1
|
Li C, Jin Y, Ma J, Sun R, Huang J, Qi Y, Wang Q, Yu J. Diagnostic value of ROCK2 protein in immune checkpoint inhibitors-associated myocarditis. Int J Cardiol 2025; 427:133104. [PMID: 40037484 DOI: 10.1016/j.ijcard.2025.133104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/17/2025] [Accepted: 02/26/2025] [Indexed: 03/06/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have advanced cancer treatment but are associated with serious cardiovascular side effects, including myocarditis, which is challenging to diagnose due to limited specificity of current biomarkers. Rho-associated kinase 2 (ROCK2) may play a role in myocarditis pathogenesis by mediating inflammation and myocardial remodeling. This study investigates the potential of ROCK2 protein detection as a diagnostic marker for ICIs-associated myocarditis. OBJECTIVES To evaluate ROCK2 protein levels in patients with ICIs-associated myocarditis and assess its diagnostic value, providing insights for improved identification and management of this condition. METHODS We conducted a study with 10 ICIs-treated gastric cancer patients diagnosed with myocarditis and a control group of 10 similar ICIs-treated patients without myocarditis. ROCK2 levels and inflammatory markers were measured via ELISA, and clinical assessments included cardiac imaging, electrocardiography, and echocardiography. Statistical analysis of group differences used independent t-tests and chi-square tests. RESULTS ROCK2 expression was significantly higher in myocarditis patients compared to controls (p < 0.001), alongside elevated inflammatory markers, specifically hypersensitive C-reactive protein (hs-CRP), interleukins1β (IL-1β) and IL-17. Diagnostic myocardial markers such as N-terminal pro B-type natriuretic peptide (NT-proBNP), and soluble suppression of tumorigenicity 2 (sST2) also showed substantial elevation. Following treatment discontinuation and glucocorticoid administration, both ROCK2 levels and inflammatory indicators declined. CONCLUSIONS Elevated ROCK2 protein levels could serve as a promising biomarker for ICIs-associated myocarditis. ROCK2 detection, combined with traditional markers, may enhance diagnostic accuracy. Further studies are warranted to explore ROCK2 inhibition as a potential therapeutic strategy for managing ICIs-associated myocarditis.
Collapse
Affiliation(s)
- Caie Li
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Yucheng Jin
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Jie Ma
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Runmin Sun
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Jiawang Huang
- The Second Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yali Qi
- Department of Oncology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Qiongying Wang
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China
| | - Jing Yu
- Department of Cardiology, The Second Hospital of Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
2
|
Wang G, Yan W, Cai J, Zhang F, Lv T, Ye M. Durvalumab-induced organizing pneumonia in extensive-stage small cell lung cancer: A case report and literature review. Radiol Case Rep 2025; 20:2253-2257. [PMID: 40129823 PMCID: PMC11930415 DOI: 10.1016/j.radcr.2025.01.072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/04/2025] [Accepted: 01/23/2025] [Indexed: 03/26/2025] Open
Abstract
The etoposide-carboplatin and anti-PD-L1 combination has become the standard-of-care for patients with extensive-stage small cell lung cancer (ES-SCLC). This combinational strategy is well tolerated with manageable immune-related adverse effects (irAEs). In this report, we presented a rare immediate irAE after one course of anti-tumor treatment. The patient with ES-SCLC was treated with first-line etoposide-carboplatin chemotherapy and Durvalumab immunotherapy. After one cycle of indicated treatment, the patient developed persistent high-grade fever with extensive consolidation, surrounding by ground glass opacifications. The lesions did not respond to empirical antibiotics and the results for pathogen testing were negative. Histological analysis of biopsy sample yielded organizing pneumonia that was very likely to associate with Durvalumab treatment. The patient was therefore treated with prednisolone that resulted in a rapid radiological improvement. The reporting of this case is imperative for informing acute onset of irAE in patients with ES-SCLC treated with anti-PD-L1 immunotherapy. Differential diagnosis of infection, tumor progression and exacerbation of underlying illness should be considered before the initiation of prednisolone therapy.
Collapse
Affiliation(s)
- Guoxin Wang
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wenjie Yan
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jun Cai
- Department of Radiology, Medical Imaging Center, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Fang Zhang
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Tangfeng Lv
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Mingxiang Ye
- Department of Respiratory Medicine, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
3
|
Ruiz JI, Zhao B, Palaskas N, Deswal A, Zhao H, McQuade J, Suarez-Almazor ME. Major Adverse Cardiovascular Events in Patients with Melanoma Receiving Immune Checkpoint Inhibitors. JOURNAL OF IMMUNOTHERAPY AND PRECISION ONCOLOGY 2025; 8:1-9. [PMID: 40224298 PMCID: PMC11987079 DOI: 10.36401/jipo-24-31] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 02/01/2024] [Accepted: 02/20/2025] [Indexed: 04/15/2025]
Abstract
Introduction Immune checkpoint inhibitors (ICIs) might increase the risk of major adverse cardiovascular events (MACEs). This study aimed to evaluate the risk of MACE in patients with melanoma after ICI initiation. Methods We conducted a before-after cohort study using claims data from Optum's deidentified Clinformatics Data Mart Database. We included adult patients with melanoma who received any approved ICI between 2011 and 2021 with a minimum of 12 months of observable data before ICI. The main outcome was time to first MACE (myocardial infarction, coronary revascularization, stroke, heart failure hospitalization) and rate of MACE before and after ICI, ascertained using International Classification of Diseases, 9th/10th Revision diagnostic codes. Hazard ratio (HR) and incidence rate ratio (IRR) were calculated. Results We identified 4024 patients with ICI-treated melanoma. Mean age was 67.4 years (SD 14.1), 36% were women; 3066 (76.2%) received monotherapy and 958 (23.8%) combination immunotherapy. A total of 160 (4%) patients had a MACE before ICI and 224 (5.6%) after ICI (HR, 1.76; 95% CI, 1.47-2.12). MACE rate in the year before ICI was 56.16 per 1000 person-years compared with 80.91 per 1000 person-years the year after ICI (IRR, 1.44; 95% CI, 1.21-1.72). Ten cases of myocarditis were observed after ICI, 50% of them had a MACE. Risk factors associated with MACE after ICI were prior MACE, other cardiovascular conditions, hypertension, and older age. Glucocorticoid use was not associated with MACE. Conclusion Our results suggest that ICI might increase the risk of MACE in patients with melanoma during the first year after ICI.
Collapse
Affiliation(s)
- Juan I. Ruiz
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bo Zhao
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicolas Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hui Zhao
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer McQuade
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria E. Suarez-Almazor
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
4
|
Ryan TD, Bates JE, Kinahan KE, Leger KJ, Mulrooney DA, Narayan HK, Ness K, Okwuosa TM, Rainusso NC, Steinberger J, Armenian SH. Cardiovascular Toxicity in Patients Treated for Childhood Cancer: A Scientific Statement From the American Heart Association. Circulation 2025; 151:e926-e943. [PMID: 40104841 DOI: 10.1161/cir.0000000000001308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
The field of cardio-oncology has expanded over the past 2 decades to address the ever-increasing issues related to cardiovascular disease in patients with cancer and survivors. There is increasing recognition that nearly all cancer treatments pose some short- or long-term risk for development of cardiovascular disease and that pediatric patients with cancer may be especially vulnerable to cardiovascular disease because of young age at treatment and expected long life span afterward. Anthracycline chemotherapy and chest-directed radiotherapy are the most well-studied cardiotoxic therapies, and dose reduction, use of cardioprotection for anthracyclines, and modern radiotherapy approaches have contributed to improved cardiovascular outcomes for survivors. Newer treatments such as small-molecule inhibitors, antibody-based cytotoxic therapy, and immunotherapy have expanded options for previously difficult-to-treat cancers but have also revealed new cardiotoxic profiles. Application of effective surveillance strategies in patients with cancer and survivors has been a focus of practitioners and researchers, whereas the prevention and treatment of extant cardiovascular disease is still developing. Incorporation of new strategies in an equitable manner and appropriate transition from pediatric to adult care will greatly influence long-term health-related outcomes in the growing population of childhood cancer survivors at risk for cardiovascular disease.
Collapse
|
5
|
Cui M, Zhou M, Zhou L, Zhou G, Liu Y. Tertiary lymphoid structures achieve 'cold' to 'hot' transition by remodeling the cold tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189312. [PMID: 40189114 DOI: 10.1016/j.bbcan.2025.189312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 03/30/2025] [Accepted: 03/31/2025] [Indexed: 04/10/2025]
Abstract
Immune checkpoint blockade (ICB) therapies have demonstrated significant clinical efficacy in immune-infiltrated tumors such as melanoma and non-small cell lung cancer. However, "cold tumors"-including ovarian cancer, pancreatic cancer, and gliomas-exhibit insufficient immune infiltration, leading to poor therapeutic responses to ICBs and limited improvement in patient prognosis. Recent studies have shown that tumor-associated tertiary lymphoid structures (TLSs) can induce strong local immune responses within the tumor microenvironment (TME), serving as important biological markers for predicting ICB therapy efficacy. Notably, preclinical and clinical studies on cold tumors have confirmed that TLSs can potently enhance ICB efficacy through TME remodeling-a breakthrough that has attracted considerable attention. Here, we systematically examine the immunological profile of cold tumors and decipher the mechanistic basis for their impaired immune cell infiltration. We further delineate the distinctive features of tumor-associated TLSs in generating antitumor immunity and establish criteria for their identification. Significantly, we emphasize the unique capability of TLSs to reprogram the immunosuppressive tumor microenvironment characteristic of cold tumors. Based on these insights, we evaluate clinical evidence supporting TLS-mediated enhancement of ICB efficacy and discuss emerging strategies for exogenous TLSs induction.
Collapse
Affiliation(s)
- Mengke Cui
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Mengfan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Lu Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China
| | - Gan Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China; National Institution of Drug Clinical Trial, Xiangya Hospital, Central South University, 110 Xiangya Road, Changsha, Hunan 410008, PR China.
| | - Yingzi Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, 87 Xiangya Road Changsha, 410008, PR China; National Laboratory of Medical Genetics, Central South University, Changsha 410078, PR China.
| |
Collapse
|
6
|
Hachem AM, Desai A, Beinart N, Ostos-Mendoza KC, Rodriguez ASL, de Leon Derby RD, Ebrahimi S, Palaskas NL. Updates in Diagnosis and Treatment of Immune Checkpoint Inhibitor Myocarditis. Curr Cardiol Rep 2025; 27:78. [PMID: 40178703 DOI: 10.1007/s11886-025-02232-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/25/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE OF REVIEW To provide an update on the literature regarding diagnosis and management of immune checkpoint inhibitor myocarditis. RECENT FINDINGS The diagnosis of immune checkpoint inhibitor myocarditis has evolved to include more reliance on performing endomyocardial biopsy to clarify the diagnosis in selected cases. Additionally, there is recognition of a spectrum of disease both clinically and on endomyocardial biopsy suggesting that there is a range of severity from mild to fulminant. The treatment of immune checkpoint inhibitor myocarditis is shifting towards increased use of additional immunosuppressive medications as steroid sparing agents. There are increased studies including two randomized controlled trials evaluating abatacept in the treatment of immune checkpoint inhibitor myocarditis. This review summarizes the latest literature regarding diagnosis and management of immune checkpoint inhibitor myocarditis and provides our experience and approach to this rare but potentially fatal condition.
Collapse
Affiliation(s)
| | - Aditya Desai
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Lakeside School, Seattle, WA, USA
| | - Noah Beinart
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keila C Ostos-Mendoza
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrey, Nuevo León, 64710, México
| | - Ana Sofia Lopez Rodriguez
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrey, Nuevo León, 64710, México
| | - Regina Diaz de Leon Derby
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Tecnologico de Monterrey, School of Medicine and Health Sciences, Monterrey, Nuevo León, 64710, México
| | - Sara Ebrahimi
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicolas L Palaskas
- University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1451, Houston, TX, 77030, USA.
| |
Collapse
|
7
|
Braghieri L, Gharaibeh A, Nkashama L, Abushouk A, Abushawer O, Mehdizadeh‐Shrifi A, Honnekeri B, Calabrese C, Menon V, Funchain P, Collier P, Sadler D, Moudgil R. Long-term cardiovascular outcomes of immune checkpoint inhibitor-related myocarditis: A large single-centre analysis. ESC Heart Fail 2025; 12:1237-1245. [PMID: 39482568 PMCID: PMC11911570 DOI: 10.1002/ehf2.15131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 09/11/2024] [Accepted: 10/03/2024] [Indexed: 11/03/2024] Open
Abstract
AIMS Immune checkpoint inhibitors (ICI) are the cornerstone of modern oncology; however, side effects such as ICI-related myocarditis (irM) can be fatal. Recently, Bonaca proposed criteria for irM; however, it is unknown if they correlate well with cardiovascular (CV) ICI-related adverse events. Additionally, whether incident irM portends worse long-term CV outcomes remains unclear. We aimed to determine the incidence of long-term CV comorbidities and CV mortality among irM patients. PATIENTS AND METHODS The ICI-related adverse event (irAE) registry was queried to identify irM patients by using Bonaca criteria. Random controls were selected after excluding patients with other concomitant irAEs. Patients' demographics, comorbidities and myocarditis presenting features were gathered. Outcomes included 2-year freedom from CV comorbidities (composite of atrial fibrillation, stroke, myocardial infarction and heart failure) and freedom from CV death. IrM was treated as a time-varying covariate. RESULTS Seventy-six patients developed irM at a median of 167 days (mean age 69, 63.2% male, 47% lung cancer). Majority of patients had new wall motion abnormalities or EKG changes on presentation. Mean LVEF was 43%, median peak TnT was 0.81, and median NTproBNP was 2057 at irM onset. Two-year freedom from CV comorbidities (67% vs 86.8%, P < 0.001) and death (93.4% vs 99.3%, P = 0.003) was lower among irM patients. Incident irM was an independent predictor of CV death (HR 8.28, P = 0.048), but not CV comorbidities (HR 2.21, P = 0.080). CONCLUSIONS This is the largest case-control study on irM highlighting worse long-term CV outcomes. Future studies are needed to establish appropriate therapeutic strategies and efficient screening strategies for irM survivors.
Collapse
Affiliation(s)
- Lorenzo Braghieri
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Ahmad Gharaibeh
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Lubika Nkashama
- Department of Internal MedicineWashU/Barnes‐Jewish HospitalSt. LouisMissouriUSA
| | | | - Osama Abushawer
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | | | - Bianca Honnekeri
- Department of Internal MedicineCleveland Clinic FoundationClevelandOhioUSA
| | - Cassandra Calabrese
- Department of Rheumatologic and Immunologic DiseaseCleveland Clinic FoundationClevelandOhioUSA
| | - Venu Menon
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| | - Pauline Funchain
- Department of Hematology & OncologyTaussig Cancer Center, Cleveland ClinicClevelandOhioUSA
| | - Patrick Collier
- Department of Cardiovascular Medicine, Division of Cardiac Imaging, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| | - Diego Sadler
- Department of Cardiovascular Medicine, Division of Cardiac Imaging, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationWestonFloridaUSA
| | - Rohit Moudgil
- Department of Cardiovascular Medicine, Heart, Vascular and Thoracic InstituteCleveland Clinic FoundationClevelandOhioUSA
| |
Collapse
|
8
|
Jensen G, Wang X, Kuempel J, Palaskas N, Chen Z, Yu W, Chen Y, Mohammad H, Luo W, Chang J. Immune checkpoint inhibitor-associated myocarditis: a historical and comprehensive review. Am J Physiol Heart Circ Physiol 2025; 328:H734-H751. [PMID: 39925096 DOI: 10.1152/ajpheart.00687.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 11/13/2024] [Accepted: 01/03/2025] [Indexed: 02/11/2025]
Abstract
The most fatal side effect associated with revolutionary immune checkpoint inhibitor (ICI) cancer therapies is myocarditis, a rare and devastating complication with a mortality rate approaching 40%. This review comprehensively examines the limited knowledge surrounding this recently recognized condition, emphasizing the absence of evidence-based therapeutic strategies, diagnostic modalities, and reliable biomarkers that hinder effective management. It explores advancements in preclinical models that are uncovering disease mechanisms and enabling the identification of therapeutic targets. These efforts have informed the design of early clinical trials aimed at reducing mortality. With the growing prevalence of ICI therapies in oncology, addressing critical gaps, such as long-term outcomes and risk stratification, has become increasingly urgent. By synthesizing current evidence, this work seeks to enhance understanding and guide the development of strategies to improve patient outcomes and ensure the continued safe use of ICIs in cancer care.
Collapse
Affiliation(s)
- Garrett Jensen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Xinjie Wang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jacob Kuempel
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Nicolas Palaskas
- Department of Cardiology, MD Anderson Cancer Center, Houston, Texas, United States
| | - Zhishi Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Wei Yu
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Yanping Chen
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Haseeb Mohammad
- Texas A&M University College of Medicine, Houston, Texas, United States
| | - Weijia Luo
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| | - Jiang Chang
- Institute for Biosciences and Technology, Center for Genomics and Precision Medicine, Texas A&M University, Houston, Texas, United States
| |
Collapse
|
9
|
Jimenez J, Amrute J, Ma P, Wang X, Das S, Dai R, Komaru Y, Herrlich A, Mack M, Lavine KJ. The immune checkpoint regulator CD40 potentiates myocardial inflammation. NATURE CARDIOVASCULAR RESEARCH 2025; 4:458-472. [PMID: 40217124 DOI: 10.1038/s44161-025-00633-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 03/05/2025] [Indexed: 04/15/2025]
Abstract
Immune checkpoint therapeutics including CD40 agonists have tremendous promise to elicit antitumor responses in patients resistant to current therapies. Conventional immune checkpoint inhibitors (PD-1, PD-L1 and CTLA-4 antagonists) are associated with serious adverse cardiac events including life-threatening myocarditis. However, little is known regarding the potential for CD40 agonists to trigger myocardial inflammation or myocarditis. Here we leverage genetic mouse models, single-cell sequencing and cell depletion studies to show that an anti-CD40 agonist antibody reshapes the cardiac immune landscape through activation of CCR2+ macrophages and subsequent recruitment of effector memory CD8+ T cells. We identify a positive feedback loop between CCR2+ macrophages (positive for the chemokine receptor CCR2) and CD8+ T cells driven by IL-12b, TNF and IFNγ signaling that promotes myocardial inflammation and show that previous exposure to CD40 agonists sensitizes the heart to secondary insults and accelerates left ventricular remodeling. Collectively, these findings highlight the potential for CD40 agonists to promote myocardial inflammation and potentiate heart failure pathogenesis.
Collapse
Affiliation(s)
- Jesus Jimenez
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
- Cardio-Oncology Center of Excellence, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Junedh Amrute
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Pan Ma
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoran Wang
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | - Shibali Das
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Yohei Komaru
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- John Cochran Division, VA Saint Louis Health Care System, St. Louis, MO, USA
| | - Andreas Herrlich
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- John Cochran Division, VA Saint Louis Health Care System, St. Louis, MO, USA
| | - Matthias Mack
- Division of Nephrology, Department of Medicine, University Hospital Regensburg, Regensburg, Germany
| | - Kory J Lavine
- Center for Cardiovascular Research, Department of Medicine, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
10
|
Kitai T, Kohsaka S, Kato T, Kato E, Sato K, Teramoto K, Yaku H, Akiyama E, Ando M, Izumi C, Ide T, Iwasaki YK, Ohno Y, Okumura T, Ozasa N, Kaji S, Kashimura T, Kitaoka H, Kinugasa Y, Kinugawa S, Toda K, Nagai T, Nakamura M, Hikoso S, Minamisawa M, Wakasa S, Anchi Y, Oishi S, Okada A, Obokata M, Kagiyama N, Kato NP, Kohno T, Sato T, Shiraishi Y, Tamaki Y, Tamura Y, Nagao K, Nagatomo Y, Nakamura N, Nochioka K, Nomura A, Nomura S, Horiuchi Y, Mizuno A, Murai R, Inomata T, Kuwahara K, Sakata Y, Tsutsui H, Kinugawa K. JCS/JHFS 2025 Guideline on Diagnosis and Treatment of Heart Failure. J Card Fail 2025:S1071-9164(25)00100-9. [PMID: 40155256 DOI: 10.1016/j.cardfail.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/01/2025]
|
11
|
Pereyra Pietri M, Farina JM, Scalia IG, Mahmoud AK, Roarke M, Wasef B, Tagle-Cornell C, Kenyon CR, Abbas MT, Ali NB, Awad KA, Javadi N, Bismee NN, Larsen CM, Herrmann J, Arsanjani R, Ayoub C. Comparison of the diagnostic and prognostic value of criteria for immune checkpoint inhibitor related myocarditis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:30. [PMID: 40149000 PMCID: PMC11948924 DOI: 10.1186/s40959-025-00327-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 03/14/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Myocarditis is a dreaded complication of immune-checkpoint inhibitor (ICI) therapy but challenging to diagnose. There are no published data comparing the two leading diagnostic criteria for ICI-related myocarditis (ICIrM) and their association with cardiovascular events. METHODS In this retrospective cohort study, we reviewed all patients who underwent ICI therapy and had cardiac troponin assessment for possible myocarditis across three tertiary institutions from 2011 to 2022. ICIrM was adjudicated by the Bonaca et al. criteria and the ESC-ICOS guidelines. A propensity matched control group was identified of patients treated with ICI without developing myocarditis. Baseline characteristics and long-term outcomes, including cardiac death, MACE (myocardial infarction, TIA/stroke, heart failure), and arrhythmias data were curated, and patients diagnosed with ICIrM by each criteria were compared to controls for cardiovascular events. RESULTS A total of 59 patients (mean age was 73.1 ± 10.2 years, 60.1% male) were identified as having a diagnosis of ICIrM by Bonaca criteria (16 definite, 13 probable and 30 possible myocarditis). Forty-seven of these patients met the ESC-ICOS guidelines criteria, and all patients meeting either set of ICIrM criteria were treated with steroid therapy. At 3-year follow up, patients diagnosed with ICIrM by the Bonaca criteria had a high risk of cardiac mortality (HR 17.84, 95%CI 2.36-134.62, p = 0.005), MACE (HR 4.90, 95%CI 2.40-10.02, p < 0.001) and arrhythmias (HR 3.33, 95%CI 1.78-6.21, p < 0.001) when compared to matched controls. ICIrM by ESC-ICOS criteria was similarly predictive of cardiac mortality, MACE, and arrhythmias (HR 15.01, 95%CI 1.96-114.76, p = 0.009, HR 5.18, 95%CI 2.33-11.53, p < 0.001, and HR 3.41, 95%CI 1.73-6.70, p < 0.001 respectively). CONCLUSION The ESC-ICOS guidelines were more restrictive than the Bonaca et al. criteria for the diagnosis of ICIrM but similar in terms of prognostic value.
Collapse
Affiliation(s)
- Milagros Pereyra Pietri
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Juan M Farina
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Isabel G Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Ahmed K Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Michael Roarke
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Beman Wasef
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Cecilia Tagle-Cornell
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Courtney R Kenyon
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Mohammed Tiseer Abbas
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Nima Baba Ali
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Kamal A Awad
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Niloofar Javadi
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Nadera N Bismee
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Carolyn M Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, 5777 East Mayo Boulevard, Phoenix, AZ, 85054, USA.
| |
Collapse
|
12
|
Li S, Du F, Zhang Y, Wang Q, Dou J, Meng X. Myocarditis prediction in locally advanced or metastatic lung cancer patients with cardiac parameters abnormalities undergoing immunotherapy: development and validation of a risk assessment model. BMC Cancer 2025; 25:541. [PMID: 40133887 PMCID: PMC11934563 DOI: 10.1186/s12885-025-13943-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 03/14/2025] [Indexed: 03/27/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized treatment for advanced lung cancer, yet their cardiotoxicity, particularly immune checkpoint inhibitor-related myocarditis, poses significant clinical challenges. This study aims to create a predictive model using cardiac biomarkers to identify patients prone to myocarditis during treatment, thereby enhancing clinical decision-making and patient outcomes. METHODS In this retrospective cohort study, 1,838 patients with locally advanced and metastatic lung cancer and abnormal baseline cardiac parameters receiving immunotherapy from June 2018 to August 2024 were analyzed, with a follow-up date cutoff of September 20, 2024. Patients were randomly divided into training (70%) and validation (30%) cohorts. Logistic regression analysis was conducted on demographic information, clinical characteristics, treatments, and cardiac parameters of these patients prior to immunotherapy. A nomogram was constructed via multivariable logistic regression, and AUC and Hosmer-Lemeshow tests were performed to verify the accuracy of the model. RESULTS Among 1,838 patients, 89 (4.84%) developed myocarditis. Independent predictors included α-HBDH > 910 U/L (OR = 10.57, 95%CI: 2.47-45.22, P = 0.001), CK-MB > 15 ng/mL (OR = 3.87, 95%CI: 1.06-14.11, P = 0.040), hs-cTnT elevation (14-28 pg/mL: OR = 4.19; 28-42 pg/mL: OR = 13.10; >42 pg/mL: OR = 25.43, P < 0.001), NT-proBNP > 3× age-adjusted upper limit (OR = 9.72, 95%CI: 1.09-86.73, P = 0.042), and Caprini score ≥ 4 (OR = 4.49, 95%CI: 2.26-8.90, P < 0.001). The nomogram demonstrated strong discrimination ability, with an AUC of 0.831 in the training cohort (sensitivity: 0.842, specificity: 0.717) and an AUC of 0.844 in the validation cohort. CONCLUSIONS This study establishes a validated risk assessment model integrating cardiac biomarkers (α-HBDH, CK-MB, hs-cTnT, NT-proBNP) and Caprini risk score to predict ICI-related myocarditis in lung cancer patients with cardiac abnormalities. The tool facilitates early identification of high-risk patients, enabling tailored monitoring and preemptive management. These findings underscore the critical role of baseline cardiac profiling in optimizing immunotherapy safety.
Collapse
Affiliation(s)
- Shanshan Li
- Shandong University Cancer Center, jinan, Shandong, China
- Department of Radiation Oncology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Feng Du
- Department of Radiation Oncology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Yan Zhang
- Department of Radiation Oncology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Qiang Wang
- Department of Radiation Oncology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Jianjian Dou
- Department of Radiation Oncology, Zibo Municipal Hospital, Zibo, Shandong, China
| | - Xiangjiao Meng
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical university and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
13
|
Miao M, Liu X, Zhang H, Dai H. Immuno-inflammatory mechanisms in cardio-oncology: new hopes for immunotargeted therapies. Front Oncol 2025; 15:1516977. [PMID: 40182041 PMCID: PMC11966441 DOI: 10.3389/fonc.2025.1516977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 02/19/2025] [Indexed: 04/05/2025] Open
Abstract
Cardio-oncology is an emerging interdisciplinary field concerned with cancer treatment-related cardiovascular toxicities (CTR-CVT) and concomitant cardiovascular diseases (CVD) in patients with cancer. Inflammation and immune system dysregulation are common features of tumors and cardiovascular disease (CVD). In addition to the mutual exacerbating effect through inflammation, tumor treatments, including immunotherapy, chemotherapy, radiation therapy, and targeted therapy, may induce immune inflammatory reactions leading to cardiovascular damage. Cancer immunotherapy is currently a new method of cancer treatment. Immunotherapeutic agents, such as immune checkpoint inhibitors (ICIs), chimeric antigen receptor T cell immunotherapy (CAR-T), mRNA vaccines, etc., can induce anti-tumor effects by enhancing the host immune response to eliminate tumor cells. They have achieved remarkable therapeutic efficacy in clinical settings but lead to many immune-related adverse events (irAEs), especially CTR-CVT. Establishing specific evaluation, diagnostic, and monitoring criteria (e.g., inflammatory biomarkers) for both immunotherapy and anti-inflammatory therapy-related cardiovascular toxicity is vital to guide clinical practice. This article explores the role of immune response and inflammation in tumor cardiology, unravels the underlying mechanisms, and provides improved methods for monitoring and treating in CTR-CVT in the field of cardio-oncology.
Collapse
Affiliation(s)
- Meiqi Miao
- Department of Cardiology, Kunshan Hospital of Chinese Medicine, Kunshan, China
| | - Xinxin Liu
- Postdoctoral Mobile Station, Heilongjiang University of Traditional Chinese Medicine, Harbin, China
- The Innovation Base, Mudanjiang Collaborative Innovation Center for the Development and Application of Northern Medicinal Resources, Mudanjiang, China
| | - Han Zhang
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Hailong Dai
- Department of Cardiology, Yan’an Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
14
|
Panchal V, Momin E, Jain S, Singh A, Ghuman G, Brar V. Immune Checkpoint Inhibitor-induced Myocarditis: A Case Report of Complete Heart Block and Challenges in a Patient on Pembrolizumab. J Innov Card Rhythm Manag 2025; 16:6215-6219. [PMID: 40125493 PMCID: PMC11927601 DOI: 10.19102/icrm.2025.16033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 10/08/2024] [Indexed: 03/25/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by modulating immune responses, leading to enhanced anti-tumor activity. ICIs, including agents targeting cytotoxic T-lymphocyte antigen 4, programmed cell death 1, and programmed cell death ligand, are now widely used in various malignancies, either as monotherapy or in combination with chemotherapy, radiotherapy, or targeted therapies. However, ICIs are associated with immune-related adverse events, affecting multiple organ systems, with myocarditis emerging as a rare but potentially fatal complication. We present a 67-year-old man with a history of prostate and renal cell carcinoma treated with pembrolizumab and lenvatinib, who developed myocarditis secondary to ICI therapy. The patient initially presented with generalized fatigue and bradycardia, later progressing to more severe symptoms, including sinus bradycardia and elevated troponin levels. An electrocardiogram revealed a sinus rhythm with first-degree atrioventricular block, non-specific intraventricular conduction delay, and elevated high-sensitivity troponin levels progressively increasing to 50,000 pg/mL. A comprehensive diagnostic workup ruled out ischemic causes, leading to the diagnosis of ICI-induced myocarditis. The patient was treated with high-dose corticosteroids, intravenous immunoglobulin, and temporary pacemaker insertion, resulting in clinical improvement. This case highlights the need for vigilance and prompt intervention in patients receiving ICI therapy, as early recognition and treatment of myocarditis are crucial to optimizing patient outcomes in this high-risk population.
Collapse
Affiliation(s)
- Viraj Panchal
- Department of Medicine, Smt. NHL Municipal Medical College and SVPISMR, Ahmedabad, Gujarat, India
| | - Elina Momin
- Department of Internal Medicine, Louisiana State University, Shreveport, Louisiana, USA
| | - Shubhika Jain
- Department of Internal Medicine, Louisiana State University, Shreveport, Louisiana, USA
| | - Anaiya Singh
- Department of Internal Medicine, Louisiana State University, Shreveport, Louisiana, USA
| | - Guntas Ghuman
- Department of Internal Medicine, Government Medical College and Hospital, Chandigarh, India
| | - Vijaywant Brar
- Department of Cardiology, Louisiana State University, Shreveport, Louisiana, USA
| |
Collapse
|
15
|
Yan L, Zheng G. Drug-induced noninfectious myocarditis: a disproportionality analysis of the FAERS database. Expert Opin Drug Saf 2025; 24:345-353. [PMID: 39472113 DOI: 10.1080/14740338.2024.2423679] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 10/21/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND This investigation aims to identify and evaluate the most common and critical drugs associated with the risk of noninfectious myocarditis utilizing the US Food and Drug Administration Adverse Event Reporting System (FAERS) database. METHODS Data pertaining to noninfectious myocarditis from 2004 Q1 to 2024 Q2 were extracted. Following data standardization, multiple signal quantification techniques, including Reporting Odds Ratio (ROR) and Proportional Reporting Ratio, were employed for analysis. RESULTS The study identified a total of 10,763 adverse event reports associated with noninfectious myocarditis. Disproportionality analysis revealed that the top 5 drugs by ROR were phendimetrazine tartrate (ROR 104.64), trimethoprim + sulfamethoxazole (ROR 67.65), aldesleukin (ROR 52.67), mesalazine (ROR 49.73), and balsalazide disodium (ROR 45.26). Notably, among the 30 drugs with the strongest ROR signals, 8 drugs lacked myocarditis risk information in their package inserts. CONCLUSION Through comprehensive analysis of the FAERS database, our study identified drugs with a strong signal for myocarditis that are not currently indicated on their labels. The findings suggest that the potential risk of myocarditis associated with these medications is significant and warrants close monitoring in clinical practice.
Collapse
Affiliation(s)
- Liyuan Yan
- Department of Cardiology, Affiliated Changshu Hospital of Nantong University, Changshu, China
| | - Guanqun Zheng
- Department of Cardiology, Affiliated Changshu Hospital of Nantong University, Changshu, China
| |
Collapse
|
16
|
Crosio S, Treglia G, Imbimbo M, Froesch P, Grazioli Gauthier L, Arangalage D, Bergamaschi L, Györik SA, Viani GM, Caretta A, Leo LA, Pedrazzini G, Moschovitis G, Pavon AG. Multimodality Imaging and Immune-Related Adverse Events During Immune Checkpoint Inhibitors Treatment: Where Do We Stand? Echocardiography 2025; 42:e70115. [PMID: 40028736 DOI: 10.1111/echo.70115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/06/2025] [Accepted: 02/23/2025] [Indexed: 03/05/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy, significantly improving survival across various malignancies. However, these therapies are associated with various types of immune-related adverse events (irAEs), including cardiotoxicity, a spectrum of rare but potentially life-threatening complications impacting significantly morbidity and mortality. Cardiovascular imaging has become key in cardio-oncology, providing essential diagnostic tools for early detection and monitoring. This review synthesizes current evidence and underlines the pivotal role of early and tailored imaging strategies in managing ICI-induced cardiotoxicity. By bridging the knowledge gap, it aims to provide targetable insights to optimize the clinical management in patients undergoing immunotherapy.
Collapse
Affiliation(s)
- Stephanie Crosio
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giorgio Treglia
- Division of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
- Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Martina Imbimbo
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Patrizia Froesch
- Oncology Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Lorenzo Grazioli Gauthier
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Dimitri Arangalage
- Department of Cardiology, Bichat-Claude Bernard Hospital and Université Paris Cité, Paris, France
| | - Luca Bergamaschi
- Department of Cardiology, IRCCS Policlinico St. Orsola-Malpighi, Department of Experimental, Diagnostic and Specialty Medicine-DIMES, University of Bologna, Bologna, Italy
| | - Sándor A Györik
- Departement of Pneumology, Hospital of Bellinzona, Bellinzona, Switzerland
| | - Giacomo Maria Viani
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Alessandro Caretta
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Laura Anna Leo
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Giovanni Pedrazzini
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Giorgio Moschovitis
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
| | - Anna Giulia Pavon
- Department of Cardiology, Cardiocentro Ticino Institute, Ente Ospedaliero Cantonale, Lugano, Switzerland
- Faculty of Biomedical Sciences, Università Della Svizzera Italiana, Lugano, Switzerland
| |
Collapse
|
17
|
Newman AAC, Dalman JM, Moore KJ. Cardiovascular Disease and Cancer: A Dangerous Liaison. Arterioscler Thromb Vasc Biol 2025; 45:359-371. [PMID: 39781742 PMCID: PMC11864891 DOI: 10.1161/atvbaha.124.319863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/10/2024] [Accepted: 12/20/2024] [Indexed: 01/12/2025]
Abstract
The field of cardio-oncology has traditionally focused on the impact of cancer and its therapies on cardiovascular health. Mounting clinical and preclinical evidence, however, indicates that the reverse may also be true: cardiovascular disease can itself influence tumor growth and metastasis. Numerous epidemiological studies have reported that individuals with prevalent cardiovascular disease have an increased incidence of cancer. In parallel, studies using preclinical mouse models of myocardial infarction, heart failure, and cardiac remodeling support the notion that cardiovascular disorders accelerate the growth of solid tumors and metastases. These findings have ushered in a new and burgeoning field termed reverse cardio-oncology that investigates the impact of cardiovascular disease pathophysiology on cancer emergence and progression. Recent studies have begun to illuminate the mechanisms driving this relationship, including shared risk factors, reprogramming of immune responses, changes in gene expression, and the release of cardiac factors that result in selective advantages for tumor cells or their local milieu, thus exacerbating cancer pathology. Here, we review the evidence supporting the relationship between cardiovascular disease and cancer, the mechanistic pathways enabling this connection, and the implications of these findings for patient care.
Collapse
Affiliation(s)
- Alexandra A C Newman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Jessie M Dalman
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- University of Michigan Medical School, Ann Arbor, MI 48104, USA
| | - Kathryn J Moore
- Cardiovascular Research Center, New York University Langone Health, New York, NY 10016, USA
- Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY 10016, USA
| |
Collapse
|
18
|
Huang YV, Sun Y, Chou H, Wagner N, Vitale MR, Bayer AL, Xu B, Lee D, Lin Z, Branche C, Waliany S, Neal JW, Wakelee HA, Witteles RM, Nguyen PK, Graves EE, Berry GJ, Alcaide P, Wu SM, Zhu H. Novel Therapeutic Approach Targeting CXCR3 to Treat Immunotherapy Myocarditis. Circ Res 2025; 136:473-490. [PMID: 39931812 PMCID: PMC11867805 DOI: 10.1161/circresaha.124.325652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/12/2025] [Accepted: 01/16/2025] [Indexed: 03/01/2025]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are successful in treating many cancers but may cause immune-related adverse events. ICI-mediated myocarditis has a high fatality rate with severe cardiovascular consequences. Targeted therapies for ICI myocarditis are currently limited. METHODS We used a genetic mouse model of PD1 deletion (MRL/Pdcd1-/-) along with a novel drug-treated ICI myocarditis mouse model to recapitulate the disease phenotype. We performed single-cell RNA-sequencing, single-cell T-cell receptor sequencing, and cellular indexing of transcriptomes and epitopes on immune cells isolated from MRL and MRL/Pdcd1-/- mice at serial time points. We assessed the impact of macrophage deletion in MRL/Pdcd1-/- mice, then inhibited CXCR3 (C-X-C motif chemokine receptor 3) in ICI-treated mice to assess the therapeutic effect on myocarditis phenotype. Furthermore, we delineated the functional and mechanistic effects of CXCR3 blockade on T-cell and macrophage interactions. We then correlated the results in human single-cell multiomics data from blood and heart biopsy data from patients with ICI myocarditis. RESULTS Single-cell multiomics demonstrated expansion of CXCL (C-X-C motif chemokine ligand) 9/10+CCR2+ macrophages and CXCR3hi (C-X-C motif chemokine receptor 3 high-expressing) CD8+ (cluster of differentiation) effector T lymphocytes in the hearts of MRL/Pdcd1-/- mice correlating with onset of myocarditis development. Both depletion of CXCL9/10+CCR2+ (C-C motif chemokine receptor) macrophages and CXCR3 blockade, respectively, led to decreased CXCR3hi CD8+ T-cell infiltration into the heart and significantly improved survival. Transwell migration assays demonstrated that the selective blockade of CXCR3 and its ligand, CXCL10, reduced CXCR3+CD8+ T-cell migration toward macrophages, implicating this interaction in T-cell cardiotropism toward cardiac macrophages. Furthermore, cardiomyocyte apoptosis was induced by CXCR3hi CD8+ T cells. Cardiac biopsies from patients with confirmed ICI myocarditis demonstrated infiltrating CXCR3+ T cells and CXCL9+/CXCL10+ macrophages. Both mouse cardiac immune cells and patient peripheral blood immune cells revealed expanded TCRs (T-cell receptors) correlating with CXCR3hi CD8+ T cells in ICI myocarditis samples. CONCLUSIONS These findings bring forth the CXCR3-CXCL9/10 axis as an attractive therapeutic target for ICI myocarditis treatment, and more broadly as a druggable pathway in cardiac inflammation.
Collapse
Affiliation(s)
- Yuhsin Vivian Huang
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Yin Sun
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Harrison Chou
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Noah Wagner
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Maria Rosaria Vitale
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | | | - Bruce Xu
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Daniel Lee
- F. Edward Hebert School of Medicine at Uniformed Services University, Bethesda, MD (D.L.)
| | - Zachary Lin
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Corynn Branche
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Sarah Waliany
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
- Massachusetts General Hospital Cancer Center, Boston, MA (S.W.)
| | - Joel W. Neal
- Division of Oncology, Stanford, CA (J.W.N., H.A.W.)
- Stanford Cancer Institute, CA (J.W.N., H.A.W.)
| | - Heather A. Wakelee
- Division of Oncology, Stanford, CA (J.W.N., H.A.W.)
- Stanford Cancer Institute, CA (J.W.N., H.A.W.)
| | - Ronald M. Witteles
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Patricia K. Nguyen
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| | | | | | - Pilar Alcaide
- Tufts University School of Medicine, Boston, MA (A.L.B., P.A.)
| | - Sean M. Wu
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| | - Han Zhu
- Stanford Cardiovascular Institute, CA (Y.V.H., Y.S., H.C., N.W., M.R.V., B.X., Z.L., C.B., R.M.W., P.K.N., S.M.W., H.Z.)
- Division of Cardiovascular Medicine, Stanford, CA (S.W., R.M.W., P.K.N., S.M.W., H.Z.)
| |
Collapse
|
19
|
Muğlu H, Sünger E, Köylü B, Tunalı D, Erol C, Selcukbiricik F, Bilici A, Olmez OF. Late-Onset Myocarditis Following Immune Checkpoint Inhibitors Therapy: A Case Series with Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:302. [PMID: 40005418 PMCID: PMC11857675 DOI: 10.3390/medicina61020302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Immune checkpoint inhibitors (ICIs) therapy has revolutionized cancer treatment. However, it is important to acknowledge that ICI therapy can lead to immune-related adverse events (irAEs), including myocarditis. While early-onset myocarditis is well-documented, late-onset cases are increasingly recognized. This case series presents four cases of late-onset ICI-associated myocarditis, emphasizing the need for long-term surveillance of this potentially fatal complication. Patients exhibited a range of cardiac symptoms, including chest pain, shortness of breath, and arrhythmias. The diagnosis was confirmed through cardiac magnetic resonance imaging (MRI) and elevated cardiac biomarkers. Treatment involved the immediate discontinuation of ICI therapy and the initiation of high-dose corticosteroids. In cases with an inadequate response, additional immunosuppressive agents were considered. This case series underscores the importance of prolonged monitoring for late-onset ICI-associated myocarditis. Further research is needed to establish optimal treatment strategies and long-term management approaches for this complex condition.
Collapse
Affiliation(s)
- Harun Muğlu
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34214, Turkey; (E.S.); (A.B.); (O.F.O.)
| | - Erdem Sünger
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34214, Turkey; (E.S.); (A.B.); (O.F.O.)
| | - Bahadır Köylü
- Department of Medical Oncology, Istanbul Koc University, Istanbul 34450, Turkey; (B.K.); (D.T.)
| | - Didem Tunalı
- Department of Medical Oncology, Istanbul Koc University, Istanbul 34450, Turkey; (B.K.); (D.T.)
| | - Cengiz Erol
- Department of Radiology, Faculty of Medicine, Medipol University, Istanbul 34214, Turkey;
| | - Fatih Selcukbiricik
- Department of Medical Oncology, Istanbul Koc University, Istanbul 34450, Turkey; (B.K.); (D.T.)
| | - Ahmet Bilici
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34214, Turkey; (E.S.); (A.B.); (O.F.O.)
| | - Omer Fatih Olmez
- Department of Medical Oncology, Faculty of Medicine, Medipol University, Istanbul 34214, Turkey; (E.S.); (A.B.); (O.F.O.)
| |
Collapse
|
20
|
Gergely TG, Drobni ZD, Sayour NV, Ferdinandy P, Varga ZV. Molecular fingerprints of cardiovascular toxicities of immune checkpoint inhibitors. Basic Res Cardiol 2025; 120:187-205. [PMID: 39023770 PMCID: PMC11790702 DOI: 10.1007/s00395-024-01068-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy by unleashing the power of the immune system against malignant cells. However, their use is associated with a spectrum of adverse effects, including cardiovascular complications, which can pose significant clinical challenges. Several mechanisms contribute to cardiovascular toxicity associated with ICIs. First, the dysregulation of immune checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1) and its ligand (PD-L1), and molecular mimicry with cardiac autoantigens, leads to immune-related adverse events, including myocarditis and vasculitis. These events result from the aberrant activation of T cells against self-antigens within the myocardium or vascular endothelium. Second, the disruption of immune homeostasis by ICIs can lead to autoimmune-mediated inflammation of cardiac tissues, manifesting as cardiac dysfunction and heart failure, arrhythmias, or pericarditis. Furthermore, the upregulation of inflammatory cytokines, particularly tumor necrosis factor-alpha, interferon-γ, interleukin-1β, interleukin-6, and interleukin-17 contributes to cardiac and endothelial dysfunction, plaque destabilization, and thrombosis, exacerbating cardiovascular risk on the long term. Understanding the intricate mechanisms of cardiovascular side effects induced by ICIs is crucial for optimizing patient care and to ensure the safe and effective integration of immunotherapy into a broader range of cancer treatment protocols. The clinical implications of these mechanisms underscore the importance of vigilant monitoring and early detection of cardiovascular toxicity in patients receiving ICIs. Future use of these key pathological mediators as biomarkers may aid in prompt diagnosis of cardiotoxicity and will allow timely interventions.
Collapse
Affiliation(s)
- Tamás G Gergely
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsófia D Drobni
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Péter Ferdinandy
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary.
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary.
| |
Collapse
|
21
|
Bloom MW, Vo JB, Rodgers JE, Ferrari AM, Nohria A, Deswal A, Cheng RK, Kittleson MM, Upshaw JN, Palaskas N, Blaes A, Brown SA, Ky B, Lenihan D, Maurer MS, Fadol A, Skurka K, Cambareri C, Chauhan C, Barac A. Cardio-Oncology and Heart Failure: a Scientific Statement From the Heart Failure Society of America. J Card Fail 2025; 31:415-455. [PMID: 39419165 DOI: 10.1016/j.cardfail.2024.08.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 10/19/2024]
Abstract
Heart failure and cancer remain 2 of the leading causes of morbidity and mortality, and the 2 disease entities are linked in a complex manner. Patients with cancer are at increased risk of cardiovascular complications related to the cancer therapies. The presence of cardiomyopathy or heart failure in a patient with new cancer diagnosis portends a high risk for adverse oncology and cardiovascular outcomes. With the rapid growth of cancer therapies, many of which interfere with cardiovascular homeostasis, heart failure practitioners need to be familiar with prevention, risk stratification, diagnosis, and management strategies in cardio-oncology. This Heart Failure Society of America statement addresses the complexities of heart failure care among patients with active cancer diagnoses and cancer survivors. Risk stratification, monitoring and management of cardiotoxicity are presented across stages A through D heart failure, with focused discussion on heart failure with preserved ejection fraction and special populations, such as survivors of childhood and young-adulthood cancers. We provide an overview of the shared risk factors between cancer and heart failure, highlighting heart failure as a form of cardiotoxicity associated with many different cancer therapeutics. Finally, we discuss disparities in the care of patients with cancer and cardiac disease and present a framework for a multidisciplinary-team approach and critical collaboration among heart failure, oncology, palliative care, pharmacy, and nursing teams in the management of these complex patients.
Collapse
Affiliation(s)
| | - Jacqueline B Vo
- Radiation Epidemiology Branch, Division of Cancer Epidemiology & Genetics, National Cancer Institute, Bethesda, MD
| | - Jo E Rodgers
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, NC
| | - Alana M Ferrari
- Division of Hematology/ Oncology, University of Virginia Health, Charlottesville, VA
| | - Anju Nohria
- Cardio-Oncology Program, Cardiovascular Division, Brigham and Women's Hospital, Boston, MA
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard K Cheng
- Division of Cardiology, University of Washington, Seattle, WA
| | - Michelle M Kittleson
- Department of Cardiology, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA
| | | | - Nicolas Palaskas
- Department of Cardiology, Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Anne Blaes
- Division of Hematology/Oncology/Transplantation, University of Minnesota, Minneapolis, MN
| | - Sherry-Ann Brown
- Department of Medicine, Medical College of Wisconsin, Milwaukee, WI; Research Collaborator, Mayo Clinic, Rochester, MN
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA; Thalheimer Center for Cardio-Oncology, Abramson Cancer Center and Division of Cardiovascular Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA
| | - Daniel Lenihan
- Saint Francis Healthcare, Cape Girardeau, MO and the International Cardio-Oncology Society, Tampa, FL
| | - Mathew S Maurer
- Division of Cardiology, Columbia University Irving Medical Center, New York, NY
| | | | | | - Christine Cambareri
- Clinical Oncology Pharmacist, Hospital of the University of Pennsylvania, Abramson Cancer Center, Philadelphia, PA
| | | | - Ana Barac
- Department of Cardiology, Inova Schar Heart and Vascular, Inova Schar Cancer, Falls Church, VA
| |
Collapse
|
22
|
Li S, Tajiri K, Yuan Z, Murakata Y, Song Z, Mizuno S, Xu D, Murakoshi N. 4E-BP3 deficiency impairs dendritic cell activation and CD4 + T cell differentiation and attenuates α-myosin-specific T cell-mediated myocarditis in mice. Basic Res Cardiol 2025; 120:225-240. [PMID: 39516410 DOI: 10.1007/s00395-024-01089-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/19/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Immune checkpoint inhibitor (ICI)-associated myocarditis is a rare but potentially fatal immune-related adverse event. Previously, we reported a case of ICI-associated myocarditis with elevated autoantibodies to 4E-binding protein 3 (4E-BP3). Recent studies have suggested that 4E-BP3 may play an important role in tumor development. However, its role in cardiac diseases including myocarditis is unknown. We investigated the role of 4E-BP3 in an autoimmune myocarditis mouse model. Myocarditis was induced in wild-type and 4E-BP3 knockout mice by immunization with murine α-myosin peptide. 4E-BP3 gene expression was upregulated in the heart of myocarditis mouse. We found that genetic deletion of 4E-BP3 attenuated myocardial inflammation, reduced fibrosis area, and improved cardiac function in myocarditis mice. Studies in bone marrow-chimeric mice demonstrated that immune cell-derived 4E-BP3 plays a pivotal role in the pathogenesis of myocarditis. Immune cell transfer experiments revealed that 4E-BP3 deficiency in dendritic cells and CD4+ T cells decreased disease severity in recipient mice. Furthermore, dendritic cells that were deficient in 4E-BP3 exhibited a diminished capacity to produce IL-6 and IL-1β. Naive CD4+ T cells lacking 4E-BP3 had a reduced ability to differentiate into T-helper (Th)1 and Th17 cells. These findings suggest that 4E-BP3 in dendritic cells and CD4+ T cells may be critically involved in the pathogenesis of α-myosin-specific T cell-mediated myocarditis. Thus, 4E-BP3 could be a possible therapeutic target for myocarditis.
Collapse
Affiliation(s)
- Siqi Li
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kazuko Tajiri
- Tsukuba Life Science Innovation Program (T-LSI), School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan.
- Department of Cardiology, National Cancer Center Hospital East, 6-5-1 Kashiwanoha, Kashiwa, Chiba, Japan.
| | - Zixun Yuan
- Stanley and Judith Frankel Institute for Heart & Brain Health, University of Michigan Medical School, Ann Arbor, MI, 48109, USA
| | - Yoshiko Murakata
- Research Center for Biosafety, Laboratory Animal and Pathogen Bank, National Institute of Infectious Diseases, Musashimurayama, Japan
| | - Zonghu Song
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Seiya Mizuno
- Laboratory Animal Resource Center and Transborder Medical Research Center, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Dongzhu Xu
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
- Tsukuba Life Science Innovation Program (T-LSI), School of Integrative and Global Majors (SIGMA), University of Tsukuba, Tsukuba, Japan
| | - Nobuyuki Murakoshi
- Department of Cardiology, Institute of Medicine, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
23
|
Deveci Ş, Uzun M, Özçelik P, Tümer Doğukan SS, Matur Z. Myositis associated with pembrolizumab presenting with myastheniform symptoms: two case reports. Anticancer Drugs 2025; 36:143-150. [PMID: 39749550 DOI: 10.1097/cad.0000000000001665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Immune checkpoint inhibitors (ICIs), such as pembrolizumab, have revolutionized cancer treatment by enhancing the immune system's response to malignancies. However, these therapies are associated with immune-related adverse events (irAEs), including neuromuscular complications such as myasthenia gravis, myositis, and myocarditis. We describe two male patients, aged 67 and 68, with small cell and non-small cell lung cancers, who developed progressive neuromuscular symptoms, including ptosis, diplopia, and generalized weakness, after receiving pembrolizumab. Clinical, biochemical, imaging, and electrophysiological findings confirmed the diagnosis of myositis with myastheniform features, with one case also involving myocarditis. Both patients underwent treatments with intravenous immunoglobulin (IVIg), pyridostigmine, and corticosteroids. The first patient, despite aggressive treatment including plasma exchange and rituximab, succumbed to complications from aspiration pneumonia. The second patient showed partial response to pyridostigmine and IVIg but later died due to metastatic cancer progression. A literature review revealed 52 cases of pembrolizumab-associated myositis with myastheniform symptoms, emphasizing its high morbidity and the need for vigilant monitoring. Pembrolizumab-associated myositis with myastheniform symptoms, especially when accompanied by myocarditis, presents a significant clinical challenge with high mortality. Early recognition and aggressive management of these irAEs are crucial to improving outcomes in cancer patients receiving ICIs.
Collapse
Affiliation(s)
- Şule Deveci
- Department of Neurology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital
| | - Mustafa Uzun
- Department of Neurology, University of Health Sciences, Basaksehir Cam and Sakura City Hospital
| | - Pinar Özçelik
- Department of Neurology, Bezmialem Vakif University, Medical Faculty, Istanbul, Turkey
| | | | - Zeliha Matur
- Department of Neurology, Bezmialem Vakif University, Medical Faculty, Istanbul, Turkey
| |
Collapse
|
24
|
Kamikawa H, Matsukawa Y, Nishii H, Naito Y, Obara K, Sahashi K, Morimoto R, Matsuo K, Ishida S, Inoue S, Miyagi S, Sakakibara A, Katsuno M, Karube K, Akamatsu S. A case of severe immune-related adverse events, myocarditis with myositis, and myasthenia gravis overlap syndrome following adjuvant nivolumab administration for muscle-invasive bladder cancer. NAGOYA JOURNAL OF MEDICAL SCIENCE 2025; 87:156-162. [PMID: 40255993 PMCID: PMC12003993 DOI: 10.18999/nagjms.87.1.156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 07/24/2024] [Indexed: 04/22/2025]
Abstract
Herein, we present a case of severe immune-related adverse events (irAEs), myocarditis with myositis, and myasthenia gravis overlap syndrome (IM3OS) in a patient receiving an immune checkpoint inhibitor (ICI), as adjuvant therapy after surgery for muscle-invasive bladder cancer. An 80-year-old woman who had undergone a total cystectomy for bladder cancer presented with ptosis, diplopia, and paralysis 18 days after receiving nivolumab, an anti-programmed cell death-1 (PD-1) monoclonal antibody, as adjuvant therapy for the first time. Initial testing revealed positive findings on the ice pack test; elevated troponin, creatine kinase, and aldolase levels; and an abnormal electrocardiogram, suggesting that the patient had developed ICI-related myocarditis, myositis, and myasthenia gravis. Despite treatment with intravenous immunoglobulin (IVIG) and high-dose corticosteroids, her condition worsened, leading to a complete atrioventricular block. After cardiac pacemaker insertion and intensive treatment with repeated high-dose corticosteroids, IVIG, plasma exchange, and tacrolimus, left ventricular function and myositis symptoms improved. However, the patient developed a respiratory infection and renal failure, leading to death on day 99. Although ICIs are considered relatively safe with few side effects, they can cause serious complications and lead to death. In particular, when severe irAEs occur in multiple organs, such as IM3OS, the prognosis is poor. Although IM3OS has no specific diagnostic biomarker, making early detection difficult, clinicians should always pay attention to patient symptoms when using ICI and evaluate other pathologies with IM3OS when conditions such as myositis or myocarditis are suspected. Further research is needed to elucidate the pathophysiology and risk factors of IM3OS.
Collapse
Affiliation(s)
- Hiroki Kamikawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
- Department of Urology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yoshihisa Matsukawa
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hisae Nishii
- Department of Urology, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Yushi Naito
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuki Obara
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kentaro Sahashi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ryota Morimoto
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuna Matsuo
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shohei Ishida
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Satoshi Inoue
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Shohei Miyagi
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Ayako Sakakibara
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kennosuke Karube
- Department of Pathology and Laboratory Medicine, Nagoya University Hospital, Nagoya, Japan
| | - Shusuke Akamatsu
- Department of Urology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
25
|
Makwana B, Malode A, Khadke S, Patel V, Shah R, Patel M, Parikh A, Dani SS, Ganatra S. Cardiac Complications of Immune Checkpoint Inhibitors and Chimeric Antigen Receptor T Cell Therapy. Cardiol Clin 2025; 43:151-167. [PMID: 39551556 DOI: 10.1016/j.ccl.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Immune checkpoint inhibitors and chimeric antigen receptor T-cell therapy have revolutionized cancer treatment but can cause life-threatening cardiovascular toxicities through immune-related adverse events. Myocarditis is the most common and potentially fatal toxicity with immune checkpoint inhibitors. T-cell therapies can potentially lead to cytokine release syndrome. Diagnosis of ICI-myocarditis requires a multimodal approach, including biomarkers, imaging, and endomyocardial biopsy, while CRS is characterized by a clinical syndrome resembling distributive shock. Management involves discontinuing the offending therapy, immunosuppression with corticosteroids for ICI-myocarditis, and interleukin-6 antagonists for CRS. Collaboration between oncologists and cardiologists is crucial for early recognition and prompt treatment.
Collapse
Affiliation(s)
- Bhargav Makwana
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Aishwarya Malode
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sumanth Khadke
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Vahin Patel
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Rushin Shah
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Manav Patel
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Aneri Parikh
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sourbha S Dani
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA
| | - Sarju Ganatra
- Division of Cardiology, Department of Internal Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA; Department of Medicine (Research), Cardio-Oncology Program, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805, USA.
| |
Collapse
|
26
|
Schoenherr C, Pietzsch S, Barca C, Müller FE, Bahr FS, Kasten M, Zeug A, Erschow S, Falk CS, Ponimaskin E, Thackeray JT, Hilfiker-Kleiner D, Ricke-Hoch M. Immune-checkpoint-inhibitor therapy directed against PD-L1 is tolerated in the heart without manifestation of cardiac inflammation in a preclinical reversible melanoma mouse model. FRONTIERS IN MOLECULAR MEDICINE 2025; 4:1487526. [PMID: 39834851 PMCID: PMC11743445 DOI: 10.3389/fmmed.2024.1487526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/10/2024] [Indexed: 01/22/2025]
Abstract
Immune-checkpoint-inhibitors (ICI) target key regulators of the immune system expressed by cancer cells that mask those from recognition by the immune system. They have improved the outcome for patients with various cancer types, such as melanoma. ICI-based therapy is frequently accompanied by immune-related adverse side effects (IRAEs). The reversible melanoma cancer mouse model (B16F10 cells stably expressing a ganciclovir (GCV)-inducible suicide gene in C57BL/6N mice: B16F10-GCV) allows chemotherapy-free tumor elimination in advanced disease stage and demonstrates almost complete recovery of the mouse heart from cancer-induced atrophy, molecular impairment and heart failure. Thus, enabling the study of anti-cancer-therapy effects. Here, we analyzed potential cardiac side effects of antibody-mediated PD-L1 inhibition in the preclinical B16F10-GCV mouse model after tumor elimination and 2 weeks recovery (50 days after tumor inoculation). Anti-PD-L1 treatment was associated with improved survival as compared to isotype control (Ctrl) treated mice. Surviving anti-PD-L1 and Ctrl mice showed similar cardiac function, dimensions and the expression of cardiac stress and hypertrophy markers. Although anti-PD-L1 treatment was associated with increased troponin I type 3 cardiac (TNNI3) blood levels, cardiac mRNA expression of macrophage markers and elevated cardiac levels of secreted inflammatory factors compared to Ctrl treatment, both groups showed a comparable density of inflammatory cells in the heart (using CXCR4-ligand 68Ga-Pentixafor in PET-CT and immunohistochemistry). Thus, anti-PD-L1 therapy improved survival in mice with advanced melanoma cancer with no major cardiac phenotype or inflammation 50 days after tumor inoculation. Without a second hit that triggers the inflammatory response, anti-PD-L1 treatment appears to be safe for the heart in the preclinical melanoma mouse model.
Collapse
Affiliation(s)
- Caroline Schoenherr
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Stefan Pietzsch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Human Genetics, Hannover Medical School, Hannover, Germany
| | - Cristina Barca
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Franziska E. Müller
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Frauke S. Bahr
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Martina Kasten
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Andre Zeug
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - Sergej Erschow
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Christine S. Falk
- Institute of Transplant Immunology, IFB-Tx, Hannover Medical School, Hannover, Germany
| | - Evgeni Ponimaskin
- Department of Cellular Neurophysiology, Hannover Medical School, Hannover, Germany
| | - James T. Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Denise Hilfiker-Kleiner
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
- Department of Cardiovascular Complications of Oncologic Therapies, Medical Faculty of the Philipps University Marburg, Marburg, Germany
| | - Melanie Ricke-Hoch
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
27
|
Kidane RD, Ruddy KJ, Lin G, Sandhu NP. Cardiovascular Health Considerations for Primary Care Physicians Treating Breast Cancer Survivors. Mayo Clin Proc 2025; 100:124-140. [PMID: 39641716 DOI: 10.1016/j.mayocp.2024.08.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Revised: 07/09/2024] [Accepted: 08/13/2024] [Indexed: 12/07/2024]
Abstract
Breast cancer (BC) survivors are at increased risk for cardiovascular disease (CVD) and require their primary care physicians to manage their long-term general medical care, including cardiovascular (CV) health. Yet, evidence exists that some primary care physicians possess insufficient knowledge about survivorship care. With the goal of bridging these knowledge gaps, a PubMed review was conducted from July 7, 2020, through October 2, 2020, with an updated PubMed review from January 3, 2024, through April 28, 2024, focusing on CV health considerations in the primary care of BC survivors. Search terms included variations of "breast cancer survivors" and "cardiovascular." In total, 152 publications were included. Breasts cancer survivors may have increased CVD risk because some anticancer therapies are cardiotoxic and risk factors for BC often also increase the risk for CVD. Multiple risk factors overlap for BC and CVD such as older age, Western diet, early menarche, physical inactivity, high body mass index, and smoking. In this review, results are summarized from studies that report the presence of CV risk factors and CVD in BC survivors. Also described are the CV effects of BC therapies (chemotherapy, hormonal agents, targeted therapies, and radiotherapy) and the type of CV evaluation (cardiac imaging and measurement of biomarkers) that these patients may need. Primary care physicians have an important role in managing the CV health of BC survivors from preventing, assessing, and managing CV risk factors to referring patients to appropriate specialists when needed.
Collapse
Affiliation(s)
- Redet D Kidane
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Kathryn J Ruddy
- Division of Medical Oncology, Mayo Clinic, Rochester, MN, USA
| | - Grace Lin
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Nicole P Sandhu
- Division of General Internal Medicine, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
28
|
Chan JSK, Chan RNC, Lee YHA, Satti DI, Dee EC, Ng K, Achim A, Ng CF, Liu T, Matthews GDK, Tse G, Vassiliou VS. Cardiovascular health of patients with cancer: Challenges abound. Trends Cardiovasc Med 2025; 35:24-31. [PMID: 38657744 DOI: 10.1016/j.tcm.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Patients with cancer have elevated cardiovascular risks compared to those without cancer. As cancer incidence increases and cancer-related mortality decreases, cardiovascular diseases in patients with a history of cancer will become increasingly important. This in turn is reflected by the exponentially increasing amount of cardio-oncology research in recent years. This narrative review aims to summarize the key existing literature in several main areas of cardio-oncology, including the epidemiology, natural history, prevention, management, and determinants of the cardiovascular health of patients with cancer, and identify relevant gaps in evidence for further research.
Collapse
Affiliation(s)
- Jeffrey Shi Kai Chan
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China
| | - Raymond Ngai Chiu Chan
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China
| | - Yan Hiu Athena Lee
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China; Division of Urology, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China; SH Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Danish Iltaf Satti
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China; Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward Christopher Dee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenrick Ng
- Department of Medical Oncology, Barts Cancer Centre, London, UK
| | - Alexandru Achim
- Department of Internal Medicine, Invasive Cardiology Division, University of Szeged, Szeged, Hungary; Department of Cardiology, "Niculae Stancioiu" Heart Institute, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Chi Fai Ng
- Division of Urology, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China; SH Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, PR China
| | - Gareth D K Matthews
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich, UK; Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich, UK
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, PR China; Kent and Medway Medical School, Canterbury, Kent CT2 7NT, UK; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, PR China.
| | - Vassilios S Vassiliou
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich, UK; Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich, UK
| |
Collapse
|
29
|
Cortellino S, D'Angelo M, Quintiliani M, Giordano A. Cancer knocks you out by fasting: Cachexia as a consequence of metabolic alterations in cancer. J Cell Physiol 2025; 240:e31417. [PMID: 39245862 DOI: 10.1002/jcp.31417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/18/2024] [Accepted: 08/09/2024] [Indexed: 09/10/2024]
Abstract
Neoplastic transformation reprograms tumor and surrounding host cell metabolism, increasing nutrient consumption and depletion in the tumor microenvironment. Tumors uptake nutrients from neighboring normal tissues or the bloodstream to meet energy and anabolic demands. Tumor-induced chronic inflammation, a high-energy process, also consumes nutrients to sustain its dysfunctional activities. These tumor-related metabolic and physiological changes, including chronic inflammation, negatively impact systemic metabolism and physiology. Furthermore, the adverse effects of antitumor therapy and tumor obstruction impair the endocrine, neural, and gastrointestinal systems, thereby confounding the systemic status of patients. These alterations result in decreased appetite, impaired nutrient absorption, inflammation, and shift from anabolic to catabolic metabolism. Consequently, cancer patients often suffer from malnutrition, which worsens prognosis and increases susceptibility to secondary adverse events. This review explores how neoplastic transformation affects tumor and microenvironment metabolism and inflammation, leading to poor prognosis, and discusses potential strategies and clinical interventions to improve patient outcomes.
Collapse
Affiliation(s)
- Salvatore Cortellino
- Laboratory of Molecular Oncology, Responsible Research Hospital, Campobasso, Italy
- Scuola Superiore Meridionale (SSM), School for Advanced Studies, Federico II University, Naples, Italy
- SHRO Italia Foundation ETS, Candiolo, Turin, Italy
| | - Margherita D'Angelo
- Department of Experimental Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine, Center for Biotechnology, College of Science and Technology, Temple University, Philadelphia, Pennsylvania, USA
- Department of Medical Biotechnologies, University of Siena, Siena, Italy
| |
Collapse
|
30
|
Pi JK, Chen XT, Zhang YJ, Chen XM, Wang YC, Xu JY, Zhou JH, Yu SS, Wu SS. Insight of immune checkpoint inhibitor related myocarditis. Int Immunopharmacol 2024; 143:113559. [PMID: 39536487 DOI: 10.1016/j.intimp.2024.113559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/20/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
As the understanding of immune-related mechanisms in the development and progression of cancer advances, immunotherapies, notably Immune Checkpoint Inhibitors (ICIs), have become integral in comprehensive cancer treatment strategies. ICIs reactivate T-cell cytotoxicity against tumors by blocking immune suppressive signals on T cells, such as Programmed Death-1 (PD-1) and Cytotoxic T-lymphocyte Antigen-4 (CTLA-4). Despite their beneficial effects, ICIs are associated with immune-related adverse events (irAEs), manifesting as autoimmune side effects across various organ systems. A particularly alarming irAE is life-threatening myocarditis. This rare but severe side effect of ICIs leads to significant long-term cardiac complications, including arrhythmias and heart failure, and has been observed to have a mortality rate of up to 50% in affected patients. This greatly limits the clinical application of ICI-based immunotherapy. In this review, we provide a comprehensive summary of the current knowledge regarding the diagnosis and management of ICI-related myocarditis. We also discuss the utility of preclinical mouse models in understanding and addressing this critical challenge.
Collapse
Affiliation(s)
- Jin-Kui Pi
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xiao-Ting Chen
- Animal Experimental Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yan-Jing Zhang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Xue-Mei Chen
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Yin-Chan Wang
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jia-Yi Xu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Jin-Han Zhou
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Shuai-Shuai Yu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China
| | - Si-Si Wu
- Core Facilities, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, PR China.
| |
Collapse
|
31
|
Beecher G, Pinal-Fernandez I, Mammen AL, Liewluck T. Immune Checkpoint Inhibitor Myopathy: The Double-Edged Sword of Cancer Immunotherapy. Neurology 2024; 103:e210031. [PMID: 39514829 DOI: 10.1212/wnl.0000000000210031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024] Open
Abstract
Immune checkpoint inhibitor (ICI) therapy has revolutionized the treatment of several malignancies, with improved survival. These monoclonal antibodies target immune checkpoints, including cytotoxic T-lymphocyte-associated protein 4 (ipilimumab and tremelimumab), programmed death 1 (nivolumab, pembrolizumab, cemiplimab, and dostarlimab), programmed death ligand 1 (atezolizumab, avelumab, and durvalumab), and lymphocyte activation gene 3 (relatlimab), and effectively augment the immune response against tumor cells. Releasing the brakes on the immune system has consequences, however, in the form of immune-related adverse events (irAEs), which may affect any organ. Neurologic irAEs represent 1%-3% of all irAEs, with immune-mediated myopathy (ICI myopathy) being the most common manifestation. Recent large patient series and systematic reviews have established the key features and highlighted new insights into ICI myopathy. ICI myopathy is characterized by an acute or subacute onset of oculobulbar and/or proximal limb weakness, with or without associated respiratory insufficiency and myocarditis. Creatine kinase elevation is common. Oculobulbar presentations with or without respiratory failure may be misattributed to neuromuscular junction disorders, particularly because acetylcholine receptor antibodies are present in up to 40% of patients; however, an electrodiagnostic evidence of a defect of neuromuscular transmission is often absent even in patients with severe weakness, highlighting that the myopathic process is the driving force behind these presentations. Muscle histopathology commonly demonstrates a unique signature of multifocal clusters of necrotic and regenerating fibers, differentiating ICI myopathy from other autoimmune myopathies. Transcriptomic analysis has uncovered distinct subgroups within ICI myopathy, revealing varying degrees of type 1 and type 2 interferon pathway activation alongside notable upregulation of the interleukin (IL)-6 pathway in affected muscle tissue. This discovery presents a promising avenue for intervention through the use of therapies that suppress the interferon pathway and target IL-6 or its receptor. Despite clinical improvements with immunomodulatory therapy, with corticosteroids the mainstay of treatment, mortality remains high, particularly in those with associated myocarditis or respiratory failure requiring intubation, where mortality occurs in up to 50%. ICI withdrawal can lead to cancer progression and death, highlighting a need for improved approaches to ICI rechallenge, performed in limited patients with variable success to date.
Collapse
Affiliation(s)
- Grayson Beecher
- From the Division of Neurology (G.B.), Department of Medicine, University of Alberta, Edmonton, Canada; Muscle Disease Section (I.P.-F., A.L.M.), National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda; Department of Neurology (I.P.-F., A.L.M.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Neurology (T.L.), Mayo Clinic, Rochester, MN
| | - Iago Pinal-Fernandez
- From the Division of Neurology (G.B.), Department of Medicine, University of Alberta, Edmonton, Canada; Muscle Disease Section (I.P.-F., A.L.M.), National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda; Department of Neurology (I.P.-F., A.L.M.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Neurology (T.L.), Mayo Clinic, Rochester, MN
| | - Andrew L Mammen
- From the Division of Neurology (G.B.), Department of Medicine, University of Alberta, Edmonton, Canada; Muscle Disease Section (I.P.-F., A.L.M.), National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda; Department of Neurology (I.P.-F., A.L.M.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Neurology (T.L.), Mayo Clinic, Rochester, MN
| | - Teerin Liewluck
- From the Division of Neurology (G.B.), Department of Medicine, University of Alberta, Edmonton, Canada; Muscle Disease Section (I.P.-F., A.L.M.), National Institute of Arthritis and Musculoskeletal and Skin Diseases, NIH, Bethesda; Department of Neurology (I.P.-F., A.L.M.), Johns Hopkins University School of Medicine, Baltimore, MD; and Department of Neurology (T.L.), Mayo Clinic, Rochester, MN
| |
Collapse
|
32
|
Kwan JM, Shen M, Akhlaghi N, Hu JR, Mora R, Cross JL, Jiang M, Mankbadi M, Wang P, Zaman S, Lee S, Im Y, Feher A, Liu YH, Ma SS, Tao W, Wei W, Baldassarre LA. Adverse cardiovascular events and cardiac imaging findings in patients on immune checkpoint inhibitors. PLoS One 2024; 19:e0314555. [PMID: 39621799 PMCID: PMC11611253 DOI: 10.1371/journal.pone.0314555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 11/12/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND There is an urgent need to better understand the diverse presentations, risk factors, and outcomes of immune checkpoint inhibitor (ICI)-associated cardiovascular toxicity. There remains a lack of consensus surrounding cardiovascular screening, risk stratification, and clinical decision-making in patients receiving ICIs. METHODS We conducted a single center retrospective cohort study including 2165 cancer patients treated with ICIs between 2013 and 2020. The primary outcome was adverse cardiovascular events (ACE): a composite of myocardial infarction, coronary artery disease, stroke, peripheral vascular disease, arrhythmias, heart failure, valvular disease, pericardial disease, and myocarditis. Secondary outcomes included all-cause mortality and the individual components of ACE. We additionally conducted an imaging substudy examining imaging characteristics from echocardiography (echo) and cardiac magnetic resonance (CMR) imaging. RESULTS In our cohort, 44% (n = 962/2165) of patients experienced ACE. In a multivariable analysis, dual ICI therapy (hazard ratio [HR] 1.23, confidence interval [CI] 1.04-1.45), age (HR 1.01, CI 1.00-1.01), male sex (HR 1.18, CI 1.02-1.36), prior arrhythmia (HR 1.22, CI 1.03-1.43), lung cancer (HR 1.17, CI 1.01-1.37), and central nervous system (CNS) malignancy (HR 1.23, CI 1.02-1.47), were independently associated with increased ACE. ACE was independently associated with a 2.7-fold increased risk of mortality (P<0.001). Dual ICI therapy was also associated with a 2.0-fold increased risk of myo/pericarditis (P = 0.045), with myo/pericarditis being associated with a 2.9-fold increased risk of mortality (P<0.001). However, the cardiovascular risks of dual ICI therapy were offset by its mortality benefit, with dual ICI therapy being associated with a ~25% or 1.3-fold decrease in mortality. Of those with echo prior to ICI initiation, 26% (n = 115/442) had abnormal left ventricular ejection fraction or global longitudinal strain, and of those with echo after ICI initiation, 28% (n = 207/740) had abnormalities. Of those who had CMR imaging prior to ICI initiation, 43% (n = 9/21) already had left ventricular dysfunction, 50% (n = 10/20) had right ventricular dysfunction, 32% (n = 6/19) had left ventricular late gadolinium enhancement, and 9% (n = 1/11) had abnormal T2 imaging. CONCLUSION Dual ICI therapy, prior arrhythmia, older age, lung and CNS malignancies were independently associated with an increased risk of ACE, and dual ICI therapy was also independently associated with an increased risk of myo/pericarditis, highlighting the utmost importance of cardiovascular risk factor optimization in this particularly high-risk population. Fortunately, the occurrence of myo/pericarditis was relatively uncommon, and the overall cardiovascular risks of dual ICI therapy appeared to be offset by a significant mortality benefit. The use of multimodal cardiac imaging can be helpful in stratifying risk and guiding preventative cardiovascular management in patients receiving ICIs.
Collapse
Affiliation(s)
- Jennifer M. Kwan
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Miles Shen
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Narjes Akhlaghi
- Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Jiun-Ruey Hu
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Ruben Mora
- Nuvance Health, Danbury, Connecticut, United States of America
| | - James L. Cross
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Matthew Jiang
- Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Michael Mankbadi
- Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Peter Wang
- Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Saif Zaman
- Yale University School of Medicine, New Haven, Connecticut, United States of America
- Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Seohyuk Lee
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Yunju Im
- Department of Biostatistics, University of Nebraska Medical Center, Omaha, Nebraska, United States of America
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, United States of America
| | - Attila Feher
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Yi-Hwa Liu
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| | - Shuangge S. Ma
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, United States of America
| | - Weiwei Tao
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, United States of America
| | - Wei Wei
- Department of Biostatistics, Yale University School of Public Health, New Haven, Connecticut, United States of America
| | - Lauren A. Baldassarre
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut, United States of America
- Yale University School of Medicine, New Haven, Connecticut, United States of America
| |
Collapse
|
33
|
Ko J, Wardill T, Tan S, Ramkumar S. Immunotherapy-mediated myocarditis and concurrent vasospastic angina in a patient with established ischaemic heart disease: a case report. Eur Heart J Case Rep 2024; 8:ytae620. [PMID: 39659463 PMCID: PMC11630064 DOI: 10.1093/ehjcr/ytae620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/12/2024] [Accepted: 11/01/2024] [Indexed: 12/12/2024]
Abstract
Background Immunotherapy has become a pillar of modern oncological management but is associated with significant immunotherapy-related adverse events (IRAEs). While myocarditis is a prominent IRAE which clinicians are increasingly aware of, immunotherapy-related coronary vasospasm is far less appreciated and can be especially difficult to elucidate in pre-existing coronary artery disease. This case demonstrates the approach to diagnosis and management of multiple cardiovascular and non-cardiovascular IRAEs. Case summary A 57-year-old male with a history of metastatic melanoma on combined immunotherapy and ischaemic heart disease with multiple previous percutaneous coronary interventions presented with typical chest pain and troponin rise. Differential diagnoses for this presentation included a non-ST elevation myocardial infarction, myocarditis, and coronary vasospasm. Coronary angiogram did not reveal any new significant obstructive coronary artery disease while cardiac MRI did not reveal any radiological features consistent with myocarditis. However, empirical treatment for IRAEs resulted in both clinical and biochemical improvement and ability to discharge the patient on steroids and coronary vasodilators, having been GTN-infusion dependent as an inpatient. Discussion Cardiovascular IRAEs are important to be aware of when managing patients on immunotherapy and more than one IRAE can occur concurrently. Given the caveats of non-invasive imaging and invasive nature of endomyocardial biopsy, the clinical history is key in establishing these crucial diagnoses which will significantly impact ongoing oncological management.
Collapse
Affiliation(s)
- Jefferson Ko
- Department of Cardio-oncology, Victorian Heart Hospital, 631 Blackburn Road, Clayton, Melbourne, VIC 3168, Australia
| | - Thomas Wardill
- Department of Cardio-oncology, Victorian Heart Hospital, 631 Blackburn Road, Clayton, Melbourne, VIC 3168, Australia
| | - Sean Tan
- Department of Cardio-oncology, Victorian Heart Hospital, 631 Blackburn Road, Clayton, Melbourne, VIC 3168, Australia
- Department of Cardio-oncology, Victorian Heart Institute, 631 Blackburn Road, Clayton, Melbourne, VIC 3168, Australia
| | - Satish Ramkumar
- Department of Cardio-oncology, Victorian Heart Hospital, 631 Blackburn Road, Clayton, Melbourne, VIC 3168, Australia
- Department of Cardio-oncology, Victorian Heart Institute, 631 Blackburn Road, Clayton, Melbourne, VIC 3168, Australia
| |
Collapse
|
34
|
Blum SM, Zlotoff DA, Smith NP, Kernin IJ, Ramesh S, Zubiri L, Caplin J, Samanta N, Martin S, Wang M, Tirard A, Song Y, Xu KH, Barth J, Sen P, Slowikowski K, Tantivit J, Manakongtreecheep K, Arnold BY, Nasrallah M, Pinto CJ, McLoughlin D, Jackson M, Chan P, Lawless A, Michaud WA, Sharova T, Nieman LT, Gainor JF, Wu CJ, Juric D, Mino-Kenudson M, Oliveira G, Sullivan RJ, Boland GM, Stone JR, Thomas MF, Neilan TG, Reynolds KL, Villani AC. Immune responses in checkpoint myocarditis across heart, blood and tumour. Nature 2024; 636:215-223. [PMID: 39506125 DOI: 10.1038/s41586-024-08105-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 09/24/2024] [Indexed: 11/08/2024]
Abstract
Immune checkpoint inhibitors are widely used anticancer therapies1 that can cause morbid and potentially fatal immune-related adverse events such as immune-related myocarditis (irMyocarditis)2-5. The pathogenesis of irMyocarditis and its relationship to antitumour immunity remain poorly understood. Here we sought to define immune responses in heart, tumour and blood in patients with irMyocarditis by leveraging single-cell RNA sequencing coupled with T cell receptor (TCR) sequencing, microscopy and proteomics analyses of samples from 28 patients with irMyocarditis and 41 unaffected individuals. Analyses of 84,576 cardiac cells by single-cell RNA sequencing combined with multiplexed microscopy demonstrated increased frequencies and co-localization of cytotoxic T cells, conventional dendritic cells and inflammatory fibroblasts in irMyocarditis heart tissue. Analyses of 366,066 blood cells revealed decreased frequencies of plasmacytoid dendritic cells, conventional dendritic cells and B lineage cells but an increased frequency of other mononuclear phagocytes in irMyocarditis. Fifty-two heart-expanded TCR clones from eight patients did not recognize the putative cardiac autoantigens α-myosin, troponin I or troponin T. Additionally, TCRs enriched in heart tissue were largely nonoverlapping with those enriched in paired tumour tissue. The presence of heart-expanded TCRs in a cycling blood CD8 T cell population was associated with fatal irMyocarditis case status. Collectively, these findings highlight crucial biology driving irMyocarditis and identify putative biomarkers.
Collapse
Affiliation(s)
- Steven M Blum
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Mass General Cancer Center, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel A Zlotoff
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Neal P Smith
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Isabela J Kernin
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Swetha Ramesh
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Leyre Zubiri
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Mass General Cancer Center, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Joshua Caplin
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Nandini Samanta
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Sidney Martin
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mike Wang
- Mass General Cancer Center, Boston, MA, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Yuhui Song
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Katherine H Xu
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jaimie Barth
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Pritha Sen
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Transplant, Oncology and Immunocompromised Host Program, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Division of Rheumatology, North Shore Physicians Group, Department of Medicine, Mass General Brigham Healthcare Center, Lynn, MA, USA
| | - Kamil Slowikowski
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kasidet Manakongtreecheep
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Mazen Nasrallah
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Rheumatology, North Shore Physicians Group, Department of Medicine, Mass General Brigham Healthcare Center, Lynn, MA, USA
| | - Christopher J Pinto
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Daniel McLoughlin
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Monica Jackson
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - PuiYee Chan
- Mass General Cancer Center, Boston, MA, USA
- Clinical Research Center, Massachusetts General Hospital, Boston, MA, USA
| | - Aleigha Lawless
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - William A Michaud
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Tatyana Sharova
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - Linda T Nieman
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Justin F Gainor
- Mass General Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Catherine J Wu
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Dejan Juric
- Mass General Cancer Center, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Mari Mino-Kenudson
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Giacomo Oliveira
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Ryan J Sullivan
- Mass General Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Genevieve M Boland
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA
| | - James R Stone
- Harvard Medical School, Boston, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Molly F Thomas
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
- Harvard Medical School, Boston, MA, USA
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Gastroenterology, Department of Medicine, Oregon Health and Sciences University, Portland, OR, USA
- Department of Cell, Developmental, and Cancer Biology, Oregon Health and Sciences University, Portland, OR, USA
| | - Tomas G Neilan
- Harvard Medical School, Boston, MA, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Kerry L Reynolds
- Mass General Cancer Center, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Krantz Family Center for Cancer Research, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
35
|
Gürdoğan M, Taylan G, Özkan U, Ebik M, Solak N, Gürlertop Y, Yalta K. Atrioventricular Block in the Setting of Immune Myocarditis: A Pragmatic Approach to Diagnosis and Treatment. Pacing Clin Electrophysiol 2024; 47:1617-1626. [PMID: 39549256 DOI: 10.1111/pace.15108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 05/01/2024] [Accepted: 10/27/2024] [Indexed: 11/18/2024]
Abstract
Immunotherapy has revolutionized cancer treatment in the last decade and has significantly improved patient survival. However, immunotherapy is associated with serious cardiac adverse events including myocarditis and conduction disturbances. In the literature, the mortality rate in patients with immunotherapy-associated myocarditis and complete AV block is reported to be approximately 60%. Current cardio-oncology guidelines provide a series of recommendations for the management of immune myocarditis (IM). However, there is no recommendation on whether or when pacemaker implantation should be performed in the setting of complete AV block associated with myocarditis. This gap in the literature has led to a trend in cardio-oncology practice to implant permanent pacemakers (PPMs) in a significant proportion of patients without waiting for a response to immunosuppressive therapy. However, in a significant proportion of patients undergoing PPM implantation, complete AV block resolves after immunosuppressive therapy. This suggests that in cases of complete AV block in the setting of IM, more robust clues are needed for PPM implantation. This review aims to present algorithms for the management of myocarditis and complete AV block, one of the most lethal complications of immunotherapy, to help fill this gap in the literature.
Collapse
Affiliation(s)
- Muhammet Gürdoğan
- Department of Cardiology, School of Medicine, Trakya University, Edirne, Turkey
| | - Gökay Taylan
- Department of Cardiology, School of Medicine, Trakya University, Edirne, Turkey
| | - Uğur Özkan
- Department of Cardiology, School of Medicine, Trakya University, Edirne, Turkey
| | - Mustafa Ebik
- Department of Cardiology, School of Medicine, Trakya University, Edirne, Turkey
| | - Nilay Solak
- Department of Cardiology, School of Medicine, Trakya University, Edirne, Turkey
| | - Yekta Gürlertop
- Department of Cardiology, School of Medicine, Trakya University, Edirne, Turkey
| | - Kenan Yalta
- Department of Cardiology, School of Medicine, Trakya University, Edirne, Turkey
| |
Collapse
|
36
|
Roumi JE, Taimeh Z. Emerging Biomarkers in Cardiac Sarcoidosis and Other Inflammatory Cardiomyopathies. Curr Heart Fail Rep 2024; 21:570-579. [PMID: 39365404 PMCID: PMC11511729 DOI: 10.1007/s11897-024-00683-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/04/2024] [Indexed: 10/05/2024]
Abstract
PURPOSE OF REVIEW Cardiac sarcoidosis and other inflammatory cardiomyopathies are disorders causing cardiac inflammation and leading to heart failure, arrythmias and cardiac arrest. Diagnosis of these entities remains challenging and multimodal. Thus, there is a growing need to develop reliable biomarkers that can aid in the diagnosis. This review aims to summarize and highlight recent findings in the field of biomarkers for cardiac sarcoidosis and inflammatory cardiomyopathy. RECENT FINDINGS Multiple categories of biomarkers including novel molecules are being investigated with the latest evidence showing promising results. Some of these biomarkers are proven to be useful as diagnostic and prognostic aids in cardiac sarcoid and inflammatory cardiomyopathy. The identification of cost-effective and accurate biomarkers is useful not only for enhancing diagnostic accuracy but also for informing therapeutic decision-making processes. This advancement would facilitate the timely institution of immunosuppressive therapies, ultimately leading to improved patient outcomes.
Collapse
Affiliation(s)
- Joseph El Roumi
- Section of Heart Failure and Transplantation Medicine; Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Miller Family Heart, Vascular & Thoracic Institute; Cleveland Clinic, 9500 Euclid Ave, J3-4, Cleveland, OH, 44195, USA
| | - Ziad Taimeh
- Section of Heart Failure and Transplantation Medicine; Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Miller Family Heart, Vascular & Thoracic Institute; Cleveland Clinic, 9500 Euclid Ave, J3-4, Cleveland, OH, 44195, USA.
| |
Collapse
|
37
|
Birch S, Otton J. Cardio-oncology and radiation oncology: How collaboration between cardiologists and oncologists can be realised in radiation oncology. J Med Imaging Radiat Oncol 2024; 68:962-973. [PMID: 38874328 DOI: 10.1111/1754-9485.13724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Accepted: 06/02/2024] [Indexed: 06/15/2024]
Abstract
Increased survivorship, improvements in cancer treatments, and the potential for cardiac side effects from cancer treatments have resulted in increased collaboration between oncologists and cardiologists and the development of cardio-oncology clinics. This collaboration is important given its role in ensuring greater patient satisfaction, aiding teams of clinicians in making complex treatment decision, and ensuring cardiac complications are diagnosed at an early stage. The particularities of implementing this collaboration in the field of radiation oncology and how this setting is different from other areas of cardio-oncology have not been well detailed in the literature. This paper will discuss what is currently understood about the need for and role of cardio-oncology and what a cardio-oncology services involves, with a particular emphasis on patient and clinician needs in the field of radiation oncology. The literature and recent guidelines do advocate for a detailed baseline assessment of cancer patients undergoing radiotherapy, especially patients with treatment or patient risk factors that increase their risk of cancer-therapy related cardiotoxicity. Advancements in cardiac imaging techniques will be discussed as these may help to diagnose cardiac side effects of certain cancer treatments, including radiotherapy, at an early stage. A multi-disciplinary and collaborative approach is well received by patients and such an approach, guided by the aim of maintaining a patient's cancer treatment wherever possible, should be the cornerstone of cardio-oncology clinics regardless of the patient's treatment regime.
Collapse
Affiliation(s)
- Samuel Birch
- Cardiology Department, St Vincent's Hospital, Sydney, New South Wales, Australia
| | - James Otton
- Victor Chang Cardiac Research Institute, Sydney, New South Wales, Australia
| |
Collapse
|
38
|
van den Berg PF, Bracun V, Noordman M, van der Meer P, Shi C, Oosting SF, Aboumsallem JP, de Wit S, Meijers WC, Jalving M, van Kruchten M, de Boer RA. Elevations of Cardiac Troponin in Patients Receiving Immune Checkpoint Inhibitors: Data From a Prospective Study. JACC. ADVANCES 2024; 3:101375. [PMID: 39583866 PMCID: PMC11584941 DOI: 10.1016/j.jacadv.2024.101375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 09/11/2024] [Accepted: 09/16/2024] [Indexed: 11/26/2024]
Abstract
Background Immune checkpoint inhibitors (ICIs) are increasingly used in the treatment of cancer. However, immune-related adverse events are prevalent in patients receiving ICI therapy. A serious immune-related adverse event is ICI-myocarditis, which is complex to diagnose given that the significance of early symptoms and biomarker trajectories, such as high-sensitivity troponin T (hs-TnT) are unclear. Objectives The purpose of the study was to evaluate kinetics of hs-TnT in cancer patients receiving ICI and to identify patients at risk of developing ICI-myocarditis. Methods This prospective, observational, single-center study included 164 patients receiving ICI therapy. Patients' history, demographics, and clinical characteristics, as well as survival statistics, were collected from electronic patient records and used to analyze associations between elevated hs-TnT (≥14 ng/L) and a significant rise in hs-TnT (100% rise from baseline, with an absolute value ≥2x upper limit of normal (ie, ≥28 ng/L) with ICI-myocarditis. Results We included 164 patients with a mean follow-up time of 1.60 ± 0.91 years. Melanoma was the most common type of cancer in the patient population, and most patients received treatment with programmed cell death protein 1 (PD-1). Twenty-six patients (16%) exhibited significant hs-TnT elevations, while 8 patients (5%) developed ICI-myocarditis. In 18 of 26 (69%) patients, ICI-myocarditis could not be diagnosed with certainty, while 10 of 26 (38%) patients had no other signs of symptoms of cardiac damage. All 8 myocarditis cases were preceded by significantly higher hs-TnT elevations than asymptomatic patients. Despite a high ICI-myocarditis incidence in our study population, cardiac mortality remained low (4%). Conclusions Significant hs-TnT elevations occur more often than previously reported, are often asymptomatic, and do not always lead to myocarditis diagnosis.
Collapse
Affiliation(s)
- Pieter F. van den Berg
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Valentina Bracun
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Michel Noordman
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Canxia Shi
- Erasmus Medical Center, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - Sjoukje F. Oosting
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | | | - Sanne de Wit
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
| | - Wouter C. Meijers
- Department of Cardiology, University Medical Center Groningen, Groningen, the Netherlands
- Erasmus Medical Center, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| | - Mathilde Jalving
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Michel van Kruchten
- Department of Medical Oncology, University Medical Center Groningen, Groningen, the Netherlands
| | - Rudolf A. de Boer
- Erasmus Medical Center, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, the Netherlands
| |
Collapse
|
39
|
Jaber Chehayeb R, Singh J, Matute-Martinez C, Chen NW, Guajardo AF, Lin D, Jayakrishnan R, Christofides A, Leveille E, Im Y, Biancon G, VanOudenhove J, Ibrahim E, Ardasheva A, Jha A, Hwa J, Halene S, Kwan JM. Clonal hematopoiesis of indeterminate potential is associated with increased risk of immune checkpoint inhibitor myocarditis in a prospective study of a cardio-oncology cohort. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:84. [PMID: 39587635 PMCID: PMC11590368 DOI: 10.1186/s40959-024-00289-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP) has been shown to increase all-cause mortality and risk of cardiomyopathy in patients with solid malignancies. CHIP has also been shown to increase T cell activation in heart failure patients. It is unclear whether CHIP can affect the risk of immune checkpoint inhibitor (ICI) myocarditis in patients with cancer treated with immunotherapy. METHODS We enrolled patients with solid tumors in a prospective study, determined CHIP status at time of enrollment through blood whole exome sequencing, and assessed incidence of ICI myocarditis from time of enrollment through December 1st, 2023. We performed a competing risk cox regression to evaluate the role of CHIP in ICI myocarditis, accounting for patient demographics, cardiac comorbidities, cardiotoxic cancer therapy, and dual ICI use in our covariates. We also generated cumulative incidence curves using subdistribution hazards to evaluate development of ICI myocarditis stratified by CHIP vs no CHIP. Chart review was performed to evaluate patient co-morbidities, lab values, imaging findings and outcomes. RESULTS Among the 88 patients receiving ICI therapy, average age was 67 ± 14 years, of which 50% harbored CHIP variants. Among all comorbidities, including diabetes, heart failure and obstructive coronary artery disease, only coronary artery calcifications were significantly increased in patients with CHIP. There were no statistically significant differences in cancer therapy or cardiovascular drugs between patients with and without CHIP. Among examined outcomes, patients with CHIP had a statistically higher rate of ICI myocarditis (overall: 57%, CHIP: 73% (32/44), no CHIP: 41% (18/44), p = 0.003) and death (CHIP: 60%, no CHIP 31%, p = 0.011). In a multivariate competing risk analysis, CHIP status doubled the risk of developing ICI myocarditis, similar to the risk of dual ICI use (CHIP status HR 2.74, 95% CI: 1.44-5.22, p = 0.002 vs dual ICI use HR 2.39, 95% CI: 1.11-5.14, p = 0.026). CONCLUSIONS This study is the first to show that CHIP independently increases risk of ICI myocarditis, with implications for risk stratification of patients prior to ICI initiation and frequency of cardiac monitoring.
Collapse
Affiliation(s)
| | | | - Carlos Matute-Martinez
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, 06511, USA
| | | | | | | | | | | | | | - Yunju Im
- University of Nebraska Medical Center, Omaha, USA
| | - Giulia Biancon
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Jennifer VanOudenhove
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Eiman Ibrahim
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | - Anastasias Ardasheva
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
| | | | - John Hwa
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, 06511, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine and Yale Cancer Center, Yale University School of Medicine, New Haven, CT, USA
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Jennifer M Kwan
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, 06511, USA.
- Yale School of Medicine, 300 George St #770B, New Haven, CT, 06511, USA.
| |
Collapse
|
40
|
Izumi R, Hashimoto T, Kisanuki H, Ikuta K, Otsuru W, Asakawa S, Yamamoto S, Misumi K, Fujino T, Shinohara K, Matsushima S, Hosokawa K, Katsuki S, Mori T, Hashisako M, Tateishi Y, Iwasaki T, Oda Y, Kinugawa S, Abe K. Clinical and pathological characteristics of immune checkpoint inhibitor-related fulminant myocarditis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:82. [PMID: 39574216 PMCID: PMC11580468 DOI: 10.1186/s40959-024-00288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 11/12/2024] [Indexed: 11/25/2024]
Abstract
The advent of immune checkpoint inhibitors (ICIs) has significantly improved cancer treatment. With the increasing use of ICIs, ICI-related myocarditis has been recognized. However, an evidence-based therapeutic strategy has not been established because of the limited knowledge on ICI-related myocarditis. Here, we present four cases of ICI-related fulminant myocarditis (FM). Three of the four cases resulted in fatal outcomes despite aggressive treatment with mechanical circulatory support and immunosuppressive therapy with corticosteroids. Given the poor prognosis of ICI-FM, the establishment of rapid and adequate therapeutic interventions on the basis of clinical and pathological evaluation is imperative.
Collapse
Affiliation(s)
- Ryo Izumi
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Toru Hashimoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan.
| | - Hiroshi Kisanuki
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Kei Ikuta
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Wataru Otsuru
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Soshun Asakawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Shoei Yamamoto
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Kayo Misumi
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Takeo Fujino
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
- Department of Advanced Cardiopulmonary Failure, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Keisuke Shinohara
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Shouji Matsushima
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Kazuya Hosokawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Shunsuke Katsuki
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Taro Mori
- Department of Anatomic Pathology, Pathological Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Diagnostic Pathology, Kyushu University Hospital, Fukuoka, Japan
| | - Mikiko Hashisako
- Department of Anatomic Pathology, Pathological Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Diagnostic Pathology, Kyushu University Hospital, Fukuoka, Japan
| | - Yuki Tateishi
- Department of Anatomic Pathology, Pathological Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Diagnostic Pathology, Kyushu University Hospital, Fukuoka, Japan
- Department of Health Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
| | - Takeshi Iwasaki
- Department of Anatomic Pathology, Pathological Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Diagnostic Pathology, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Pathological Sciences, Faculty of Medical Sciences, Kyushu University, Fukuoka, Japan
- Division of Diagnostic Pathology, Kyushu University Hospital, Fukuoka, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| | - Kohtaro Abe
- Department of Cardiovascular Medicine, Faculty of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Clinical Research Building B 416, Higashi-Ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
41
|
Kurnik M, Kolar Kus P, Krepek M, Vlaović J, Podbregar M. Fatal rhabdomyolysis and fulminant myocarditis with malignant arrhythmias after one dose of ipilimumab and nivolumab. Immunotherapy 2024; 16:1203-1210. [PMID: 39548815 PMCID: PMC11758944 DOI: 10.1080/1750743x.2024.2427563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/06/2024] [Indexed: 11/18/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) related myocarditis is a rare complication of modern immunotherapy. It can present as an asymptomatic subclinical condition or full-blown fulminant myocarditis with malignant arrythmias and cardiogenic shock. Myositis/rhabdomyolysis and/or myasthenic symptoms can be present concomitantly. We present a case of fatal fulminant myocarditis presenting with cardiac arrythmias and severe systolic dysfunction, with accompanying rhabdomyolysis after the first dose of ipilimumab and nivolumab immunotherapy. First working diagnosis of subacute late presenting acute myocardial infarction (ACS) was incorrect and the correct diagnosis was established only after additional testing and consultation. Treatment consisted of high-dose corticosteroids, intravenous immunoglobulins, sedation with mechanical ventilation, antibiotic coverage, hemodialysis, and sustained low-efficiency daily diafiltration (SLEDD) with CytoSorb or TheraNova membranes, and intra-aortic balloon pump mechanical cardiac support. No tangible improvement in the condition was observed during the whole treatment period and the patient died on the sixth day of intensive care treatment.
Collapse
Affiliation(s)
- Marko Kurnik
- Department of Internal Intensive Medicine, General Hospital Celje, Celje, Slovenia
| | - Petra Kolar Kus
- Department of Internal Intensive Medicine, General Hospital Celje, Celje, Slovenia
| | - Mihela Krepek
- Department of Internal Intensive Medicine, General Hospital Celje, Celje, Slovenia
| | - Janko Vlaović
- Department of Internal Intensive Medicine, General Hospital Celje, Celje, Slovenia
| | - Matej Podbregar
- Department of Internal Intensive Medicine, General Hospital Celje, Celje, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
42
|
Griffiths CD, Shah M, Shao W, Borgman CA, Janes KA. Three modes of viral adaption by the heart. SCIENCE ADVANCES 2024; 10:eadp6303. [PMID: 39536108 PMCID: PMC11559625 DOI: 10.1126/sciadv.adp6303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Accepted: 10/08/2024] [Indexed: 11/16/2024]
Abstract
Viruses elicit long-term adaptive responses in the tissues they infect. Understanding viral adaptions in humans is difficult in organs such as the heart, where primary infected material is not routinely collected. In search of asymptomatic infections with accompanying host adaptions, we mined for cardio-pathogenic viruses in the unaligned reads of nearly 1000 human hearts profiled by RNA sequencing. Among virus-positive cases (~20%), we identified three robust adaptions in the host transcriptome related to inflammatory nuclear factor κB (NF-κB) signaling and posttranscriptional regulation by the p38-MK2 pathway. The adaptions are not determined by the infecting virus, and they recur in infections of human or animal hearts and cultured cardiomyocytes. Adaptions switch states when NF-κB or p38-MK2 is perturbed in cells engineered for chronic infection by the cardio-pathogenic virus, coxsackievirus B3. Stratifying viral responses into reversible adaptions adds a targetable systems-level simplification for infections of the heart and perhaps other organs.
Collapse
Affiliation(s)
- Cameron D. Griffiths
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Millie Shah
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - William Shao
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Cheryl A. Borgman
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
| | - Kevin A. Janes
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA 22908, USA
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
| |
Collapse
|
43
|
Villatore A, Bosi C, Pomaranzi C, Cigliola A, Tateo V, Mercinelli C, Vignale D, Rizzo S, Necchi A, Peretto G. Myocarditis Following Pembrolizumab Plus Axitinib, and Belzutifan Plus Lenvatinib for Renal Cell Carcinoma: A Case Report. Cardiovasc Toxicol 2024; 24:1168-1173. [PMID: 39085529 DOI: 10.1007/s12012-024-09906-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Cardiac toxicity is an adverse event of several classes of anti-cancer drugs. Herein, we present the case of a 52-year-old woman with metastatic renal cell carcinoma (RCC), previously treated with debulking surgery, pembrolizumab (immune checkpoint inhibitor) in combination with axitinib (tyrosine kinase inhibitor (TKI)), followed by lenvatinib (TKI) and belzutifan (HIF-2α inhibitor), who developed myocarditis proven by cardiac magnetic resonance and endomyocardial biopsy. The case was notable for reporting a not-yet described adverse event during treatment with belzutifan plus lenvatinib, the etiology of which was of unobvious determination given the pre-exposure to pembrolizumab, a known cause of drug-related myocarditis. We surmise that myocarditis was a delayed adverse event related to pembrolizumab (8 months after treatment interruption), although we emphasize that only attentive monitoring of cardiac adverse events of patients exposed to belzutifan and lenvatinib in the context of large clinical trials may rule out any causal implication of these drugs.
Collapse
Affiliation(s)
- Andrea Villatore
- Vita-Salute San Raffaele University, Milan, Italy.
- Disease Unit for Myocarditis & Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Hospital, Milan, Italy.
| | - Carlo Bosi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | | | - Antonio Cigliola
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Valentina Tateo
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Chiara Mercinelli
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Davide Vignale
- Experimental Imaging Center, Radiology Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Stefania Rizzo
- Cardiovascular Pathology Unit, Azienda Ospedaliera Di Padova, Padua, Italy
| | - Andrea Necchi
- Vita-Salute San Raffaele University, Milan, Italy
- Department of Medical Oncology, IRCCS San Raffaele Hospital, Milan, Italy
| | - Giovanni Peretto
- Vita-Salute San Raffaele University, Milan, Italy
- Disease Unit for Myocarditis & Arrhythmogenic Cardiomyopathies, IRCCS San Raffaele Hospital, Milan, Italy
| |
Collapse
|
44
|
Rossi S, Farina A, Malvaso A, Dinoto A, Fionda L, Cornacchini S, Florean I, Zuliani L, Garibaldi M, Lauletta A, Baccari F, Zenesini C, Rinaldi R, Mariotto S, Damato V, Diamanti L, Gastaldi M, Vogrig A, Marchioni E, Guarino M. Clinical Course of Neurologic Adverse Events Associated With Immune Checkpoint Inhibitors: Focus on Chronic Toxicities. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200314. [PMID: 39298719 PMCID: PMC11413993 DOI: 10.1212/nxi.0000000000200314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/29/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND AND OBJECTIVES The clinical course and the risk of chronicity of neurologic immune-related adverse events (n-irAEs) associated with immune checkpoint inhibitors (ICIs) are not well documented. This study aimed to characterize the clinical course of n-irAEs and assess the prevalence of chronic events. METHODS This nationwide, multicenter, retrospective study included patients with n-irAEs identified at 7 Italian hospitals. The clinical course of n-irAEs was categorized into fulminant (if resulted in death within 12 weeks), monophasic (if resolved within 12 weeks), and chronic (if persisted beyond 12 weeks). Chronic n-irAEs were further subdivided into active (if there was indirect evidence of ongoing inflammation [i.e., required ongoing immunosuppression, relapsed on steroid tapering, or exhibited neurologic progression]) and inactive (if patients had neurologic sequelae without ongoing inflammation). Comparisons between groups and time-to-death analyses were performed. RESULTS Sixty-six patients were included (median age: 69 years [IQR 62-75]; 53 [80%] men). n-irAEs involved the peripheral nervous system in 48 patients (73%), the central nervous system in 14 (21%), and both in 4 (6%). Twelve patients (18%) had a fulminant course, with the risk being significantly higher in those with concurrent myocarditis (OR 5.4; 95% CI [1.02-28.31]). Among 54 patients with a nonfulminant course, 23 (43%) had a monophasic n-irAE and 31 (57%) had a chronic n-irAE, of which 16 of 31 (52%) were chronic active (due to ongoing immunosuppression [69%], relapses at corticosteroid tapering [19%], or neurologic disease progression [12%]) and 15 of 31 (48%) were chronic inactive. In patients with chronic inactive n-irAEs, neurologic sequelae included cerebellar ataxia (33%), neuromuscular weakness (27%), visual loss (13%), sensory disturbances (13%), focal neurologic signs (7%), and cognitive impairment (7%). Compared with patients with monophasic events, those with chronic n-irAEs had a higher rate of severe neurologic disability at the last evaluation (p < 0.01), shorter survival (p < 0.01), and higher overall mortality (p < 0.01), primarily due to cancer progression. DISCUSSION More than half of the patients with n-irAEs who survived the acute phase developed a chronic condition. Patients with chronic n-irAEs were at higher risk of death, mainly due to cancer progression. Future studies are needed to further characterize chronic n-irAEs and identify optimal long-term management strategies.
Collapse
Affiliation(s)
- Simone Rossi
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Antonio Farina
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Antonio Malvaso
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Alessandro Dinoto
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Laura Fionda
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Sara Cornacchini
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Irene Florean
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Luigi Zuliani
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Matteo Garibaldi
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Antonio Lauletta
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Flavia Baccari
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Corrado Zenesini
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Rita Rinaldi
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Sara Mariotto
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Valentina Damato
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Luca Diamanti
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Matteo Gastaldi
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Alberto Vogrig
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Enrico Marchioni
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| | - Maria Guarino
- From the IRCCS Istituto delle Scienze Neurologiche di Bologna (S.R., R.R., M. Guarino), Italy; Reference Centre for Paraneoplastic Neurological Syndromes and Autoimmune Encephalitis (A.F), Hospices Civils de Lyon, Neurological Hospital, Bron, MeLiS - UCBL-CNRS UMR 5284 - INSERM U1314, Universitè Claude Bernard Lyon 1, France; IRCCS Mondino Foundation (A.M., L.D., E.M.), Pavia; Neurology Unit (A.D., S.M.), Department of Neurosciences, Biomedicine, and Movement Sciences, University of Verona, Italy; Neuromuscular and Rare Disease Centre (L.F.), Neurology Unit, Sant'Andrea University Hospital, Rome, Italy; Department of Neurosciences Drugs and Child Health (S.C., V.D.), University of Florence; Clinical Neurology Unit (I.F., A.V.), Department of Medicine (DMED), University of Udine; Neurology Unit (L.Z.), AULSS8 Berica, San Bortolo Hospital, Vicenza; Department of Neuroscience (M. Garibaldi, A.L.), Mental Health and Sensory Organs (NESMOS), SAPIENZA University of Rome, Sant'Andrea Hospital; Epidemiology and Statistics Unit (F.B., C.Z.), IRCCS Istituto delle Scienze Neurologiche di Bologna; Department of Neurology 2, Careggi University Hospital, Florence, Italy; Neuroimmunology Laboratory (M.Gastaldi), IRCCS Mondino Foundation, Pavia, Italy; and Clinical Neurology (A.V.), Department of Head-Neck and Neuroscience, Azienda Sanitaria Universitaria Friuli Centrale (ASUFC)
| |
Collapse
|
45
|
Abulizi A, Yan G, Xu Q, Muhetaer R, Wu S, Abudukelimu K, Chen X, Liu C, Li J. Cardiovascular adverse events and immune-related adverse events associated with PD-1/PD-L1 inhibitors for head and neck squamous cell carcinoma (HNSCC). Sci Rep 2024; 14:25919. [PMID: 39472591 PMCID: PMC11522629 DOI: 10.1038/s41598-024-75099-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 10/01/2024] [Indexed: 11/02/2024] Open
Abstract
While some literature has provided limited information about the potential cardiovascular risk and immune-related adverse events (irAEs) risk associated with PD-1/PD-L1 inhibitors in the treatment of Head and Neck Squamous Cell Carcinoma (HNSCC), the exact relevance is still uncertain. To assess the pharmacovigilance (PV), constituent ratio, severity, and reaction outcomes of major adverse cardiovascular events (MACE) and immune-related adverse events (irAEs) related to PD-1/PD-L1 inhibitors for HNSCC reported to the United States Food and Drug Administration Adverse Event Reporting System (FAERS). We analyzed reports of cardiovascular adverse events and irAEs associated with drug therapy for HNSCC submitted to FAERS from the 1st quarter 2015 to the 3rd quarter of 2023. Three PD-1/PD-L1 inhibitors were identified: nivolumab, pembrolizumab and durvalumab. Our primary composite endpoint was the PV of MACE and irAEs related to PD-1/PD-L1 inhibitors in the treatment of HNSCC, and the secondary endpoint was PV of other cardiovascular events. The software implemented was STATA 17.0 MP. 19,372 suspected drug-adverse event reports related to drug treatment in patients with HNSCC were identified, of which 916 reports were cardiovascular events, including 555 reports of MACE and 361 reports of other cardiovascular events. The PV signal regarding MACE was detected in durvalumab (PRR = 2.12, 95% CI: 1.24-3.61; χ2 = 7.71; ROR = 2.19, 95% CI: 1.24-3.86; IC = 1.01; IC025 = 0.07) but not in nivolumab and pembrolizumab. The constituent ratio of MACE in all adverse events caused by nivolumab (OR = 0.38, 95% CI: 0.19-0.73) and pembrolizumab (OR = 0.48, 95% CI: 0.23-0.99) was significantly decreased, compared with durvalumab. A PV signal about other cardiovascular events was detected in durvalumab (PRR = 3.04, 95% CI: 1.73-5.31; χ2 = 16.13; ROR = 3.15, 95% CI: 1.74-5.70; IC = 1.46; IC025 = 0.48), but it was not detected in nivolumab or pembrolizumab. The constituent ratio of other cardiovascular events in all adverse events caused by nivolumab (OR = 0.25, 95% CI: 0.13-0.48) and pembrolizumab (OR = 0.40, 95% CI: 0.20-0.80) was significantly decreased, compared with durvalumab. The constituent ratio of other cardiovascular events in all adverse events caused by nivolumab (OR = 0.61, 95% CI: 0.38-0.99) was significantly decreased, compared with pembrolizumab. There were 40 cases of hypertension. A PV signal about hypertension was detected in pembrolizumab (PRR = 3.72, 95% CI: 1.87-7.43; χ2 = 15.99; ROR = 3.75, 95% CI: 1.87-7.51; IC = 1.53, IC025 = 0.45), but it was not detected in nivolumab. The constituent ratio of hypertension in all adverse events caused by nivolumab (OR = 0.09, 95% CI: 0.09-0.39) was significantly decreased, compared with pembrolizumab. There were 737 cases of irAEs. A PV signal about irAEs was detected in nivolumab (PPR = 1.27, 95% CI: 1.05-1.53; χ2 = 6.38; ROR = 1.28, 95% CI: 1.06-1.56; IC = 0.29, IC025 = -0.00) and pembrolizumab (PPR = 2.20, 95% CI: 1.79-2.71; χ2 = 56.55; ROR = 2.31, 95% CI: 1.84-2.88; IC = 1.03; IC025 = 0.68), but it was not detected in durvalumab. The constituent ratio of irAEs in all adverse events caused by nivolumab (OR = 0.58, 95% CI: 0.44-0.76) significantly decreased, compared with pembrolizumab. By comparing the PV signals, constituent ratio, severity, and reaction outcome of the three drugs, we suppose that nivolumab can be used as the safest PD-1/PD-L1 inhibitor for HNSCC.
Collapse
Affiliation(s)
- Adila Abulizi
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Guangpeng Yan
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Qian Xu
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Reyihanguli Muhetaer
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Shihan Wu
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Kudelaiti Abudukelimu
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China
| | - Xi Chen
- School of Health, Brooks College, Sunnyvale, USA
- Department of Epidemiology and Statistics, School of Public Health, Medical College, Zhejiang University, Hangzhou, China
| | - Chengjiang Liu
- Department of General Medicine, Anhui Medical University, Hefei, 230000, China
| | - Jun Li
- Department of Maxillofacial Surgery, People's Hospital of Xinjiang Uygur Autonomous Region, Xinjiang, China.
| |
Collapse
|
46
|
Gutierrez C, Rajendram P, Idowu O. Novel Cancer Therapeutics: Perioperative Implications and Challenges. Anesth Analg 2024:00000539-990000000-01013. [PMID: 39453847 DOI: 10.1213/ane.0000000000007210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2024]
Abstract
Since the introduction of immunotherapy and targeted therapies, patients not only have adequate tumoral response to these treatments, but their quality of life has improved due to milder toxicities. However, due to their wide mechanisms of action, the toxicity profile for these therapies is broad, can have an insidious onset, and their recognition can be challenging. Rarely, some of these toxicities can cause significant morbidity if not diagnosed early and lead to intensive care unit (ICU) admission and death. Anesthesiologists are likely to encounter not only a wide spectrum of these toxicities but also a wide range of severity. In some cases, they could be the first to make the diagnosis and therefore need to be prepared to rapidly assess, establish differentials, perform a diagnostic workup, and evaluate the impact the toxicity could have on the patients' care during the perioperative period. In this article, we set to review toxicities of novel cancer therapies such as checkpoint inhibitors and targeted therapies, that could present in the perioperative setting. This article will help as a guide for anesthesiologists to recognize their clinical presentation, the approach to their diagnosis, and their impact on patient care.
Collapse
Affiliation(s)
- Cristina Gutierrez
- From the Department of Critical Care Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Prabalini Rajendram
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Olakunle Idowu
- Department of Anesthesiology and Perioperative Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
47
|
Cheema PK, Iafolla MAJ, Abdel-Qadir H, Bellini AB, Chatur N, Chandok N, Comondore VR, Cunningham M, Halperin I, Hu AB, Jaskolka D, Darvish-Kazem S, Khandaker MH, Kitchlu A, Sachdeva JS, Shapera S, Woolnough NRJ, Nematollahi M. Managing Select Immune-Related Adverse Events in Patients Treated with Immune Checkpoint Inhibitors. Curr Oncol 2024; 31:6356-6383. [PMID: 39451777 PMCID: PMC11506662 DOI: 10.3390/curroncol31100473] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
The increased use of immune checkpoint inhibitors (ICIs) across cancer programs has created the need for standardized monitoring and management of immune-related adverse events (irAEs). Delayed recognition without appropriate treatment can have serious and life-threatening consequences. The management of irAEs presents a unique set of challenges that must be addressed at a multidisciplinary level. Although various national and international guidelines and working groups provide high-level recommendations for the management of irAEs, practical guidance is lacking. Furthermore, timely collaboration between specialists requires institutional protocols that enable the early recognition, assessment, and treatment of irAEs. Such protocols should be developed by institution specialists and include algorithms for all healthcare providers involved in the care of patients treated with ICIs. At William Osler Health System in Brampton, Ontario, practical step-by-step multidisciplinary treatment approaches with recommendations for the management of irAEs were developed in collaboration with experts across Canada. Here, we provide an in-depth description of the approaches, outlining baseline investigations prior to the initiation of ICIs, as well as the monitoring and management of irAEs based on symptoms, severity, and involved organ systems. We encourage other centres to adapt and modify our approaches according to their specific needs and requirements.
Collapse
Affiliation(s)
- Parneet K. Cheema
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Marco A. J. Iafolla
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Husam Abdel-Qadir
- Women’s College Hospital Research Institute, Toronto, ON M5S 1B2, Canada;
- Division of Cardiology, Peter Munk Cardiac Centre, Toronto General Hospital, University Health Network, Toronto, ON M5G 2N2, Canada
| | - Andrew B. Bellini
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Nazira Chatur
- Division of Gastroenterology, Faculty of Medicine, Vancouver General Hospital (Sanders), University of British Columbia, Vancouver, BC V5Z 1M9, Canada;
| | - Natasha Chandok
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Vikram R. Comondore
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Morven Cunningham
- Toronto Centre for Liver Disease, University Health Network, Toronto, ON M5G 2C4, Canada;
| | - Ilana Halperin
- Division of Endocrinology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada;
| | - Anne B. Hu
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Diana Jaskolka
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Saeed Darvish-Kazem
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Masud H. Khandaker
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Abhijat Kitchlu
- Division of Nephrology, Department of Medicine, University Health Network, University of Toronto, Toronto, ON M5G 2C4, Canada;
| | - Jasdip S. Sachdeva
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Shane Shapera
- Department of Medicine, University of Toronto, Toronto, ON M5G 2N2, Canada;
| | - Nicholas R. J. Woolnough
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| | - Massey Nematollahi
- William Osler Health System, Brampton, ON L6R 3J7, Canada; (M.A.J.I.); (A.B.B.); (N.C.); (V.R.C.); (A.B.H.); (D.J.); (S.D.-K.); (M.H.K.); (J.S.S.); (N.R.J.W.); (M.N.)
| |
Collapse
|
48
|
Zhong Y, Li Z, Tao J, Yuan J, Fu Z. Drug-induced myocarditis: a real-world pharmacovigilance study using the FDA adverse event reporting system database. Expert Opin Drug Saf 2024:1-8. [PMID: 39400122 DOI: 10.1080/14740338.2024.2416933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 10/15/2024]
Abstract
BACKGROUND Myocarditis is a rare but potentially life-threatening inflammation of the heart muscle that can be caused by various drugs. This study aimed to comprehensively evaluate the risk of drug-induced myocarditis using data from the FDA Adverse Event Reporting System (FAERS) database. METHODS We queried the FAERS database for reports of myocarditis from Q1 2004 to Q4 2023. The reporting odds ratio (ROR) and proportional reporting ratio (PRR) were calculated to detect disproportionality signals for drugs associated with myocarditis. RESULTS A total of 8,212 myocarditis-related reports were identified in the FAERS database. The most frequently reported drugs were clozapine (N = 1269), followed by nivolumab (N = 621), pembrolizumab (N = 358), mesalazine (252), and olanzapine (N = 191). Disproportionality analysis revealed strong signals for the top 50 drugs, including mesalazine (ROR 48.01, 95% CI 42.29-54.49), cemiplimab (ROR 38.84, 95% CI 26.71-56.47), clozapine (ROR 35.21, 95% CI 33.13-37.39), nivolumab (ROR 23.21, 95% CI 21.38-25.2), atezolizumab (ROR 20.75, 95% CI 17.91-24.05) and pembrolizumab (ROR 19.90, 95% CI 17.89-22.13). CONCLUSIONS Our findings suggest a potential risk of drug-induced myocarditis associated with various medications. Close monitoring for signs and symptoms of myocarditis is crucial, especially in patients with risk factors or those receiving these drugs. Further investigations are warranted to establish causality and identify risk factors.
Collapse
Affiliation(s)
- Yunxiang Zhong
- Department of Pharmacy, Dongguan Binhaiwan Center Hospital, Dongguan, China
| | - Zhiping Li
- Department of Pharmacy, Dongguan Binhaiwan Center Hospital, Dongguan, China
| | - Jinyi Tao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiao Yuan
- Department of Pharmacy, Dongguan Binhaiwan Center Hospital, Dongguan, China
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
49
|
Kalinoski H, Daoud A, Rusinkevich V, Jurčová I, Talor MV, Welsh RA, Hughes D, Zemanová K, Stříž I, Hooper JE, Kautzner J, Peichl P, Melenovský V, Won T, Čiháková D. Injury-induced myosin-specific tissue-resident memory T cells drive immune checkpoint inhibitor myocarditis. Proc Natl Acad Sci U S A 2024; 121:e2323052121. [PMID: 39378095 PMCID: PMC11494310 DOI: 10.1073/pnas.2323052121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 09/10/2024] [Indexed: 10/10/2024] Open
Abstract
Cardiac myosin-specific (MyHC) T cells drive the disease pathogenesis of immune checkpoint inhibitor-associated myocarditis (ICI-myocarditis). To determine whether MyHC T cells are tissue-resident memory T (TRM) cells, we characterized cardiac TRM cells in naive mice and established that they have a distinct phenotypic and transcriptional profile that can be defined by their upregulation of CD69, PD-1, and CXCR6. We then investigated the effects of cardiac injury through a modified experimental autoimmune myocarditis mouse model and an ischemia-reperfusion injury mouse model and determined that cardiac inflammation induces the recruitment of autoreactive MyHC TRM cells, which coexpress PD-1 and CD69. To investigate whether the recruited MyHC TRM cells could increase susceptibility to ICI-myocarditis, we developed a two-hit ICI-myocarditis mouse model where cardiac injury was induced, mice were allowed to recover, and then were treated with anti-PD-1 antibodies. We determined that mice who recover from cardiac injury are more susceptible to ICI-myocarditis development. We found that murine and human TRM cells share a similar location in the heart and aggregate along the perimyocardium. We phenotyped cells obtained from pericardial fluid from patients diagnosed with dilated cardiomyopathy and ischemic cardiomyopathy and established that pericardial T cells are predominantly CD69+ TRM cells that up-regulate PD-1. Finally, we determined that human pericardial macrophages produce IL-15, which supports and maintains pericardial TRM cells.
Collapse
Affiliation(s)
- Hannah Kalinoski
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
| | - Vitali Rusinkevich
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Ivana Jurčová
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Monica V. Talor
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Robin A. Welsh
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - David Hughes
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
- Department of Chemical and Biomolecular Engineering, Whiting School of Engineering, Johns Hopkins University, Baltimore, MD21205
| | - Kateřina Zemanová
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Ilja Stříž
- Department of Clinical and Transplant Immunology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Jody E. Hooper
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Josef Kautzner
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Petr Peichl
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Vojtěch Melenovský
- Department of Cardiology, Institute for Clinical and Experimental Medicine, Prague140 21, Czech Republic
| | - Taejoon Won
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD21205
- Department of Pathology, School of Medicine, Johns Hopkins University, Baltimore, MD21205
| |
Collapse
|
50
|
Yu J, Long B, Li Z, Tian X, Li D, Long J, Wang Y, Chen Y, Zhang F, Liu H, Qian C, Shan J. Central memory CD4+ T cells play a protective role against immune checkpoint inhibitor-associated myocarditis. Cardiovasc Res 2024; 120:1442-1455. [PMID: 38850163 DOI: 10.1093/cvr/cvae133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Revised: 03/04/2024] [Accepted: 05/11/2024] [Indexed: 06/10/2024] Open
Abstract
AIMS The widespread use of immune checkpoint inhibitors (ICIs) has demonstrated significant survival benefits for cancer patients and also carries the risk of immune-related adverse events. ICI-associated myocarditis is a rare and serious adverse event with a high mortality rate. Here, we explored the mechanism underlying ICI-associated myocarditis. METHODS AND RESULTS Using the peripheral blood of patients with ICI therapy and of ICI-treated mice with transplanted tumours, we dissect the immune cell subsets and inflammatory factors associated with myocarditis. Compared to the control group, patients with myocarditis after ICI therapy showed an increase in NK cells and myeloid cells in the peripheral blood, while T cells significantly decreased. Among T cells, there was an imbalance of CD4/CD8 ratio in the peripheral blood of myocarditis patients, with a significant decrease in central memory CD4+ T (CD4+ TCM) cells. RNA sequencing revealed that CD4+ TCM cells in myocarditis patients were immunosuppressive cell subsets, which highly express the immunosuppressive factor IL-4I1. To elucidate the potential mechanism of the decrease in CD4+ TCM cells, protein array was performed and revealed that several inflammatory factors gradually increased with the severity of myocarditis in the myocarditis group, such as IL-1B/CXCL13/CXCL9, while the myocardial protective factor IL-15 decreased. Correlation analysis indicated a positive correlation between IL-15 and CD4+ TCM cells, with high expression of IL-15 receptor IL15RA. Furthermore, in vivo studies using an anti-PDL1 antibody in a mouse tumour model indicated a reduction in CD4+ TCM cells and an increase in effector memory-expressing CD45RA CD8+ T (TEMRA) cells, alongside evidence of cardiac fibrosis. Conversely, combining anti-PDL1 antibody treatment with IL-15 led to a resurgence of CD4+ TCM cells, a reduction in CD8+ TEMRA cells, and a mitigated risk of cardiac fibrosis. CONCLUSION Our data highlight CD4+ TCM cells' crucial role in cardiac protection during ICI therapy. IL-15, IL-4I1, and CD4+ TCM cells can serve as therapeutic targets to reduce ICI-associated myocarditis in cancer patients.
Collapse
Affiliation(s)
- Jiajun Yu
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Bo Long
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Ziyong Li
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Xiaolong Tian
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Dairong Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Jianling Long
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Yujue Wang
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Yue Chen
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Fang Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Haixia Liu
- Department of Cardio-Oncology, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Cheng Qian
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| | - Juanjuan Shan
- School of Medicine, Chongqing University, Chongqing 400030, China
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, No. 181, Hanyulu, Shapingba, Chongqing 400030, China
| |
Collapse
|