2,842
views
0
recommends
+1 Recommend
1 collections
    0
    shares

      2023 Journal Citation Reports Journal Impact Factor is 0.9. Scopus Citescore 0.8. 

      Interested in becoming a CVIA published author?

      • Platinum Open Access with no APCs. 
      • Fast peer review/Fast publication online after article acceptance.

      Submissions should be made electronically at: https://mc04.manuscriptcentral.com/cvia-journal.

      Please refer to the Author Guidelines at https://cvia-journal.org/instructions-to-authors/ before submission.

       

      scite_
      0
      0
      0
      0
      Smart Citations
      0
      0
      0
      0
      Citing PublicationsSupportingMentioningContrasting
      View Citations

      See how this article has been cited at scite.ai

      scite shows how a scientific paper has been cited by providing the context of the citation, a classification describing whether it supports, mentions, or contrasts the cited claim, and a label indicating in which section the citation was made.

       
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Diagnosing Immune Checkpoint Inhibitor-Induced Myocarditis: Insights, Challenges, and Uncertainties

      Published
      review-article
      Bookmark

            Abstract

            Recent advancements in immunotherapy have substantially improved overall survival and quality of life among patients with cancer. Notably, immune checkpoint inhibitors (ICIs) have emerged as a revolutionary strategy, particularly in the management of advanced cancers. However, the success of ICIs is accompanied by the challenge of immune-related adverse events. Although rare, cardiovascular adverse events associated with ICIs are associated with high fatality rates and rapid clinical progression, thereby necessitating timely intervention. This review explores the histopathologic characteristics of ICI-induced myocarditis, shedding light on the complexities of diagnosis and management. Several studies examining the histopathologic features of ICI-induced myocarditis have emphasized the roles of macrophages and the potential utility of ancillary tests such as immunohistochemistry. Quantifying CD68+ macrophage abundance may enhance diagnostic sensitivity, thereby providing valuable insights into clinical outcomes. In conclusion, this review underscores the need for a nuanced approach to diagnosing ICI-induced myocarditis. The comprehensive exploration of histopathologic characteristics, ancillary tests, and emerging diagnostic markers provides valuable guidance for practicing pathologists. As the population of ICI-treated patients with cancer continues to grow, optimizing immunohistochemistry panels and refining diagnostic criteria will be crucial to address the unique challenges posed by ICI-induced myocarditis.

            Main article text

            Abbreviations: EmBx, endomyocardial biopsy; ICI, immune checkpoint inhibitor; IHC, Immunohistochemistry.

            Introduction

            Recent advances in cancer therapy have substantially improved overall survival [1] and quality of life [2] among patients with cancer. By harnessing the immune response against tumor cells, immune checkpoint inhibitors (ICI) have revolutionized cancer management strategies, particularly for advanced cancer. However, although ICIs offer a promising avenue to improve clinical outcomes in cancer patients, many of their adverse effects, driven by the same immunologic mechanisms that exert cytotoxic effect on tumor cells, may unleash an uncontrolled immune response to the body’s native cells, thus leading to adverse events.

            Cardiovascular adverse events, including myocarditis, pericarditis, and vasculitis, are uncommon manifestations of ICI-related toxicity and are estimated to affect fewer than 1% of patients [3]. Although rare, ICI-related cardiovascular adverse events are associated with high fatality rates (reported range 25%–40%) [4] and a rapid clinical course, thus requiring emergent intervention and timely diagnosis. However, the definitions of cardiotoxicity can be highly variable and obscure, and diagnosing myocarditis is clinically challenging in patients with cancer under active treatment. The true incidence and prevalence of cardiac adverse events are likely to be underreported [5].

            Endomyocardial biopsy is widely recognized as the gold standard for diagnosing myocarditis (Figure 1) [6]. Despite being considered a relatively safe procedure (complication rate ∼1.9% [7]), its invasiveness is inherently associated with a risk of complications. A lack of expertise (among both treating clinicians and interpreting pathologists) at many institutions further renders endomyocardial biopsy an underused diagnostic tool. The interpretation of endomyocardial biopsies is additionally complicated by variability in tissue preparation methods, the availability of ancillary studies (immunohistochemistry, immunofluorescence, and electron microscopy), and differences in guidelines/reporting (Dallas Criteria in North America vs. European Society of Cardiology [ESC] in Europe) [8]. Furthermore, no guidelines have been established for the histopathologic diagnosis of ICI-induced myocarditis. Several recent studies have described the complex and heterogeneous histopathologic findings in ICI-induced myocarditis, and demonstrated the utility of ancillary tests. The aim of this brief review is to summarize the salient features and provide a reference for practicing pathologists to interpret endomyocardial biopsies in the setting of clinically suspected ICI-associated myocarditis.

            Next follows the figure caption
            Figure 1

            Histopathologic diagnosis of ICI-related myocarditis. Endomyocardial biopsy is considered the gold standard, and the current diagnostic approach integrates the characterization of myocyte injury (by morphology) and immune cell infiltration (by immunohistochemistry).

            Diagnostic Challenge: Heterogeneity in Clinical Presentation

            ICI-associated myocarditis can have a variety of clinical presentations. In mild cases, patients may present with asymptomatic cardiac biomarker elevation (troponin and BNP) [9]. In a recent multicenter Franco-German study, Troponin-T has been shown to be a more sensitive biomarker than Troponin I and creatine kinase in diagnosing ICI-associated myocarditis [10]. The most concerning cases involve severe decompensation with end-organ damage, and common clinical presentations include chest pain, dyspnea, and palpitations [11]. The primary diagnostic challenge in clinical practice is that similar symptoms may be observed in other cardiac (viral myocarditis, coronary artery disease, and pericarditis) and non-cardiac etiologies (pulmonary embolism and pneumonitis) [9]. Although ECG may contribute to diagnosing myocarditis, the findings are usually not specific [12]. Cardiac magnetic resonance imaging is considered the most effective non-invasive study for diagnosing myocarditis [13], and recent updates to the Lake Louise Criteria have improved this method’s diagnostic accuracy [14]. Coronary angiography is frequently performed with endomyocardial biopsy to rule out coronary artery disease. Cardiac magnetic resonance [15] or endocardial voltage mapping [16] guidance may improve the diagnostic yield of endomyocardial biopsy.

            Lessons from Recent Case Series

            An early systemic study compared the histopathology findings of ICI myocarditis (including nine endomyocardial biopsies and one autopsy) with those of cardiac transplant allograft rejection. Champion and Stone [17] successfully used a CD3+ cell count (>50 cells/HPF) to dichotomize the study population into high- and low-grade groups. Furthermore, ICI myocarditis was found to feature more lymphohistiocytic inflammation, rather than a lymphocytic pattern in patients with acute cellular rejection after cardiac transplantation. Interestingly, low-grade ICI myocarditis cases were associated with elevated PD-L1 positive macrophages. PD-L1 staining in myocytes was 100% sensitive in ICI myocarditis cases. However, its specificity was suboptimal: approximately half the control group (grade 2R acute cellular rejection) also showed positive PD-L1 staining. Nonetheless, use of a higher threshold (>10 cells/10 HPF) enhanced its diagnostic utility in distinguishing high-grade ICI (two of three cases) from low-grade ICI myocarditis or acute cellular rejection. Recently, Jimenez et al. [18] have demonstrated that characterizing the presence of interstitial macrophages, in addition to using the Dallas criteria, can enhance the diagnostic yield of ICI-associated myocarditis; these findings correlate well with clinical parameters. Incorporation of histiocytic inflammation (>50 CD68+ cells/HPF) would have led to reclassification of 7/15 patients into the definitive category. These two studies highlight the role of macrophages in diagnosing ICI-associated myocarditis.

            Another multicenter collaboration [19] among six clinical centers in Germany and the United States has further underscored the heterogeneities in the clinical and histopathologic presentation of ICI-related myocarditis. On the basis of clinical criteria, eight patients with melanoma treated with ICI with reported cardiologic adverse events were included in the study. Histopathologic findings were reported on six patients, including four biopsies and two autopsies. Whereas several cases showed non-specific findings, such as interstitial fibrosis or scattered inflammatory cells, cardiomyocyte damage was reported in one biopsy with lymphohistiocytic infiltrates, and one autopsy case exhibited diffuse myocarditis with multinucleated giant cells, lymphocytes, and eosinophils. Although viral testing was not explicitly described in this autopsy case, a more recent study [20] has demonstrated the presence of enterovirus in an endomyocardial biopsy from a patient with ICI-related giant cell myocarditis, with immunohistochemical evidence of viral protein synthesis in cardiomyocytes surrounded by inflammatory infiltrates. A plethora of preclinical and clinical studies have suggested that CTLA-4 inhibition is associated with giant cell myocarditis with predominant CD4+ cell infiltration, whereas PD-1 inhibition is more frequently associated with lymphocytic myocarditis mediated by CD8+ T cell infiltration [21]. These studies have suggested that the complex interactions among the immune system, infections agents, and ICIs in the myocardium remain a challenging area for further investigation.

            Palaskas et al. [22] have proposed a grading system modified from the Dallas criteria and World Heart Federation criteria according to additional immunohistochemistry studies. Similarly to the results reported by Champion and Stone, PD-L1 staining was observed in infiltrating macrophages and damaged cardiomyocytes. Although the proposed classification system did not show a significant stratification of overall mortality, this study demonstrated a spectrum in the density of inflammatory cell infiltrates in patients with clinically suspected ICI myocarditis. With an aggressive institutional protocol aimed at early diagnosis of myocarditis in the setting of rising troponin levels, this single-center study captured milder histologic findings than observed in other case series. Four patients with histologic evidence of myocardial inflammation continued on ICI therapy. Those four patients included three patients with grade 1A (10–20 inflammatory cells/HPF without myocyte injury) and one patient with grade 1B (>20 inflammatory cells/HPF without myocyte injury); no fatalities were reported. Whether this mild inflammation is a harbinger of evolving myocarditis or merely reflects a self-limited process warrants further investigation. Notably, an imminent decision point would be the cessation of ICI therapy and/or immunomodulatory agents in light of mild inflammation. To address these questions, future studies following a multidisciplinary approach will be necessary to maximize the therapeutic effects of ICIs and circumvent the collateral damage from systemic toxicity [23].

            In a smaller cohort, Balanescu et al. [24] have observed extensive lymphohistiocytic inflammation with evidence of myocyte necrosis in two of three patients clinically diagnosed with myocarditis. In areas of cardiomyocyte damage, positive C4d and PD-L1 were present, thus further supporting the utility of these two markers in the assessment of myocyte injury. One of the three patients did not show histopathologic evidence of myocardial inflammation – a finding attributed to tissue heterogeneity and sampling bias.

            A rare syndromic presentation seen in ICI-treated patients is ICI-induced myocarditis with myositis and/or myasthenia gravis overlap syndrome (IM3OS). Pathak et al. [25] have systematically reviewed 60 recent cases reported in prior studies, only 15 of which received cardiac biopsies. Whereas most cases showed lymphocytic infiltrates with predominantly CD4+ and CD8+ T-cells and CD68+ macrophages, cardiomyocyte injury was observed in only six cases, thus again underscoring the known limitations of the Dallas criteria [26]. Endocardial fibrosis [27] has been described in a case of advanced thymic malignancy, although echocardiography did not reveal any imaging evidence of fibrosis. Muscle biopsies from 18 cases showed various degrees of myofiber necrosis and atrophy, with predominantly lymphocytes and macrophages. Interestingly, whereas negative staining for CD20 was seen in two case reports [3, 28], a single-center study [29] from Japan has reported CD 20+ cell infiltration in two patients with a clinical diagnosis of myositis overlapping with myocarditis.

            Roles of Macrophages

            In the native myocardium, most immune cells are tissue resident macrophages [30, 31], which are key regulators of cardiac inflammation [32]. Increases in macrophages are observed in a variety of physiological and pathologic conditions, including aging, hemodynamic stress [33], acute myocardial infarction, heart failure, and COVID-19 [34]. Cardiac macrophages and recruited monocytes play divergent roles, including both exacerbation and attenuation of tissue injury [35]. According to current clinical diagnostic guidelines [28], tissue diagnosis alone is confirmatory for definite myocarditis [11]; however, the Dallas criteria do not explicitly account for the presence of macrophages as a component of inflammatory infiltrates, and the ESC guidelines [36] specify “up to 4 monocytes/mm2 with the presence of CD3 positive T-lymphocytes ≥7 cells/mm2”. Future investigations are warranted to explore and establish the roles of macrophages and associated diagnostic markers, including CD68 and CD163 (a marker with a higher specificity for the macrophage/histiocyte/monocyte lineage [37]) in the workup of ICI-associated myocarditis.

            Myocyte Injury

            Myocyte injury is conventionally assessed according to histologic features of myocytolysis. For instance, the International Society for Heart and Lung Transplantation has described clearing of sarcoplasm/nuclei, enlarged nuclei, and conspicuous nucleoli [38], although in daily practice, identification of the presence of myocyte injury can be subjective and controversial [8, 39]. In a recent trans-Atlantic survey [8], approximately half the participating cardiovascular pathologists indicated that they would make a diagnosis of myocarditis in the absence of myocyte injury.

            Two main systems are used for the diagnosis of myocarditis. The Dallas criteria [40], widely used by North American pathologists, emphasize qualitative, morphologic features (edema, myocyte damage, and inflammatory infiltrates). The cardiovascular pathology community has advocated for redefining the Dallas criteria to encompass additional modalities including immunohistochemistry, polymerase chain reaction, and cardiac antibody assessment [26]. Assessment of myocyte injury has been shown to be a major area of discrepancy in distinguishing borderline myocarditis from myocarditis or non-myocarditis [39].

            In contrast, the ESC guidelines [38] use a more quantitative method with immunohistochemical assessment. Compared with the Dallas Criteria, the ESC guidelines are more conducive to diagnosing ICI-associated myocarditis, although certain revisions might be necessary to account for the lymphohistiocytic cell populations. Immunohistochemical markers for cardiomyocyte injury, such as C4d and C9 [41], and the accumulation of fibronectin and depletion of troponin T [42], might enhance sensitivity in diagnosing myocyte damage and potentially decrease interobserver variability.

            Antigen Presentation

            Infiltrating immune cells are key mediators in the tumor microenvironment. A case report by Johnson et al. [43] included two patients with melanoma who developed fatal myocarditis after treatment with ICI. Pre- and post-treatment biopsies were compared; both patients’ pre-treatment biopsies showed minimal to modest amounts of immune infiltrates, and postmortem assessment revealed prominent lymphocytic infiltrates. Interestingly, one of the two patients had clonal expansion of T-cells; the clonal T-cell populations sampled from the myocardium were identical to those infiltrating the tumor and skeletal muscle. These findings suggested the possibility of a shared epitope/antigen between the tumor and striated muscle. The anti-tumor response might have caused collateral damage in skeletal muscles and cardiomyocytes. Various strategies have been developed to enhance antigen presentation in cancer immunotherapy [44, 45], and the potential for fine-tuning antigen presentation to minimize off-target effects while designing more tumor-specific immune response merits further investigation. In such endeavors, an organoid-based screening tool [46] and engineered heart tissue [47] might provide a physiologic platform recapitulating the complex interactions among tumors, immune cells, and the myocardium.

            Immune Profiling: Bench to Bedside

            A pioneering mechanistic study has investigated the immunoproteasome, a key modulator in ICI-related myocarditis, acting through a troponin I-directed autoimmune pathway [48]. Through mitigating cytokine production, immunoproteasome inhibitors have been shown to attenuate the ICI-related heart-specific autoimmune response in mouse models. In two patients with ICI-related myocarditis, bulk RNA-sequencing analyses have demonstrated enrichment of various markers indicating immunoproteasome expression, TLR-mediated activation of monocytes, chemokine and cytokine responses, as well as T- and B-cell activation.

            Recent advances in single-cell profiling techniques, including time-of-flight mass cytometry (CyTOF), single-cell RNA sequencing (scRNA-seq), single-cell T-cell receptor sequencing (scTCR-seq), and cellular indexing of transcriptomes and epitopes by sequencing (CITE-seq), offer further options to study tumor cells and their immune microenvironment in detail [49]. By bridging gene expression data and the histologic characteristics of cell groups in a tissue section, spatially resolved transcriptomics and proteomics will provide another powerful tool for deciphering the molecular mechanisms of ICI-induce myocarditis and personalizing onco-immunotherapy.

            Conclusion

            Although most ICI-associated cardiac adverse events are characterized within the first 90 days of therapy initiation, increasing evidence demonstrates late-onset cardiac adverse events with more pronounced left ventricular systolic dysfunction and heart failure, and less frequent supraventricular arrhythmias [50]. Fulminant myocarditis has been reported as many as 2 years after ICI initiation [51]. Although endomyocardial biopsy is usually performed in patients with high clinical suspicion for ICI-induced myocarditis, other differential diagnoses including immune-related sarcoidosis [52], viral myocarditis [12], and vaccination [53] must be considered.

            Optimizing the IHC panel for ICI-induced myocarditis is crucial to address the rising prevalence of ICI-treated patients with cancer in an aging population. Although no consensus exists, many studies [17, 18] have described lymphohistiocytic inflammation as the key finding in ICI-induced myocarditis. Macrophage markers (CD68, CD163, and PU.1) and lymphocytic markers (CD3 and CD20) may help reveal the inflammatory cell population. Overall, collaborative efforts among cardiovascular pathologists, clinicians, radiologists, and scientists are needed to establish a robust, consensus-driven diagnostic framework to diagnose ICI-induced myocarditis and ultimately improve the clinical outcomes of patients with cancer.

            Acknowledgements

            The authors thank Dr Xiongbin Lu at Indiana University for technical assistance.

            Conflict of Interest

            Haodong Xu is the editorial board member of Cardiovascular Innovations and Applications. Haodong Xu was not involved in the peer review or decision-making process of the manuscript. The other authors have no conflict of interest to declare.

            Citation Information

            References

            1. , , , , , , et al. Association of sex, age, and Eastern Cooperative Oncology Group performance status with survival benefit of cancer immunotherapy in randomized clinical trials: a systematic review and meta-analysis. JAMA Netw Open 2020;3(8):e2012534.

            2. , , , , , . Association of quality-of-life outcomes in cancer drug trials with survival outcomes and drug class. JAMA Oncol 2022;8(6):879–86.

            3. Fulminant myocarditis with combination immune checkpoint blockade | NEJM. Accessed 2023 Nov 1. Available from: https://www.nejm.org/doi/full/10.1056/NEJMoa1609214 .

            4. , , , , , , et al. Diagnostic value of the International Society of Cardio-Oncology definition for suspected immune checkpoint inhibitor–associated myocarditis. J Am Heart Assoc 2023;12(8):e029211.

            5. , , , , , , et al. Managing toxicities associated with immune checkpoint inhibitors: consensus recommendations from the Society for Immunotherapy of Cancer (SITC) Toxicity Management Working Group. J Immunother Cancer 2017;5(1):95.

            6. , , , . Diagnostic use of the endomyocardial biopsy: a consensus statement. Virchows Arch 2013;463(1):1–5.

            7. , , , , , , et al. Endomyocardial biopsy via the femoral access - still safe and valuable diagnostic tool. BMC Cardiovasc Disord 2016;16(1):222.

            8. , , , , , , et al. Diagnosing myocarditis in endomyocardial biopsies: survey of current practice. Cardiovasc Pathol 2023;64:107494.

            9. , , , , , , et al. Clinical strategy for the diagnosis and treatment of immune checkpoint inhibitor–associated myocarditis: a narrative review. JAMA Cardiol 2021;6(11):1329–37.

            10. , , , , , , et al. Cardiomuscular biomarkers in the diagnosis and prognostication of immune checkpoint inhibitor myocarditis. Circulation 2023;148(6):473–86.

            11. , , , , , , et al. Myocarditis in the setting of cancer therapeutics. Circulation 2019;140(1):80–91.

            12. , , , , . Immune checkpoint inhibitor myocarditis: pathophysiological characteristics, diagnosis, and treatment. J Am Heart Assoc 2020;9(2):e013757.

            13. , , . Diagnostic approach for suspected acute myocarditis: considerations for standardization and broadening clinical spectrum. J Am Heart Assoc 2023;12(17):e031454.

            14. , , , , , , et al. Cardiovascular magnetic resonance in nonischemic myocardial inflammation. J Am Coll Cardiol 2018;72(24):3158–76.

            15. , , , , , , et al. Real-time magnetic resonance imaging guidance improves the diagnostic yield of endomyocardial biopsy. JACC Basic Transl Sci 2016;1(5):376–83.

            16. , , , , , , et al. Diagnostic yield of electroanatomic voltage mapping in guiding endomyocardial biopsies. Circulation 2020;142(13):1249–60.

            17. , . Immune checkpoint inhibitor associated myocarditis occurs in both high-grade and low-grade forms. Mod Pathol 2020;33(1):99–108.

            18. , , , , , , et al. Clinicopathological classification of immune checkpoint inhibitor-associated myocarditis: possible refinement by measuring macrophage abundance. Cardiooncology 2023;9(1):14.

            19. , , , , , , et al. Cardiotoxicity associated with CTLA4 and PD1 blocking immunotherapy. J Immunother Cancer 2016;4(1):50.

            20. , , , , , , et al. Late-onset giant cell myocarditis due to enterovirus during treatment with immune checkpoint inhibitors. JACC CardioOncology 2020;2(3):511–4.

            21. , , , , , , et al. Immune checkpoint inhibitors mediated lymphocytic and giant cell myocarditis: uncovering etiological mechanisms. Front Cardiovasc Med 2021;8:721333.

            22. , , , , , , et al. Immune checkpoint inhibitor myocarditis: elucidating the spectrum of disease through endomyocardial biopsy. Eur J Heart Fail 2021;23(10):1725–35.

            23. , . Winning the battle, but losing the war: mechanisms and morphology of cancer-therapy-associated cardiovascular toxicity. Cardiovasc Pathol 2017;30:55–63.

            24. , , , , , , et al. Immunomodulatory treatment of immune checkpoint inhibitor-induced myocarditis: pathway toward precision-based therapy. Cardiovasc Pathol 2020;47:107211.

            25. , , , , , . Immune checkpoint inhibitor–induced myocarditis with myositis/myasthenia gravis overlap syndrome: a systematic review of cases. Oncologist 2021;26(12):1052–61.

            26. . Diagnosis of myocarditis: death of Dallas criteria. Circulation 2006;113(4):593–5.

            27. , , , , , , et al. Checkpoint inhibitor-induced myocarditis and myasthenia gravis in a recurrent/metastatic thymic carcinoma patient: a case report. Eur Heart J Case Rep 2020;4(3):1–8.

            28. , , , , , , et al. Fatal myocarditis and rhabdomyositis in a patient with stage IV melanoma treated with combined ipilimumab and nivolumab. Curr Oncol 2019;26(3):e418–21.

            29. , , , , , , et al. Immune checkpoint inhibitor-related myositis overlapping with myocarditis: an institutional case series and a systematic review of literature. Front Pharmacol 2022;13:884776.

            30. , , , , , , et al. Transcriptional and cellular diversity of the human heart. Circulation 2020;142(5):466–82.

            31. , , , , , , et al. Single-cell transcriptomics reveals cell-type-specific diversification in human heart failure. Nat Cardiovasc Res 2022;1(3):263–80.

            32. , , , . The quest for new approaches in myocarditis and inflammatory cardiomyopathy. J Am Coll Cardiol 2016;68(21):2348–64.

            33. , . The dynamic role of cardiac macrophages in aging and disease. Curr Cardiol Rep 2022;24:925–33.

            34. , , , , , , et al. COVID-19 and myocarditis: a systematic review and overview of current challenges. Heart Fail Rev 2022;27(1):251–61.

            35. , , . Exploring cardiac macrophage heterogeneity in the healthy and diseased myocardium. Curr Opin Immunol 2021;68:54–63.

            36. , , , , , , et al. Current state of knowledge on aetiology, diagnosis, management, and therapy of myocarditis: a position statement of the European Society of Cardiology Working Group on Myocardial and Pericardial Diseases. Eur Heart J 2013;34(33):2636–48.

            37. , , . CD163: a specific marker of macrophages in paraffin-embedded tissue samples. Am J Clin Pathol 2004;122(5):794–801.

            38. , , , , , , et al. Revision of the 1990 working formulation for the standardization of nomenclature in the diagnosis of heart rejection. J Heart Lung Transplant 2005;24(11):1710–20.

            39. , , , , , , et al. Myocarditis and endomyocardial biopsy: achieving consensus diagnosis on 100 cases. Cardiovasc Pathol 2023;62:107492.

            40. . Myocarditis: the Dallas criteria. Hum Pathol 1987;18(6):619–24.

            41. , , , , , . The utility of C4d, C9, and troponin T immunohistochemistry in acute myocardial infarction. Arch Pathol Lab Med 2010;134:256–63.

            42. , . The role of immunohistochemical markers in the diagnosis of early myocardial infarction. Cureus 2022;14(2):e22391.

            43. , , , , , , et al. Fulminant myocarditis with combination immune checkpoint blockade. N Engl J Med 2016;375(18):1749–55.

            44. , , , , , , et al. A facile approach to enhance antigen response for personalized cancer vaccination. Nat Mater 2018;17(6):528–34.

            45. , , . Engineering nanoparticles toward the modulation of emerging cancer immunotherapy. Adv Healthc Mater 2021;10(5):2000845.

            46. , , , , , , et al. An organoid-based screen for epigenetic inhibitors that stimulate antigen presentation and potentiate T-cell-mediated cytotoxicity. Nat Biomed Eng 2021;5(11):1320–35.

            47. , , , . Engineered heart tissues and induced pluripotent stem cells: macro- and microstructures for disease modeling, drug screening, and translational studies. Adv Drug Deliv Rev 2016;96:234–44.

            48. , , , , , , et al. Heart-specific immune responses in an animal model of autoimmune-related myocarditis mitigated by an immunoproteasome inhibitor and genetic ablation. Circulation 2020;141(23):1885–902.

            49. , , , , , , et al. Applications of single-cell and bulk RNA sequencing in onco-immunology. Eur J Cancer 2021;149:193–210.

            50. , , , , , , et al. Late cardiac adverse events in patients with cancer treated with immune checkpoint inhibitors. J Immunother Cancer 2020;8(1):e000261.

            51. , , , , , , et al. Late-onset immunotherapy-induced myocarditis 2 years after checkpoint inhibitor initiation. JACC CardioOncol 2022;4(5):727–30.

            52. , , , , , , et al. Immune-related sarcoidosis observed in combination ipilimumab and nivolumab therapy. JAAD Case Rep 2016;2(3):264–8.

            53. , . Lymphohistiocytic myocarditis possibly due to moderna mRNA-1273 vaccine. Am J Clin Pathol 2022;158(2):167–72.

            Author and article information

            Journal
            CVIA
            Cardiovascular Innovations and Applications
            CVIA
            Compuscript (Ireland )
            2009-8782
            2009-8618
            03 August 2024
            : 9
            : 1
            : e936
            Affiliations
            [1] 1Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
            [2] 2Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA
            [3] 3Department of Pathology, Wake Forest University School of Medicine, Winston Salem, NC, USA
            Author notes
            Correspondence: Liang Lu, Department of Pathology and Laboratory Medicine, Dartmouth Hitchcock Medical Center, One Medical Center Drive, Lebanon, NH 03756, USA, Tel: +1-603-650-7211, Fax: +1-603-650-7214, E-mail: Liang.Lu@ 123456hitchcock.org
            Article
            cvia.2024.0039
            10.15212/CVIA.2024.0039
            d6f78ef7-5c2d-4f8e-b88a-0471dd164c63
            2024 The Authors.

            Creative Commons Attribution 4.0 International License

            History
            : 20 January 2024
            : 14 April 2024
            : 09 June 2024
            Page count
            Figures: 1, References: 53, Pages: 8
            Categories
            Review

            General medicine,Medicine,Geriatric medicine,Transplantation,Cardiovascular Medicine,Anesthesiology & Pain management
            onco-cardiology/cardio-oncology,immune checkpoint inhibitor,Cardiovascular histopathology

            Comments

            Comment on this article