanced lung adenocarcinoma: A case report. Front. Cardiovasc. Med. 2022, 9, 955527. [Google Scholar] [CrossRef]
  105. Chahine, J.; Collier, P.; Maroo, A.; Tang, W.W.; Klein, A.L. Myocardial and Pericardial Toxicity Associated With Immune Checkpoint Inhibitors in Cancer Patients. JACC Case Rep. 2020, 2, 191–199. [Google Scholar] [CrossRef] [PubMed]
  106. Hu, Y.; Lu, W.; Tang, B.; Zhao, Z.; An, Z. Urinary incontinence as a possible signal of neuromuscular toxicity during immune checkpoint inhibitor treatment: Case report and retrospective pharmacovigilance study. Front. Oncol. 2022, 12, 954468. [Google Scholar] [CrossRef] [PubMed]
  107. 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-Oncology 2020, 2, 800–804. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, Y.; Huang, A.; Yang, Q.; Yu, J.; Li, G. Case report: A successful re-challenge report of GLS-010 (Zimberelimab), a novel fully humanized mAb to PD-1, in a case of recurrent endometrial cancer. Front. Immunol. 2022, 13, 987345. [Google Scholar] [CrossRef]
  109. Erritzøe-Jervild, M.; Scheie, D.; Stenør, C. Checkpoint inhibitor induced myositis–The value of MRI STIR. Eneurologicalsci 2023, 30, 100442. [Google Scholar] [CrossRef]
  110. Arponen, O.; Skyttä, T. Immune checkpoint inhibitor-induced myocarditis not visible with cardiac magnetic resonance imaging but detected with PET-CT: A case report. Acta Oncol. 2020, 59, 490–492. [Google Scholar] [CrossRef]
  111. Hu, X.; Wei, Y.; Shuai, X. Case Report: Glucocorticoid Effect Observation in a Ureteral Urothelial Cancer Patient With ICI-Associated Myocarditis and Multiple Organ Injuries. Front. Immunol. 2021, 12, 5314. [Google Scholar] [CrossRef]
  112. Martinez-Calle, N.; Rodriguez-Otero, P.; Villar, S.; Mejías, L.; Melero, I.; Prosper, F.; Marinello, P.; Paiva, B.; Idoate-Gastearena, M.; Miguel, J.S. Anti-PD1 associated fulminant myocarditis after a single pembrolizumab dose: The role of occult pre-existing autoimmunity. Haematologica 2018, 103, e318–e321. [Google Scholar] [CrossRef]
  113. Gibson, R.; DeLaune, J.; Szady, A.; Markham, M. Suspected autoimmune myocarditis and cardiac conduction abnormalities with nivolumab therapy for non-small cell lung cancer. BMJ Case Rep. 2016, 2016, bcr2016216228. [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]
  115. 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] [PubMed]
  116. Tan, J.L.; Mugwagwa, A.N.; Cieslik, L.; Joshi, R. Nivolumab-induced myocarditis complicated by complete atrioventricular block in a patient with metastatic non-small cell lung cancer. BMJ Case Rep. 2019, 12, e229963. [Google Scholar] [CrossRef] [PubMed]
  117. 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] [PubMed]
  118. Agrawal, N.; Khunger, A.; Vachhani, P.; Colvin, T.A.; Hattoum, A.; Spangenthal, E.; Curtis, A.B.; Dy, G.K.; Ernstoff, M.S.; Puzanov, I. Cardiac Toxicity Associated with Immune Checkpoint Inhibitors: Case Series and Review of the Literature. Case Rep. Oncol. 2019, 12, 260–276. [Google Scholar] [CrossRef]
  119. 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]
  120. Takai, M.; Kato, D.; Iinuma, K.; Maekawa, Y.M.; Nakane, K.; Tsuchiya, T.; Yokoi, S.; Koie, T. Simultaneous pembrolizumab-induced myasthenia gravis and myocarditis in a patient with metastatic bladder cancer: A case report. Urol. Case Rep. 2020, 31, 101145. [Google Scholar] [CrossRef]
  121. 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]
  122. Barham, W.; Guo, R.; Park, S.S.; Herrmann, J.; Dong, H.; Yan, Y. Case Report: Simultaneous Hyperprogression and Fulminant Myocarditis in a Patient With Advanced Melanoma Following Treatment With Immune Checkpoint Inhibitor Therapy. Front. Immunol. 2021, 11, 561083. [Google Scholar] [CrossRef]
  123. Lie, G.; Weickhardt, A.; Kearney, L.; Lam, Q.; John, T.; Liew, D.; Arulananda, S. Nivolumab resulting in persistently elevated troponin levels despite clinical remission of myocarditis and myositis in a patient with malignant pleural mesothelioma: Case report. Transl. Lung Cancer Res. 2020, 9, 360–365. [Google Scholar] [CrossRef]
  124. Gallegos, C.; Rottmann, D.; Nguyen, V.Q.; A Baldassarre, L. Myocarditis with checkpoint inhibitor immunotherapy: Case report of late gadolinium enhancement on cardiac magnetic resonance with pathology correlate. Eur. Heart J. Case Rep. 2019, 3, yty149. [Google Scholar] [CrossRef]
  125. Iniesta, M.S.; López, L.L.; Costa, F.C.; 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]
  126. Ji, H.; Wen, Z.; Liu, B.; Chen, H.; Lin, Q.; Chen, Z. Sintilimab induced ICIAM in the treatment of advanced HCC: A case report and analysis of research progress. Front. Immunol. 2022, 13, 995121. [Google Scholar] [CrossRef]
  127. Kimura, T.; Fukushima, S.; Miyashita, A.; Aoi, J.; Jinnin, M.; Kosaka, T.; Ando, Y.; Matsukawa, M.; Inoue, H.; Kiyotani, K.; et al. Myasthenic crisis and polymyositis induced by one dose of nivolumab. Cancer Sci. 2016, 107, 1055–1058. [Google Scholar] [CrossRef] [Green Version]
  128. Nierstedt, R.T.; Yeahia, R.; Barnett, K.M. Unanticipated Myocarditis in a Surgical Patient Treated With Pembrolizumab: A Case Report. A A Pract. 2020, 14, e01177. [Google Scholar] [CrossRef]
  129. Esfahani, K.; Buhlaiga, N.; Thébault, P.; Lapointe, R.; Johnson, N.A.; Miller, W.H., Jr. Alemtuzumab for Immune-Related Myocarditis Due to PD-1 Therapy. N. Engl. J. Med. 2019, 380, 2375–2376. [Google Scholar] [CrossRef] [PubMed]
  130. 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] [PubMed] [Green Version]
  131. Jespersen, M.S.; Fanø, S.; Stenør, C.; Møller, A.K. A case report of immune checkpoint inhibitor-related steroid-refractory myocarditis and myasthenia gravis-like myositis treated with abatacept and mycophenolate mofetil. Eur. Heart J. Case Rep. 2021, 5, ytab342. [Google Scholar] [CrossRef] [PubMed]
  132. 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] [Green Version]
  133. Chen, R.; Peng, L.; Qiu, Z.; Wang, Y.; Wei, F.; Zhou, M.; Zhu, F. Case Report: Cardiac Toxicity Associated With Immune Checkpoint Inhibitors. Front. Cardiovasc. Med. 2021, 8, 727445. [Google Scholar] [CrossRef]
  134. Yang, Z.-X.; Chen, X.; Tang, S.-Q.; Zhang, Q. Sintilimab-Induced Myocarditis Overlapping Myositis in a Patient With Metastatic Thymoma: A Case Report. Front. Cardiovasc. Med. 2021, 8, 1943. [Google Scholar] [CrossRef]
  135. Aghel, N.; Gustafson, D.; Di Meo, A.; Music, M.; Prassas, I.; Seidman, M.A.; Hansen, A.R.; Thavendiranathan, P.; Diamandis, E.P.; Delgado, D.; et al. Recurrent Myocarditis Induced by Immune-Checkpoint Inhibitor Treatment Is Accompanied by Persistent Inflammatory Markers Despite Immunosuppressive Treatment. JCO Precis. Oncol. 2021, 5, 485–491. [Google Scholar] [CrossRef] [PubMed]
  136. McDowall, L.M.; Fernando, S.L.; Ange, N.; Yun, J.; Chia, K.K. Immune checkpoint inhibitor–mediated myocarditis and ventricular tachycardia storm. Heart Case Rep. 2019, 5, 497–500. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Liang, S.; Yang, J.; Lin, Y.; Li, T.; Zhao, W.; Zhao, J.; Dong, C. Immune Myocarditis Overlapping With Myasthenia Gravis Due to Anti-PD-1 Treatment for a Chordoma Patient: A Case Report and Literature Review. Front. Immunol. 2021, 12, 682262. [Google Scholar] [CrossRef] [PubMed]
  138. Katano, H.; Kano, M.; Nakamura, T.; Kanno, T.; Asanuma, H.; Sata, T. A novel real-time PCR system for simultaneous detection of human viruses in clinical samples from patients with uncertain diagnoses. J. Med. Virol. 2010, 83, 322–330. [Google Scholar] [CrossRef] [PubMed]
  139. Matsui, H.; Kawai, T.; Sato, Y.; Ishida, J.; Kadowaki, H.; Akiyama, Y.; Yamada, Y.; Nakamura, M.; Yamada, D.; Akazawa, H.; et al. A Fatal Case of Myocarditis Following Myositis Induced by Pembrolizumab Treatment for Metastatic Upper Urinary Tract Urothelial Carcinoma. Int. Heart J. 2020, 61, 1070–1074. [Google Scholar] [CrossRef]
  140. Arangalage, D.; Pavon, A.G.; Özdemir, B.C.; Michielin, O.; Schwitter, J.; Monney, P. Acute cardiac manifestations under immune checkpoint inhibitors—Beware of the obvious: A case report. Eur. Heart J. Case Rep. 2021, 5, ytab262. [Google Scholar] [CrossRef]
  141. Yin, N.; Liu, X.; Ye, X.; Song, W.; Lu, J.; Chen, X. PD-1 inhibitor therapy causes multisystem immune adverse reactions: A case report and literature review. Front. Oncol. 2022, 12, 4695. [Google Scholar] [CrossRef]
  142. Wang, Q.; Hu, B. Successful therapy for autoimmune myocarditis with pembrolizumab treatment for nasopharyngeal carcinoma. Ann. Transl. Med. 2019, 7, 247. [Google Scholar] [CrossRef]
  143. Cohen, M.; Mustafa, S.; Elkherpitawy, I.; Meleka, M. A Fatal Case of Pembrolizumab-Induced Myocarditis in Non–Small Cell Lung Cancer. JACC Case Rep. 2020, 2, 426–430. [Google Scholar] [CrossRef]
  144. Sessums, M.; Yarrarapu, S.; Guru, P.K.; Sanghavi, D.K. Atezolizumab-induced myositis and myocarditis in a patient with metastatic urothelial carcinoma. BMJ Case Rep. 2020, 13, e236357. [Google Scholar] [CrossRef]
  145. Kushnareva, E.; Stepanova, M.; Artemeva, E.; Shuginova, T.; Kushnarev, V.; Simakova, M.; Moiseenko, F.; Moiseeva, O. Case Report: Multiple Causes of Cardiac Death After the First Infusion of Atezolizumab: Histopathological and Immunohistochemical Findings. Front. Immunol. 2022, 13, 871542. [Google Scholar] [CrossRef] [PubMed]
  146. Su, L.; Liu, C.; Wu, W.; Cui, Y.; Wu, M.; Chen, H. Successful Therapy for Myocarditis Concomitant With Complete Heart Block After Pembrolizumab Treatment for Head and Neck Squamous Cell Carcinoma: A Case Report With Literature Review. Front. Cardiovasc. Med. 2022, 9, 1161. [Google Scholar] [CrossRef] [PubMed]
  147. 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] [PubMed]
  148. 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]
  149. Monge, C.; Maeng, H.; Brofferio, A.; Apolo, A.B.; Sathya, B.; Arai, A.; Gulley, J.L.; Bilusic, M. Myocarditis in a patient treated with Nivolumab and PROSTVAC: A case report. J. Immunother. Cancer 2018, 6, 150. [Google Scholar] [CrossRef] [Green Version]
  150. 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, 1851. [Google Scholar] [CrossRef]
  151. Wang, C.; Lin, J.; Wang, Y.; Hsi, D.H.; Chen, J.; Liu, T.; Zhou, Y.; Ren, Z.; Zeng, Z.; Cheng, L.; et al. Case Series of Steroid-Resistant Immune Checkpoint Inhibitor Associated Myocarditis: A Comparative Analysis of Corticosteroid and Tofacitinib Treatment. Front. Pharmacol. 2021, 12, 770631. [Google Scholar] [CrossRef]
  152. Komatsu, M.; Hirai, M.; Kobayashi, K.; Hashidate, H.; Fukumoto, J.; Sato, A.; Usuda, H.; Tanaka, K.; Takahashi, K.; Kuwabara, S. A rare case of nivolumab-related myasthenia gravis and myocarditis in a patient with metastatic gastric cancer. BMC Gastroenterol. 2021, 21, 333. [Google Scholar] [CrossRef]
  153. Chen, Y.; Chen, Y.; Xie, J.; Liu, D.; Hong, X. Multisystem immune-related adverse events due to toripalimab: Two cases-based review. Front. Cardiovasc. Med. 2022, 9, 1036603. [Google Scholar] [CrossRef]
  154. Nishikawa, T.; Tamiya, M.; Ohta-Ogo, K.; Ikeda, Y.; Hatakeyama, K.; Honma, K.; Yasui, T.; Shioyama, W.; Oka, T.; Inoue, T.; et al. A Case of Lung Cancer with Very-Late-Onset Immune Checkpoint Inhibitor-Related Myocarditis. CJC Open 2022, 4, 651–655. [Google Scholar] [CrossRef]
  155. Deharo, F.; Carvelli, J.; Cautela, J.; Garcia, M.; Sarles, C.; de Paula, A.M.; Bourenne, J.; Gainnier, M.; Bichon, A. Immune Checkpoint Inhibitor-Induced Myositis/Myocarditis with Myasthenia Gravis-like Misleading Presentation: A Case Series in Intensive Care Unit. J. Clin. Med. 2022, 11, 5611. [Google Scholar] [CrossRef] [PubMed]
  156. Tsuruda, T.; Yoshikawa, N.; Kai, M.; Yamaguchi, M.; Toida, R.; Kodama, T.; Kajihara, K.; Kawabata, T.; Nakamura, T.; Sakata, 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]
  157. Bawek, S.J.; Ton, R.; McGovern-Poore, M.; Khoncarly, B.; Narvel, R. Nivolumab-Induced Myasthenia Gravis Concomitant With Myocarditis, Myositis, and Hepatitis. Cureus 2021, 13, e18040. [Google Scholar] [CrossRef] [PubMed]
  158. Chatzantonis, G.; Evers, G.; Meier, C.; Bietenbeck, M.; Florian, A.; Klingel, K.; Bleckmann, A.; Yilmaz, A. Immune Checkpoint Inhibitor-Associated Myocarditis. JACC Case Rep. 2020, 2, 630–635. [Google Scholar] [CrossRef]
  159. Lorente-Ros, Á.; Rajjoub-Al-Mahdi, E.-A.; Ruiz, J.M.M.; García, S.R.; Pérez, R.O.; Golfín, C.F.; Álvarez-García, J.; Gómez, J.L.Z. Checkpoint Immunotherapy-Induced Myocarditis and Encephalitis Complicated With Complete AV Block. JACC Case Rep. 2022, 4, 1032–1036. [Google Scholar] [CrossRef]
  160. Bi, H.; Ren, D.; Wang, Q.; Ding, X.; Wang, H. Immune checkpoint inhibitor-induced myocarditis in lung cancer patients: A case report of sintilimab-induced myocarditis and a review of the literature. Ann. Palliat. Med. 2021, 10, 793–802. [Google Scholar] [CrossRef] [PubMed]
  161. Hernández, A.P.; Clemente, M.B.; Garcίa, D.E.; Calvo, R.V.; Garcίa, B.N.; Domίnguez, J.F.O.; Antón, C.S.; García, M.M.; Cubero, J.S.; Domínguez, F. Checkpoint inhibitor-induced fulminant myocarditis, complete atrioventricular block and myasthenia gravis—A case report. Cardiovasc. Diagn. Ther. 2021, 11, 1013–1019. [Google Scholar] [CrossRef]
  162. Norwood, T.G.; Lenneman, C.A.; Westbrook, B.C.; Litovsky, S.H.; McKee, S.B.; Conry, R.M. Evolution of Immune Checkpoint Blockade–Induced Myocarditis Over 2 Years. JACC Case Rep. 2020, 2, 203–209. [Google Scholar] [CrossRef]
  163. Liu, Z.; Fan, Y.; Guo, J.; Bian, N.; Chen, D. Fulminant myocarditis caused by immune checkpoint inhibitor: A case report and possible treatment inspiration. ESC Heart Fail. 2022, 9, 2020–2026. [Google Scholar] [CrossRef]
  164. Nakagomi, Y.; Tajiri, K.; Shimada, S.; Li, S.; Inoue, K.; Murakata, Y.; Murata, M.; Sakai, S.; Sato, K.; Ieda, M. Immune Checkpoint Inhibitor-Related Myositis Overlapping With Myocarditis: An Institutional Case Series and a Systematic Review of Literature. Front. Pharmacol. 2022, 13, 884776. [Google Scholar] [CrossRef]
  165. Barry, T.; Gallen, R.; Freeman, C.; Agasthi, P.; Pedrotty, D.; Yang, M.; Jokerst, C.E.; Mookadam, F.; Hardaway, B.W.; LeMond, L.; et al. Successful Treatment of Steroid-Refractory Checkpoint Inhibitor Myocarditis with Globulin Derived-Therapy: A Case Report and Literature Review. Am. J. Med. Sci. 2021, 362, 424–432. [Google Scholar] [CrossRef] [PubMed]
  166. Ye, Y.; Li, Y.; Zhang, S.; Han, G. Teriprizumab-induced myocarditis in a patient with cholangiocarcinoma: A case report. J. Int. Med. Res. 2022, 50, 03000605221133259. [Google Scholar] [CrossRef] [PubMed]
  167. Inayat, F.; Masab, M.; Gupta, S.; Ullah, W. New drugs and new toxicities: Pembrolizumab-induced myocarditis. BMJ Case Rep. 2018, 2018, bcr-2017. [Google Scholar] [CrossRef]
  168. Xing, Q.; Zhang, Z.; Zhu, B.; Lin, Q.; Shen, L.; Li, F.; Xia, Z.; Zhao, Z. Case Report: Treatment for steroid-refractory immune-related myocarditis with tofacitinib. Front. Immunol. 2022, 13, 944013. [Google Scholar] [CrossRef] [PubMed]
  169. Doms, J.; Prior, J.; Peters, S.; Obeid, M. Tocilizumab for refractory severe immune checkpoint inhibitor-associated myocarditis. Ann. Oncol. 2020, 31, 1273–1275. [Google Scholar] [CrossRef]
  170. Hu, C.; Zhao, L.; Zhou, C.; Wang, H.; Jiang, S.; Li, Y.; Peng, Y.; Deng, C.; Ma, F.; Pan, Y.; et al. Pacemakers and methylprednisolone pulse therapy in immune-related myocarditis concomitant with complete heart block. Open Med. 2022, 17, 2109–2116. [Google Scholar] [CrossRef]
  171. Matsuo, K.; Ishiguro, T.; Najama, T.; Shimizu, Y.; Kobayashi, Y.; Mutou, M. Nivolumab-induced Myocarditis Successfully Treated with Corticosteroid Therapy: A Case Report and Review of the Literature. Intern. Med. 2019, 58, 2367–2372. [Google Scholar] [CrossRef] [Green Version]
  172. Naganuma, K.; Horita, Y.; Matsuo, K.; Miyama, Y.; Mihara, Y.; Yasuda, M.; Nakano, S.; Hamaguchi, T. An Autopsy Case of Late-onset Fulminant Myocarditis Induced by Nivolumab in Gastric Cancer. Intern. Med. 2022, 61, 2867–2871. [Google Scholar] [CrossRef]
  173. Läubli, H.; Balmelli, C.; Bossard, M.; Pfister, O.; Glatz, K.; Zippelius, A. Acute heart failure due to autoimmune myocarditis under pembrolizumab treatment for metastatic melanoma. J. Immunother. Cancer 2015, 3, 11. [Google Scholar] [CrossRef] [Green Version]
  174. 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 therapy for metastatic renal cell carcinoma: A case report. J. Med. Case Rep. 2021, 15, 508. [Google Scholar] [CrossRef]
  175. 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]
  176. Delgado-Lazo, V.; Abdelmottaleb, W.; Popescu-Martinez, A. Pembrolizumab-Induced Myocarditis and Pancreatitis in a Patient With Colon Cancer: A Case Report. Cureus 2022, 14, e26034. [Google Scholar] [CrossRef]
  177. Mahmood, S.S.; Chen, C.L.; Shapnik, N.; Krishnan, U.; Singh, H.S.; Makker, V. Myocarditis with tremelimumab plus durvalumab combination therapy for endometrial cancer: A case report. Gynecol. Oncol. Rep. 2018, 25, 74–77. [Google Scholar] [CrossRef] [PubMed]
  178. Ono, R.; Iwai, Y.; Yamazaki, T.; Takahashi, H.; Hori, Y.; Fukushima, K.; Saotome, T. Nivolumab-induced Myositis and Myocarditis with Positive Anti-titin Antibody and Anti-voltage-gated Potassium Channel Kv1.4 Antibody. Intern. Med. 2022, 61, 2973–2979. [Google Scholar] [CrossRef] [PubMed]
  179. 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] [Green Version]
  180. Onderko, L.L.; Heinrich, R.; Gosling, K.; Downs, T.; Afari, M.E. Myocarditis Following Immune Checkpoint Inhibition With Pembrolizumab: Management in a Context of Steroid Intolerance. CJC Open 2022, 4, 854–857. [Google Scholar] [CrossRef]
  181. Tamura, Y.; Tamura, Y.; Taniguchi, H.; Imanaka-Yoshida, K. Acute Myocarditis After Discontinuation of Immune Checkpoint Inhibitor Therapy. Circ. J. 2023, 87, 376. [Google Scholar] [CrossRef]
  182. Nishikawa, T.; Kunimasa, K.; Ohta-Ogo, K.; Ikeda, Y.; Yasui, T.; Shioyama, W.; Oka, T.; Honma, K.; Hatakeyama, K.; Kumagai, T.; et al. Sinus Node Dysfunction Co-occurring with Immune Checkpoint Inhibitor-associated Myocarditis. Intern. Med. 2022, 61, 2161–2165. [Google Scholar] [CrossRef]
  183. Zomborska, E.; Kasperova, S.; Slopovsky, J.; Pazderova, N.; Kasperova, B.; Penz, P.; Nyitrayova, O.; Porsok, S.; Mladosievicova, B.; Mego, M. Fatal myocarditis after the first dose of nivolumab. Klin. Onkol. 2022, 6, 486–492. [Google Scholar] [CrossRef]
  184. Czarnecka, A.M.; Kleibert, M.; Płachta, I.; Rogala, P.; Wągrodzki, M.; Leszek, P.; Rutkowski, P. Myocarditis Induced by Immunotherapy in Metastatic Melanoma—Review of Literature and Current Guidelines. J. Clin. Med. 2022, 11, 5182. [Google Scholar] [CrossRef]
  185. Finke, D.; Heckmann, M.B.; Herpel, E.; Katus, H.A.; Haberkorn, U.; Leuschner, F.; Lehmann, L.H. Early Detection of Checkpoint Inhibitor-Associated Myocarditis Using 68Ga-FAPI PET/CT. Front. Cardiovasc. Med. 2021, 8, 614997. [Google Scholar] [CrossRef] [PubMed]
  186. Ramayya, T.; Mitchell, J.D.; Hartupee, J.C.; Lavine, K.; Ridley, C.H.; Kotkar, K.D.; Jimenez, J.; Lin, C.-Y.; Alvarez-Cardona, J.A.; Krone, R.K.; et al. Delayed Diagnosis and Recovery of Fulminant Immune Checkpoint Inhibitor-Associated Myocarditis on VA-ECMO Support. JACC Cardio-Oncology 2022, 4, 722–726. [Google Scholar] [CrossRef] [PubMed]
  187. Cham, J.; Ng, D.; Nicholson, L. Durvalumab-induced myocarditis, myositis, and myasthenia gravis: A case report. J. Med. Case Rep. 2021, 15, 278. [Google Scholar] [CrossRef]
  188. Rhee, J.Y.; Torun, N.; Neilan, T.G.; Guidon, A.C. Consider Myocarditis When Patients Treated with Immune Checkpoint Inhibitors Present with Ocular Symptoms. Oncologist 2022, 27, e402–e405. [Google Scholar] [CrossRef]
  189. Osinga, T.E.; Oosting, S.F.; van der Meer, P.; de Boer, R.A.; Kuenen, B.C.; Rutgers, A.; Bergmann, L.; Munnink, T.H.O.; Jalving, M.; van Kruchten, M. Immune checkpoint inhibitor–associated myocarditis. Neth. Heart J. 2022, 30, 295–301. [Google Scholar] [CrossRef]
  190. Nguyen, A.T.; Berry, G.J.; Witteles, R.M.; Le, D.T.; Wu, S.M.; Fisher, G.A.; Zhu, H. Late-Onset Immunotherapy-Induced Myocarditis 2 Years After Checkpoint Inhibitor Initiation. JACC Cardio-Oncology 2022, 4, 727–730. [Google Scholar] [CrossRef]
  191. Ben Zadok, O.I.; Ben-Avraham, B.; Nohria, A.; Orvin, K.; Nassar, M.; Iakobishvili, Z.; Neiman, V.; Goldvaser, H.; Kornowski, R.; Ben Gal, T. Immune-Checkpoint Inhibitor-Induced Fulminant Myocarditis and Cardiogenic Shock. JACC Cardio-Oncology 2019, 1, 141–144. [Google Scholar] [CrossRef]
  192. 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 ruxolitinib: Proof of concept. J. Immunother. Cancer 2022, 10, e004699. [Google Scholar] [CrossRef] [PubMed]
  193. Kuhnly, N.M.; Coviello, J.S. Immune Checkpoint Inhibitor–Related Myocarditis: Recognition, Surveillance, and Management. Clin. J. Oncol. Nurs. 2022, 26, 54–60. [Google Scholar] [CrossRef]
  194. Chen, Y.; Jia, Y.; Liu, Q.; Shen, Y.; Zhu, H.; Dong, X.; Huang, J.; Lu, J.; Yin, Q. Myocarditis related to immune checkpoint inhibitors treatment: Two case reports and literature review. Ann. Palliat. Med. 2021, 10, 8512–8517. [Google Scholar] [CrossRef]
  195. 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]
  196. Heinzerling, L.; Ott, P.A.; Hodi, F.S.; Husain, A.N.; Tajmir-Riahi, A.; Tawbi, H.; Pauschinger, M.; Gajewski, T.F.; Lipson, E.J.; Luke, J.J. Cardiotoxicity associated with CTLA4 and PD1 blocking immunotherapy. J. Immunother. Cancer 2016, 4, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  197. Shalata, W.; Abu-Salman, A.; Steckbeck, R.; Jacob, B.M.; Massalha, I.; Yakobson, A. Cardiac Toxicity Associated with Immune Checkpoint Inhibitors: A Systematic Review. Cancers 2021, 13, 5218. [Google Scholar] [CrossRef] [PubMed]
  198. Koelzer, V.H.; Rothschild, S.I.; Zihler, D.; Wicki, A.; Willi, B.; Willi, N.; Voegeli, M.; Cathomas, G.; Zippelius, A.; Mertz, K.D. Systemic inflammation in a melanoma patient treated with immune checkpoint inhibitors—An autopsy study. J. Immunother. Cancer 2016, 4, 13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  199. Pathak, R.; Katel, A.; Massarelli, E.; Villaflor, V.M.; Sun, V.; Salgia, R. Immune Checkpoint Inhibitor–Induced Myocarditis with Myositis/Myasthenia Gravis Overlap Syndrome: A Systematic Review of Cases. Oncologist 2021, 26, 1052–1061. [Google Scholar] [CrossRef] [PubMed]
  200. Aldrich, J.; Pundole, X.; Tummala, S.; Palaskas, N.; Andersen, C.R.; Shoukier, M.; Abdel-Wahab, N.; Deswal, A.; Suarez-Almazor, M.E. Inflammatory Myositis in Cancer Patients Receiving Immune Checkpoint Inhibitors. Arthritis Rheumatol. 2020, 73, 866–874. [Google Scholar] [CrossRef]
  201. Bonaca, M.P.; Olenchock, B.A.; Salem, J.-E.; Wiviott, S.D.; Ederhy, S.; Cohen, A.; Stewart, G.C.; Choueiri, T.K.; Di Carli, M.; Allenbach, Y.; et al. Myocarditis in the Setting of Cancer Therapeutics. Circulation 2019, 140, 80–91. [Google Scholar] [CrossRef]
  202. Thuny, F.; Bonaca, M.P.; Cautela, J. What Is the Evidence of the Diagnostic Criteria and Screening of Immune Checkpoint Inhibitor–Induced Myocarditis? JACC Cardio-Oncology 2022, 4, 624–628. [Google Scholar] [CrossRef]
  203. Herrmann, J.; Lenihan, D.; Armenian, S.; Barac, A.; Blaes, A.; Cardinale, D.; Carver, J.; Dent, S.; Ky, B.; Lyon, A.R.; et al. Defining cardiovascular toxicities of cancer therapies: An International Cardio-Oncology Society (IC-OS) consensus statement. Eur. Heart J. 2021, 43, 280–299. [Google Scholar] [CrossRef]
  204. Friedrich, M.G.; Sechtem, U.; Schulz-Menger, J.; Holmvang, G.; Alakija, P.; Cooper, L.T.; White, J.A.; Abdel-Aty, H.; Gutberlet, M.; Prasad, S.; et al. Cardiovascular Magnetic Resonance in Myocarditis: A JACC White Paper. J. Am. Coll. Cardiol. 2009, 53, 1475–1487. [Google Scholar] [CrossRef] [Green Version]
  205. Hu, J.-R.; Florido, R.; Lipson, E.J.; Naidoo, J.; Ardehali, R.; Tocchetti, C.G.; Lyon, A.R.; Padera, R.F.; Johnson, D.B.; Moslehi, J. Cardiovascular toxicities associated with immune checkpoint inhibitors. Cardiovasc. Res. 2019, 115, 854–868. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Li, K.; Ho, J.; Recaldin, B.; Gong, M.; Li, G.; Liu, T.; Wu, W.; Wong, M.; Xia, Y.; Dong, M.; et al. Acute Cellular Rejection and Infection Rates in Alemtuzumab vs Traditional Induction Therapy Agents for Lung and Heart Transplantation: A Systematic Review and Meta-analysis. Transplant. Proc. 2018, 50, 3723–3731. [Google Scholar] [CrossRef] [PubMed]
  207. Song, W.; Zheng, Y.; Dong, M.; Zhong, L.; Bazoukis, G.; Perone, F.; Li, G.; Ng, C.F.; Baranchuk, A.; Tse, G.; et al. Electrocardiographic Features of Immune Checkpoint Inhibitor-Associated Myocarditis. Curr. Probl. Cardiol. 2023, 2, 101478. [Google Scholar] [CrossRef]
  208. Patel, R.P.; Parikh, R.; Gunturu, K.S.; Tariq, R.Z.; Dani, S.S.; Ganatra, S.; Nohria, A. Cardiotoxicity of Immune Checkpoint Inhibitors. Curr. Oncol. Rep. 2021, 23, 79. [Google Scholar] [CrossRef]
  209. Dolladille, C.; Ederhy, S.; Sassier, M.; Cautela, J.; Thuny, F.; Cohen, A.A.; Fedrizzi, S.; Chrétien, B.; DA Silva, A.; Plane, A.-F.; et al. Immune Checkpoint Inhibitor Rechallenge After Immune-Related Adverse Events in Patients With Cancer. JAMA Oncol. 2020, 6, 865–871. [Google Scholar] [CrossRef]
  210. Lyon, A.R.; Yousaf, N.; Battisti, N.M.L.; Moslehi, J.; Larkin, J. Immune checkpoint inhibitors and cardiovascular toxicity. Lancet Oncol. 2018, 19, e447–e458. [Google Scholar] [CrossRef] [PubMed]
  211. Poto, R.; Marone, G.; Pirozzi, F.; Galdiero, M.R.; Cuomo, A.; Formisano, L.; Bianco, R.; Della Corte, C.M.; Morgillo, F.; Napolitano, S.; et al. How can we manage the cardiac toxicity of immune checkpoint inhibitors? Expert Opin. Drug Saf. 2021, 6, 685–694. [Google Scholar] [CrossRef]
  212. Sarocchi, M.; Grossi, F.; Arboscello, E.; Bellodi, A.; Genova, C.; Bello, M.G.D.; Rijavec, E.; Barletta, G.; Biello, F.; Ghigliotti, G.; et al. Serial Troponin for Early Detection of Nivolumab Cardiotoxicity in Advanced Non-Small Cell Lung Cancer Patients. Oncol. 2018, 23, 936–942. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  213. Lyon, A.R.; López-Fernández, T.; Couch, L.S.; Asteggiano, R.; Aznar, M.C.; Bergler-Klein, J.; Boriani, G.; Cardinale, D.; Cordoba, R.; Cosyns, B.; et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur. Heart J. 2022, 43, 4229–4361. [Google Scholar] [CrossRef]
  214. Vasyuk, Y.A.; Gendlin, G.E.; Emelina, E.I.; Shupenina, E.Y.; Ballyuzek, M.F.; Barinova, I.V.; Vitsenya, M.V.; Davydkin, I.L.; Dundua, D.P.; Duplyakov, D.V.; et al. Сonsensus statement of Russian experts on the prevention, diagnosis and treatment of cardiotoxicity of anticancer therapy. Russ. J. Cardiol. 2021, 26, 4703. [Google Scholar] [CrossRef]
Table 1. ICIs in oncology.
Table 1. ICIs in oncology.
Blocked
Checkpoint Protein
International Non-Proprietary NameFirst Approvals *Indications for Use (as of 2023) *
CTLA-4IpilimumabFDA, 2011
EMA, 2011
Melanoma, renal cell carcinoma, colorectal cancer, hepatocellular carcinoma, non-small cell lung cancer, malignant pleural mesothelioma, esophageal cancer
TremelimumabFDA, 2022Hepatocellular carcinoma
PD-1Cadonilimab [5]CNMPA, 2022Cervical cancer
Camrelizumab [6,7]CNMPA, 2019Hodgkin’s lymphoma, hepatocellular carcinoma, non-small cell lung cancer, esophageal cancer
CemiplimabFDA, 2018
EMA, 2019
Cutaneous squamous cell carcinoma, basal cell carcinoma, non-small cell lung cancer
NivolumabFDA, 2014
EMA, 2015
Melanoma, non-small cell lung cancer, malignant pleural mesothelioma, renal cell carcinoma, classical Hodgkin lymphoma, squamous cell carcinoma of the head and neck, urothelial carcinoma, colorectal cancer, hepatocellular carcinoma, esophageal cancer, gastric cancer, gastroesophageal junction cancer, esophageal adenocarcinoma
PembrolizumabFDA, 2014
EMA, 2015
Melanoma, non-small cell lung cancer, head and neck squamous cell cancer, classical Hodgkin lymphoma, primary mediastinal large b-cell lymphoma, urothelial carcinoma, microsatellite instability-high or mismatch repair deficient cancer, microsatellite instability-high or mismatch repair deficient colorectal cancer, gastric cancer, esophageal cancer, cervical cancer, hepatocellular carcinoma, Merkel cell carcinoma, renal cell carcinoma, endometrial carcinoma, tumor mutational burden-high cancer, cutaneous squamous cell carcinoma, triple-negative breast cancer, adult classical Hodgkin lymphoma and adult primary mediastinal large b-cell lymphoma
Sintilimab [8]CNMPA, 2018
EMA, 2020
Melanoma, esophageal cancer, nasopharyngeal cancer, small cell lung cancer, soft tissue sarcoma, peripheral T-cell lymphoma
Tislelizumab [9]CNMPA, 2019
EMA, 2020
Non-small cell lung cancer, esophageal squamous cell carcinoma
Toripalimab [10]CNMPA, 2018
EMA, 2022
Melanoma, esophageal squamous cell carcinoma, nasopharyngeal cancer, urothelial carcinoma
PD-L1AtezolizumabFDA, 2016
EMA, 2017
Urothelial carcinoma, non-small cell lung cancer, non-small cell lung cancer, hepatocellular carcinoma, melanoma, alveolar soft part sarcoma
DurvalumabFDA, 2017
EMA, 2018
Non-small
cell lung cancer, small cell lung cancer, biliary tract cancer, hepatocellular carcinoma
LAG-3RelatlimabFDA, 2022
EMA, 2022
Melanoma (in combination with nivolumab)
CNMPA: China National Medical Products Administration; FDA: Food and Drug Administration; EMA: European Medicines Agency. *—information from sites: IUPHAR/BPS Guide to PHARMACOLOGY, https://www.guidetopharmacology.org; Drugs@FDA: FDA-Approved Drugs, https://www.accessdata.fda.gov/scripts/cder/daf/index.cfm; EMA, Medicine, https://www.ema.europa.eu/en/medicines/field_ema_web_categories%253Aname_field/Human (accessed on 13 January 2023).
Table 2. IrAE grading system.
Table 2. IrAE grading system.
The Organ(s)Grade 1Grade 2Grade 3Grade 4
Acute kidney injury (creatinine increased)<1.5× baseline or upper limit of normal1.5–3.0× baseline or upper limit of normal3.0–6.0× baseline or upper limit of normal
or
>3.0× baseline, but dialysis not indicated
>6.0× baseline or upper limit of normal
or
Dialysis indicated
ArthritisMild pain with inflammation, redness, or joint swellingModerate pain with inflammation, redness, or joint swelling
Restriction of instrumental ADL
Severe pain with inflammation, redness, or joint swelling
Irreversible joint injury
Restriction of self-care
ColitisAsymptomatic
<4 bowel movements in 24 h
Abdominal pain
Mucus or blood in feces
4–6 bowel movements in 24 h
Severe abdominal pain
Peritoneal signs
>7 bowel movements in 24 h
Life-threatening consequences
Liver damage
(ALT increased)
<3.0× upper limit of normal
Asymptomatic
3.0–5.0× upper limit of normal
Asymptomatic
5.0–20.0× upper limit of normal
Symptomatic
Compensated cirrhosis
20.0× upper limit of normal
Liver function decompensation (ascites, dysfunction of hemostasis, toxic damage to the central nervous system, coma)
Pituitary lesionAsymptomatic
Mild symptoms
Moderate symptoms limiting age-appropriate instrumental ADLSevere or medically significant limiting self-care ADLLife-threatening consequences
Skin rashLess than 10% of body surface area affected10–30% of body surface area affectedMore than 30% of body surface area affectedLife-threatening manifestations
Generalized exfoliation, skin ulcers
Myasthenia gravisNo or mild manifestationsModerate manifestations
Limiting age-appropriate instrumental ADL
Severe manifestations
Limiting self-care ADL
Life-threatening consequences, dysfunction of the respiratory muscles
MyositisPainPain associated with mild or moderate weakness; Limitation of instrumental ADLPain with severe weakness; limitation of self-serviceLife-threatening consequences; urgent intervention indicated
MyocarditisAsymptomatic
Cardiac enzyme elevation or abnormal electrocardiogram (EСG)
Symptoms with moderate activityManifestations at rest and with minimal exertionLife-threatening consequences
ArrhythmiasAsymptomatic, intervention
not indicated
Non-urgent medical
intervention indicated
Symptomatic, urgent
intervention indicated (pacemaker, ablation, etc.)
Life-threatening
consequences; embolus
requiring urgent intervention
Atrioventricular block
complete
Non-emergency intervention
indicated
Partially medication controlled, or controlled with
device (e.g., pacemaker)
Life-threatening
consequences; urgent
intervention indicated
Conduction disorderIntervention not neededNon-emergency medical interventionEmergency interventionLife-threatening consequences
Heart failureAsymptomatic with laboratory (e.g., BNP (brain natriuretic peptide)) or cardiac imaging changesSymptoms with moderate activity or exertionSymptoms at rest or with minimal activity or exertionEmergency intervention indicated
Ventricular tachycardia Non-urgent medical intervention indicatedSymptomatic, urgent intervention indicatedLife-threatening consequences; hemodynamic compromise
Pericardial effusion AsymptomaticSymptomaticEmergency intervention indicated
PericarditisAsymptomaticSymptomatic, mild or moderate manifestationsPhysiologic consequences, severe manifestationsLife-threatening consequence
PneumonitisAsymptomatic
Only one lobe of the lung is affected or less than 25% of the lung
Symptomatic (dyspnea, cough or chest pain)
Two or more lobes of the lung are affected or 25–50% of the lung
Severe symptoms (new or worsening hypoxia)
All lung lobes of the lung are affected or >50% of the lung
Oxygen therapy
Life-threatening respiratory compromise
ADL: activity of daily living.
Table 3. Demographic and clinical characteristics of individual case reports.
Table 3. Demographic and clinical characteristics of individual case reports.
CharacteristicsPatients N (%)
Median age (year)67 (IQR, 60–74)
Male sex (%)143 (58.6%)
Female sex (%)99 (40.6%)
Gender not specified (%)2 (0.8%)
Types of ICI
Pembrolizumab75 (30.7%)
Nivolumab49 (20.1%)
Nivolumab plus ipilimumab41 (16.8%)
Sintilimab20 (9.3%)
Camrelizumab15 (6.1%)
Durvalumab8 (3.3%)
Toripalimab7 (2.9%)
PD-L1 inhibitor (investigational)6 (2.6%)
Atezolizumab5 (2%)
Tislelizumab5 (2%)
Others13 (5.3%)
Table 4. Days until presentation of ICI-associated cardiotoxicity, organized by treatment.
Table 4. Days until presentation of ICI-associated cardiotoxicity, organized by treatment.
ICISintilimabNivolumabPembrolizumabIpilimumab + NivolumabDurvalumabAtezolizumabToripalimabTislelizumabCamrelizumab
Number of cases20497541857515
Median days until manifestation212326193414202025
Table 5. Electrocardiographic findings.
Table 5. Electrocardiographic findings.
FindingsN (%)
Sinus arrhythmia
Sinus tachycardia24 (12%)
Sinus bradycardia5 (2.5%)
Sick sinus syndrome2 (1%)
Atrial arrhythmia
Atrial extrasystole3 (1.5%)
Atrial flutter3 (1.5%)
Atrial fibrillation16 (8%)
Ventricular arrhythmia
Ventricular extrasystole11 (5.5%)
Ventricular tachycardia26 (13%)
Atrioventricular block
1-degree atrioventricular block8 (4%)
2-degree atrioventricular block12 (6%)
3-degree atrioventricular block51 (25.4%)
Bundle branch block
Left bundle branch block12 (6%)
Right bundle branch block37 (18.4%)
ST segment changes
ST segment elevation33 (6.4%)
ST segment depression12 (6%)
T wave inversion21 (10.4%)
Non-classified
New Q wave1 (0.5%)
Low voltage8 (4%)
No changes30 (14.9%)
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Zotova, L. Immune Checkpoint Inhibitors-Related Myocarditis: A Review of Reported Clinical Cases. Diagnostics 2023, 13, 1243. https://doi.org/10.3390/diagnostics13071243

AMA Style

Zotova L. Immune Checkpoint Inhibitors-Related Myocarditis: A Review of Reported Clinical Cases. Diagnostics. 2023; 13(7):1243. https://doi.org/10.3390/diagnostics13071243

Chicago/Turabian Style

Zotova, Liudmila. 2023. "Immune Checkpoint Inhibitors-Related Myocarditis: A Review of Reported Clinical Cases" Diagnostics 13, no. 7: 1243. https://doi.org/10.3390/diagnostics13071243

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