Abstract
Background
Immune checkpoint inhibitors (ICIs) have revolutionized the management of multiple tumors, due to improved efficacy, quality of life, and safety. While most immune-related adverse events (irAEs) are mild and easily managed, in rare cases such events may be life-threatening, especially those affecting the neuromuscular and cardiac system. The management of neuromuscular/cardiac irAEs is not clear due to the lack of consistent data. Therefore, we carried out a pooled analysis of collected cases from selected Italian centers and individual data from published case reports and case series, in order to improve our understanding of these irAEs.
Patients and methods
We collected retrospective data from patients treated in six Italian centers with ICIs (programmed cell death protein 1 or programmed death-ligand 1 and/or cytotoxic T-lymphocyte antigen 4 inhibitor) for any solid tumor who experienced neuromuscular and/or cardiovascular toxicity. Then, we carried out a search of case reports and series of neuromuscular/cardiac irAEs from ICIs with any solid tumor.
Results
This analysis includes cases from Italian institutions (n = 18) and the case reports identified in our systematic literature search (n = 120), for a total of 138 patients. Among these patients, 50 (36.2%) had complete resolution of their neuromuscular/cardiac irAEs, in 21 (15.2%) cases there was a clinical improvement with mild sequelae, and 53 (38.4%) patients died as a result of the irAEs. Factors significantly associated with worse outcomes were early irAE onset, within the first two cycles of ICI (Fisher P < 0.0001), clinical manifestation of both myositis and myocarditis when compared with patients who developed only myositis or myocarditis (chi-square P = 0.0045), and the development of arrhythmia (Fisher P = 0.0070).
Conclusions
To the best of our knowledge, this is the largest collection of individual cases of immune-related myocarditis/myositis. Early irAE onset, concurrent development of myositis and myocarditis, as well as occurrence of arrhythmias are associated with worse outcomes and should encourage an aggressive immunomodulatory treatment.
Key words: immune checkpoint inhibitors, myasthenia gravis, myositis, myocarditis, melanoma, immunotherapy
Highlights
-
•
Myocarditis and myositis represent rare but potentially lethal irAEs.
-
•
There is no standard management of these irAEs due to the lack of consistent data.
-
•
This is the largest pooled analysis of individual cases of myocarditis/myositis related to ICIs.
-
•
Early onset and the concurrent manifestation of myositis and myocarditis are associated with worse outcomes.
Introduction
In recent years, cancer therapy has been revolutionized by the advent of immunotherapy. Since their introduction, immune checkpoint inhibitors (ICIs) have progressively become the standard of care for many advanced and early-stage solid tumors,1 leading to unprecedented results in terms of efficacy,2,3 quality of life,4 and safety.5
The mechanism of action of this class of drugs [programmed cell death protein 1 (PD-1)/ programmed death-ligand 1 (PD-L1) inhibitors and cytotoxic T-lymphocyte antigen 4 (CTLA-4) inhibitors] leads to the inactivation of an immunological escape pathway used by tumor cells, reactivating the anti-neoplastic effector activity by adaptive immunity.6
However, such reactivation may lead to a series of adverse events of autoimmune pathogenesis, directed mainly against the endocrine glands, the skin, but also the kidneys, lungs and gastrointestinal tract, and potentially affecting any organ or tissue.7,8 In most cases, these adverse events, also called immune-related adverse events (irAEs), are clinically easy to manage and paucisymptomatic.7,8 However, in some rare cases irAEs may be life-threatening, especially those affecting the neuromuscular system and muscle tissue, including the myocardial tissue,7,8 identified as myositis and myocarditis, respectively.
The onset of muscular and cardiac irAEs can be acute and lead the patient to the emergency department with acute heart failure or respiratory insufficiency constituting a serious diagnostic and therapeutic challenge. The rapid identification and management of these clinical presentations is very important not only for the emergency physicians, but also for oncologists, cardiologists, or neurologists, who can be still unfamiliar with the management of these rare serious events.9
To face this knowledge lag, scientific societies expressed guidelines for the management of these toxicities.7,8,10, 11, 12 However, these guidelines are sometimes conflicting and are only based on expert opinion, due to a lack of consistent data. Therefore, we collected case reports of neuromuscular and cardiac irAEs from selected Italian centers and carried out a systematic literature search to collect case reports and case series on this topic to improve our understanding of these irAEs.
Patients and methods
Search strategy, study selection, and data extraction
We collected retrospective data from patients, reported by the referring physicians, treated in six Italian centers with ICIs (PD-1 or PD-L1 and/or CTLA-4 inhibitor) for any solid tumor who experienced neuromuscular and/or cardiovascular toxicity up to 4 July 2021. Approval of local Ethics Committee (Liguria, Italy) was obtained. Then, we carried out a computerized search of case reports and case series of neuromuscular and/or cardiologic toxicity from ICIs with any solid tumor using the PubMed search engine with the search string reported in the Supplementary Material, available at https://doi.org/10.1016/j.esmoop.2023.100791. The search included cases published from 1 January 2010 to 4 July 2021.
Case reports and case series published in English and reporting neuromuscular and/or cardiovascular toxicity in patients receiving ICIs (PD-1 or PD-L1 inhibitors and/or CTLA-4 inhibitors) were evaluated.
The following information was extracted from each report: first author and year of publication, sex and age of patients, type of cancer, cancer treatment, previous medical conditions, clinical manifestations of the adverse event, diagnostic work-up, management, outcome of the event, and cancer objective response to the ICI.
Data were independently extracted by two investigators (A.Bou. and A.Bot.) to ensure homogeneity of collection and to rule out the effect of subjectivity in data gathering and entry. Disagreements were resolved by iteration, discussion, and consensus.
Statistical analysis
Descriptive and statistical analyses were carried out on the whole of our case series and the reports extracted from the literature. Contingency analyses were carried out by using Fisher’s exact test (for multiple groups chi-square test was employed instead). Group analyses for continuous variables were carried out by using the Mann–Whitney test.
For the purpose of statistical analyses, we evaluated complete remission, partial remission, death, as well as the lack of improvement without reported death. Since our analysis was based on case reports and case series, where the final outcome of the patient might be unknown, when division in sub-groups was functional to the analysis, we associated complete and partial remissions (assuming that they can both be considered favorable outcomes). Similarly, we considered both death and complete lack of improvement as unfavorable outcomes. Notably, only few patients experienced neither death nor at least partial improvement of the irAE.
The analyses were carried out using GraphPad Prism version 9.3.1 for Windows (GraphPad Software, San Diego, www.graphpad.com); the same software was employed for developing graphs.
Results
Patient characteristics
Patients identified within Italian institutions
We identified a total of 18 patients diagnosed with solid tumors who received at least one dose of ICI at six Italian centers from January 2017 to December 2020 who experienced at least one neuromuscular or cardiac irAE.
The included patients were affected by non-small-cell lung cancer (NSCLC; n = 5), melanoma (n = 11), cutaneous squamous cell carcinoma (n = 1), and renal cell carcinoma (n = 1) and had been treated with nivolumab (n = 8), pembrolizumab (n = 3), nivolumab plus ipilimumab (n = 3), durvalumab (n = 2), cemiplimab (n = 1), and ipilimumab (n = 1). Their median age was 72 years.
Selected case reports
In our systematic literature search, 71 full-texts of case reports or case series of patients with solid tumors who manifested neuromuscular or cardiac irAEs were selected, and data from 120 patients were extracted.13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, 25, 26, 27, 28, 29, 30, 31, 32, 33, 34, 35, 36, 37, 38, 39, 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
The included patients had melanoma (n = 56), NSCLC (n = 30), renal cell carcinoma (n = 6), head and neck squamous cell carcinoma (n = 4), pancreatic cancer (n = 4), thymic cancer (n = 4), thymoma (n = 1), urothelial carcinoma (n = 5), cutaneous squamous cell carcinoma (n = 2), endometrial cancer (n = 2), hepatocellular carcinoma (n = 2), colorectal cancer (n = 1), cholangiocarcinoma (n = 1), mesothelioma (n = 1), and prostate cancer (n = 1). These patients received the ICIs pembrolizumab (n = 37), nivolumab (n = 28), nivolumab plus ipilimumab (n = 25), ipilimumab (n = 9), atezolizumab (n = 4), durvalumab plus tremelimumab (n = 3), atezolizumab plus cobimetinib (n = 2), avelumab (n = 2), durvalumab (n = 4), nivolumab plus vaccine (n = 2), cemiplimab (n = 1), chemotherapy plus pembrolizumab (n = 1), and sintilimab (n = 2). The median age was 70 years. For 110 patients, data on toxicity outcomes were reported and for 50 patients, information on the best overall response to ICI was available. The identified studies are summarized in Figure 1.
Figure 1.
The PRISMA flowchart summarizing the process for the identification of the eligible studies. ICI, immune checkpoint inhibitor.
Outcome analysis
The outcome analysis of muscular/cardiac irAEs has been conducted including both cases from the Italian institutions (n = 18) and the case reports identified in our systematic literature search (n = 120), for a total of 138 patients.
Among all these patients, 50 (36.2%) had complete resolution of their neuromuscular/cardiac irAEs, and in 21 (15.2%) patients there was a clinical improvement with mild sequelae. A total of 53 (38.4%) patients died as a result of the neuromuscular/cardiac irAEs. In four (2.9%) patients no clinical improvements of the irAEs were observed; their cause of death was progressive disease (n = 1), complications and side effects of irAE treatments (n = 1), or not reported (n = 2). Finally, the toxicity outcomes of 10 (7.3%) patients were not reported. Individual patients’ outcomes have been summarized in Supplementary Table S1 and Figure S1, available at https://doi.org/10.1016/j.esmoop.2023.100791. No differences in terms of irAE outcomes were observed on the basis of sex or age (Fisher P = 0.9999 and Mann–Whitney P = 0.3792, respectively, Supplementary Table S2, Figures S2 and S3, available at https://doi.org/10.1016/j.esmoop.2023.100791).
No significant difference in the occurrence or outcome of cardiac/muscular irAEs was observed in patients with history of cardiovascular, respiratory, autoimmune, endocrine, or metabolic diseases. The relevant findings involving medical history and cardiac/muscular irAEs outcomes are reported in Table 1 and Figure 2.
Table 1.
Patients’ outcomes based on history of pre-existing cardiovascular disease, endocrine/metabolic disease, chronic respiratory disease (asthma or chronic obstructive pulmonary disease), or autoimmune disease before treatment with immune checkpoint blockade
| N | Complete remission, n | Partial remission, n | Neither improvement nor death, n | Death, n | Not reported, n | |
|---|---|---|---|---|---|---|
| Cardiovascular disease | 60 | 18 | 12 | 1 | 29 | 0 |
| No cardiovascular disease | 41 | 21 | 6 | 3 | 11 | 0 |
| Cardiovascular disease history not reported | 37 | 11 | 3 | 0 | 13 | 10 |
| Endocrine/metabolic disease | 38 | 15 | 10 | 1 | 12 | 0 |
| No endocrine/metabolic disease | 62 | 24 | 7 | 3 | 28 | 0 |
| Endocrine/metabolic disease history not reported | 38 | 11 | 4 | 0 | 13 | 10 |
| Chronic respiratory disease | 10 | 4 | 1 | 1 | 4 | 0 |
| No chronic respiratory disease | 87 | 34 | 15 | 3 | 35 | 0 |
| Chronic respiratory disease history not reported | 41 | 12 | 5 | 0 | 14 | 10 |
| Autoimmune disease | 7 | 5 | 2 | 0 | 0 | 0 |
| No autoimmune disease | 93 | 35 | 14 | 4 | 40 | 0 |
| Autoimmune disease history not reported | 38 | 10 | 5 | 0 | 13 | 10 |
Figure 2.
Outcomes of cardiac/muscular irAEs based on: (A) cardiovascular (CV) disease history (Fisher P = 0.1529), (B) endocrine or metabolic (endo/met) disease history (Fisher P = 0.1487), (C) respiratory disease history (Fisher P = 0.7477), and (D) autoimmune disease history (Fisher P = 0.0169). irAEs, immune-related adverse events.
Cancer type and outcomes
Most of the included patients had melanoma (n = 67), of whom 62 were cutaneous, while 4 patients had uveal melanoma and 1 had mucosal melanoma. A total of 27 (40.3%) melanoma patients had complete remission of the neuromuscular/cardiac irAE, while in 12 (17.9%) patients there was a partial improvement of the event; by contrast, 25 (37.3%) patients died due to irAEs and in one case the clinical manifestation of the irAE did not improve and did not cause death. The outcomes of the irAEs experienced by two patients were not reported. Notably, in the subgroup of patients with melanoma, nine had resected disease and were under adjuvant ICI treatment. Among these patients, six (66.6%) died as a consequence of the irAEs.
The second most frequent cancer type was NSCLC. Among the 35 patients with NSCLC, 10 (28.6%) totally recovered, 5 (14.3%) had improvement of the irAE, 15 (42.9%) died, and 3 (8.6%) did not improve or die according to the report; data regarding the irAEs outcomes were not reported for 2 patients with NSCLC.
With regard to patients with other malignancies (n = 36), 13 (36.1%) had complete remission, 4 (11.1%) had partial improvement, 13 (36.1%) died, and none had persisting event with no improvement, while outcomes of 6 patients were not reported. Globally, no difference in terms of outcomes was observed based on primary tumor (chi-square P = 0.3750), as reported in Table 2 and Figure 3.
Table 2.
Outcomes of the cardiac/muscular irAEs according to primary tumor
| N | Complete remission, n | Partial remission, n | Neither improvement nor death, n | Death, n | Not reported, n | |
|---|---|---|---|---|---|---|
| Melanoma (four uveal, one mucosal) | 67 | 27 | 12 | 1 | 25 | 2 |
| NSCLC | 35 | 10 | 5 | 3 | 15 | 2 |
| RCC | 7 | 2 | 1 | 0 | 4 | 0 |
| HNSCC | 4 | 1 | 0 | 0 | 2 | 1 |
| Cholangiocarcinoma | 1 | 0 | 0 | 0 | 0 | 1 |
| CRC | 1 | 0 | 1 | 0 | 0 | 0 |
| CSCC | 3 | 0 | 0 | 0 | 3 | 0 |
| Endometrial cancer | 2 | 1 | 1 | 0 | 0 | 0 |
| HCC | 2 | 2 | 0 | 0 | 0 | 0 |
| Mesothelioma | 1 | 0 | 0 | 0 | 1 | 0 |
| Pancreatic cancer | 4 | 1 | 1 | 0 | 0 | 2 |
| Prostate cancer | 1 | 1 | 0 | 0 | 0 | 0 |
| Thymoma | 1 | 1 | 0 | 0 | 0 | 0 |
| Thymic cancer | 4 | 1 | 0 | 0 | 2 | 1 |
| Urothelial cancer | 5 | 3 | 0 | 0 | 1 | 1 |
| Total | 138 | 50 | 21 | 4 | 53 | 10 |
CRC, colorectal carcinoma; CSCC, cutaneous squamous cell carcinoma; HCC, hepatocellular carcinoma; HNSCC, head-neck squamous cell carcinoma; irAEs, immune-related adverse events; NSCLC, non-small-cell lung cancer; RCC, renal cell carcinoma.
Figure 3.
Outcomes of cardiac/muscular irAEs based on: (A) primary tumor (chi-square P = 0.3750), (B) use of single-agent ICI (anti-PD-1 or PD-L1) or dual ICIs (anti-PD-1/PD-L1 plus anti-CTLA-4) (Fisher P = 0.6796), (C) time of onset of the muscular/cardiac irAE (Fisher P < 0.0001), and (D) clinical manifestations of the muscular/cardiac irAE (chi-square P = 0.0045).
CTLA-4, cytotoxic T-lymphocyte antigen 4; ICI, immune checkpoint inhibitor; irAEs, immune-related adverse events; NSCLC, non-small-cell lung cancer; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1.
Immunotherapy regimen and outcomes
We also investigated the neuromuscular/cardiac events outcomes in patients receiving single-agent ICI (PD-1/PD-L1 or CTLA-4 inhibitor) and in patients receiving a combination of ICI (PD-1/PD-L1 inhibitor plus CTLA-4 inhibitor). Among a total of 102 patients treated with single-agent ICI, data on the adverse events outcomes were reported in 92 cases. Among them, 37 (40.2%) patients totally recovered from the irAEs, 15 (16.3%) had a partial resolution, 4 (4.4%) did not have any improvement, and 36 (39.1%) died from the adverse events.
In the ICI combination group (n = 36), a total of 13 (36.1%) patients had complete resolution of the adverse events, while 6 (16.7%) had a partial resolution and 17 (47.2%) died as a result of the adverse events. When the outcomes of cardiac/muscular irAEs of single-agent ICI and dual ICIs were compared, no significant difference was observed (Fisher P = 0.6796). Data are summarized in Table 3 and Figure 3.
Table 3.
Outcomes of the cardiac/muscular irAEs according to immunotherapy regimen
| N | Complete remission, n | Partial remission, n | Neither improvement nor death, n | Death, n | Not reported, n | |
|---|---|---|---|---|---|---|
| Monotherapy (PD-1/PD-L1or CTLA-4 inhibitor) | 102 | 37 | 15 | 4 | 36 | 10 |
| Combination (PD-1/PD-L1 + CTLA-4 inhibitor) | 31 | 10 | 6 | 0 | 15 | 0 |
| Combination (PD-1 inhibitor + chemotherapy) | 1 | 0 | 0 | 0 | 1 | 0 |
| Combination (PD-1 inhibitor + vaccine) | 2 | 1 | 0 | 0 | 1 | 0 |
| Combination (PD-L1 inhibitor + MEK inhibitor) | 2 | 2 | 0 | 0 | 0 | 0 |
CTLA-4, cytotoxic T-lymphocyte antigen 4; irAEs, immune-related adverse events; MEK, mitogen-activated protein kinase kinase; PD-1, programmed cell death protein 1; PD-L1, programmed death-ligand 1.
Clinical manifestation of irAEs and outcomes
Among 132 patients for whom the number of cycles before the onset of irAE was reported, in 96 (72.7%) of them the muscular and/or cardiac adverse event occurred within the first two cycles of therapy. In this subgroup of patients, data on irAEs outcome were reported in 89 patients: muscular/cardiac adverse events were lethal in 48 (53.9%), while 30 (33.7%) and 9 (10.1%) patients had complete and partial resolution of irAEs manifestations, respectively. In the group of patients whose toxicity onset was observed after the second cycle of ICI (n = 36), data on outcome of adverse events were reported in 33 patients, of whom 17 (51.5%) totally recovered, 11 (33.3%) partially recovered, and 3 (9.1%) died due to the adverse events. When early onset (within the first two cycles) and late onset (after two cycles) were compared, a statistically significant difference in terms of outcome was detected (Fisher P < 0.0001), as earlier onset of the irAE was associated with worse outcomes (Figure 3; Table 4).
Table 4.
Outcomes of the cardiac/muscular irAEs according to time of onset (within two cycles or after two cycles), clinical manifestation (only myositis, only myocarditis, or both), and instrumental findings
| N | Complete remission, n | Partial remission, n | Neither improvement nor death, n | Death, n | Not reported, n | |
|---|---|---|---|---|---|---|
| ≤2 cycles | 96 | 30 | 9 | 2 | 48 | 7 |
| >2 cycles | 36 | 17 | 11 | 2 | 3 | 3 |
| Only myositis | 46 | 21 | 8 | 1 | 10 | 6 |
| Only myocarditis | 49 | 18 | 10 | 0 | 21 | 0 |
| Both myositis and myocarditis | 43 | 11 | 3 | 3 | 22 | 4 |
| LVEF ≤45% | 22a | 6 | 6 | 1 | 9 | 0 |
| LVEF> 45% | 54a | 20 | 9 | 2 | 22 | 1 |
| Arrhythmia at ECG | 47b | 11 | 6 | 2 | 27 | 1 |
| No arrhythmia at ECG | 37b | 16 | 8 | 1 | 10 | 2 |
| Ischemic signs at ECG | 19b | 8 | 2 | 0 | 9 | 0 |
| No ischemic signs at ECG | 65b | 19 | 9 | 3 | 27 | 7 |
ECG, electrocardiogram; irAEs, immune-related adverse events; LVEF, left ventricular ejection fraction.
Only 76 patients had data on echocardiographic assessment of LVEF.
Only 84 patients had data on ECG.
Among the patients who manifested only myocarditis (n = 49), 18 (36.7%) patients had complete resolution of the adverse event, 10 (20.4%) patients achieved partial clinical improvement of the event, and 21 (42.8%) patients died. With regard to patients who had only myositis (n = 46), the outcomes were known for 40 patients. Among these, 21 (52.5%) patients had complete resolution of myositis, 8 (20.0%) patients had partial clinical improvement, 1 (2.5%) patient did not achieve any improvement, and 10 (25.0%) patients died due to myositis.
With regard to patients who experienced both myositis and myocarditis and whose outcome was known (n = 39 out of 43), the following data were reported: 11 (28.2%) patients had complete recovery from the adverse events, 3 (7.7%) patients had a partial clinical improvement of the event, 3 (7.7%) patients did not have any clinical improvement but did not die, while a total of 22 (56.4%) patients died. The patients who developed both myositis and myocarditis experienced significantly worse outcomes compared to patients who developed only myositis or myocarditis (chi-square P = 0.0045), as reported in Table 4 and Figure 3.
When myositis was present and the involved muscular districts were reported (n = 85), they predominantly included limb muscles (n = 55), axial muscles (n = 45), oculomotor muscles (n = 36), diaphragm (n = 23), and muscles of the ENT (ear, nose, and throat) district (n = 27), as reported in Supplementary Figure S4, available at https://doi.org/10.1016/j.esmoop.2023.100791. Notably, each patient could have more involved muscular districts at once. Due to the diagnostic heterogeneity in the reported cases, and the underreporting of acetylcholine receptor auto-antibodies values, no cases of ‘myasthenia gravis’ were classified in our analysis.
Laboratory findings and outcomes
In a total of 93 patients, the creatine kinase enzyme (CK) blood levels have been assessed at the onset of the irAE. In most cases, the peak value was reported, while in some cases the value was not reported and CK was defined as ‘elevated’, ‘normal’, or above various cut-off values. A total of 78 (83.9%) patients had increased CK (considering the upper normal limit of 200 U/l). The median CK value was 1900 U/l (grade 3 according to the Common Terminology Criteria for Adverse Events v.5). No significant difference in terms of outcomes was observed on the basis of peak CK value (Mann–Whitney P = 0.2995).
With regard to troponin, only data for 54 patients were available. Among these, 10 researchers reported the use of troponin T, while for other 33 cases the use of troponin I was reported and in 11 cases the specific troponin employed was not reported. Considering these limitations and considering an upper normal limit value of 0.04 ng/ml, most patients (n = 48, 88.9%) had troponin values above the upper normal limit. The median value of troponin was 2.965 ng/ml. Notably, patients with higher troponin values had numerically higher risk of unfavorable outcomes of the muscular/cardiac irAE, albeit significance was not reached (Mann–Whitney P = 0.0830). The irAEs outcomes based on CK and troponin values are summarized in Figure 4 and Supplementary Table S3, available at https://doi.org/10.1016/j.esmoop.2023.100791.
Figure 4.
Outcomes of cardiac/muscular irAEs based on: (A) creatine kinase (CK) values (Mann–Whitney P = 0.2995) and troponin values (Mann–Whitney P = 0.0830), (B) left ventricular ejection fraction (LVEF) (chi-square P = 0.9999), (C) finding of arrhythmia at electrocardiogram (ECG) (chi-square P = 0.0070), and (D) ischemic signs at ECG (chi-square P = 0.9999). The outliers of the box-and-whisker plot are above the maximum value of the ‘y’ axis in order to make the plot more easily readable. irAEs, immune-related adverse events.
Instrumental findings and outcomes
In patients with myocarditis, we analyzed the cardiovascular diagnostic work-up that was undertaken.
In 76 patients who had echocardiographic assessment of left ventricular ejection fraction (LVEF) at the onset of myocarditis, 22 (28.9%) patients had LVEF ≤45% and 54 (71.1%) patients had LVEF >45%. Among patients with reduced LVEF and known outcome data, in 6 (27.3%) patients myocarditis resolved without sequelae, whereas in 9 (40.9%) cases the event led to patient’s death. In a total of six (27.3%) cases of myocarditis improved with sequelae and in one (4.5%) patient, the adverse event did not improve and did not lead to patient’s death, with no additional information on patient’s outcome. In contrast, in patients with preserved LVEF and known outcome data (n = 53), 20 (37.8%) had complete resolution of the adverse event and in 9 (16.9%) patients there was partial remission of the adverse event, whereas in 2 cases (3.8%) there was no improvement nor worsening of the event, and in 22 (41.5%) the adverse event led to patient’s death. When patients’ outcomes were compared on the basis of LVEF (cut-off value: 45%), no significant differences were observed (Fisher P = 0.9999).
An electrocardiogram (ECG) was carried out in 84 patients. No arrhythmia was found in 37 (44.0%) patients, whereas new-onset cardiac conduction disturbances were found in 47 (56.0%) patients (including atrioventricular blocks, grade 3 atrioventricular block in 17 patients). The irAE outcomes were not reported in one case with arrhythmia at ECG and two cases with no arrhythmia. Among patients with new-onset arrhythmia, toxicity completely resolved in 11 (23.9%) and resulted in death in 27 (58.8%) patients, while 6 (13.0%) patients achieved a clinical improvement of the adverse event and in 2 (4.3%) patients there was no improvement of the myocarditis. In contrast, among patients with no arrhythmia, where reported, myocarditis completely resolved in 16 (45.7%), whereas it led to death in 10 (28.6%) patients or improved with some sequelae in 8 (22.9%) patients. When we compared the outcomes of patients with or without arrhythmia, we identified a statistically significant difference favoring patients without arrhythmia (Fisher P = 0.0070).
Ischemic signs were detected in 19 patients, whereas no ischemic signs were detected in 65 patients. In the group of patients without ischemic signs on ECG, 19 (32.8%) patients had complete remission from myocarditis, 9 (15.4%) patients had clinical improvement, 3 (5.2%) patients did not have any clinical improvement and 27 (46.6%) patients died due to the adverse events (data were not reported for 7 patients). In this subgroup, a coronary study was carried out in 11 patients; at coronarography, coronary abnormalities were detected in the case of one single patient who had a history of coronary artery disease. Among 19 patients with ischemic signs on ECG, 8 (42.1%) had complete remission, 2 (10.5%) had clinical improvement, and 9 (47.4%) died due to myocarditis. In this subgroup, coronary angiography was carried out in 11 patients, all without alterations. When patients with and without ischemic signs at ECG were compared in terms of outcomes, no significant differences were observed (Fisher P = 0.9999). The irAEs outcomes based on ECG and echocardiography are summarized in Table 4 and Figure 4.
With regard to cardiac magnetic resonance, we identified 25 patients who underwent the exam, of which only 12 had late gadolinium enhancement. Among the patients for whom magnetic resonance data were available (n = 15), only in 5 (33.3%) cases with mild alterations in terms of left ventricular posterior wall thickening were reported. No significant differences were observed on the basis of magnetic resonance findings (Fisher P = 0.9999), albeit the number of assessable patients was extremely limited. Data on cardiac magnetic resonance are reported in Supplementary Table S4 and Figure S5, available at https://doi.org/10.1016/j.esmoop.2023.100791.
Finally, electromyography data were available for 35 patients; notably, almost all the patients (31 out of 35, 88.6%) had some degree of conduction alteration. Due to the extremely limited number of patients with negative electromyography, no comparison was carried out.
Treatment of the immune-related adverse events and outcomes
A total of 106 patients were reported to have received corticosteroid therapy; of these, 59 patients received exclusive steroidal therapy, without other immunosuppressive agents. Among those who received exclusive steroid therapy, 24 (40.7%) had complete resolution from irAE, 13 (22.0%) clinically improved without reaching complete resolution of the irAEs, whereas 20 (33.9%) died.
We also analyzed the corticosteroid dosage used (where reported): in a total of 35 patients, a corticosteroid dosage of no >1 mg/kg daily was administered. Of these, 13 (37.1%) had complete remission from symptoms, 8 (22.9%) had clinical improvement without reaching complete remission, and 12 (34.3%) died due to the adverse event. In a total of 23 patients, a maximum daily dose of corticosteroid between 1 and 2 mg/kg was used. Of these, toxicity completely resolved in 9 (39.1%) patients, improved without complete remission in 5 (21.7%) patients, and led to death in 8 (34.8%) patients. In contrast, a total of 39 patients received daily doses of corticosteroid ≥2 mg/kg, with complete remission in 14 (35.9%) patients, partial clinical improvement in 5 (12.8%), and death in 20 (51.3%) cases. No differences in outcomes were observed on the basis of maximum daily dose of corticosteroid (chi-square P = 0.5269). Notably, when we compared the outcomes of patients treated exclusively with corticosteroids or corticosteroids followed by or associated with other agents, we observed a non-significant difference in outcome favoring patients who received corticosteroids alone (Fisher P = 0.0913).
With specific regard to other immunosuppressive agents, a total of 14 patients received a disease-modifying antirheumatic drug (DMARD), with 8 (57.1%) of the patients in complete remission from the adverse event and 5 (35.7%) of the patients deceased. Among the eight patients to whom a biologic immunosuppressant drug was administered, one (12.5%) patient had a complete remission, two (25.0%) had a partial remission, and five (62.5%) patients died. A total of 32 patients also received intravenous immunoglobulin (IVIG), with 12 (37.5%) of the patients in complete remission, and a 50% (n = 16) death rate. A total of 16 patients also received plasma exchange, with only 2 (12.5%) patients in complete remission from toxicity and 11 (68.8%) deceased patients. When the collective outcomes were based on the employed agents, no significant difference was observed (chi-square P = 0.1070), albeit DMARDs achieved a notable benefit. Outcomes of the irAEs based on immunomodulatory treatments were summarized in Figure 5 and Table 5.
Figure 5.
Outcomes of cardiac/muscular irAEs based on: (A) the maximum daily reported dose of CTS (prednisone or equivalent dose) (chi-square P = 0.5269), (B) exclusive use of CTS versus association with other agents (Fisher P = 0.0913), (C) use of specific immunomodulatory agents (chi-square P = 0.1070), and (D) best tumor response (chi-square P = 0.3300). BI, biologic immunosuppressant; CR, complete response; CTS, corticosteroids; DMARD, disease-modifying antirheumatic drug; IVIG, intravenous immunoglobulins; irAEs, immune-related adverse events; PD, progressive disease; PE, plasma exchange; PR, partial response; SD, stable disease.
Table 5.
Outcomes of the cardiac/muscular irAEs according to immunosuppressive treatments and according to best overall tumor response among patients with available treatment and tumor response data based on RECIST
| N | Complete remission, n | Partial remission, n | Neither improvement nor death, n | Death, n | Not reported, n | |
|---|---|---|---|---|---|---|
| CTS treatment plus other agents | 55 | 19 | 6 | 2 | 27 | 1 |
| Only CTS | 59 | 24 | 13 | 2 | 20 | 0 |
| CTS ≤ 1 mg/kg | 35 | 13 | 8 | 2 | 12 | 0 |
| CTS >1 and <2 mg/kg | 23 | 9 | 5 | 1 | 8 | 0 |
| CTS ≥ 2 mg/kg | 39 | 14 | 5 | 0 | 20 | 0 |
| Biologic immunosuppressant | 8 | 1 | 2 | 0 | 5 | 0 |
| DMARDS | 14 | 8 | 1 | 0 | 5 | 0 |
| IVIG | 32 | 12 | 3 | 1 | 16 | 0 |
| Plasma exchange | 16 | 2 | 1 | 1 | 11 | 1 |
| Complete response | 6 | 3 | 1 | 0 | 2 | 0 |
| Partial response | 23 | 11 | 7 | 0 | 5 | 0 |
| Stable disease | 15 | 8 | 3 | 0 | 4 | 0 |
| Progressive disease | 15 | 6 | 1 | 1 | 6 | 1 |
CTS, corticosteroids; DMARD, disease-modifying antirheumatic drug; irAEs, immune-related adverse events; IVIG, intravenous immunoglobulin.
Cancer best overall response and outcomes
Of a total of 58 patients whose toxicity outcome was known and objective response to ICI was assessed on the in vivo or post-mortem context (based on autoptic findings), 15 (25.9%) had progressive disease, 15 (25.9%) had stable disease, 23 (39.7%) had a partial response, and in 6 (10.3%) a complete response was observed (these were 3 patients with NSCLC, 2 patients with melanoma, and 1 patient with thymic cancer).
Among patients who had progressive disease, where data on the irAEs outcomes were reported (n = 14), a total of six (42.9%) patients had complete remission of the adverse event, one (7.1%) patient had clinical improvement without complete remission, one (7.1%) patient had no clinical improvement, and six (42.9%) patients died due to the irAEs. Among patients who had stable disease, partial, or complete response (n = 44), a total of 22 (50.0%) patients had complete remission of the irAEs, 11 (25.0%) had clinical improvement without complete remission, and 11 (25.0%) died due to neuromuscular/cardiac event. No differences in terms of irAEs outcomes were observed on the basis of tumor response (chi-square P = 0.3300), albeit patients with controlled disease (response or stable disease) achieved numerically better outcomes compared with patients experiencing disease progression as best response. Outcomes of the irAEs based on tumor response to immunotherapy are summarized in Figure 5 and Table 5.
Additional analyses have been carried out on patients’ medical history, biopsy findings, and autoimmunity markers in Supplementary Analyses a, b, and c, in Supplementary Table S5, and in Supplementary Figure S6, available at https://doi.org/10.1016/j.esmoop.2023.100791.
Discussion
Immunotherapy is acknowledged as a generally safe and well-tolerated approach in cancer treatment, especially when employed as a single agent across various solid tumors.84 Most irAEs are mild or moderate at their onset, and their management is relatively easy due to the use of corticosteroids. Furthermore, across the recent years, physicians have developed extensive expertise in the management of irAEs, thus becoming keen on the timely detection and prompt management of such events, usually with complete remission. However, cardiomuscular irAEs, with specific reference to myocarditis and myositis, still represent an alarming and potentially life-threatening event during treatment with ICIs.85 Indeed, these events are globally rare, as the global incidence of myocarditis is reportedly <1%, although an incidence increase is observed with dual immune checkpoint blockade compared to single-agent blockade. However, their severity has induced international guidelines to include data on epidemiology and management. Notably, while some guidelines specifically refer to myocarditis, in other cases the two events are analyzed together as they are considered part of the same spectrum of toxicity.80, 81, 82
In our work, we carried out a retrospective, multicentric analysis of 18 patients who had received ICIs for solid tumors and experienced myocarditis/myositis; furthermore, we collected suitable case reports of immune-related myocarditis and myositis from the literature, thus reaching a total number of 138 assessable patients. To the best of our knowledge, this is the largest collection of individual cases of myocarditis/myositis during treatment with ICIs for solid tumors. Due to the strict correlation between myocarditis and myositis, we decided to analyze the occurrence of both events.
The first observation of our study is represented by the global outcomes; considering that 53 out of 138 patients (38.4%) died, our findings confirm the severity of myocarditis/myositis. With regard to pre-existing conditions, no specific comorbidity was consistently associated with more or less favorable outcomes, with the exception of the few patients with known autoimmune disease. However, no conclusion can be drawn from this subgroup due to their extremely limited number (n = 6). By contrast, the clinical manifestations of the irAE were significantly associated with outcomes; more specifically, an earlier onset (within the first two cycles) and the coexistence of both myocarditis and myositis were associated with a higher risk of death. Furthermore, our analysis found no differences in the frequency of irAEs in patients treated with single-agent ICI versus dual ICIs. This may be attributed to the fact that, currently, fewer patients are generally treated with ICI combinations (than ICI monotherapy), thus resulting in fewer reported cases.
With regard to diagnostic work-up, the American Society of Clinical Oncology Clinical Practice Guideline suggests that upon the occurrence of myocarditis-related symptoms, ECG, troponin dosing, B-natriuretic peptide (BNP) dosing, echocardiogram, and chest X-ray should be carried out, while cardiac magnetic resonance can be considered.86 The required exams for myositis include CK, transaminases, and lactate dehydrogenase (LDH), while electromyography and auto-antibody testing are suggested. Tissue biopsy is encouraged, when feasible.86 In our analysis, higher troponin values were numerically associated with increased risk of death, albeit the association was not statistically significant; such findings are consistent with current guidelines.81, 82 Additionally, it has been suggested that troponin dosing should be carried out at baseline, before receiving ICIs, and especially when dual blockade is employed;86 this approach might help interpret subsequent troponin values during treatment with ICIs, since different causes of troponin increase might be present, especially among patients with lung cancer.87
In addition to troponin and CK, we tried to assess other circulating biomarkers, including liver transaminases and BNP; unfortunately, we were not able to collect consistent data from the various case reports, especially when we considered the peak values. With regard to autoimmunity, a relevant variety of different auto-antibodies was detected, preventing consistent assessments; furthermore, the baseline value of auto-antibodies before receiving ICIs, which might be an interesting information for screening approaches, was not consistently available.
With regard to instrumental exams, most patients were negative for ischemia at ECG or impaired ejection fraction at echocardiography, and none of these parameters influenced the toxicity outcomes, with the exception of arrhythmia at ECG, which was significantly associated with worse outcomes. It has been observed that patients treated with ICIs might experience severe cardiac/muscular irAEs without prior cardiovascular signs, potentially complicated by the development of rhythm alterations.88 When we evaluated cardiac magnetic resonance findings among patients with myocarditis, we noticed that in most cases the exam was negative, suggesting that the exam might not be pivotal for the management of myocarditis, despite proving to be a highly specific imaging test.89 However, we also noticed that only few patients underwent magnetic resonance, and most of them had a favorable outcome. It is possible that in many cases the magnetic resonance might have been proposed only to patients with stable or mild symptoms, eventually in order to confirm the diagnostic hypothesis of myocarditis, while patients with unfavorable outcomes might have died before undergoing magnetic resonance or the exam might have not been proposed due to a clear diagnosis with other exams. Conversely, most patients who underwent electromyography for myositis had evidence of conduction alterations, which is consistent with symptom development. Finally, a non-negligible proportion of patients underwent tissue biopsy, consistently with guidelines. In most cases, inflammatory infiltrate, mostly based on lymphoid cells, was reported. Overall, the diagnosis of immune-related myositis and myocarditis is therefore necessarily clinical and is based on: (i) correlation with ICI therapy; (ii) clinical symptoms of acute heart failure and/or arrhythmia for myocarditis and of myalgia and/or myasthenic signs for myositis and myasthenia gravis; and (iii) CK and/or troponin elevation.
Once myocarditis and/or myositis are detected, the therapeutic approach suggested by the available guidelines includes ICI interruption and immunomodulatory treatments, starting with corticosteroids. However, apart from interruption (ultimately leading to discontinuation in severe cases), the relatively scarce scientific evidence does not allow to define structured guidelines, and even the revisors state that treatment recommendations are based on anecdotal evidence and the risk of death due to the event. Indeed, most indications rely on the use of high-dose steroids, referral to a specialist such as cardiologists or neurologists, and use of ‘second-line’ treatments when immediate response to high-dose corticosteroids is not observed, including, among others, mycophenolate, infliximab (albeit it is contraindicated in case of moderate-severe cardiac failure), or antithymocyte globulin.86 However, the approaches in common clinical practice may vary on the basis of expertise and readily available immunosuppressive agents.
In this case series, when patients exclusively received corticosteroids, their outcomes were numerically, albeit non-statistically, better than when corticosteroids were followed by other agents. While this finding might appear surprising at the first glance, it is easily explained by the fact that patients not responding to corticosteroids in the first place are subsequently treated with other agents. No other relevant conclusion could be drawn with regard to the effect of immunomodulatory agents on the outcomes of irAEs, mostly due to the high variability of employed therapeutic approaches and different dosing of corticosteroids over time for each individual patient. Furthermore, the analysis of the case reports did not allow a proper identification of the average duration of the steroidal treatment, and consequently it was neither possible to identify its timing nor to estimate the appropriate time to consider treatment failure.
We are aware that this analysis is characterized by several limitations. In the first place, our data have been collected from published case reports and case series; indeed, case reports are usually emblematic in their nature, and do not usually represent the normal distribution of cases among the population. Additionally, case reports from different authors lack the consistency of a systematic data collection from populations of consecutive patients, and different clinicians might have adopted different diagnostic and therapeutic approaches at the irAE onset. Finally, the outcome data of some irAEs have not been defined, as some patients achieved improvement of their irAE but not resolution, while few patients had stability of the event, without improvement or death. However, these issues reflect the limits of the data from the literature.
Theoretically, prospective multicentric studies designed to monitor the onset of irAEs among patients treated with ICIs might address many of the current questions regarding the proper management of myocarditis and myositis. However, such studies would require large numbers of enrolled patients and long follow-up time; until then, the currently available guidelines should be used. In order to provide additional information, we included an algorithm summarizing suggested indications for the management of neuromuscular and/or cardiac irAEs according to the currently available literature (Figure 6).
Figure 6.
Suggested management of immune-related neuromuscular and/or cardiac irAEs. irAEs, immune-related adverse events.
In conclusion, myocarditis and myositis represent rare but potentially lethal irAEs. Our data support the current management guidelines published by international scientific societies. Early irAE onset, concurrent development of both myositis and myocarditis, and occurrence of arrhythmias are associated with worse outcomes and should encourage an aggressive immunomodulatory treatment.
Acknowledgements
The authors thank the following institutions for research grants supporting our studies involving immunotherapy in non-small-cell lung cancer: Italian Ministry of Health (5 × 1000 funds; Ricerca Corrente 2022-2024); Bristol-Myers Squibb (BMS 209-828; fundings in support of the management of patients treated with immune checkpoint inhibitors).
Funding
This work was supported by the Italian Ministry of Health (5 × 1000 funds) [grant number Ricerca Corrente 2022-2024]; and Bristol-Myers Squibb [grant number BMS 209-828].
Disclosure
FS received honoraria for presentations or lectures from Sanofi Genzyme, Roche, BMS, Novartis, Merck, Sunpharma, MSD, Pierre Fabre, advisory boards for Novartis, Philogen, Sunpharma, MSD. GB received contracts from AstraZeneca, GSK, honoraria from Roche and Pierre Fabre, advisory boards for Pierre Fabre and Roche. FDR received honoraria from MSD, Sunpharma, Pierre Fabre, Novartis, BMS. RM received honoraria for presentations or lectures from BMS, Novartis, Ipsen, Pierre Fabre, MSD, Roche, Sanofi, AAA, advisory boards for Novartis, BMS, Ipsen, Pierre Fabre, MSD. ML received honoraria from Roche, Lilly, Novartis, Pfizer, Sandoz, Libbs, Knight, Takeda, advisory boards for Roche, Lilly, Novartis, AstraZeneca, MSD, Exact Sciences, Seagen, Gilead, Pfizer, and received honoraria from AstraZeneca, BMS, Boehringer-Ingelheim, MSD, Roche. All other authors have declared no conflicts of interest.
Supplementary data
References
- 1.Rijavec E., Genova C., Alama A., et al. Role of immunotherapy in the treatment of advanced non-small-cell lung cancer. Future Oncol. 2014;10(1):79–90. doi: 10.2217/fon.13.145. [DOI] [PubMed] [Google Scholar]
- 2.Ribas A., Wolchok J.D. Cancer immunotherapy using checkpoint blockade. Science. 2018;359(6382):1350–1355. doi: 10.1126/science.aar4060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Twomey J.D., Zhang B. Cancer immunotherapy update: FDA-approved checkpoint inhibitors and companion diagnostics. AAPS J. 2021;23(2):39. doi: 10.1208/s12248-021-00574-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Boutros A., Bruzzone M., Tanda E.T., et al. Health-related quality of life in cancer patients treated with immune checkpoint inhibitors in randomised controlled trials: a systematic review and meta-analysis. Eur J Cancer. 2021;159:154–166. doi: 10.1016/j.ejca.2021.10.005. [DOI] [PubMed] [Google Scholar]
- 5.Nishijima T.F., Shachar S.S., Nyrop K.A., Muss H.B. Safety and tolerability of PD-1/PD-L1 inhibitors compared with chemotherapy in patients with advanced cancer: a meta-analysis. Oncologist. 2017;22(4):470–479. doi: 10.1634/theoncologist.2016-0419. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Pardoll D.M. The blockade of immune checkpoints in cancer immunotherapy. Nat Rev Cancer. 2012;12(4):252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Haanen J.B.A.G., Carbonnel F., Robert C., et al. Management of toxicities from immunotherapy: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 2017;28(suppl 4):iv119–iv142. doi: 10.1093/annonc/mdx225. [DOI] [PubMed] [Google Scholar]
- 8.Schneider B.J., Naidoo J., Santomasso B.D., et al. Management of immune-related adverse events in patients treated with immune checkpoint inhibitor therapy: ASCO Guideline Update. JCO. 2021;39(36):4073–4126. doi: 10.1200/JCO.21.01440. [DOI] [PubMed] [Google Scholar]
- 9.Yeung S.J., Qdaisat A., Chaftari P., et al. Diagnosis and management of immune-related adverse effects of immune checkpoint therapy in the emergency department. J Am Coll Emerg Physicians Open. 2020;1(6):1637–1659. doi: 10.1002/emp2.12209. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Kostine M., Finckh A., Bingham C.O., et al. EULAR points to consider for the diagnosis and management of rheumatic immune-related adverse events due to cancer immunotherapy with checkpoint inhibitors. Ann Rheum Dis. 2021;80(1):36–48. doi: 10.1136/annrheumdis-2020-217139. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Guidon A.C., Burton L.B., Chwalisz B.K., et al. Consensus disease definitions for neurologic immune-related adverse events of immune checkpoint inhibitors. J Immunother Cancer. 2021;9(7) doi: 10.1136/jitc-2021-002890. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Neilan T.G., Rothenberg M.L., Amiri-Kordestani L., et al. Myocarditis associated with immune checkpoint inhibitors: an expert consensus on data gaps and a call to action. Oncologist. 2018;23(8):874–878. doi: 10.1634/theoncologist.2018-0157. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Veccia A., Kinspergher S., Grego E., et al. Myositis and myasthenia during nivolumab administration for advanced lung cancer: a case report and review of the literature. Anticancer Drugs. 2020;31(5):540–544. doi: 10.1097/CAD.0000000000000903. [DOI] [PubMed] [Google Scholar]
- 14.Monge C., Maeng H., Brofferio A., et al. Myocarditis in a patient treated with nivolumab and PROSTVAC: a case report. J Immunother Cancer. 2018;6(1):150. doi: 10.1186/s40425-018-0473-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Kudo F., Watanabe Y., Iwai Y., et al. Advanced lung adenocarcinoma with nivolumab-associated dermatomyositis. Intern Med. 2018 Aug 1;57(15):2217–2221. doi: 10.2169/internalmedicine.9381-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.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 doi: 10.1136/bcr-2016-216228. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Kang K.H., Grubb W., Sawlani K., et al. Immune checkpoint-mediated myositis and myasthenia gravis: a case report and review of evaluation and management. Am J Otolaryngol. 2018;39(5):642–645. doi: 10.1016/j.amjoto.2018.06.003. [DOI] [PubMed] [Google Scholar]
- 18.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 Reports Immunol. 2019;2019 doi: 10.1155/2019/2539493. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Chen Y.H., Liu F.C., Hsu C.H., Chian C.F. Nivolumab-induced myasthenia gravis in a patient with squamous cell lung carcinoma: case report. Medicine (Baltimore) 2017;96(27) doi: 10.1097/MD.0000000000007350. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.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(2):155–158. doi: 10.1097/CMR.0000000000000314. [DOI] [PubMed] [Google Scholar]
- 21.Kadota H., Gono T., Shirai Y., Okazaki Y., Takeno M., Kuwana M. Immune checkpoint inhibitor-induced myositis: a case report and literature review. Curr Rheumatol Rep. 2019;21(4):10. doi: 10.1007/s11926-019-0811-3. [DOI] [PubMed] [Google Scholar]
- 22.Kamo H., Hatano T., Kanai K., et al. Pembrolizumab-related systemic myositis involving ocular and hindneck muscles resembling myasthenic gravis: a case report. BMC Neurol. 2019;19(1):184. doi: 10.1186/s12883-019-1416-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Luecke E., Ganzert C., Vielhaber S., et al. Immune checkpoint inhibitor-induced fatal myositis in a patient with squamous cell carcinoma and a history of thymoma. Clin Lung Cancer. 2020;21(4):e246–e249. doi: 10.1016/j.cllc.2020.01.008. [DOI] [PubMed] [Google Scholar]
- 24.Gallegos C., Rottmann D., Nguyen V.Q., Baldassarre L.A. 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(1):yty149. doi: 10.1093/ehjcr/yty149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Kosche C., Stout M., Sosman J., Lukas R.V., Choi J.N. Dermatomyositis in a patient undergoing nivolumab therapy for metastatic melanoma: a case report and review of the literature. Melanoma Res. 2020;30(3):313–316. doi: 10.1097/CMR.0000000000000642. [DOI] [PubMed] [Google Scholar]
- 26.Johnson D.B., Balko J.M., Compton M.L., et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med. 2016;375(18):1749–1755. doi: 10.1056/NEJMoa1609214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Matson D.R., Accola M.A., Rehrauer W.M., Corliss R.F. Fatal myocarditis following treatment with the PD-1 inhibitor nivolumab. J Forensic Sci. 2018;63(3):954–957. doi: 10.1111/1556-4029.13633. [DOI] [PubMed] [Google Scholar]
- 28.Khoury Z.H., Hausner P.F., Idzik-Starr C.L., et al. Combination nivolumab/ipilimumab immunotherapy for melanoma with subsequent unexpected cardiac arrest: a case report and review of literature. J Immunother. 2019;42(8):313–317. doi: 10.1097/CJI.0000000000000282. [DOI] [PubMed] [Google Scholar]
- 29.Fukasawa Y., Sasaki K., Natsume M., et al. Nivolumab-induced myocarditis concomitant with myasthenia gravis. Case Rep Oncol. 2017;10(3):809–812. doi: 10.1159/000479958. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Jain V., Mohebtash M., Rodrigo M.E., Ruiz G., Atkins M.B., Barac A. Autoimmune myocarditis caused by immune checkpoint inhibitors treated with antithymocyte globulin. J Immunother. 2018;41(7):332–335. doi: 10.1097/CJI.0000000000000239. [DOI] [PubMed] [Google Scholar]
- 31.Wang Q., Hu B. Successful therapy for autoimmune myocarditis with pembrolizumab treatment for nasopharyngeal carcinoma. Ann Transl Med. 2019;7(11):247. doi: 10.21037/atm.2019.04.73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Tadokoro T., Keshino E., Makiyama A., et al. Acute lymphocytic myocarditis with anti-PD-1 antibody nivolumab. Circ Heart Fail. 2016;9(10) doi: 10.1161/CIRCHEARTFAILURE.116.003514. [DOI] [PubMed] [Google Scholar]
- 33.Konstantina T., Konstantinos R., Anastasios K., et al. 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. doi: 10.1016/j.lungcan.2019.06.015. [DOI] [PubMed] [Google Scholar]
- 34.Kimura T., Fukushima S., Miyashita A., et al. Myasthenic crisis and polymyositis induced by one dose of nivolumab. Cancer Sci. 2016;107(7):1055–1058. doi: 10.1111/cas.12961. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Diamantopoulos P.T., Tsatsou K., Benopoulou O., Anastasopoulou A., Gogas H. Inflammatory myopathy and axonal neuropathy in a patient with melanoma following pembrolizumab treatment. J Immunother. 2017;40(6):221–223. doi: 10.1097/CJI.0000000000000172. [DOI] [PubMed] [Google Scholar]
- 36.Hardy T., Yin M., Chavez J.A., et al. Acute fatal myocarditis after a single dose of anti-PD-1 immunotherapy, autopsy findings: a case report. Cardiovasc Pathol. 2020;46 doi: 10.1016/j.carpath.2020.107202. [DOI] [PubMed] [Google Scholar]
- 37.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. doi: 10.1016/j.gore.2018.05.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Pushkarevskaya A., Neuberger U., Dimitrakopoulou-Strauss A., Enk A., Hassel J.C. Severe ocular myositis after ipilimumab treatment for melanoma: a report of 2 cases. J Immunother. 2017;40(7):282–285. doi: 10.1097/CJI.0000000000000178. [DOI] [PubMed] [Google Scholar]
- 39.Valenti-Azcarate R., Esparragosa Vazquez I., Toledano Illan C., Idoate Gastearena M.A., Gállego Pérez-Larraya J. Nivolumab and Ipilimumab-induced myositis and myocarditis mimicking a myasthenia gravis presentation. Neuromuscul Disord. 2020;30(1):67–69. doi: 10.1016/j.nmd.2019.10.006. [DOI] [PubMed] [Google Scholar]
- 40.Sutaria R., Patel P., Danve A. Autoimmune myositis and myasthenia gravis resulting from a combination therapy with nivolumab and ipilimumab for metastatic melanoma. Eur J Rheumatol. 2019;6(3):153–154. doi: 10.5152/eurjrheum.2019.18159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Tauber M., Cohen R., Laly P., Josselin L., André T., Mekinian A. Severe necrotizing myositis associated with long term anti-neoplastic efficacy following nivolumab plus ipilimumab combination therapy. Clin Rheumatol. 2019;38(2):601–602. doi: 10.1007/s10067-018-4373-y. [DOI] [PubMed] [Google Scholar]
- 42.John S., Antonia S.J., Rose T.A., et al. Progressive hypoventilation due to mixed CD8+ and CD4+ lymphocytic polymyositis following tremelimumab - durvalumab treatment. J Immunother Cancer. 2017;5(1):54. doi: 10.1186/s40425-017-0258-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Katsume Y., Isawa T., Toi Y., et al. Complete atrioventricular block associated with pembrolizumab-induced acute myocarditis: the need for close cardiac monitoring. Intern Med. 2018;57(21):3157–3162. doi: 10.2169/internalmedicine.0255-17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Hibino M., Maeda K., Horiuchi S., Fukuda M., Kondo T. Pembrolizumab-induced myasthenia gravis with myositis in a patient with lung cancer. Respirol Case Rep. 2018;6(7) doi: 10.1002/rcr2.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Liu Y., Liu Z., Zeng X., et al. Fatal myositis and spontaneous haematoma induced by combined immune checkpoint inhibitor treatment in a patient with pancreatic adenocarcinoma. BMC Cancer. 2019;19(1):1193. doi: 10.1186/s12885-019-6372-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Ansari-Gilani K., Tirumani S.H., Smith D.A., et al. Myocarditis associated with immune checkpoint inhibitor therapy: a case report of three patients. Emerg Radiol. 2020;27(4):455–460. doi: 10.1007/s10140-020-01765-6. [DOI] [PubMed] [Google Scholar]
- 47.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(6) doi: 10.1213/XAA.0000000000001177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Peters N., Greally M., Breen K., Fabre A., Blazkova S. Immunotherapy- a double edged sword; a case of fatal myocarditis and complete response. Ir Med J. 2019;112(5):937. [PubMed] [Google Scholar]
- 49.Robinson S.D., Lai C., Hotton G., Anand G. Life threatening pembrolizumabinduced myositis in a patient treated for advanced adenocarcinoma of the lung. Acute Med. 2019;18(3):197–199. [PubMed] [Google Scholar]
- 50.Norwood T.G., Westbrook B.C., Johnson D.B., et al. Smoldering myocarditis following immune checkpoint blockade. J Immunother Cancer. 2017;5(1):91. doi: 10.1186/s40425-017-0296-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Nakanishi S., Nishida S., Miyazato M., Goya M., Saito S. A case report of nivolumab-induced myasthenia gravis and myositis in a metastatic renal cell carcinoma patient. Urol Case Rep. 2020;29 doi: 10.1016/j.eucr.2019.101105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Heinzerling L., Ott P.A., Hodi F.S., et al. Cardiotoxicity associated with CTLA4 and PD1 blocking immunotherapy. J Immunother Cancer. 2016;4:50. doi: 10.1186/s40425-016-0152-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.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. doi: 10.1186/s40425-015-0057-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Hsu C.Y., Su Y.W., Chen S.C. Sick sinus syndrome associated with anti-programmed cell death-1. J Immunother Cancer. 2018;6(1):72. doi: 10.1186/s40425-018-0388-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Berner A.M., Sharma A., Agarwal S., Al-Sam S., Nathan P. Fatal autoimmune myocarditis with anti-PD-L1 and tyrosine kinase inhibitor therapy for renal cell cancer. Eur J Cancer. 2018;101:287–290. doi: 10.1016/j.ejca.2018.06.021. [DOI] [PubMed] [Google Scholar]
- 56.Narváez J., Juarez-López P., LLuch J., et al. Rheumatic immune-related adverse events in patients on anti-PD-1 inhibitors: fasciitis with myositis syndrome as a new complication of immunotherapy. Autoimmun Rev. 2018;17(10):1040–1045. doi: 10.1016/j.autrev.2018.05.002. [DOI] [PubMed] [Google Scholar]
- 57.Liu S.Y., Huang W.C., Yeh H.I., et al. Sequential blockade of PD-1 and PD-L1 causes fulminant cardiotoxicity-from case report to mouse model validation. Cancers (Basel) 2019;11(4):E580. doi: 10.3390/cancers11040580. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Saibil S.D., Bonilla L., Majeed H., et al. Fatal myocarditis and rhabdomyositis in a patient with stage IV melanoma treated with combined ipilimumab and nivolumab. Curr Oncol. 2019;26(3):e418–e421. doi: 10.3747/co.26.4381. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.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(6):e4869. doi: 10.7759/cureus.4869. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Guo C.W., Alexander M., Dib Y., et al. A closer look at immune-mediated myocarditis in the era of combined checkpoint blockade and targeted therapies. Eur J Cancer. 2020;124:15–24. doi: 10.1016/j.ejca.2019.09.009. [DOI] [PubMed] [Google Scholar]
- 61.Matas-García A., Milisenda J.C., Selva-O’Callaghan A., et al. Emerging PD-1 and PD-1L inhibitors-associated myopathy with a characteristic histopathological pattern. Autoimmun Rev. 2020;19(2) doi: 10.1016/j.autrev.2019.102455. [DOI] [PubMed] [Google Scholar]
- 62.Sobol I., Chen C.L., Mahmood S.S., Borczuk A.C. Histopathologic characterization of myocarditis associated with immune checkpoint inhibitor therapy. Arch Pathol Lab Med. 2020;144(11):1392–1396. doi: 10.5858/arpa.2019-0447-OA. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Agrawal N., Khunger A., Vachhani P., et al. Cardiac toxicity associated with immune checkpoint inhibitors: case series and review of the literature. Case Rep Oncol. 2019;12(1):260–276. doi: 10.1159/000498985. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Möhn N., Sühs K.W., Gingele S., et al. Acute progressive neuropathy-myositis-myasthenia-like syndrome associated with immune-checkpoint inhibitor therapy in patients with metastatic melanoma. Melanoma Res. 2019;29(4):435–440. doi: 10.1097/CMR.0000000000000598. [DOI] [PubMed] [Google Scholar]
- 65.Shirai T., Sano T., Kamijo F., et al. Acetylcholine receptor binding antibody-associated myasthenia gravis and rhabdomyolysis induced by nivolumab in a patient with melanoma. JPN J Clin Oncol. 2016;46(1):86–88. doi: 10.1093/jjco/hyv158. [DOI] [PubMed] [Google Scholar]
- 66.Tomoaia R., Beyer R.Ș., Pop D., Minciună I.A., Dădârlat-Pop A. Fatal association of fulminant myocarditis and rhabdomyolysis after immune checkpoint blockade. Eur J Cancer. 2020;132:224–227. doi: 10.1016/j.ejca.2020.03.003. [DOI] [PubMed] [Google Scholar]
- 67.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 Oncol Stem Cell Ther. 2022;15(1):63–67. doi: 10.1016/j.hemonc.2020.04.006. [DOI] [PubMed] [Google Scholar]
- 68.Puzanov I., Subramanian P., Yatsynovich Y.V., et al. Clinical characteristics, time course, treatment and outcomes of patients with immune checkpoint inhibitor-associated myocarditis. J Immunother Cancer. 2021;9(6) doi: 10.1136/jitc-2021-002553. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Tan R.Y.C., Toh C.K., Takano A. Continued response to one dose of nivolumab complicated by myasthenic crisis and myositis. J Thorac Oncol. 2017;12(7):e90–e91. doi: 10.1016/j.jtho.2017.02.024. [DOI] [PubMed] [Google Scholar]
- 70.Jeyakumar N., Etchegaray M., Henry J., et al. 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 Reports Immunol. 2020;2020 doi: 10.1155/2020/5126717. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Hayakawa N., Kikuchi E., Suzuki S., Oya M. Myasthenia gravis with myositis induced by pembrolizumab therapy in a patient with metastatic urothelial carcinoma. Int Cancer Conf J. 2020;9(3):123–126. doi: 10.1007/s13691-020-00408-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Xing Q., Zhang Z.W., Lin Q.H., et al. 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(5):250. doi: 10.21037/atm.2020.01.79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.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 doi: 10.3389/fimmu.2021.621858. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.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. 2020;11 doi: 10.3389/fimmu.2020.561083. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.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(8):1003–1006. doi: 10.1111/imj.14946. [DOI] [PubMed] [Google Scholar]
- 76.Salido Iniesta M., López López L., Carreras Costa F., Sionis A. A different type of acute myocarditis: a case report of acute autoimmune myocarditis mediated by anti-PD-1 T lymphocyte receptor (pembrolizumab) Eur Heart J Case Rep. 2020;4(5):1–6. doi: 10.1093/ehjcr/ytaa214. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Pagkopoulou E., Simopoulou T., Maragkouli E., Perifanou-Sotiri S., Kotsakis A., Bogdanos D.P. Arthritis and myositis in a patient treated with programmed cell death-1 (PD-1) inhibitor pembrolizumab for lung cancer. Mediterr J Rheumatol. 2020;31(3):355–357. doi: 10.31138/mjr.31.3.355. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Diamantopoulos P.T., Tsatsou K., Benopoulou O., et al. Concomitant development of neurologic and cardiac immune-related adverse effects in patients treated with immune checkpoint inhibitors for melanoma. Melanoma Res. 2020;30(5):484–491. doi: 10.1097/CMR.0000000000000681. [DOI] [PubMed] [Google Scholar]
- 79.Cham J., Ng D., Nicholson L. Durvalumab-induced myocarditis, myositis, and myasthenia gravis: a case report. J Med Case Rep. 2021;15(1):278. doi: 10.1186/s13256-021-02858-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.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 doi: 10.3389/fcvm.2021.797009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Yanase T., Moritoki Y., Kondo H., Ueyama D., Akita H., Yasui T. Myocarditis and myasthenia gravis by combined nivolumab and ipilimumab immunotherapy for renal cell carcinoma: a case report of successful management. Urol Case Rep. 2021;34 doi: 10.1016/j.eucr.2020.101508. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kobayashi M., Saiki M., Omori C., et al. Myositis induced by durvalumab in a patient with non-small cell lung cancer: a case report. Thorac Cancer. 2020;11(12):3614–3617. doi: 10.1111/1759-7714.13709. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Matsui H., Kawai T., Sato Y., 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(5):1070–1074. doi: 10.1536/ihj.20-162. [DOI] [PubMed] [Google Scholar]
- 84.Zullo L., Rossi G., Dellepiane C., et al. Safety and efficacy of immune checkpoint inhibitors in non-small-cell lung cancer: focus on challenging populations. Immunotherapy. 2021;13(6):509–525. doi: 10.2217/imt-2020-0226. [DOI] [PubMed] [Google Scholar]
- 85.Thuny F., Alexandre J., Salem J.E., et al. Management of immune checkpoint inhibitor-induced myocarditis: the French working group’s plea for a pragmatic approach. JACC CardioOncol. 2021;3(1):157–161. doi: 10.1016/j.jaccao.2020.12.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Brahmer J.R., Lacchetti C., Schneider B.J., 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(17):1714–1768. doi: 10.1200/JCO.2017.77.6385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Sarocchi M., Grossi F., Arboscello E., et al. Serial troponin for early detection of nivolumab cardiotoxicity in advanced non-small cell lung cancer patients. Oncologist. 2018;23(8):936–942. doi: 10.1634/theoncologist.2017-0452. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Escudier M., Cautela J., Malissen N., et al. Clinical features, management, and outcomes of immune checkpoint inhibitor-related cardiotoxicity. Circulation. 2017;136(21):2085–2087. doi: 10.1161/CIRCULATIONAHA.117.030571. [DOI] [PubMed] [Google Scholar]
- 89.Cau R., Solinas C., De Silva P., et al. Role of cardiac MRI in the diagnosis of immune checkpoint inhibitor-associated myocarditis. Intl J Cancer. 2022;151(11):1860–1873. doi: 10.1002/ijc.34169. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.






