1
|
Stojanović SD, Thum T, Bauersachs J. Anti-senescence therapies: a new concept to address cardiovascular disease. Cardiovasc Res 2025; 121:730-747. [PMID: 40036821 DOI: 10.1093/cvr/cvaf030] [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: 11/05/2024] [Revised: 12/16/2024] [Accepted: 01/22/2025] [Indexed: 03/06/2025] Open
Abstract
Accumulation of senescent cells is an increasingly recognized factor in the development and progression of cardiovascular (CV) disease (CVD). Senescent cells of different types display a pro-inflammatory and matrix remodelling molecular programme, known as the 'senescence-associated secretory phenotype' (SASP), which has roots in (epi)genetic changes. Multiple therapeutic options (senolytics, anti-SASP senomorphics, and epigenetic reprogramming) that delete or ameliorate cellular senescence have recently emerged. Some drugs routinely used in the clinics also have anti-senescence effects. However, multiple challenges hinder the application of novel anti-senescence therapeutics in the clinical setting. Understanding the biology of cellular senescence, advantages and pitfalls of anti-senescence treatments, and patients who can profit from these interventions is necessary to introduce this novel therapeutic modality into the clinics. We provide a guide through the molecular machinery of senescent cells, systematize anti-senescence treatments, and propose a pathway towards senescence-adapted clinical trial design to aid future efforts.
Collapse
Affiliation(s)
- Stevan D Stojanović
- Department of Cardiology and Angiology, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- PRACTIS Clinician Scientist Program, Dean's Office for Academic Career Development, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
- Center for Translational Regenerative Medicine, Hannover Medical School, Carl Neuberg Str. 1, Hannover 30625, Germany
| |
Collapse
|
2
|
Watson H, Holley N, Nkongho TN, Patel B. New onset of hypertension associated with immune checkpoint inhibitor therapy in cancer patients. Immunotherapy 2025:1-9. [PMID: 40380783 DOI: 10.1080/1750743x.2025.2504868] [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: 01/13/2025] [Accepted: 05/08/2025] [Indexed: 05/19/2025] Open
Abstract
AIM To evaluate whether immune checkpoint inhibitor (ICI) therapy is associated with a higher incidence of hypertension (HTN) among cancer patients compared to those not treated with ICIs. MATERIALS & METHODS This retrospective cohort study utilized data from the TriNetX Research Network, a global database of de-identified electronic health records. Adult patients (≥18 years) with lung, breast, colon, kidney, or skin cancer were categorized based on ICI treatment. Patients with preexisting hypertension were excluded. Propensity score matching (1:1) based on demographics and comorbidities yielded two balanced cohorts of 24,956 patients each. The primary outcome was the incidence of hypertension within one year of cancer diagnosis. RESULTS The incidence of hypertension was significantly higher in the ICI group (13.2%) compared to the non-ICI group (9.7%). The risk ratio was 1.356 (95% CI: 1.271-1.446), and the odds ratio was 1.410 (95% CI: 1.311-1.516), both with p < 0.001. Kaplan-Meier analysis showed lower hypertension-free survival in the ICI group (log-rank p < 0.001; HR = 1.071). CONCLUSIONS ICI therapy is significantly associated with an increased risk of developing hypertension. These findings support the need for routine cardiovascular monitoring in patients receiving ICI treatment.
Collapse
Affiliation(s)
- HangYu Watson
- Department of Medicine, West Virginia University, Morgantown, WV, USA
| | - Nolan Holley
- Department of Medicine, West Virginia University, Morgantown, WV, USA
| | | | - Brijesh Patel
- Department of Cardiology, Indiana University, Indianapolis, IN, USA
| |
Collapse
|
3
|
Nonaka M, Hirakata M, Sakai C, Tomikawa E, Izawa A, Nishi T, Koga Y, Takahashi K, Shimozono R, Ohshima K, Narumi H, Miyoshi T, Oshida K, Uchida M, Uezono Y. TXB-001, A Newly-Developed Polymer-Conjugated Anthracycline, Alleviates Anthracycline-Induced Cardiotoxicity. Cardiovasc Toxicol 2025:10.1007/s12012-025-09994-2. [PMID: 40327285 DOI: 10.1007/s12012-025-09994-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 04/05/2025] [Indexed: 05/07/2025]
Abstract
Anthracycline anti-cancer drugs, which are used in cancer chemotherapy, frequently cause cardiotoxicity, the incidence of which depends on cumulative doses. TXB-001 is a new candidate polymer-conjugated pirarubicin (THP) with higher THP purity and content compared to previous P-THP (polymerized THP) and is expected to exhibit lower cardiotoxicity and higher efficacy against cancer cells. We examined the effects of TXB-001 on cardiac function and the pharmacokinetics after its intravenous administration compared with those of existing anthracyclines (doxorubicin (DOX), DOXIL (liposomal formulation of DOX), THP) in mice. Echocardiography and electrocardiography showed that DOX caused cardiac dysfunction in mice, with associated changes in organ weights, blood chemical parameters, and mRNA/protein expressions. DOXIL and THP induced similar, but weaker changes than DOX. TXB-001 did not significantly affect cardiac function or associated changes under the conditions of this study. The results of the pharmacokinetic evaluation revealed that the distributions of DOXIL and TXB-001 from plasma to heart tissue were lower than those of DOX and THP, while the distribution of TXB-001 was lower than that of DOXIL. Furthermore, TXB-001 did not show cardiac accumulation in contrast to DOXIL. In addition, the anthracycline exposure level of TXB-001 in the heart was lower than those of DOX, DOXIL, and THP, with less exposure being regarded as one reason for the low or no cardiotoxicity of TXB-001 in mice. Collectively, these results suggest the potential of TXB-001 as an anti-cancer drug with fewer side effects than anthracyclines, particularly cardiotoxicity. Novel TXB-001 may become an effective anti-cancer drug with fewer cardiotoxicity.
Collapse
Affiliation(s)
- Miki Nonaka
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan.
| | - Mikito Hirakata
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Chizuka Sakai
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Emi Tomikawa
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Akiko Izawa
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Tatsuya Nishi
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Yoko Koga
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Kei Takahashi
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Rieko Shimozono
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Kaori Ohshima
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
- Department of Pathology, Immunology, and Microbiology Graduate School of Medicine, The University of Tokyo, 7-3-1, Hongo, Bunkyo-Ku, Tokyo, 113-0033, Japan
| | - Hideki Narumi
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Tomoya Miyoshi
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Keiyu Oshida
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan
| | - Masashi Uchida
- Pharmaceutical Research Laboratories, Toray Industries, Inc, 6-10-1 Tebiro, Kamakura-City, Kanagawa, 248-8555, Japan.
| | - Yasuhito Uezono
- Department of Pain Control Research, The Jikei University School of Medicine, 3-25-8, Nishi-Shimbashi, Minato-Ku, Tokyo, 105-8461, Japan
- Supportive and Palliative Care Research Support Office, National Center Hospital East, 6-5-1, Kashiwanoha, Kashiwa-City, Chiba, 277-8577, Japan
| |
Collapse
|
4
|
Fankhauser RG, Johnson DB, Moslehi JJ, Balko JM. Preclinical mouse models of immune checkpoint inhibitor-associated myocarditis. NATURE CARDIOVASCULAR RESEARCH 2025; 4:526-538. [PMID: 40335724 DOI: 10.1038/s44161-025-00640-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 03/10/2025] [Indexed: 05/09/2025]
Abstract
In this Review, we present a comprehensive analysis of preclinical models used to study immune checkpoint inhibitor-associated myocarditis (hereafter ICI-myocarditis), a potentially lethal immune-related adverse event. We begin by providing an overview of immune checkpoint inhibitors, highlighting how their efficacy in cancer treatment is counterbalanced by their predisposition to cause immune-related adverse events. Next, we draw from human data to identify disease features that an effective mouse model should ideally mimic. After that, we present a critical evaluation of a wide variety of existing mouse models including genetic, pharmacological and humanized models. We summarize insights gathered about the underlying mechanisms of ICI-myocarditis and the role of mouse models in these discoveries. We conclude with a perspective on the future of preclinical models, highlighting potential model improvements and research directions that could strengthen our understanding of ICI-myocarditis, ultimately improving patient outcomes.
Collapse
Grants
- 5R01HL156021-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL155990-04 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01HL141466-05 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH P01 HL141084 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- NIH R01 HL160688 U.S. Department of Health & Human Services | NIH | National Heart, Lung, and Blood Institute (NHLBI)
- 5R01CA227481-05 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- 5P30CA068485-29 U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
- T32GM007347 U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS)
- 25PRE1375723 American Heart Association (American Heart Association, Inc.)
Collapse
Affiliation(s)
- Reilly G Fankhauser
- Medical Scientist Training Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Douglas B Johnson
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Javid J Moslehi
- Section of Cardio-Oncology and Immunology, Cardiovascular Research Institute, University of California San Francisco School of Medicine, San Francisco, CA, USA
| | - Justin M Balko
- Department of Medicine, Division of Hematology and Oncology, Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA.
- Breast Cancer Research Program, Vanderbilt-Ingram Cancer Center, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
5
|
Luo Y, Liu J, Qu P, Han S, Li X, Wang Y, Su X, Zeng J, Li J, Deng S, Liang Q, Hou L, Cheng P. The crosstalk of breast cancer and ischemic heart disease. Cell Death Discov 2025; 11:185. [PMID: 40251177 PMCID: PMC12008236 DOI: 10.1038/s41420-025-02428-6] [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: 08/06/2024] [Revised: 03/07/2025] [Accepted: 03/21/2025] [Indexed: 04/20/2025] Open
Abstract
In recent years, the continuous optimization of anti-tumor therapy has greatly improved the cancer-specific survival rate for patients with breast cancer (BC). The prevention and treatment of breast cancer-related heart diseases have become a new breakthrough in improving the long-term survival for BC patient. The cardiac damages caused by BC treatment are increasingly prominent among BC patients, of which ischemic heart disease (IHD) is the most prominent. Besides, the systemic inflammatory response activated by tumor microenvironment c an induce and exacerbate IHD and increase the risk of myocardial infarction (MI). Conversely, IHD can also exert detrimental effects on tumors. MI not only increases the risk of BC, but also induces specialized immune cell to BC and accelerates the progression of BC. Meanwhile, the treatment of IHD can also promote BC metastasis and transition to more aggressive phenotypes. Although BC and IHD are diseases of two independent systems, their crosstalk increases the difficulty of anti-cancer treatment and IHD management, which reduces the survival for both diseases. Therefore, this review mainly explores the mutual influence and underlying mechanisms between BC and IHD, aiming to provide insights for improving the long-term survival for patients with BC or IHD.
Collapse
Affiliation(s)
- Yunbo Luo
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Jun Liu
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Peng Qu
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
| | - Shiqi Han
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Xue Li
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China
| | - Yali Wang
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Xiaohan Su
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China
| | - Jiao Zeng
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Jinsui Li
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Shishan Deng
- Department of Academician (expert) Workstation, Biological Targeting Laboratory of Breast Cancer, Breast and Thyroid Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, P. R. China
| | - Qi Liang
- Department of Clinical Laboratory, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, People's Republic of China.
- School of Laboratory Medicine, North Sichuan Medical College, Nanchong, 637007, People's Republic of China.
- Translational Medicine Research Center, North Sichuan Medical College, Nanchong, 637007, People's Republic of China.
| | - Lingmi Hou
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, 610041, China.
| | - Panke Cheng
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China.
- Ultrasound in Cardiac Electrophysiology and Biomechanics Key Laboratory of Sichuan Province, Chengdu, 610072, P.R. China.
| |
Collapse
|
6
|
Huang S, Kang Y, Liu T, Xiong Y, Yang Z, Zhang Q. The role of immune checkpoints PD-1 and CTLA-4 in cardiovascular complications leading to heart failure. Front Immunol 2025; 16:1561968. [PMID: 40255399 PMCID: PMC12006013 DOI: 10.3389/fimmu.2025.1561968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Accepted: 02/13/2025] [Indexed: 04/22/2025] Open
Abstract
Immune checkpoints, such as PD-1 and CTLA-4, are crucial regulators of immune responses, acting as gatekeepers to balance immunity against foreign antigens and self-tolerance. These checkpoints play a key role in maintaining cardiac homeostasis by preventing immune-mediated damage to critical organs like the heart. In this study, we explored the involvement of PD-1 and CTLA-4 in cardiovascular complications, particularly atherosclerosis and myocarditis, which can lead to heart failure. We conducted a comprehensive analysis using animal models and clinical data to assess the effects of immune checkpoint inhibition on cardiac function. Our findings indicate that disruption of PD-1 and CTLA-4 pathways exacerbates myocardial inflammation, accelerates atherosclerotic plaque formation, and promotes the development of heart failure. Additionally, we observed that immune checkpoint inhibition in these models led to increased infiltration of T lymphocytes, higher levels of pro-inflammatory cytokines, and enhanced tissue damage. These results suggest that PD-1 and CTLA-4 are critical in preserving cardiac health, and their inhibition can result in severe cardiovascular toxicity. Our study emphasizes the need for careful monitoring of cardiovascular health in patients undergoing immune checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Shoulian Huang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Cardiology, The Second People’s Hospital of Yibin, Yibin, Sichuan, China
| | - Yu Kang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Liu
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Xiong
- Institute of Cardiovascular Diseases & Department of Cardiology, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zixuan Yang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing Zhang
- Department of Cardiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Gray R, Manisty C, Cheng R, Dastidar A, Mamas M, Ghosh A. Immune checkpoint inhibitors: Unravelling atherosclerotic cardiovascular risk. Atherosclerosis 2025; 403:119147. [PMID: 40037087 DOI: 10.1016/j.atherosclerosis.2025.119147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 02/02/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Affiliation(s)
| | | | - Richard Cheng
- Division of Cardiology, University of Washington, USA
| | - Amardeep Dastidar
- Bristol Heart Institute, NIHR Biomedical Research Centre Cardiovascular & North Bristol NHS Trust, Bristol, UK
| | - Mamas Mamas
- Keele Cardiovascular Research Group, Keele University, UK
| | | |
Collapse
|
8
|
Gasimova Z, Khandanpour C, Luley K, von Bubnoff NCC. [Which Immunotherapies are Standard Today? And What Side Effects Should be Expected?]. Zentralbl Chir 2025; 150:170-174. [PMID: 40199376 DOI: 10.1055/a-2544-9630] [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: 04/10/2025]
Abstract
In comparison to earlier treatment standards (including chemotherapy, radiation therapy, and surgery), immunotherapy has significantly improved patients' survival and quality of life. Immunotherapy is ultimately a broad term that encompasses a variety of therapeutics. Immunotherapeutic approaches have firmly established themselves as a new pillar of cancer care, ranging from neoadjuvant and adjuvant strategies to the metastatic phase in various entities. In this review article, we highlight which immunotherapies are currently standard in oncological care, with a strong focus on immune checkpoint inhibitors (ICIs), their limitations, and the commonly occurring side effects.
Collapse
Affiliation(s)
| | - Cyrus Khandanpour
- Klinik für Hämatoonkologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Deutschland
| | - Kim Luley
- Klinik für Hämatoonkologie, Universitätsklinikum Schleswig-Holstein, Campus Lübeck, Lübeck, Deutschland
| | | |
Collapse
|
9
|
Zhang Y, Cao Z, Jia H, Feng Y, Sun X, Wu H, Xu B, Wei Z. Immune checkpoint inhibitor induces cardiac injury by impairing efferocytosis of macrophages via MerTK cleavage. Int Immunopharmacol 2025; 150:114263. [PMID: 39938164 DOI: 10.1016/j.intimp.2025.114263] [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: 08/01/2024] [Revised: 02/01/2025] [Accepted: 02/06/2025] [Indexed: 02/14/2025]
Abstract
Cancer immunotherapy is a well-established therapeutic approach for various types of cancer. However, its clinical utility is usually limited by cardiovascular adverse events. Immune Checkpoint Inhibitors (ICIs) can induce diverse forms of cardiotoxicity, with myocarditis being the most fatal complication. The underlying mechanism of its occurrence remains elusive. Therefore, this study aims to elucidate the impact of programmed death-1 (PD-1) inhibitor on myocarditis development in mice. Myeloid-epithelial-reproductive tyrosine kinase (MerTK) receptors, located on the surface of macrophages, play a pivotal role in phagocytic regulation. We established a mouse model of autoimmune myocarditis by injecting 6-week-old normal male BALB/c mice with PD-1 inhibitor and cardiac troponin I peptide fragments, which resulted in elevated levels of serum soluble MerTK (SolMer) and reduced numbers of MerTK-CD68 double-positive macrophages, accompanied by cardiac injury in mice. In vitro, PD-1 inhibitor promotes a disintegrin and metalloproteinase17 (ADAM17)-mediated shed of the MerTK, forming SolMer, through MKK3/P38 MAPK pathway, leading to downregulation of MerTK expression on the macrophage surface. This results in the inhibition of efferocytosis and impairment of tissue repair function, ultimately contributing to myocarditis development. TAPI-0 inhibited the activity of ADAM17, while SB203580 inhibited the phosphorylation of P38 MAPK. Both inhibitors effectively restored the inhibition of efferocytosis induced by the PD-1 inhibitor. In vitro, when the PD-1 receptor on the surface of RAW264.7 macrophages was knocked down and then stimulated with a PD-1 inhibitor, no further significant alterations in the pathway were elicited. In conclusion, the PD-1 inhibitor induces the shedding of MerTK in macrophages by binding to the PD-1 receptor on the surface of macrophages and activating the MKK3/P38 MAPK/ADAM17 pathway, leading to impaired efferocytosis. Elucidation of this molecular mechanism holds promise for improved prognosis and therapeutic strategies in cancer patients.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China
| | - Zhenzhu Cao
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China
| | - Huihui Jia
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China
| | - Yuting Feng
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China
| | - Xuan Sun
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China
| | - Han Wu
- Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China.
| | - Biao Xu
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China; Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China.
| | - Zhonghai Wei
- Department of Cardiology, Nanjing Drum Tower Hospital, Nanjing Drum Tower Hospital Clinical College, Nanjing University of Chinese Medicine, 358 Zhongshan Road 210008 Nanjing, China; Department of Cardiology, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing 210008 China.
| |
Collapse
|
10
|
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] [Download PDF] [Figures] [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
|
11
|
Huang R, Shen ZY, Huang D, Zhao SH, Dan LX, Wu P, Tang QZ, Ma ZG. Microbiota-indole-3-propionic acid-heart axis mediates the protection of leflunomide against αPD1-induced cardiotoxicity in mice. Nat Commun 2025; 16:2651. [PMID: 40108157 PMCID: PMC11923180 DOI: 10.1038/s41467-025-58107-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 03/12/2025] [Indexed: 03/22/2025] Open
Abstract
Anti-programmed death 1 (αPD1) immune checkpoint blockade is used in combination for cancer treatment but associated with cardiovascular toxicity. Leflunomide (Lef) can suppress the growth of several tumor and mitigate cardiac remodeling in mice. However, the role of Lef in αPD1-induced cardiotoxicity remains unclear. Here, we report that Lef treatment inhibits αPD1-related cardiotoxicity without compromising the efficacy of αPD1-mediated immunotherapy. Lef changes community structure of gut microbiota in αPD1-treated melanoma-bearing mice. Moreover, mice receiving microbiota transplants from Lef+αPD1-treated melanoma-bearing mice have better cardiac function compared to mice receiving transplants from αPD1-treated mice. Mechanistically, we analyze metabolomics and identify indole-3-propionic acid (IPA), which protects cardiac dysfunction in αPD1-treated mice. IPA can directly bind to the aryl hydrocarbon receptor and promote phosphoinositide 3-kinase expression, thus curtailing the cardiomyocyte response to immune injury. Our findings reveal that Lef mitigates αPD1-induced cardiac toxicity in melanoma-bearing mice through modulation of the microbiota-IPA-heart axis.
Collapse
Affiliation(s)
- Rong Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zhuo-Yu Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Dan Huang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Shu-Hong Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Ling-Xuan Dan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Pan Wu
- Department of Adult Internal Medicine, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, PR China
| | - Qi-Zhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China
| | - Zhen-Guo Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, PR China.
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, PR China.
| |
Collapse
|
12
|
Chen Y, Luo Y, Liu Y, Qiu X, Luo D, Liu A. Mediation of macrophage M1 polarization dynamics change by ubiquitin-autophagy-pathway regulated NLRP3 inflammasomes in PD-1 inhibitor-related myocardial inflammatory injury. Inflamm Res 2025; 74:56. [PMID: 40097836 DOI: 10.1007/s00011-024-01979-1] [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: 08/22/2024] [Revised: 11/01/2024] [Accepted: 11/08/2024] [Indexed: 03/19/2025] Open
Abstract
OBJECTIVE AND DESIGN Immune checkpoint inhibitors (ICIs)-induced cardiac injury is a life-threatening immune-related adverse events (irAEs). However, the current understanding of its pathogenesis and therapeutic options is relatively limited. We aimed to provide a comprehensive overview of the myocardial inflammatory injury process and underlying mechanisms associated with ICIs. MATERIAL OR SUBJECTS We conducted a descriptive analysis of lung cancer patients with ICIs-induced myocarditis at our institution and validated our clinical findings using single-cell sequencing (scRNA-seq) data. Furthermore, we established animal and cellular models to investigate the underlying mechanisms. RESULTS Our findings revealed that lung cancer patients with ICIs-induced myocarditis exhibit an early increase in peripheral blood monocytes, which migrate to the heart and differentiate into macrophages, ultimately leading to myocardial inflammation. Mechanistically, programmed death-1 (PD-1) inhibition triggers myocardial inflammation through the activation of the signal transducer and activator of transcription 1 (STAT1)/nuclear factor kappa-B (NF-κB)/oligomerization domain (NOD)-like receptor thermal protein domain-associated protein 3 (NLRP3) signaling pathway and drives the polarization of macrophages towards the M1 phenotype. However, the ubiquitin (Ub)-autophagy pathway degrades NLRP3 inflammasomes, resulting in the gradual resolution of inflammation and the transition of M1 macrophages to the M2 phenotype. Finally, mouse experiments confirmed that NLRP3 inhibition using MCC950 effectively alleviates myocardial inflammatory injury. CONCLUSIONS We recommend monitoring fluctuations in peripheral blood monocyte counts in lung cancer patients undergoing ICIs treatment. Additionally, MCC950 holds potential as a therapeutic agent for ICIs-induced cardiac inflammation injury.
Collapse
Affiliation(s)
- Yanxin Chen
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Department of Radiotherapy, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, 410000, Hunan Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yuxi Luo
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Yunwei Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Xingpeng Qiu
- College of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Daya Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Anwen Liu
- Department of Oncology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Jiangxi Key Laboratory of Clinical Translational Cancer Research, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
- Radiation Induced Heart Damage Institute, Jiangxi Medical College, Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| |
Collapse
|
13
|
Bello MO, Wadid M, Malode A, Patel V, Shah A, Vyas A, Ahmad HA, Tarun T, Dani S, Ahmad J, Zarwan C, Ganatra S. Atrial Fibrillation in Patients with Breast Cancer: A Literature Review. Cardiol Ther 2025; 14:1-15. [PMID: 39714744 PMCID: PMC11893935 DOI: 10.1007/s40119-024-00394-1] [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: 09/13/2024] [Accepted: 12/04/2024] [Indexed: 12/24/2024] Open
Abstract
In addition to traditional risk factors, patients with breast cancer are at an increased risk of atrial fibrillation due to cancer itself and certain cancer therapies. Atrial fibrillation in these patients adds to their morbidity and mortality. The precise mechanisms leading to the increased atrial fibrillation in patients with breast cancer are not well understood. The main goal of atrial fibrillation management in this population is to facilitate uninterrupted cancer treatment while addressing the arrhythmia and other cardiovascular sequelae of cancer treatment. Rhythm control is often challenging to implement in patients with breast cancer during active antineoplastic therapy because of the need for uninterrupted anticoagulation, potential drug-drug interactions between cancer treatments and antiarrhythmic medications, and the increased likelihood of atrial fibrillation recurrence. Prevention of thromboembolism and anticoagulation can also be challenging in patients with breast cancer as a result of the increased risk of cancer-related procoagulant state and coagulopathies. The integration of a cardio-oncology team and a multidisciplinary approach are crucial for better outcomes. The therapeutic interventions should be tailored toward individual patients' profiles through a shared decision-making approach. The precise mechanisms leading to the increased atrial fibrillation in patients with breast cancer are not well understood, highlighting the gaps in our knowledge. More research is required to reduce these gaps, refine risk stratification, and optimize treatment strategies in these patients.
Collapse
Affiliation(s)
- Mozidat Olamide Bello
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Mark Wadid
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Aishwarya Malode
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Vahin Patel
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Anuj Shah
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Ankit Vyas
- Department of Vascular Medicine, Ochsner Clinic Foundation, New Orleans, LA, USA
| | | | - Tushar Tarun
- Division of Cardiovascular Medicine, Department of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Sourbha Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Javaria Ahmad
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA
| | - Corrine Zarwan
- Division of Hematology/Oncology, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA.
| |
Collapse
|
14
|
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
|
15
|
Chen W, Wang Y, Zhao M, Zhang H, Zong Y, Zhao X. Incidence of and Risk Factors for Anti-PD-1/PD-L1- Associated Diarrhea and Colitis: A Retrospective Cohort Study of the Chinese Population. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:353. [PMID: 40005469 PMCID: PMC11857649 DOI: 10.3390/medicina61020353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/19/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: The prevalence of and risk factors for immune checkpoint inhibitor-associated diarrhea and colitis (IMDC) in the Chinese population are unclear. This study aimed to estimate IMDC incidence and identify potential risk factors. Materials and Methods: We reviewed the electronic medical records from Beijing Friendship Hospital (2015-2022) to identify the patients treated with immune checkpoint inhibitors. The primary outcome was IMDC occurrence. The demographics, cancer type, baseline labs, and concurrent medications were analyzed. The univariable and multivariable analyses validated the associated factors. Results: Among 1186 patients (median follow-up: 217 days), the IMDC incidence was 4.6%, with colitis at 0.67%. Digestive system tumors increased the IMDC risk (OR 2.79, 95% CI 1.42-5.75, p = 0.004), while platinum agents decreased it (OR 0.41, 95% CI 0.21-0.78, p = 0.008). PPIs, antibiotics, NSAIDs, and glucocorticoids showed no significant association. Colitis was the third most common irAE, leading to ICI discontinuation (15.6%). Conclusions: IMDC prevalence is 4.6% in the Chinese population, the third most frequent irAE causing ICI discontinuation. Digestive tumors and platinum agents are risk and protective factors, respectively, while other medications show no significant impact.
Collapse
Affiliation(s)
- Wei Chen
- Beijing Digestive Disease Center, National Clinical Research Center for Digestive Disease, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (W.C.); (Y.Z.)
| | - Yan Wang
- Liver Research Center, National Clinical Research Center for Digestive Disease, Key Laboratory on Translational Medicine on Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.W.); (M.Z.)
| | - Mengyu Zhao
- Liver Research Center, National Clinical Research Center for Digestive Disease, Key Laboratory on Translational Medicine on Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.W.); (M.Z.)
| | - Hong Zhang
- Department of Pharmacy, Wenzhou Integrated Traditional Chinese and Western Medicine Hospital, Wenzhou 325000, China;
| | - Ye Zong
- Beijing Digestive Disease Center, National Clinical Research Center for Digestive Disease, Beijing Key Laboratory for Precancerous Lesion of Digestive Disease, Department of Gastroenterology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (W.C.); (Y.Z.)
| | - Xinyan Zhao
- Liver Research Center, National Clinical Research Center for Digestive Disease, Key Laboratory on Translational Medicine on Cirrhosis, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China; (Y.W.); (M.Z.)
| |
Collapse
|
16
|
Yang TY, Chuang MH, Lin HM, Wu VC, Pan HC, Chou Y, Chen JY. Clinical outcomes after immune checkpoint inhibitor-associated acute kidney injury: a cohort study. BMJ Open 2025; 15:e092752. [PMID: 39920047 PMCID: PMC11808918 DOI: 10.1136/bmjopen-2024-092752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 12/11/2024] [Indexed: 02/09/2025] Open
Abstract
OBJECTIVES Immune checkpoint inhibitors (ICPi) have significantly improved survival for patients with advanced cancers. However, the occurrence of ICPi-associated acute kidney injury (AKI) and its clinical impact remains unclear. This study evaluates the effects of ICPi-associated AKI (ICPi-AKI) on mortality, kidney and cardiovascular outcomes in patients undergoing ICPi treatments. DESIGN This multicentre retrospective cohort study with propensity score matching to balance baseline characteristics. The International Classification of Diseases, 10th Revision codes were used to identify individuals with cancer and treated with ICPi concurrently. Kaplan-Meier analyses coupled with log-rank tests were conducted to estimate the survival probabilities. SETTING Data were sourced from the TriNetX database spanning records from 25 March 2011 to 5 April 2024. PARTICIPANTS Patients with cancer aged ≥18 years treated with ICPi with or without AKI occurrence. PRIMARY AND SECONDARY OUTCOME MEASURES The primary outcome was all-cause mortality, and secondary outcomes included major adverse kidney events (MAKE), major adverse cardiovascular events (MACE), the composite of MAKE or MACE with death, and end-stage renal disease. RESULTS The study identified 926 patients with cancer who developed ICPi-AKI (mean age, 67.1±11.8 years; 57.4% men). The control group consisted of 48 147 patients treated with ICPi but did not develop AKI (mean age, 65.3±13.1 years; 53.7% men). After matching, the ICPi-AKI group exhibited a higher risk of all-cause mortality (HR=1.27; 95% CI 1.02 to 1.61), MAKE (HR=3.83; 95% CI 1.72 to 8.40), MACE (HR=1.35; 95% CI 1.03 to 1.75)) compared with the non-ICPi-AKI group. Subgroup analyses confirmed these findings across various patient's characteristics. CONCLUSION Individuals with ICPi-AKI are associated with an increased risk of all-cause mortality, MAKE and MACE. Enhancing awareness and timely intervention for ICPi-AKI are crucial for improving prognosis and reducing complications among patients with cancer.
Collapse
Affiliation(s)
- Ting-Ya Yang
- Department of Family Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Min-Hsiang Chuang
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Hong-Min Lin
- Department of Family Medicine, Chi Mei Medical Center, Tainan, Taiwan
| | - Vin-Cent Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Heng-Chih Pan
- Department of Internal Medicine, Keelung Chang Gung Memorial Hospital, Keelung, Taiwan
| | - Yun Chou
- Department of Pediatrics, Chi Mei Medical Center, Tainan, Taiwan
- Department of Early Childhood Care and Education, Shu-Zen Junior College of Medicine and Management, Kaohsiung, Taiwan
| | - Jui-Yi Chen
- Division of Nephrology, Department of Internal Medicine, Chi Mei Medical Center, Tainan, Taiwan
- Department of Health and Nutrition, Chia Nan University of Pharmacy and Science, Tainan, Taiwan
| |
Collapse
|
17
|
Li D, Chen Y, Zhang B, Heng X, Yin J, Zhao P, Sun N, Shao C. Praeruptorin A screened by a ferrous ion probe inhibited DMT1 and ferroptosis to attenuate Doxorubicin-induced cardiomyopathy. Eur J Med Chem 2025; 283:117108. [PMID: 39615370 DOI: 10.1016/j.ejmech.2024.117108] [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: 10/09/2024] [Revised: 11/15/2024] [Accepted: 11/24/2024] [Indexed: 01/03/2025]
Abstract
Doxorubicin (DOX)-induced cardiomyopathy (DIC) greatly limits its clinical application of the anticancer drug. Therefore, there is an immediate necessity to undertake intervention studies to minimize DIC, encompassing the screening of regulatory compounds and delving into the underlying regulatory mechanisms. A growing body of research suggests that ferroptosis is an essential process in the development of DIC. Here, we demonstrated that DOX causes elevated iron levels in cardiomyocytes and mouse hearts, and leads to ferroptosis and cardiac insufficiency. Next, we performed high-throughput screening of a library of herbal small molecule compounds for novel compounds that inhibit ferroptosis, using Fe2+ levels as a screening index for DIC prevention and treatment drugs. We found that Praeruptorin A (PA) was able to reduce Fe2+ concentration in cardiomyocytes, inhibit ferroptosis, and alleviate DIC and cardiac dysfunction in mice. Concurrently, PA exhibits a synergistic effect with DOX in suppressing the proliferation of carcinoma of breast MCF-7 cell in nude mice. Mechanistically, we found that PA inhibited the expression of divalent metal transporter protein 1 (DMT1), suppressed Fe2+ overload in cardiomyocytes, and inhibited ferroptosis, thereby alleviating DIC. Our study demonstrated the feasibility of high-throughput screening targeting the Fe2+ concentration, and elucidated the role and mechanism of PA in alleviating DIC, which provides a new possibility.
Collapse
Affiliation(s)
- Dujuan Li
- Wuxi School of Medicine & Wuxi Mental Health Center, Jiangnan University, Wuxi, 214122, China
| | - Yan Chen
- Wuxi School of Medicine & Wuxi Mental Health Center, Jiangnan University, Wuxi, 214122, China
| | - Bo Zhang
- School of Pharmacy, Changzhou University, Changzhou, 213164, China
| | - Xinyu Heng
- Wuxi School of Medicine & Wuxi Mental Health Center, Jiangnan University, Wuxi, 214122, China
| | - Jiajun Yin
- Wuxi School of Medicine & Wuxi Mental Health Center, Jiangnan University, Wuxi, 214122, China
| | - Peilin Zhao
- Wuxi School of Medicine & Wuxi Mental Health Center, Jiangnan University, Wuxi, 214122, China
| | - Ning Sun
- Wuxi School of Medicine & Wuxi Mental Health Center, Jiangnan University, Wuxi, 214122, China
| | - Chenwen Shao
- Wuxi School of Medicine & Wuxi Mental Health Center, Jiangnan University, Wuxi, 214122, China.
| |
Collapse
|
18
|
Wang RH, Chen Y, Lou YL, Lu YL, Xu HM. Characteristics, Incidence, and Management of Immune Checkpoint Inhibitors Related Cardiovascular Adverse Events in Real-World Practice-A Retrospective Study in Chinese Han Population. Ther Clin Risk Manag 2025; 21:125-135. [PMID: 39980984 PMCID: PMC11840335 DOI: 10.2147/tcrm.s477417] [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: 05/08/2024] [Accepted: 01/25/2025] [Indexed: 02/22/2025] Open
Abstract
Purpose This study aimed to elaborate on the incidence, clinical features, and management of immune checkpoint inhibitors (ICIs) related cardiovascular adverse events (CVAEs) in real-world practice. Patients and Methods We performed a retrospective chart review study on patients receiving at least one dose of ICI therapy at a Chinese tertiary hospital from March 2020 to March 2021. CVAEs were identified through clinical assessment and the Naranjo algorithm. The management and outcomes of CVAEs were monitored over a median follow-up duration of 8 months. Results Among the included 203 patients, 4.4% (9/203) developed CVAEs, including heart failure (n = 3), arrhythmia (n = 2), myocarditis (n = 2), and pericardial disease (n = 2), with a proportion (6/9) tending to be severe (grade 3 or grade 4). CVAEs were more common in older patients (mean age: 73.6 ± 9.2 years) and those with hypertension (p = 0.02) or heart failure (p = 0.01). Adherence to the American Society of Clinical Oncology (ASCO) guidelines for managing CVAEs was low (44%), with most cases showing partial resolution by the last follow-up. Conclusion We reported that the incidence of ICI-related CVAEs in the Chinese institution was higher than that in some prior studies. Adherence to guidelines for managing ICI-related CVAEs is found to be suboptimal in real-world practice and highlighted as a needed improvement.
Collapse
Affiliation(s)
- Rong-Hua Wang
- Department of Pharmacy, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Yin Chen
- Department of Pharmacy, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Ya-Ling Lou
- Department of Pharmacy, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Yu-Liang Lu
- Department of Cardiology, HuZhou Central Hospital (The Fifth School of Clinical Medicine of Zhejiang Chinese Medical University), Huzhou, People’s Republic of China
| | - Hui-Min Xu
- Department of Pharmacy, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, People’s Republic of China
| |
Collapse
|
19
|
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
|
20
|
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
|
21
|
Lerchner T, Mincu RI, Bühning F, Vogel J, Klingel K, Meetschen M, Schlosser T, Haubold J, Umutlu L, Dobrev D, Totzeck M, Rassaf T, Michel L. Cardiac magnetic resonance imaging in patients with suspected myocarditis from immune checkpoint inhibitor therapy - A real-world observational study. IJC HEART & VASCULATURE 2025; 56:101581. [PMID: 39882168 PMCID: PMC11775410 DOI: 10.1016/j.ijcha.2024.101581] [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: 09/29/2024] [Revised: 11/08/2024] [Accepted: 11/10/2024] [Indexed: 01/31/2025]
Abstract
Background and aims Cardiotoxicity from immune checkpoint inhibitor (ICI) therapy is a challenge in clinical practice, and the assessment of ICI-related myocarditis (ICI-M) is often complicated by a variable phenotype. Cardiac magnetic resonance imaging (CMR) is used frequently, but evidence is poor. Here, we aim to assess the role of CMR in the assessment of suspected ICI-M in a real-world clinical setting. Methods All patients receiving CMR at our centre for suspected ICI-M between September 2019 and January 2024 were included and retrospectively analysed. CMR parameters were correlated with clinical, laboratory and echocardiographic parameters and stratified for presence of myocarditis as per final diagnosis. Results A total of 55 patients who received CMR for suspected ICI-M were analysed, including 25 patients with ICI-M and 30 patients with non-myocarditis cardiotoxicity (non-M). The mean age (ICI-M versus (vs.) non-M) was 65.7 ± 13.6 vs. 67.3 ± 9.9 (p = 0.61) years, 32.0 % vs. 26.7 % (p = 0.67) were female, and 40.0 % vs. 26.7 % (p = 0.29) had pre-existing coronary heart disease. Cardiac biomarkers and echocardiographic data did not differ between the groups. In CMR analysis, presence of LGE was associated with ICI-M (56.0 % in ICI-M vs. 26.7 % in non-M, p = 0.03). Myocardial oedema was generally rare and not associated with ICI-M. Conclusion In this real-life assessment of routine clinical practice, the diagnostic assessment of ICI-M is challenged by low sensitivity of common diagnostic measures, often requiring a multimodal approach. Presence of LGE in CMR is associated with ICI-M, but sensitivity and specificity are low. Prospective data to improve diagnostic criteria is needed.
Collapse
Affiliation(s)
- Tobias Lerchner
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Raluca I. Mincu
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Florian Bühning
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Julia Vogel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Karin Klingel
- Cardiopathology, Institute for Pathology and Neuropathology, Tübingen, Germany
| | - Mathias Meetschen
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Germany
- Institute of Artificial Intelligence in Medicine, University Hospital Essen, Germany
| | - Thomas Schlosser
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Germany
| | - Johannes Haubold
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Germany
- Institute of Artificial Intelligence in Medicine, University Hospital Essen, Germany
| | - Lale Umutlu
- Department of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, Germany
| | - Dobromir Dobrev
- Institute of Pharmacology, West German Heart and Vascular Center, University Hospital Essen, Germany
- Department of Medicine and Research Center, Montreal Heart Institute and Université de Montréal, Montréal, QC, Canada
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Germany
| |
Collapse
|
22
|
Panuccio G, Correale P, d'Apolito M, Mutti L, Giannicola R, Pirtoli L, Giordano A, Labate D, Macheda S, Carabetta N, Abdelwahed YS, Landmesser U, Tassone P, Tagliaferri P, De Rosa S, Torella D. Immuno-related cardio-vascular adverse events associated with immuno-oncological treatments: an under-estimated threat for cancer patients. Basic Res Cardiol 2025; 120:153-169. [PMID: 39225869 PMCID: PMC11790807 DOI: 10.1007/s00395-024-01077-7] [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: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy represents an emergent and heterogeneous group of anticancer treatments harnessing the human immune-surveillance system, including immune-checkpoint inhibitor monoclonal antibodies (mAbs), Chimeric Antigen Receptor T Cells (CAR-T) therapy, cancer vaccines and lymphocyte activation gene-3 (LAG-3) therapy. While remarkably effective against several malignancies, these therapies, often in combination with other cancer treatments, have showed unforeseen toxicity, including cardiovascular complications. The occurrence of immuno-mediated adverse (irAEs) events has been progressively reported in the last 10 years. These irAEs present an extended range of severity, from self-limiting to life-threatening conditions. Although recent guidelines in CardioOncology have provided important evidence in managing cancer treatments, they often encompass general approaches. However, a specific focus is required due to the particular etiology, unique risk factors, and associated side effects of immunotherapy. This review aims to deepen the understanding of the prevalence and nature of cardiovascular issues in patients undergoing immunotherapy, offering insights into strategies for risk stratification and management.
Collapse
Affiliation(s)
- Giuseppe Panuccio
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany.
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Pierpaolo Correale
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Maria d'Apolito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Applied Sciences and Biotechnology, Università dell'Aquila, L'Aquila, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, 53100, Siena, Italy
| | - Demetrio Labate
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Sebastiano Macheda
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Youssef S Abdelwahed
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
23
|
Leven AS, Wagner N, Nienaber S, Messiha D, Tasdogan A, Ugurel S. Changes in tumor and cardiac metabolism upon immune checkpoint. Basic Res Cardiol 2025; 120:133-152. [PMID: 39658699 PMCID: PMC11790718 DOI: 10.1007/s00395-024-01092-8] [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: 07/01/2024] [Revised: 11/06/2024] [Accepted: 11/25/2024] [Indexed: 12/12/2024]
Abstract
Cardiovascular disease and cancer are the leading causes of death in the Western world. The associated risk factors are increased by smoking, hypertension, diabetes, sedentary lifestyle, aging, unbalanced diet, and alcohol consumption. Therefore, the study of cellular metabolism has become of increasing importance, with current research focusing on the alterations and adjustments of the metabolism of cancer patients. This may also affect the efficacy and tolerability of anti-cancer therapies such as immune-checkpoint inhibition (ICI). This review will focus on metabolic adaptations and their consequences for various cell types, including cancer cells, cardiac myocytes, and immune cells. Focusing on ICI, we illustrate how anti-cancer therapies interact with metabolism. In addition to the desired tumor response, we highlight that ICI can also lead to a variety of side effects that may impact metabolism or vice versa. With regard to the cardiovascular system, ICI-induced cardiotoxicity is increasingly recognized as one of the most life-threatening adverse events with a mortality of up to 50%. As such, significant efforts are being made to assess the specific interactions and associated metabolic changes associated with ICIs to improve both efficacy and management of side effects.
Collapse
Affiliation(s)
- Anna-Sophia Leven
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany.
| | - Natalie Wagner
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Stephan Nienaber
- Clinic III for Internal Medicine, University of Cologne, Faculty of Medicine and University Hospital Cologne, Cologne, Germany
| | - Daniel Messiha
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre, University of Duisburg-Essen, Essen, Germany
| | - Alpaslan Tasdogan
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| | - Selma Ugurel
- Department of Dermatology, Venereology and Allergology, University Hospital Essen, University Duisburg-Essen, Essen, Germany
- German Cancer Consortium (DKTK), Partner Site Essen/Düsseldorf, Essen, Germany
- National Center for Tumor Diseases (NCT)-West, Campus Essen, and Research Alliance Ruhr, Research Center One Health, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
24
|
Wang X, Luan X, Yin W, Wang Y, Li X, Chen R, Zhang G, Zhao R, Dong X, Zhang Z, Fan Y, Li Z, Chu X, Wang S. Advancements in Diagnosis and Treatment of Cardiac Sarcomas: A Comprehensive Review. Curr Treat Options Oncol 2025; 26:103-127. [PMID: 39885109 DOI: 10.1007/s11864-024-01287-0] [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] [Accepted: 12/08/2024] [Indexed: 02/01/2025]
Abstract
OPINION STATEMENT Cardiac sarcomas are rare, aggressive malignancies originating from various cardiac cell types, presenting significant challenges in both diagnosis and treatment. This comprehensive review explores recent advancements in diagnosis and treatment of cardiac angiosarcoma, fibrosarcoma, leiomyosarcoma, and rhabdomyosarcoma. And we briefly discuss the exceedingly rare occurrence of cardiac osteosarcoma and present our perspectives on its treatment. Development of these tumors is influenced by genetic mutations, environmental factors, and chromosomal abnormalities, necessitating a multidisciplinary approach for accurate diagnosis and management. Advanced imaging techniques, biomarkers, and immunohistochemical analysis assist in confirming the diagnosis and guiding treatment decisions. Surgical resection, adjuvant therapies, and personalized treatment strategies based on genetic profiling offer promising avenues for improving patient outcomes. Emerging therapeutic approaches, such as targeted therapies and immunotherapies, have shown promising progress in recent years. Despite these advancements, the prognosis for patient with cardiac sarcomas remains poor, highlighting the urgent need for continued research to refine treatment methods and enhance long-term survival outcomes. Ongoing efforts and clinical trials are essential for advancing the management of these rare and aggressive tumors, ultimately improving quality of life for affected patients.
Collapse
Affiliation(s)
- Xuezhe Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xinchi Luan
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Wenwen Yin
- Department of Pulmonology, The Sixth Affiliated Hospital of Qingdao University, Weihai, Shandong, China
| | - Yilin Wang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xiaoxuan Li
- Department of Oncology, Key Laboratory of Cancer Molecular and Translational Research, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ruolan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guoliang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruizhe Zhao
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xue Dong
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Zhishang Zhang
- Department of Joint Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Yuchen Fan
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Zhaodong Li
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China
| | - Xianming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| | - Shuang Wang
- School of Basic Medicine, Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
25
|
Tong X, Tang R, Xu J, Wang W, Du Q, Shi S, Yu X. Cancer type-specific adverse events of immune checkpoint inhibitors: A systematic review and meta-analysis. Heliyon 2025; 11:e41597. [PMID: 39866435 PMCID: PMC11757769 DOI: 10.1016/j.heliyon.2024.e41597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 11/11/2024] [Accepted: 12/30/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND The distribution of adverse events (AEs) triggered by immune checkpoint inhibitors (ICIs) across different cancer types has never been demonstrated. METHODS Randomised controlled trials exclusively assessing ICI monotherapy in cohorts of over 100 patients were considered. Our primary outcome was a comprehensive summary of the distribution of all-grade treatment-related adverse events (TRAEs) as well as serious TRAEs (CTCAE grade 3 or higher) across different malignancies. The study is registered with PROSPERO CRD42023387934. FINDINGS 75 trials that enrolled over 100 patients were included. While investigating the incidence of each TRAE across various cancers, we found special linkages existed between certain TRAEs and particular cancer types. In anti-PD-1 monotherapy group, melanoma patients experienced the most frequent fatigue (31.1 %, 95 % CI 29.7%-32.5 %); the incidences of severe pneumonitis and other respiratory disorders were highest in Hodgkin lymphoma (4.1 %, 95 % CI 1.5%-8.6 %; 4.1 %, 95 % CI 1.5%-8.6 %, respectively). Among individuals undergoing single-agent anti-PD-L1, higher frequency of all-grade pruritus occurred in 19.0 % of renal cell carcinoma (RCC) patients (95 % CI 15.2%-23.2 %), and the highest probability of developing other severe musculoskeletal disorders was observed in patients with RCC (6.2 %, 95 % CI 4.0%-9.0 %). In anti-CTLA-4 monotherapy, the incidences of both all-grade and severe diarrhea occurred most frequently in prostate cancer patients (41.9 %, 95 % CI 37.9%-47.9; 14.8 %, 95 % CI 11.5%-18.7 %, respectively). INTERPRETATION This is the first comprehensive study addressing the distribution of various TRAEs across cancer types. Our research emphasizes the significance of considering cancer-specific TRAEs when using ICIs for treatment.
Collapse
Affiliation(s)
- Xuhui Tong
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Rong Tang
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Jin Xu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Wei Wang
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Qiong Du
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Pharmacy, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Si Shi
- Shanghai Pancreatic Cancer Institute, Shanghai, China
- Pancreatic Cancer Institute, Fudan University, Shanghai, China
| | - Xianjun Yu
- Department of Pancreatic Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Tzuberi M, Brzezinski RY, Flint N, Slieman M, Zornitzki L, Viskin D, Hemed AR, Waissengrin B, Barak R, Golomb I, Wolf I, Golan N, Topilsky Y, Banai S, Kapusta L, Laufer-Perl M. Safety and efficacy of immune checkpoint inhibitors in patients with pre-treatment reduced left ventricular function. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:2. [PMID: 39780280 PMCID: PMC11707996 DOI: 10.1186/s40959-024-00297-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025]
Abstract
AIMS Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment outcomes. However, the response varies across different populations, and their use may lead to life-threatening cardiovascular (CV) events. While pre-treatment reduced left ventricular ejection fraction (LVEF) is considered a marker for high-risk cardiotoxicity and a contraindication for anthracycline and HER2-targeted therapies, there is limited evidence on the safety and efficacy of ICIs therapy in patients presenting with pre-treatment reduced LVEF. The study aims to evaluate the safety and efficacy of ICIs therapy in patients with pre-treatment reduced LVEF. METHODS Retrospective single center cohort of patients treated with ICIs therapy, who performed pre-treatment LVEF assessment. The primary endpoint was to evaluate the safety of ICIs among this population, assessed by CV events (composite of myocarditis, acute coronary syndrome, heart failure, and arrhythmias). The secondary endpoint was to evaluate the efficacy of ICIs, assessed by all-cause mortality and progression-free survival (PFS). RESULTS The cohort included 307 patients, with 30 (10%) presenting with pre-treatment reduced LVEF, with a mean LVEF of 39 ± 7%. While a significantly higher incidence of CV events was observed in the reduced LVEF group (37% vs. 14%, p = 0.004), following a multivariate Cox regression analysis including baseline CV diseases and risk factors, pre-treatment reduced LVEF did not remain a significant independent predictor (p = 0.358). No significant differences were observed between the groups regarding all-cause mortality and PFS. CONCLUSIONS Pre-treatment reduced LVEF was not identified as an independent marker for clinical outcomes in patients treated with ICIs therapy.
Collapse
Affiliation(s)
- Maor Tzuberi
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Rafael Y Brzezinski
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Nir Flint
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Moaad Slieman
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Lior Zornitzki
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Dana Viskin
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Anna Rozenfeld Hemed
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Barliz Waissengrin
- Division of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Renana Barak
- Division of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Inbal Golomb
- Division of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Ido Wolf
- Division of Oncology, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Netanel Golan
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Yan Topilsky
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Shmuel Banai
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
| | - Livia Kapusta
- Pediatric Cardiology Unit, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel
- Department of Pediatrics Cardiology, Amalia Children's Hospital, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Michal Laufer-Perl
- Division of Cardiology, Tel Aviv Sourasky medical Center, Tel Aviv, Israel.
- School of Medicine, Tel Aviv University, 6 Weizmann Street, Tel Aviv, Israel.
| |
Collapse
|
27
|
Ayoub C, Appari L, Pereyra M, Farina JM, Chao CJ, Scalia IG, Mahmoud AK, Abbas MT, Baba NA, Jeong J, Lester SJ, Patel BN, Arsanjani R, Banerjee I. Multimodal Fusion Artificial Intelligence Model to Predict Risk for MACE and Myocarditis in Cancer Patients Receiving Immune Checkpoint Inhibitor Therapy. JACC. ADVANCES 2025; 4:101435. [PMID: 39759436 PMCID: PMC11699614 DOI: 10.1016/j.jacadv.2024.101435] [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: 06/12/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 01/07/2025]
Abstract
Background Immune checkpoint inhibitor (ICI) therapy has dramatically improved the prognosis for some cancers but can be associated with myocarditis, adverse cardiovascular events, and mortality. Objectives The aim of this study was to develop an artificial intelligence (AI) model to predict the increased likelihood for the development of ICI-related myocarditis and adverse cardiovascular events. Methods Cancer patients treated with ICI at a tertiary institution from 2011 to 2022 were reviewed. Baseline characteristics, laboratory values, electrocardiograms, and cardiovascular clinical outcomes were extracted. A composite outcome of ICI-related myocarditis and major adverse cardiovascular events (transient ischemic attack/stroke, new diagnosis of heart failure, myocardial infarction, and cardiac death) was used to develop a multimodal joint fusion AI model by combining baseline tabular data with electrocardiogram (ECG) in a single end-to-end model. ECG data were parsed using 1-D convolution and tubular data using multilayer perceptron. Results Of 2,258 cancer patients who had ICI therapy and troponin measurement (mean age 68.5 ± 11.5 years, 59.7% male), the composite of cardiovascular clinical adverse events, including ICI-related myocarditis and major adverse cardiovascular events, occurred in 264 (11.7%) unique patients, with 428 events overall (including 59 [3%] ICI-related myocarditis events and 59 [3%] cardiac deaths). The proposed joint fusion model outperformed individual ECG and baseline electronic medical record data and laboratory value models with an area under the operating characteristics curve of 0.72 (0.64 true positive rate and 0.98 negative predictive value). Conclusion A multimodal fusion AI model to predict myocarditis and adverse cardiovascular events in cancer patients starting ICI therapy had good prognostic performance. It may have clinical utility in identifying at-risk patients who may benefit from closer surveillance.
Collapse
Affiliation(s)
- Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Lalith Appari
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Juan M. Farina
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Chieh-Ju Chao
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Isabel G. Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Ahmed K. Mahmoud
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | | | - Nima Ali Baba
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Jiwoong Jeong
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Steven J. Lester
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | | | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, Arizona, USA
| | - Imon Banerjee
- Department of Radiology, Mayo Clinic, Phoenix, Arizona, USA
| |
Collapse
|
28
|
Xing J, Ma X, Yu Y, Xiao Y, Chen L, Yuan W, Wang Y, Liu K, Guo Z, Tang H, Fan K, Jiang W. A Cardiac-Targeting and Anchoring Bimetallic Cluster Nanozyme Alleviates Chemotherapy-Induced Cardiac Ferroptosis and PANoptosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405597. [PMID: 39467094 PMCID: PMC11714205 DOI: 10.1002/advs.202405597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 08/25/2024] [Indexed: 10/30/2024]
Abstract
Doxorubicin (DOX), a potent antineoplastic agent, is commonly associated with cardiotoxicity, necessitating the development of strategies to reduce its adverse effects on cardiac function. Previous research has demonstrated a strong correlation between DOX-induced cardiotoxicity and the activation of oxidative stress pathways. This work introduces a novel antioxidant therapeutic approach, utilizing libraries of tannic acid and N-acetyl-L-cysteine-protected bimetallic cluster nanozymes. Through extensive screening for antioxidative enzyme-like activity, an optimal bimetallic nanozyme (AuRu) is identified that possess remarkable antioxidant characteristics, mimicking catalase-like enzymes. Theoretical calculations reveal the surface interactions of the prepared nanozymes that simulate the hydrogen peroxide decomposition process, showing that these bimetallic nanozymes readily undergo OH⁻ adsorption and O₂ desorption. To enhance cardiac targeting, the atrial natriuretic peptide is conjugated to the AuRu nanozyme. These cardiac-targeted bimetallic cluster nanozymes, with their anchoring capability, effectively reduce DOX-induced cardiomyocyte ferroptosis and PANoptosis without compromising tumor treatment efficacy. Thus, the therapeutic approach demonstrates significant reductions in chemotherapy-induced cardiac cell death and improvements in cardiac function, accompanied by exceptional in vivo biocompatibility and stability. This study presents a promising avenue for preventing chemotherapy-induced cardiotoxicity, offering potential clinical benefits for cancer patients.
Collapse
Affiliation(s)
- Junyue Xing
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Xiaohan Ma
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Yanan Yu
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Yangfan Xiao
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Lu Chen
- Department of Cardiovascular Diseases the First Clinical Medical CollegeShanxi Medical UniversityTaiyuanShanxi030001China
| | - Weining Yuan
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Yingying Wang
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Keyu Liu
- School of Clinical MedicineShandong Second Medical UniversityWeifangShandong261053China
| | - Zhiping Guo
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Hao Tang
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
| | - Kelong Fan
- CAS Engineering Laboratory for NanozymeKey Laboratory of Biomacromolecules (CAS)CAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijing100101China
- Nanozyme Laboratory in ZhongyuanHenan Academy of Innovations in Medical ScienceZhengzhouHenan451163China
| | - Wei Jiang
- National Health Commission Key Laboratory of Cardiovascular Regenerative MedicineCentral China Subcenter of National Center for Cardiovascular DiseasesHenan Cardiovascular Disease CenterFuwai Central‐China Cardiovascular HospitalCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhou450046China
- Henan Key Laboratory of Chronic Disease ManagementCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Zhengzhou Key Laboratory of Cardiovascular AgingCentral China Fuwai Hospital of Zhengzhou UniversityZhengzhouHenan451464China
- Academy of Medical SciencesTianjian Laboratory of Advanced Biomedical SciencesZhengzhou UniversityZhengzhouHenanChina
| |
Collapse
|
29
|
Zhang J, Jiang X, Liu N, Qi Z, Mi X, Fang Y, Zhang W, Yang Z, Ou W, Lin X, Hou J. Clinical characteristics and prognosis of pancreatitis associated with immune checkpoint inhibitors. Clin Transl Oncol 2025; 27:333-339. [PMID: 38995514 DOI: 10.1007/s12094-024-03573-7] [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: 04/07/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024]
Abstract
BACKGROUND AND OBJECTIVE Immune checkpoint inhibitors (ICIs) have shown remarkable efficacy against various cancers in clinical practice. However, ICIs can cause immune checkpoint inhibitor-associated pancreatic injury, often leading to drug withdrawal, and then patients must go to specialized treatment. The patients, their primary tumors are sensitive to ICIs therapy, may experience treatment delays due to such adverse reactions. Therefore, there is a need for systematic clinical researches on immune-related pancreatic toxicity to provide a clinical basis for its prevention and treatment. METHODS This study involved the collection of data from patients treated with ICIs and addressed pancreatic injury with preemptive treatment before continuing ICIs therapy. Then, we also statistically analyzed the incidence of pancreatic injury in patients with different courses and combined treatment, and the success rate of rechallenge treatment. RESULTS The study included 62 patients, with 33.9% (21/62) experiencing varying degrees of pancreatic injury. Patients with pancreatic injury, 10 cases evolved into pancreatitis, representing 47.6% (10/21) in the pancreatic injury subgroup and 16.1% (10/62) of the total patient cohort. Preemptive treatment was administered to 47.6% (10/21) of patients with pancreatitis, the effective rate was 100%. Among these patients, 70% (7/10) underwent successful rechallenge with ICIs. The occurrence of pancreatic injury was positively correlated with the treatment duration (P < 0.05) but showed no significant correlation with combination therapies (P > 0.05). CONCLUSION The likelihood of pancreatic injury increased with longer treatment durations with ICIs; no significant association was found between the incidence of ICIs-related pancreatic damage and combination therapies. Preemptive treatment for immune-related pancreatitis is feasible, allowing some patients to successfully undergo rechallenge with ICIs therapy.
Collapse
Affiliation(s)
- Junzi Zhang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xianzhuo Jiang
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, Jilin, China
| | - Ning Liu
- General Surgery of the First Clinical Hospital of Jilin Academy of Chinese Medicine Sciences, Changchun, Jilin, China
| | - Zhaoxue Qi
- Department of Secretory Metabolism, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xuguang Mi
- Department of Central Laboratory, Jilin Provincial People's Hospital, Changchun, Jilin, China
| | - Yanqiu Fang
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, Jilin, China
| | - Wenqi Zhang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Zhen Yang
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Wenjie Ou
- Changchun University of Chinese Medicine, Changchun, Jilin, China
| | - Xiuying Lin
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, Jilin, China
| | - Junjie Hou
- Department of Tumor Comprehensive Therapy, Jilin Provincial People's Hospital, Changchun, Jilin, China.
| |
Collapse
|
30
|
Sun CL. Breaking Barriers in Cardiovascular and Cancer Care With Multimodal AI. JACC. ADVANCES 2025; 4:101433. [PMID: 39759438 PMCID: PMC11697234 DOI: 10.1016/j.jacadv.2024.101433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/07/2025]
Affiliation(s)
- Christopher L.F. Sun
- Telfer School of Management, University of Ottawa, Ottawa, Ontario, Canada
- University of Ottawa Heart Institute, University of Ottawa, Ottawa, Ontario, Canada
| |
Collapse
|
31
|
Xu Q, Hu J, Wang Y, Wang Z. The role of tumor types in immune-related adverse events. Clin Transl Oncol 2024:10.1007/s12094-024-03798-6. [PMID: 39738878 DOI: 10.1007/s12094-024-03798-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/13/2024] [Indexed: 01/02/2025]
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that block inhibitors of T cell activation and function. With the widespread use of ICIs in cancer therapy, immune-related adverse events (irAEs) have gradually emerged as urgent clinical issues. Tumors not only exhibit high heterogeneity, and their response to ICIs varies, with "hot" tumors showing better anti-tumor effects but also a higher susceptibility to irAEs. The manifestation of irAEs displays a tumor-heterogeneous pattern, correlating with the tumor type in terms of the affected organs, incidence, median onset time, and severity. Understanding the mechanisms underlying the pathogenic patterns of irAEs can provide novel insights into the prevention and management of irAEs, guide the development of biomarkers, and contribute to a deeper understanding of the toxicological characteristics of ICIs. In this review, we explore the impact of tumor type on the therapeutic efficacy of ICIs and further elucidate how these tumor types influence the occurrence of irAEs. Finally, we assess key candidate biomarkers and their relevance to proposed irAE mechanisms. This paper also outlines management strategies for patients with various types of tumors, based on their disease patterns.
Collapse
Affiliation(s)
- Qian Xu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Jing Hu
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China
| | - Yan Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| | - Zhaohui Wang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
- Hubei Provincial Engineering Research Center of Immunological Diagnosis and Therapy for Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430000, Hubei, China.
| |
Collapse
|
32
|
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
|
33
|
Wu S, Jamal F. Cardiooncology in the ICU - Cardiac Urgencies in Cancer Care. J Intensive Care Med 2024:8850666241303461. [PMID: 39632745 DOI: 10.1177/08850666241303461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024]
Abstract
Cardiovascular disease is an increasing risk of morbidity and mortality in cancer patients, related to an growing number of aging survivors with pre-existing cardiovascular disease and the use of traditional and novel cancer therapies with cardiotoxic effects. While many cardiac complications are chronic processes that develop over time, there are many acute processes that may arise in hospitalized patients. It is important for hospitalists and critical care physicians to be familiar with the recognition and management of these conditions in this unique population. This article reviews the presentation and management of common cardiac urgencies in critically ill cancer patients including acute decompensated heart failure, acute coronary syndromes, arrhythmias, hypertensive crises, pulmonary embolism, pericardial tamponade and myocarditis.
Collapse
Affiliation(s)
- Stephanie Wu
- Department of Medicine, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| | - Faizi Jamal
- Department of Medicine, City of Hope Comprehensive Cancer Center, Duarte, California, USA
| |
Collapse
|
34
|
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
|
35
|
Gamero MT, Patel A, Storozynsky E. The Good (Tumor Killing) and the Bad (Cardiovascular Complications) of Immunologic Checkpoint Inhibitors. Curr Cardiol Rep 2024; 26:1487-1498. [PMID: 39441327 DOI: 10.1007/s11886-024-02147-x] [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: 09/23/2024] [Indexed: 10/25/2024]
Abstract
PURPOSE OF REVIEW This review details the significant advancement in knowledge of Immune-checkpoint inhibitor (ICI) and its potential deleterious cardiac immune-related adverse effects (irAE). We explore their mechanisms on the cardiac tissue, providing guidance on risk factors, clinical presentations, diagnostic strategies along with treatment. RECENT FINDINGS Recent findings have provided insights of cardiac irAEs that exist beyond the previously well-known ICI-induced myocarditis. We have a better understanding of the wide variety of cardiac irAEs pathologies both early and late onset. Moreover, there is more data on mechanisms of cardiotoxicity and patient and therapy-related risk factors, supporting closer routine cardiac monitoring with biomarkers and imaging for prevention and early detection. Diagnosing cardiac irAEs is a challenge given its broad clinical presentation. A high-level of suspicion in addition to early work-up is crucial to prevent serious cardiac events. A multi-disciplinary team including Cardiologists and Oncologists is essential for closely monitor patients' cardiac status on ICI therapy. There is a need of updated guidelines to establish clear recommendations in patients on ICIs.
Collapse
Affiliation(s)
- Maria T Gamero
- Department of Medicine, Division of Cardiovascular Disease, Jefferson Heart Institute, Thomas Jefferson University Hospital, Philadelphia, PA, USA.
| | - Avish Patel
- Department of Medicine, Division of Cardiovascular Disease, Jefferson Heart Institute, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Eugene Storozynsky
- Department of Medicine, Division of Cardiovascular Disease, Jefferson Heart Institute, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| |
Collapse
|
36
|
Tamaki N, Manabe O, Hirata K. Cardiovascular imaging in cardio-oncology. Jpn J Radiol 2024; 42:1372-1380. [PMID: 39207643 PMCID: PMC11588866 DOI: 10.1007/s11604-024-01636-x] [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: 06/12/2024] [Accepted: 07/23/2024] [Indexed: 09/04/2024]
Abstract
Advances in cancer treatment have improved in patient survival rate. On the other hand, management of cardiovascular complications has been increasingly required in cancer patients. Thus, cardio-oncology has attracted the attention by both oncologists and cardiologists. Cardiovascular imaging has played a key role for non-invasive assessment of cardiovascular alterations complimentary to biomarkers and clinical assessment. Suitable imaging selection and interpretation may allow early diagnosis of cardiovascular injury with potential implications for therapeutic management and improved outcomes after cancer therapy. Echocardiography has been commonly used to evaluate cardiac dysfunction in cardio-oncology area. Cardiac CT is valuable for assessing structural abnormalities of the myocardium, coronary arteries, and aorta. Molecular imaging has an important role in the assessment of the pathophysiology and future treatment strategy of cardiovascular dysfunction. Cardiac MRI is valuable for characterization of myocardial tissue. PET and SPECT molecular imaging has potential roles for quantitative assessment of cardiovascular disorders. Particularly, FDG-PET is considered as an elegant approach for simultaneous assessment of tumor response to cancer therapy and early detection of possible cardiovascular involvement as well. This review describes the promising potential of these non-invasive cardiovascular imaging modalities in cardio-oncology.
Collapse
Affiliation(s)
- Nagara Tamaki
- Kyoto College of Medical Science, Sonobe, Kyoto, Japan.
- Department of Radiology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| | - Osamu Manabe
- Department of Radiology, Jichi Medical University Saitama Medical Center, Saitama, Japan
| | - Kenji Hirata
- Department of Diagnostic Imaging, Faculty of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
37
|
Menounos S, Shen H, Tipirneni S, Bhaskar SMM. Decoding the Nexus: Cellular and Molecular Mechanisms Linking Stroke and Neurotoxic Microenvironments in Brain Cancer Patients. Biomolecules 2024; 14:1507. [PMID: 39766214 PMCID: PMC11673144 DOI: 10.3390/biom14121507] [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: 10/11/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 01/06/2025] Open
Abstract
Stroke is an often underrecognized albeit significant complication in patients with brain cancer, arising from the intricate interplay between cancer biology and cerebrovascular health. This review delves into the multifactorial pathophysiological framework linking brain cancer to elevated stroke risk, with particular emphasis on the crucial role of the neurotoxic microenvironment (NTME). The NTME, characterized by oxidative stress, neuroinflammation, and blood-brain barrier (BBB) disruption, creates a milieu that promotes and sustains vascular and neuronal injury. Key pathogenic factors driving brain cancer-related stroke include cancer-related hypercoagulability, inflammatory and immunological mechanisms, and other tumor-associated processes, including direct tumor compression, infection-related sequelae, and treatment-related complications. Recent advances in genomic and proteomic profiling present promising opportunities for personalized medicine, enabling the identification of biomarkers-such as oncogenes and tumor suppressor genes-that predict stroke susceptibility and inform individualized therapeutic strategies. Targeting the NTME through antioxidants to alleviate oxidative stress, anti-inflammatory agents to mitigate neuroinflammation, and therapies aimed at reinforcing the BBB could pave the way for more effective stroke prevention and management strategies. This integrative approach holds the potential to reduce both the incidence and severity of stroke, ultimately improving clinical outcomes and quality of life for brain cancer patients. Further research and well-designed clinical trials are essential to validate these strategies and integrate them into routine clinical practice, thereby redefining the management of stroke risk in brain cancer patients.
Collapse
Affiliation(s)
- Spiro Menounos
- Global Health Neurology Lab, Sydney, NSW 2150, Australia; (S.M.); (H.S.); (S.T.)
- School of Clinical Medicine, Medicine & Health, University of New South Wales (UNSW), St George and Sutherland Clinical Campuses, Sydney, NSW 2150, Australia
| | - Helen Shen
- Global Health Neurology Lab, Sydney, NSW 2150, Australia; (S.M.); (H.S.); (S.T.)
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW 2170, Australia
| | - Shraddha Tipirneni
- Global Health Neurology Lab, Sydney, NSW 2150, Australia; (S.M.); (H.S.); (S.T.)
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW 2170, Australia
| | - Sonu M. M. Bhaskar
- Global Health Neurology Lab, Sydney, NSW 2150, Australia; (S.M.); (H.S.); (S.T.)
- UNSW Medicine and Health, University of New South Wales (UNSW), South West Sydney Clinical Campuses, Sydney, NSW 2170, Australia
- NSW Brain Clot Bank, NSW Health Pathology, Sydney, NSW 2170, Australia
- Ingham Institute for Applied Medical Research, Clinical Sciences Stream, Liverpool, NSW 2170, Australia
- Department of Neurology & Neurophysiology, Liverpool Hospital and South West Sydney Local Health District, Liverpool, NSW 2150, Australia
- National Cerebral and Cardiovascular Center (NCVC), Department of Neurology, Division of Cerebrovascular Medicine and Neurology, Suita 564-8565, Osaka, Japan
| |
Collapse
|
38
|
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
|
39
|
Jan YJA, Chiang CH, Osataphan S, Lawless AR, Reynolds KL, Sullivan RJ. Body mass index and type 2 diabetes mellitus as metabolic determinants of immune checkpoint inhibitors response in melanoma. J Immunother Cancer 2024; 12:e009769. [PMID: 39510794 PMCID: PMC11552572 DOI: 10.1136/jitc-2024-009769] [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] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have improved survival outcomes in melanoma. Studies exploring the correlations between body mass index (BMI), type 2 diabetes (T2DM) and the outcomes of ICI treatment have yielded inconsistent results. In this study, we aim to investigate the effects of BMI and T2DM on survival outcomes of patients with melanoma receiving ICIs. METHODS A retrospective multicenter cohort of patients with melanoma treated with ICIs was analyzed. Overall survival was evaluated with Kaplan-Meier survival analysis, univariate Cox and multivariate Cox proportional hazards model. Propensity-score matching (1:1) analysis between overweight and non-overweight groups was done and survival analyses and Cox analyses were performed again. Subgroup analyses and secondary analyses stratifying patients with different weights and T2DM statuses were also performed. RESULTS A total of 2,078 patients were included, of whom 1,412 were overweight (BMI≥25 kg/m2) and 666 were non-overweight (BMI<25 kg/m2). Overweight patients had better overall survival compared with non-overweight (median 71.7 vs 36.7 months, p<0.001). Patients with T2DM had worse overall survival compared with patients without T2DM (median 28.5 vs 67.3 months, p<0.001). After propensity-score matching (666 overweight were matched to 666 non-overweight), overweight patients remained to have better overall survival compared with non-overweight (median 67.7 vs 36.7 months, p<0.001). Patients with T2DM had worse survival in univariate Cox (HR 1.71, (95% CI: 1.20 to 2.43)) and multivariate Cox (HR 1.58, (95% CI: 1.08 to 2.31)) analyses. Overweight patients without T2DM had the best survival outcomes compared with other weight and T2DM combinations. CONCLUSION In patients with melanoma treated with ICIs, being overweight had better survival outcomes compared with non-overweight. Having T2DM was associated with worse survival compared with those without T2DM. Further studies are needed to investigate the underlying mechanisms of these associations.
Collapse
Affiliation(s)
- Yu Jen Alexander Jan
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Harvard Medical School, Boston, Massachusetts, USA
| | - Cho-Han Chiang
- Harvard Medical School, Boston, Massachusetts, USA
- Department of Medicine, Mount Auburn Hospital, Cambridge, Massachusetts, USA
| | - Soravis Osataphan
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Medical Oncology, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Aleigha R Lawless
- Division of Hematology and Medical Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kerry L Reynolds
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ryan J Sullivan
- Harvard Medical School, Boston, Massachusetts, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
40
|
Vergara A, De Felice M, Cesaro A, Gragnano F, Pariggiano I, Golia E, De Pasquale A, Blasi E, Fimiani F, Monda E, Limongelli G, Calabrò P. Immune-Checkpoint Inhibitor-Related Myocarditis: Where We Are and Where We Will Go. Angiology 2024; 75:909-920. [PMID: 37699402 DOI: 10.1177/00033197231201929] [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] [Indexed: 09/14/2023]
Abstract
Immune checkpoint inhibitors (ICIs) are specific monoclonal antibodies directed against inhibitory targets of the immune system, mainly represented by programmed death-1 (PD1) ligand-1 (PD-L1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4), thus enabling an amplified T-cell-mediated immune response against cancer cells. These drugs have significantly improved prognosis in patients with advanced metastatic cancer (e.g., melanoma, non-small cell lung cancer, renal cell carcinoma). However, uncontrolled activation of anti-tumor T-cells could trigger an excessive immune response, possibly responsible for multi-organ damage, including, among others, lymphocytic myocarditis. The incidence of ICIs-induced myocarditis is underestimated and the patients affected are poorly characterized. The diagnosis and management of this condition are mainly based on expert opinion and case reports. EKG and ultrasound are tests that can help identify patients at risk of myocarditis during treatment by red flags, such as QRS complex enlargement and narrowing of global longitudinal strain (GLS). Therapy of ICI-related myocarditis is based on immunosuppressors, monoclonal antibodies and fusion proteins. A future strategy could involve the use of microRNAs. This review considers the current state of the art of immune-related adverse cardiovascular events, focusing on histological and clinical features, diagnosis and management, including current treatments and future pharmacological targets.
Collapse
Affiliation(s)
- Andrea Vergara
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Marco De Felice
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", Caserta, Italy
- Division of Oncology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Arturo Cesaro
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Felice Gragnano
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Ivana Pariggiano
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Enrica Golia
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Antonio De Pasquale
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Ettore Blasi
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| | - Fabio Fimiani
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli "V. Monaldi", Naples, Italy
| | - Emanuele Monda
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli "V. Monaldi", Naples, Italy
| | - Giuseppe Limongelli
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Unit of Inherited and Rare Cardiovascular Diseases, A.O.R.N. Dei Colli "V. Monaldi", Naples, Italy
| | - Paolo Calabrò
- Department of Translational Medical Sciences, University of Campania 'Luigi Vanvitelli', Caserta, Italy
- Division of Clinical Cardiology, A.O.R.N. 'Sant'Anna e San Sebastiano', Caserta, Italy
| |
Collapse
|
41
|
Oyakawa T, Muraoka N, Iida K, Fujita A, Yokoyama K, Ishikawa H, Murakami H. Relevance of surveillance manual for the early detection of immune checkpoint inhibitor-induced myocarditis: A case series. Asia Pac J Oncol Nurs 2024; 11:100598. [PMID: 39582548 PMCID: PMC11582373 DOI: 10.1016/j.apjon.2024.100598] [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: 07/30/2024] [Accepted: 09/18/2024] [Indexed: 11/26/2024] Open
Abstract
Objective The European Cardio-Oncology Guidelines recommend regular electrocardiography and troponin testing during immune checkpoint inhibitors (ICIs) treatment, but their efficacy for monitoring ICI treatment remains unclear. This study aimed to evaluate the effectiveness of a surveillance protocol for early detection of ICI-induced myocarditis. Methods Between May 2014 and May 2024, patients who began treatment with ICIs at our hospital and developed ICI-induced myocarditis were included in this study. We created a straightforward management flowchart for myocarditis. The protocol was based on monitoring troponin T levels. We confirmed the efficacy of our surveillance protocol using a case series of ICI-induced myocarditis. Results During the observation period, 3481 patients were newly started on ICIs at our hospital. Five patients were previously diagnosed with myocarditis, and five patients were diagnosed with myocarditis after the implementation of the surveillance protocol. The manual enabled the early detection of myocarditis, and the mortality rate for myocarditis at our hospital improved from 60% to 0%. The incidence of conduction system disorders significantly reduced from 100% to 0% (P < 0.01). After the surveillance protocol was initiated, there were no cases of myocarditis requiring immunosuppressive drugs beyond steroids. Conclusions This study confirmed the relevance of a troponin-based surveillance protocol for the early detection of ICI-induced myocarditis. The implementation of the surveillance protocol reduced mortality from myocarditis and significantly reduced serious complications of conduction system disorders. Although this study is a small case series of patients who developed myocarditis, we confirm the effectiveness of surveillance for myocarditis.
Collapse
Affiliation(s)
- Takuya Oyakawa
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Nao Muraoka
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | - Kei Iida
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
- Mishimatoukai Hospital, Shizuoka, Japan
| | - Ayano Fujita
- Division of Cardio-oncology, Shizuoka Cancer Center, Shizuoka, Japan
| | | | | | - Haruyasu Murakami
- Division of Advanced Medical Development, Shizuoka Cancer Center, Shizuoka, Japan
| |
Collapse
|
42
|
Wang C, Fan P, Wang Q. Evolving therapeutics and ensuing cardiotoxicities in triple-negative breast cancer. Cancer Treat Rev 2024; 130:102819. [PMID: 39216183 DOI: 10.1016/j.ctrv.2024.102819] [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: 05/22/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
Defined as scarce expression of hormone receptors and human epidermal growth factor receptor 2, triple-negative breast cancer (TNBC) is labeled as the most heterogeneous subtype of breast cancer with poorest prognosis. Despite rapid advancements in precise subtyping and tailored therapeutics, the ensuing cancer therapy-related cardiovascular toxicity (CTR-CVT) could exert detrimental impacts to TNBC survivors. Nowadays, this interdisciplinary issue is incrementally concerned by cardiologists, oncologists and other pertinent experts, propelling cardio-oncology as a booming field focusing on the whole-course management of cancer patients with potential cardiovascular threats. Here in this review, we initially profile the evolving molecular subtyping and therapeutic landscape of TNBC. Further, we introduce various monitoring approaches of CTR-CVT. In the main body, we elaborate on typical cardiotoxicities ensuing anti-TNBC treatments in detail, ranging from chemotherapy (especially anthracyclines), surgery, anesthetics, radiotherapy to immunotherapy, with future perspectives on promising directions in the era of artificial intelligence and traditional Chinese medicine.
Collapse
Affiliation(s)
- Chongyu Wang
- Department of Medicine, Xinglin College, Nantong University, Nantong 226007, Jiangsu, China
| | - Pinchao Fan
- The First Clinical Medical College, Nanjing Medical University, Nanjing 211166, Jiangsu, China; Sir Run Run Hospital, Nanjing Medical University, Nanjing 211112, Jiangsu, China
| | - Qingqing Wang
- Department of Thyroid and Breast Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
43
|
Liu L, Yao W, Wang M, Wang B, Kong F, Fan Z, Fan G. A systematic review of cardiovascular toxicities induced by cancer immune therapies: Underlying mechanisms, clinical manifestations and therapeutic approaches. Semin Cancer Biol 2024; 106-107:179-191. [PMID: 39442678 DOI: 10.1016/j.semcancer.2024.10.004] [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: 04/29/2024] [Revised: 09/26/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Immunotherapy has revolutionized the management of various types of cancers, even those previously deemed untreatable. Nonetheless, these medications have been associated with inflammation and damage across various organs. These challenges are exemplified by the adverse cardiovascular impacts of cancer immunotherapy, which need comprehensive understanding, clarification, and management integrated into the overall care of cancer patients. Numerous anticancer immunotherapies have been linked to the prevalence and severity of cardiovascular toxicity. These challenges emphasize the importance of conducting fundamental and applied research to elucidate disease causes, discover prognostic indicators, enhance diagnostic methods, and create successful therapies. Despite the acknowledged importance of T cells, there remains a knowledge gap regarding the inciting antigens, the reasons for their recognition, and the mechanisms of how they contribute to cardiac cell injury. In this review, we summarize the molecular mechanism, epidemiology, diagnosis, pathophysiology and corresponding treatment of cardiovascular toxicity induced by immunotherapy, including immune checkpoint inhibitors (ICIs), adoptive cell therapies (ACT), and bi-specific T-cell engagers (BiTEs) among others. By elucidating these aspects, we aim to provide a better understanding of immunotherapies in cancer treatment and offer guidance for their clinical application.
Collapse
Affiliation(s)
- Li Liu
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| | - Wentao Yao
- Department of Urology, Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou, China
| | - Mi Wang
- Department of Cardiology Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Baohui Wang
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fanming Kong
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| | - Zhongguo Fan
- Department of Cardiology Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Guanwei Fan
- First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China; National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China.
| |
Collapse
|
44
|
Guan Z, Yao T, Liu G, Liu J, Guo L, Li Z, Ma J. Peripheral biomarkers to assess risk, severity, and prognosis of immune checkpoint inhibitor-associated myocarditis: a retrospective clinical study. Front Cardiovasc Med 2024; 11:1465743. [PMID: 39512372 PMCID: PMC11540693 DOI: 10.3389/fcvm.2024.1465743] [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: 07/16/2024] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Background Immune checkpoint inhibitor-associated myocarditis (ICI myocarditis) is an infrequent but potentially fatal immune-related adverse event. This study aimed to identify valuable indicators for risk prediction and evaluation of disease severity and outcomes. Methods A total of 79 patients with severe or mild ICI myocarditis and 158 controls without post-ICI immune-related adverse events were enrolled in this retrospective study. The clinical application value of a series of simple biomarkers were tested. Results Higher levels of the systemic immune-inflammation index (SII), neutrophil-to-eosinophil ratio (NER), aspartate transferase-to-albumin ratio (AAR), and lactic dehydrogenase-to-albumin ratio (LAR) at myocarditis onset were associated with severe disease conditions. In the receiver operating characteristic analysis, biomarkers areas under the curve (AUC) close to or greater than 0.8 were LAR (AUC: 0.810) and AAR (AUC: 0.806). Patients with higher SII, AAR, and LAR also exhibited poorer overall survival. The SII, NER, AAR, and LAR before the last ICI treatment increased relative to baseline in patients with ICI myocarditis, whereas no significant changes in the tested biomarkers were observed in the control group. For SII, AAR, and LAR, high ratios of the biomarker levels before the last ICI to baseline was associated with the incidence of myocarditis. Conclusions Surveillance of these economical biomarkers during ICI therapy might contribute to the risk prediction of ICI myocarditis, as well as the assessment of disease severity and prognosis.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jingtao Ma
- Department of Cardiology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
45
|
Quagliariello V, Canale ML, Bisceglia I, Iovine M, Giordano V, Giacobbe I, Scherillo M, Gabrielli D, Maurea C, Barbato M, Inno A, Berretta M, Tedeschi A, Oliva S, Greco A, Maurea N. Glucagon-like Peptide 1 Receptor Agonists in Cardio-Oncology: Pathophysiology of Cardiometabolic Outcomes in Cancer Patients. Int J Mol Sci 2024; 25:11299. [PMID: 39457081 PMCID: PMC11508560 DOI: 10.3390/ijms252011299] [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: 09/06/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Cancer patients, especially long cancer survivors, are exposed to several cardio-metabolic diseases, including diabetes, heart failure, and atherosclerosis, which increase their risk of cardiovascular mortality. Therapy with glucagon-like peptide 1 (GLP1) receptor agonists demonstrated several beneficial cardiovascular effects, including atherosclerosis and heart failure prevention. Cardiovascular outcome trials (CVOTs) suggest that GLP-1 RA could exert cardiorenal benefits and systemic anti-inflammatory effects in patients with type-2 diabetes through the activation of cAMP and PI3K/AkT pathways and the inhibition of NLRP-3 and MyD88. In this narrative review, we highlight the biochemical properties of GLP-1 RA through a deep analysis of the clinical and preclinical evidence of the primary prevention of cardiomyopathies. The overall picture of this review encourages the study of GLP-1 RA in cancer patients with type-2 diabetes, as a potential primary prevention strategy against heart failure and atherosclerosis.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | | | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy;
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Vienna Giordano
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Marino Scherillo
- Division of Cardiology, Hospital San Pio Benevento (BN), 82100 Benevento, Italy;
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy;
| | - Carlo Maurea
- Department of Medicine, University of Salerno, 84084 Fisciano, Italy;
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy;
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy;
| | - Andrea Tedeschi
- Cardiology Unit of Emergency Department, Guglielmo da Saliceto Hospital, 29121 Piacenza, Italy;
| | - Stefano Oliva
- UOSD Cardiologia di Interesse Oncologico IRCCS Istituto Tumori “Giovanni Paolo II”, 70124 Bari, Italy;
| | - Alessandra Greco
- Divisione di Cardiologia, Fondazione IRCCS San Matteo Hospital, Viale Golgi 19, 27100 Pavia, Italy;
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (M.I.); (V.G.); (I.G.); (M.B.); (N.M.)
| |
Collapse
|
46
|
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
|
47
|
Milutinovic S, Jancic P, Jokic V, Petrovic M, Dumic I, Rodriguez AM, Tanasijevic N, Begosh-Mayne D, Stanojevic D, Escarcega RO, Lopez-Mattei J, Cao X. Pembrolizumab-Associated Cardiotoxicity: A Retrospective Analysis of the FDA Adverse Events Reporting System. Pharmaceuticals (Basel) 2024; 17:1372. [PMID: 39459012 PMCID: PMC11510316 DOI: 10.3390/ph17101372] [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: 08/02/2024] [Revised: 10/08/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been successfully used in the previous decade for the treatment of a variety of malignancies. Adverse events (AEs) can cause many symptoms, most notably cardiac. We analyzed the frequency of these adverse events, comparing pembrolizumab and other ICIs. METHODS Using the Food and Drug Administration (FDA) adverse event reporting database (FAERS), we searched for all adverse events of interest reported for every ICI included in this study. After obtaining the data, we conducted a disproportionality analysis using the reporting odds ratio (ROR) and the information component (IC). RESULTS A total of 6719 ICI-related cardiac adverse events of interest were reported in the database. Serious outcomes were reported in 100% of the cases, with 34.3% of the cases ending fatally. Compared with all other medications in the database, pembrolizumab use was more frequently associated with myocarditis, pericardial disease, heart failure, and atrial fibrillation. No difference was found in cardiotoxicity between different ICIs. CONCLUSIONS Although infrequent, cardiac AEs in pembrolizumab use are associated with serious outcomes and high mortality. Prospective studies are needed to further research the connection between ICI use and cardiotoxicity.
Collapse
Affiliation(s)
- Stefan Milutinovic
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
| | - Predrag Jancic
- Faculty of Medicine, University of Belgrade, 11000 Belgrade, Serbia
| | - Vera Jokic
- Montefiore New Rochelle Hospital, New Rochelle, NY 10801, USA
| | - Marija Petrovic
- Cardiology Fellowship Program, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Igor Dumic
- Department of Hospital Medicine, Mayo Clinic Health System, Eau Claire, WI 54703, USA
| | - Ambar Morales Rodriguez
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
| | | | - Dustin Begosh-Mayne
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
| | - Dragana Stanojevic
- Clinic for Cardiology, University Clinical Center Nis, 18000 Nis, Serbia
| | - Ricardo O. Escarcega
- Internal Medicine Residency Program at Lee Health, Florida State University College of Medicine, Cape Coral, FL 33909, USA
- Lee Health Heart Institute, Fort Myers, FL 33908, USA
| | | | - Xiangkun Cao
- Lee Health Heart Institute, Fort Myers, FL 33908, USA
| |
Collapse
|
48
|
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
|
49
|
Nielsen DL, Juhl CB, Nielsen OH, Chen IM, Herrmann J. Immune Checkpoint Inhibitor-Induced Cardiotoxicity: A Systematic Review and Meta-Analysis. JAMA Oncol 2024; 10:1390-1399. [PMID: 39172480 PMCID: PMC11342217 DOI: 10.1001/jamaoncol.2024.3065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 04/17/2024] [Indexed: 08/23/2024]
Abstract
Importance Immune checkpoint inhibitors (ICIs) improve outcomes in a wide range of cancers; however, serious adverse effects, including cardiovascular adverse effects (CVAEs), can occur. Objective To determine the incidence of CVAEs and analyze data on the management of myocarditis in patients exposed to ICIs. Data Sources PubMed, Embase, and Cochrane Central Register of Controlled Trials from inception were searched on April 4, 2023. Study Selection Two separate studies were performed. Key inclusion criteria for study 1 were phases 1 to 4 trials involving adults with malignant neoplasms treated with an ICI and toxicity data; for study 2, publications (case reports and retrospective analyses) on clinical manifestations and treatment of patients with ICI-induced CVAEs. Studies with dose escalation or fewer than 11 patients in each group and all case reports, retrospective analyses, letters, reviews, and editorials were excluded from study 1. Studies not published in English were excluded from study 2. Data Extraction and Synthesis The PRISMA guidelines and Cochrane Handbook for Systematic Reviews were followed. Data were extracted independently by 2 researchers. A meta-analysis of the incidence of CVAEs in clinical trials and a systematic review of the evidence for the management of myocarditis were performed. Data were pooled using a random-effects model. Main Outcomes and Measures In study 1, the primary outcome was incidence CVAEs in clinical trials with ICIs and ICI combination therapies. Study 2 examined evidence supporting specific management strategies that may decrease the mortality rate of myocarditis. The primary outcomes were planned before data collection began. Results In study 1, a total of 83 315 unique participants in 589 unique trials were included in the meta-analysis. Incidence of CVAEs induced by anti-programmed cell death 1 and/or programmed cell death ligand 1 was 0.80% (95% CI, 0%-1.66%) in clinical trials, with no differences between the compounds, except for cemiplimab, which was associated with a higher risk of CVAEs. Incidence of CVAEs following ipilimumab treatment was 1.07% (95% CI, 0%-2.58%). The incidence of myocarditis was significantly higher following treatment with dual ICIs. However, CVAE incidence was not higher with dual ICIs, ICI combination with chemotherapy, or tyrosine kinase inhibitors. Evidence from randomized clinical trials on recommended monitoring and treatment strategies for ICI-induced myocarditis was lacking. Study 2 showed that myocarditis-associated mortality occurred in 83 of 220 patients (37.7%). Prospective data from 40 patients with myocarditis indicated that systematic screening for respiratory muscle involvement, coupled with active ventilation, prompt use of abatacept, and the addition of ruxolitinib, may decrease the mortality rate. Conclusions and Relevance Immune checkpoint inhibitor-induced CVAEs and/or myocarditis were recorded in 1.07% of patients in clinical trials. The CVAE mortality risk remains high, justifying the need for monitoring and management strategies for which evidence from randomized clinical trials is absent. Early recognition, ICI therapy cessation, prompt initiation of corticosteroid therapy, and escalation of therapy are all crucial elements for achieving optimal outcomes. Prospective clinical trials or at least prospective registration of treatments and outcomes are highly warranted.
Collapse
Affiliation(s)
- Dorte Lisbet Nielsen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Carsten Bogh Juhl
- Department of Sports Science and Clinical Biomechanics, University of Southern Denmark, Odense, Denmark
- Department of Physiotherapy and Occupational Therapy, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Ole Haagen Nielsen
- Department of Gastroenterology, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Inna Markovna Chen
- Department of Oncology, Herlev and Gentofte Hospital, University of Copenhagen, Copenhagen, Denmark
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota
| |
Collapse
|
50
|
Cheng X, Lin J, Wang B, Huang S, Liu M, Yang J. Clinical characteristics and influencing factors of anti-PD-1/PD-L1-related severe cardiac adverse event: based on FAERS and TCGA databases. Sci Rep 2024; 14:22199. [PMID: 39333574 PMCID: PMC11436968 DOI: 10.1038/s41598-024-72864-4] [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: 01/26/2024] [Accepted: 09/11/2024] [Indexed: 09/29/2024] Open
Abstract
Combining the FDA Adverse Event Reporting System (FAERS) and the Cancer Genome Atlas (TCGA) databases, we aim to explore the factors that influence anti-programmed cell death protein-1 inhibitors/programmed death-ligand-1 (PD-1/PD-L1) related severe cardiac adverse events (cAEs). We obtained anti-PD-1/PD-L1 adverse event reports from January 2014 to December 2022 from the FAERS database. Disproportionality analysis was performed to find anti-PD-1/PD-L1-related cAEs using the proportional reporting ratio (PRR). We were exploring influencing factors based on multivariate logistic regression analysis. Finally, we utilized a strategy that combines FAERS and TCGA databases to explore the potential immune and genetic influencing factors associated with anti-PD-1/PD-L1-related severe cAEs. Reports of severe cAEs accounted for 7.10% of the overall anti-PD-1/PD-L1 adverse event reports in the FAERS database. Immune-mediated myocarditis (PRR = 77.01[59.77-99.23]) shows the strongest toxic signal. The elderly group (65-74: OR = 1.34[1.23-1.47], ≥ 75: OR = 1.64[1.49-1.81]), male (OR = 1.14[1.05-1.24]), anti-PD-L1 agents (OR = 1.17[1.03-1.33]), patients with other adverse events (OR = 2.38[2.17-2.60]), and the concomitant use of proton pump inhibitor (OR = 1.29[1.17-1.43]), nonsteroidal anti-inflammatory drugs (OR = 1.17[1.04-1.31]), or antibiotics (OR = 1.24[1.08-1.43]) may increase the risk of severe cAEs. In addition, PD-L1 mRNA (Rs = 0.71, FDR = 2.30 × 10- 3) and low-density lipoprotein receptor-related protein 3 (LRP3) (Rs = 0.82, FDR = 2.17 × 10- 2) may be immune and genetic influencing factors for severe cAEs. Severe cAEs may be related to antigen receptor-mediated signalling pathways. In this study, we found that age, gender, anti-PD-1/PD-L1 agents, concomitant other adverse events, concomitant medication, PD-L1 mRNA, and LRP3 may be influencing factors for anti-PD-1/PD-L1-related severe cAEs. However, our findings still require a large-scale prospective cohort validation.
Collapse
Affiliation(s)
- Xitong Cheng
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Jierong Lin
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Bitao Wang
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Shunming Huang
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China
- College of Pharmacy, Fujian Medical University, Fuzhou, China
| | - Maobai Liu
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China.
- College of Pharmacy, Fujian Medical University, Fuzhou, China.
- Department of Pharmacy, Union Hospital Affiliated to Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, 350001, China.
| | - Jing Yang
- Department of Pharmacy, Fujian Medical University Union Hospital, Fuzhou, China.
- College of Pharmacy, Fujian Medical University, Fuzhou, China.
- Department of Pharmacy, Union Hospital Affiliated to Fujian Medical University, No.29, Xinquan Road, Gulou District, Fuzhou, 350001, China.
| |
Collapse
|