1
|
Rolling CC, Lewirt S, Beitzen-Heineke A, Beckmann L, Bokemeyer C, Alsdorf W, Voigtlaender M, Langer F. Checkpoint Inhibitors, CAR T Cells, and the Hemostatic System: What Do We Know So Far? Hamostaseologie 2025; 45:175-187. [PMID: 40334710 DOI: 10.1055/a-2528-5071] [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: 05/09/2025] Open
Abstract
Immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells are novel therapeutic strategies that enhance anticancer immunity by activating or engineering cancer-targeting T cells. The resulting hyperinflammation carries several side effects, ranging from autoimmune-like symptoms to cytokine release syndrome (CRS), with potentially severe consequences. Recent findings indicate that ICIs increase the risk of venous and arterial thromboembolic adverse events. Patients with prior VTE might be at higher risk of developing new events under ICI while other risk factors vary across studies. So far, data on CAR T-linked coagulopathies are limited. Hypofibrinogenemia in the presence of CRS is the most commonly observed dysregulation of hemostatic parameters. A rare but particularly severe adverse event is the development of disseminated intravascular coagulation activation, which can occur in the setting of CRS and may be linked to immune effector cell-associated hemophagocytic lymphohistiocytosis. While the increasing number of studies on thromboembolic complications and coagulation alterations under ICIs and CAR T therapies are concerning, these results might be influenced by the retrospective study design and the heterogeneous patient populations. Importantly, numerous promising new T cell-based immunotherapies are currently under investigation for various cancers and are expected to become very prominent therapy options in the near future. Therefore, coagulopathies and thrombosis under T cell-directed immuno- and anti-cancer therapies is important. Our review provides an overview of the current understanding of ICI- and CAR T-associated thromboembolism. We discuss pathogenic mechanisms of inflammation-associated coagulation activation and explore potential biomarkers for VTE.
Collapse
Affiliation(s)
- Christina C Rolling
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Samuel Lewirt
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Antonia Beitzen-Heineke
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, New York University Grossman School of Medicine, New York, New York, United States
| | - Lennart Beckmann
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Minna Voigtlaender
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Langer
- Department of Oncology, Hematology and BMT with Section of Pneumology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
2
|
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
|
3
|
Guan T, Monteiro O, Chen D, Luo Z, Chi K, Li Z, Liang Y, Lu Z, Jiang Y, Yang J, Lin W, Yi M, Zhang K, Ou C. Long-term and short-term cardiovascular disease mortality among patients of 21 non-metastatic cancers. J Adv Res 2025; 69:215-224. [PMID: 38537701 PMCID: PMC11954795 DOI: 10.1016/j.jare.2024.03.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 03/05/2024] [Accepted: 03/23/2024] [Indexed: 04/04/2024] Open
Abstract
INTRODUCTION Previous studies on cardiovascular disease (CVD) death risk in cancer patients mostly focused on overall cancer, age subgroups and single cancers. OBJECTIVES To assess the CVD death risk in non-metastatic cancer patients at 21 cancer sites. METHODS A total of 1,672,561 non-metastatic cancer patients from Surveillance, Epidemiology, and End Results (SEER) datebase (1975-2018) were included in this population-based study, with a median follow-up of 12·7 years. The risk of CVD deaths was assessed using proportions, competing-risk regression, absolute excess risks (AERs), and standardized mortality ratios (SMRs). RESULTS In patients with localized cancers, the proportion of CVD death and cumulative mortality from CVD in the high-competing risk group (14 of 21 unique cancers) surpassed that of primary neoplasm after cancer diagnosis. The SMRs and AERs of CVD were found higher in patients with non-metastatic cancer than the general US population (SMR 1·96 [95 %CI, 1·95-1·97]-19·85[95 %CI, 16·69-23·44]; AER 5·77-210·48), heart disease (SMR 1·94[95 %CI, 1·93-1·95]-19·25[95 %CI, 15·76-23·29]; AER 4·36-159·10) and cerebrovascular disease (SMR 2·05[95 %CI, 2·02-2·08]-24·71[95 %CI, 16·28-35·96]; AER 1·01-37·44) deaths. In the high-competing risk group, CVD-related SMR in patients with localized stage cancer increased with survival time but followed a reverse-dipper pattern in the low-competing risk group (7 of 21 cancers). The high-competing risk group had higher CVD-related death risks than the low-competing risk group. CONCLUSION The CVD death risk in patients with non-metastatic cancer varied by cancer stage, site and survival time. The risk of CVD mortality is higher in 14 out of 21 localized cancers (high-competing cancers). Targeted strategies for CVD management in non-metastatic cancer patients are needed.
Collapse
Affiliation(s)
- Tianwang Guan
- Cancer Center, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China; Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510515, China
| | - Olivia Monteiro
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, Macao 999078, China
| | - Dongting Chen
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Zehao Luo
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Kaiyi Chi
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China; Guangdong Eye Institute, Department of Ophthalmology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou 510080, China
| | - Zhihao Li
- Department of Epidemiology, School of Public Health, Southern Medical University, Guangzhou 510515, China
| | - Yinglan Liang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Zhenxing Lu
- The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China
| | - Yanting Jiang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Jinming Yang
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Wenrui Lin
- Department of Clinical Medicine, Clinical Medical School, Guangzhou Medical University, Guangzhou 510180, China
| | - Min Yi
- Department of Endocrinology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510180, China
| | - Kang Zhang
- Faculty of Medicine, Medical Sciences Division, Macau University of Science and Technology, Avenida da Harmonia, Praia Park, Coloane, Macao 999078, China; The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China.
| | - Caiwen Ou
- Guangdong Provincial Key Laboratory of Cardiac Function and Microcirculation, Guangzhou 510515, China; The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan 523059, China.
| |
Collapse
|
4
|
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
|
5
|
Tolu-Akinnawo O, Ogunniyi KE. Accelerated Coronary Artery Disease in a Patient With Advanced Lung Cancer: Interplay of Cancer, Inflammation, and Treatment Effects. Cureus 2025; 17:e77769. [PMID: 39981449 PMCID: PMC11841481 DOI: 10.7759/cureus.77769] [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] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Cardiovascular disease (CVD) and lung cancer are among the most prevalent causes of death worldwide, representing substantial public health challenges. The relationship between coronary artery disease (CAD) and lung cancer is potentially multifaceted, influenced by common risk factors and the adverse cardiac effects of cancer treatments. However, cases of accelerated CAD occurring within a year of cancer treatment initiation are rarely reported. We present a case of a Black American male undergoing combination therapy (chemotherapy/radiotherapy/immunotherapy) for small-cell lung cancer, who developed ST-elevation myocardial infarction (STEMI) seven months after beginning treatment. Although we do not have the official records, the patient reported that he had previously undergone a cardiac workup (including an echo/stress test) at an outside facility a year prior, due to persistent dyspnea, which was unremarkable. The patient underwent successful percutaneous coronary intervention and was discharged on apixaban and prasugrel. This study underscores the importance of maintaining a high index of suspicion for acute coronary syndromes (ACS) in patients receiving lung cancer treatments, emphasizes the need for early recognition of warning signs, and highlights the critical role of risk factor management and enhanced surveillance in this vulnerable population.
Collapse
|
6
|
Li H, Zheng Y, Li B, Zhi Y, Chen M, Zeng J, Jiao Q, Tao Y, Liu X, Shen Z, Zhang J, Zhao W, Chen D. Association among major adverse cardiovascular events with immune checkpoint inhibitors: A systematic review and meta-analysis. J Intern Med 2025; 297:36-46. [PMID: 39537368 DOI: 10.1111/joim.20028] [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] [Indexed: 11/16/2024]
Abstract
BACKGROUND This meta-analysis aimed to determine the incidence and overall risk of major adverse cardiovascular events (MACEs) related to immune checkpoint inhibitors (ICIs). METHODS We systematically searched all cohort studies, including the available MACE data in cancer patients receiving ICIs, in PubMed, Embase, and the Cochrane Library, from their inception to September 5, 2023. The primary outcome was the incidence of MACEs associated with ICI exposure, and the secondary outcome was the overall risk of MACEs associated with ICI exposure versus non-ICI exposure controls. Risk ratios with 95% confidence intervals were used in the random- or fixed-effects models. RESULTS Overall, 26 cohort studies met the inclusion criteria, involving 109,883 cancer patients. In the median follow-up period ranging from 3.3 to 55.2 months, the incidence of MACEs associated with ICI exposure was 8.22%, ranging from 0.55% to 3.98%, among the nine MACEs, including myocarditis, tachyarrhythmia, pericarditis, pericardial effusions, cardiovascular death, myocardial infarction, heart failure, stroke, and conduction disorder. The incidence of MACE associated with non-ICI exposure was 3.84%, ranging from 0.81% to 4.72%. The risks of all-grade MACEs and pericardial effusions were significantly higher in the ICI group than in the non-ICI controls. ICI treatment, age, male sex, and prior radiation therapy were significantly associated with MACEs. CONCLUSION The risk of MACEs during ICI treatment in patients with cancer is more common than is currently recognized. ICI use is closely associated with an increased risk of MACEs. Patients at risk were older, male, and had a history of radiation therapy.
Collapse
Affiliation(s)
- Haixia Li
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanfei Zheng
- Center for Studies in Constitution Research of Traditional Chinese Medicine, Basic Medical School, Beijing University of Chinese Medicine, Beijing, China
| | - Bin Li
- Acupuncture and Moxibustion Department, Beijing Hospital of Traditional Chinese Medicine, Beijing Key Laboratory of Acupuncture Neuromodulation, Capital Medical University, Beijing, China
| | - Yinghao Zhi
- Department of Rehabilitation, Wenzhou Traditional Chinese Medicine Hospital of Zhejiang Chinese Medical University, Wenzhou, China
| | - Mingxian Chen
- Department of Gastroenterology, Tongde Hospital of Zhejiang Province, Hangzhou, China
| | - Jing Zeng
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qian Jiao
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuxuan Tao
- Department of Cardiology, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xinmei Liu
- School of Graduates, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zican Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jiahui Zhang
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| | - Weizhe Zhao
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Dong Chen
- Department of Oncology, Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China
| |
Collapse
|
7
|
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
|
8
|
Polomski EAS, Kapiteijn EW, Heemelaar JC, van der Kolk AV, Kalisvaart TM, van de Burgt A, Dibbets-Schneider P, van Velden FHP, Seijkens TTP, Stöger JL, Jukema JW, de Geus-Oei LF, Antoni ML. Arterial inflammation on [ 18F]FDG PET/CT in melanoma patients treated with and without immune checkpoint inhibitors: CHECK-FLAME I. Atherosclerosis 2024; 398:118595. [PMID: 39307054 DOI: 10.1016/j.atherosclerosis.2024.118595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/04/2024] [Accepted: 09/05/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND AND AIMS Immune checkpoint inhibitors (ICIs) revolutionized cancer treatment. However, ICIs may increase the immune response to non-tumor cells, possibly resulting in increased arterial inflammation, raising the risk of atherosclerotic events. Nevertheless, malignancies may induce a pro-inflammatory state and the association between ICIs and arterial inflammation remains to be clarified. This study aims to assess differences in increase in arterial inflammation between patients with advanced melanoma treated with ICIs compared to a control group without ICIs. METHODS Patients with advanced melanoma who underwent [18F]FDG PET/CT scans at baseline, 6 months (T1) and 18 months (T2) were included in this retrospective observational study. Arterial inflammation was evaluated in eight segments by calculating the target-to-background ratio (TBR). The primary study outcome was the difference in increase in mean TBRmax between patients treated with and without ICIs. RESULTS We included 132 patients of whom 72.7 % were treated with ICIs. After exclusion for the use of anti-inflammatory medication, patients treated with ICIs showed a significant increase in mean TBRmax between baseline and T1 from 1.29 ± 0.12 to 1.33 ± 0.13 (p = 0.017), while in the control group, no change in mean TBRmax (1.30 ± 0.12 to 1.28 ± 0.10, p = 0.22) was observed (p = 0.027). During longer follow-up, mean TBRmax remained stable in both groups. Arterial inflammation increased significantly after ICI therapy in patients without active inflammation (p < 0.001) and in patients without calcifications (p = 0.013). CONCLUSIONS A significant increase in arterial inflammation as measured on [18F]FDG PET/CT was observed in patients with advanced melanoma treated with ICIs only in the first six months after initiation of therapy, whereas no changes were observed in the control group. Moreover, arterial inflammation was mainly increased in patients without pre-existing inflammatory activity and with non-calcified lesions.
Collapse
Affiliation(s)
- Elissa A S Polomski
- Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Ellen W Kapiteijn
- Department of Medical Oncology, Leiden University Medical Center, Leiden, the Netherlands
| | - Julius C Heemelaar
- Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Anne V van der Kolk
- Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, the Netherlands
| | - Timo M Kalisvaart
- Section Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alina van de Burgt
- Section Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Department of Nuclear Medicine, Alrijne Hospital, Leiden, the Netherlands
| | - Petra Dibbets-Schneider
- Section Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Floris H P van Velden
- Section Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Tom T P Seijkens
- Department of Medical Oncology, Antoni van Leeuwenhoek - Netherlands Cancer Institute, Amsterdam, the Netherlands; Department of Medical Biochemistry, Amsterdam University Medical Center, Amsterdam, the Netherlands
| | - J Lauran Stöger
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - J Wouter Jukema
- Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, the Netherlands; Netherlands Heart Institute, Utrecht, the Netherlands
| | - Lioe-Fee de Geus-Oei
- Section Nuclear Medicine, Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands; Biomedical Photonic Imaging Group, University of Twente, Enschede, the Netherlands; Department of Radiation Science & Technology, Delft University of Technology, Delft, the Netherlands
| | - M Louisa Antoni
- Department of Cardiology, Heart Lung Center, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
9
|
Tocchetti CG, Farmakis D, Koop Y, Andres MS, Couch LS, Formisano L, Ciardiello F, Pane F, Au L, Emmerich M, Plummer C, Gulati G, Ramalingam S, Cardinale D, Brezden-Masley C, Iakobishvili Z, Thavendiranathan P, Santoro C, Bergler-Klein J, Keramida K, de Boer RA, Maack C, Lutgens E, Rassaf T, Fradley MG, Moslehi J, Yang EH, De Keulenaer G, Ameri P, Bax J, Neilan TG, Herrmann J, Mbakwem AC, Mirabel M, Skouri H, Hirsch E, Cohen-Solal A, Sverdlov AL, van der Meer P, Asteggiano R, Barac A, Ky B, Lenihan D, Dent S, Seferovic P, Coats AJS, Metra M, Rosano G, Suter T, Lopez-Fernandez T, Lyon AR. Cardiovascular toxicities of immune therapies for cancer - a scientific statement of the Heart Failure Association (HFA) of the ESC and the ESC Council of Cardio-Oncology. Eur J Heart Fail 2024; 26:2055-2076. [PMID: 39087551 DOI: 10.1002/ejhf.3340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 05/22/2024] [Accepted: 06/03/2024] [Indexed: 08/02/2024] Open
Abstract
The advent of immunological therapies has revolutionized the treatment of solid and haematological cancers over the last decade. Licensed therapies which activate the immune system to target cancer cells can be broadly divided into two classes. The first class are antibodies that inhibit immune checkpoint signalling, known as immune checkpoint inhibitors (ICIs). The second class are cell-based immune therapies including chimeric antigen receptor T lymphocyte (CAR-T) cell therapies, natural killer (NK) cell therapies, and tumour infiltrating lymphocyte (TIL) therapies. The clinical efficacy of all these treatments generally outweighs the risks, but there is a high rate of immune-related adverse events (irAEs), which are often unpredictable in timing with clinical sequalae ranging from mild (e.g. rash) to severe or even fatal (e.g. myocarditis, cytokine release syndrome) and reversible to permanent (e.g. endocrinopathies).The mechanisms underpinning irAE pathology vary across different irAE complications and syndromes, reflecting the broad clinical phenotypes observed and the variability of different individual immune responses, and are poorly understood overall. Immune-related cardiovascular toxicities have emerged, and our understanding has evolved from focussing initially on rare but fatal ICI-related myocarditis with cardiogenic shock to more common complications including less severe ICI-related myocarditis, pericarditis, arrhythmias, including conduction system disease and heart block, non-inflammatory heart failure, takotsubo syndrome and coronary artery disease. In this scientific statement on the cardiovascular toxicities of immune therapies for cancer, we summarize the pathophysiology, epidemiology, diagnosis, and management of ICI, CAR-T, NK, and TIL therapies. We also highlight gaps in the literature and where future research should focus.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Dimitrios Farmakis
- Department of Cardiology, Attikon University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Yvonne Koop
- Department of Cardiovascular Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht University, Utrecht, The Netherlands
- Dutch Heart Foundation, The Hague, The Netherlands
| | - Maria Sol Andres
- Royal Brompton Hospital, Part of Guy's and St Thomas' NHS Foundation Trust, London, UK
| | - Liam S Couch
- Division of Cardiovascular Medicine, Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Fortunato Ciardiello
- Department of Precision Medicine, University of Campania 'Luigi Vanvitelli', Naples, Italy
| | - Fabrizio Pane
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy
| | - Lewis Au
- Skin and Renal Unit, Royal Marsden NHS Foundation Trust, London, UK
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, VIC, Australia
| | - Max Emmerich
- The Francis Crick Institute, London, UK
- The Royal Marsden Hospital, London, UK
- St. John's Institute of Dermatology, Guy's and St Thomas' Hospital, London, UK
| | - Chris Plummer
- Department of Cardiology, Freeman Hospital, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Geeta Gulati
- Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Cardiology, Division of Medicine, Oslo University Hospital, Ullevål, Oslo, Norway
- Division of Research and Innovation, Akershus University Hospital, Lørenskog, Norway
| | - Sivatharshini Ramalingam
- Royal Brompton Hospital, Part of Guy's and St Thomas' NHS Foundation Trust, London, UK
- Oxted Health Centre, Oxted, UK
| | - Daniela Cardinale
- CardioOncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | | | - Zaza Iakobishvili
- Department of Community Cardiology, Tel Aviv Jaffa District, Clalit Health Services, Tel Aviv, Israel
- Department of Cardiology, Assuta Ashdod University Hospital, Ashdod, Israel
- Faculty of Health Sciences, Ben Gurion University of the Negev, Be'er Sheva, Israel
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University of Toronto, Toronto, ON, Canada
| | - Ciro Santoro
- Department of Advanced Biomedical Science, Federico II University Hospital, Naples, Italy
| | | | - Kalliopi Keramida
- Cardiology Department, General Anti-Cancer, Oncological Hospital, Agios Savvas, Athens, Greece
| | - Rudolf A de Boer
- Department of Cardiology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center (CHFC), University Clinic Würzburg, Würzburg, Germany
- Medical Clinic 1, University Clinic Würzburg, Würzburg, Germany
| | - Esther Lutgens
- Department of Cardiovascular Medicine and Immunology, Mayo Clinic, Rochester, MN, USA
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Essen, Germany
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Javid Moslehi
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA, USA
| | - Gilles De Keulenaer
- Department of Pharmaceutical Sciences, University of Antwerp, Antwerp, Belgium
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, Genoa, Italy
- IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Jeroen Bax
- Department of Cardiology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joerg Herrmann
- Cardio-Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Amam C Mbakwem
- College of Medicine, University of Lagos/Lagos University Teaching Hospital Idi Araba, Lagos, Nigeria
| | | | - Hadi Skouri
- Cardiology Division, Sheikh Shakhbout Medical City, Khalifa University, Abu Dhabi, UAE
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Turin, Turin, Italy
| | - Alain Cohen-Solal
- Paris Cité University INSERM U 948 MASCOT Research Unit Cardiology, Lariboisere Universitaire Hospital, AP-HP, Paris, France
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology, University of Newcastle, Hunter Medical Research Institute, Calvary Mater Newcastle, Hunter New England Health, Newcastle, NSW, Australia
- Cardiovascular Department, John Hunter Hospital, New Lambton Heights, NSW, Australia
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Riccardo Asteggiano
- Internal Medicine, Department of Medicine and Surgery, University of Insubria, Varese, Italy
- LARC, Laboratorio Analisi e Ricerca Clinica, Turin, Italy
| | - Ana Barac
- Inova Schar Heart and Vascular Institute, Falls Church, VA, USA
| | - Bonnie Ky
- Division of Cardiology, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Daniel Lenihan
- Cape Cardiology, St Francis Healthcare, Cape Girardeau, MO, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC, USA
| | - Petar Seferovic
- University Medical Center, Medical Faculty University of Belgrade, Serbian Academy of Sciences and Arts, Belgrade, Serbia
| | | | - Marco Metra
- Cardiology, ASST Spedali Civili, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Giuseppe Rosano
- Department of Human Sciences and Promotion of Quality of Life, San Raffaele Open University of Rome, Rome, Italy
- Cardiology, San Raffaele Cassino Hospital, Cassino, FR, Italy
| | - Thomas Suter
- Bern University Hospital, University of Bern, Bern, Switzerland
| | - Teresa Lopez-Fernandez
- Cardio-Oncology Unit, Cardiology Department, IdiPAZ Research Institute, La Paz University Hospital, Madrid, Spain
- Cardiology Department, Quironsalud Madrid University Hospital, Madrid, Spain
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| |
Collapse
|
10
|
Gergely TG, Drobni ZD, Kallikourdis M, Zhu H, Meijers WC, Neilan TG, Rassaf T, Ferdinandy P, Varga ZV. Immune checkpoints in cardiac physiology and pathology: therapeutic targets for heart failure. Nat Rev Cardiol 2024; 21:443-462. [PMID: 38279046 DOI: 10.1038/s41569-023-00986-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/12/2023] [Indexed: 01/28/2024]
Abstract
Immune checkpoint molecules are physiological regulators of the adaptive immune response. Immune checkpoint inhibitors (ICIs), such as monoclonal antibodies targeting programmed cell death protein 1 or cytotoxic T lymphocyte-associated protein 4, have revolutionized cancer treatment and their clinical use is increasing. However, ICIs can cause various immune-related adverse events, including acute and chronic cardiotoxicity. Of these cardiovascular complications, ICI-induced acute fulminant myocarditis is the most studied, although emerging clinical and preclinical data are uncovering the importance of other ICI-related chronic cardiovascular complications, such as accelerated atherosclerosis and non-myocarditis-related heart failure. These complications could be more difficult to diagnose, given that they might only be present alongside other comorbidities. The occurrence of these complications suggests a potential role of immune checkpoint molecules in maintaining cardiovascular homeostasis, and disruption of physiological immune checkpoint signalling might thus lead to cardiac pathologies, including heart failure. Although inflammation is a long-known contributor to the development of heart failure, the therapeutic targeting of pro-inflammatory pathways has not been successful thus far. The increasingly recognized role of immune checkpoint molecules in the failing heart highlights their potential use as immunotherapeutic targets for heart failure. In this Review, we summarize the available data on ICI-induced cardiac dysfunction and heart failure, and discuss how immune checkpoint signalling is altered in the failing heart. Furthermore, we describe how pharmacological targeting of immune checkpoints could be used to treat heart failure.
Collapse
Affiliation(s)
- Tamás G Gergely
- 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
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Marinos Kallikourdis
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
- Adaptive Immunity Lab, Humanitas Research Hospital IRCCS, Milan, Italy
| | - Han Zhu
- Division of Cardiovascular Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Wouter C Meijers
- Erasmus MC, Cardiovascular Institute, Thorax Center, Department of Cardiology, Rotterdam, The Netherlands
| | - Tomas G Neilan
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- 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
|
11
|
Sharma A, Alexander G, Chu JH, Markopoulos A, Maloul G, Ayub MT, Fidler MJ, Okwuosa TM. Immune Checkpoint Inhibitors and Cardiotoxicity: A Comparative Meta-Analysis of Observational Studies and Randomized Controlled Trials. J Am Heart Assoc 2024; 13:e032620. [PMID: 38761070 PMCID: PMC11179795 DOI: 10.1161/jaha.123.032620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 02/20/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have uncommon associations with cardiotoxicity, yet these cardiotoxic effects are associated with high mortality. An accurate assessment of risk for cardiotoxicity is essential for clinical decision-making, but data from randomized controlled trials often differ from real-world observational studies. METHODS AND RESULTS A systematic search of PubMed, Embase, Cochrane Library, and Scopus was performed, including phase II and III randomized controlled trials (RCTs) and observational studies (OSs) reporting myocarditis or pericardial disease, myocardial infarction, or stroke with an immunotherapy. Odds ratios (ORs) were used to pool results between ICIs and other cancer therapy in RCTs and OSs. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guideline was followed. In total, 54 RCTs (N=38 264) and 24 OSs (N=12 561 455) were included. In RCTs, ICI use resulted in higher risk of myocarditis (OR, 3.55 [95% CI, 2.10-5.98]), pericardial disease (OR, 2.73 [95% CI, 1.57-4.77]), and myocardial infarction (OR, 1.83 [95% CI, 1.03-3.25]), compared with non-ICI (placebo or chemotherapy). In OSs, ICI use was not associated with myocarditis, pericardial disease, or myocardial infarction compared with controls; however, combination ICIs demonstrated higher risk of myocarditis compared with single ICI use (OR, 3.07 [95% CI, 1.28-7.39]). Stroke risk was not increased with use of ICIs in RCTs. CONCLUSIONS We demonstrated increased risk of ICI myocarditis, pericardial disease, and myocardial infarction in RCTs but not OSs. Results of this study suggest there are differences between ICI cardiotoxicity risk, possibly suggesting differences in diagnoses and management, in clinical trials versus the OSs.
Collapse
Affiliation(s)
- Akash Sharma
- Department of Medicine University at Buffalo-Catholic Health System Buffalo NY
- Center for Global Health Research Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai India
| | - Grace Alexander
- Department of Internal Medicine University of Iowa Hospitals & Clinics Iowa City IA
| | - Jian H Chu
- Division of Cardiology, Department of Medicine University of Oklahoma Oklahoma City OK
| | | | | | - Muhammad Talha Ayub
- Heart and Vascular Institute, University of Pittsburgh Medical Center Pittsburgh PA
| | - Mary J Fidler
- Division of Hematology/Oncology/Stem cell transplant Rush University Medical Center Chicago IL
| | - Tochukwu M Okwuosa
- Division of Cardiology, Department of Internal Medicine Rush University Medical Center Chicago IL
| |
Collapse
|
12
|
Kong Y, Wang X, Qie R. Immunotherapy-associated cardiovascular toxicities: insights from preclinical and clinical studies. Front Oncol 2024; 14:1347140. [PMID: 38482205 PMCID: PMC10932998 DOI: 10.3389/fonc.2024.1347140] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 02/12/2024] [Indexed: 11/02/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have become a widely accepted and effective treatment for various types of solid tumors. Recent studies suggest that cardiovascular immune-related adverse events (irAEs) specifically have an incidence rate ranging from 1.14% to more than 5%. Myocarditis is the most common observed cardiovascular irAE. Others include arrhythmias, pericardial diseases, vasculitis, and a condition resembling takotsubo cardiomyopathy. Programmed cell death-1 (PD-1)/programmed cell death ligand-1 (PD-L1) pathway, cytotoxic T-lymphocyte antigen-4 (CTLA-4) pathway, and the recently discovered lymphocyte-activation gene 3 (LAG-3) pathway, play a critical role in boosting the body's natural immune response against cancer cells. While ICIs offer significant benefits in terms of augmenting immune function, they can also give rise to unwanted inflammatory side effects known as irAEs. The occurrence of irAEs can vary in severity, ranging from mild to severe, and can impact the overall clinical efficacy of these agents. This review aims to summarize the underlying mechanisms of cardiovascular irAE from both preclinical and clinical studies for a better understanding of cardiovascular irAE in clinical application.
Collapse
Affiliation(s)
- Youqian Kong
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xiaoyu Wang
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Rui Qie
- First Affiliated Hospital, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
13
|
de Jesus M, Chanda A, Grabauskas T, Kumar M, Kim AS. Cardiovascular disease and lung cancer. Front Oncol 2024; 14:1258991. [PMID: 38410099 PMCID: PMC10896114 DOI: 10.3389/fonc.2024.1258991] [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: 07/14/2023] [Accepted: 01/19/2024] [Indexed: 02/28/2024] Open
Abstract
Lung cancer is the second most common cancer worldwide and the leading cause of cancer-related death. While survival rates have improved with advancements in cancer therapeutics, additional health challenges have surfaced. Cardiovascular disease (CVD) is a leading cause of morbidity and mortality in patients with lung cancer. CVD and lung cancer share many risk factors, such as smoking, hypertension, diabetes, advanced age, and obesity. Optimal management of this patient population requires a full understanding of the potential cardiovascular (CV) complications of lung cancer treatment. This review outlines the common shared risk factors, the spectrum of cardiotoxicities associated with lung cancer therapeutics, and prevention and management of short- and long-term CVD in patients with non-small cell (NSCLC) and small cell (SCLC) lung cancer. Due to the medical complexity of these patients, multidisciplinary collaborative care among oncologists, cardiologists, primary care physicians, and other providers is essential.
Collapse
Affiliation(s)
- Mikhail de Jesus
- Department of Cardiology, University of Connecticut Hartford Hospital, Hartford, CT, United States
| | - Anindita Chanda
- Department of Internal Medicine, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Titas Grabauskas
- University of Connecticut School of Medicine, Farmington, CT, United States
| | - Manish Kumar
- Department of Cardiology, Pat & Jim Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Agnes S Kim
- Department of Cardiology, Pat & Jim Calhoun Cardiology Center, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
14
|
Inno A, Veccia A, Madonia G, Berti A, Bortolotti R, Incorvaia L, Russo A, Caffo O, Gori S. Risk of cardiovascular toxicity with combination of immune-checkpoint inhibitors and angiogenesis inhibitors: a meta-analysis. Front Cardiovasc Med 2024; 11:1309100. [PMID: 38370161 PMCID: PMC10869562 DOI: 10.3389/fcvm.2024.1309100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 01/22/2024] [Indexed: 02/20/2024] Open
Abstract
Introduction Combinations of immune checkpoint inhibitors (ICIs) and angiogenesis inhibitors (AIs) have been investigated for the treatment of several tumor types. Both ICIs and AIs may lead to cardiovascular adverse events, and their combination may potentially increase the risk for cardiovascular toxicity. In the present meta-analysis, we aim to assess the cardiovascular toxicity of ICIs plus AIs vs. AIs alone. Secondary objectives are non-cardiovascular adverse events and efficacy. Methods Systematic review was performed according to PRISMA statement. Phase II and III randomized clinical trials were identified by searching the MEDLINE/PubMed, Cochrane Library and ASCO Meeting abstracts, from inception to June 2022. The pooled risks for overall response rate (ORR), 1-year progression-free survival (PFS), adverse events (AEs), immune-related AEs, (irAEs), hypertension, and vascular events defined as stroke, myocardial infarction and pulmonary embolisms, were calculated. Results In terms of cardiovascular toxicity, we found higher risk for severe hypertension among patients treated with ICIs plus AIs as compared with those receiving AIs (OR 1.24, 95% CI: 1.01-1.53), but no significant difference was found for any-grade hypertension, and for vascular events. There was also no difference in terms of overall AEs, whereas the incidence of irAEs was increased in the ICIs plus AIs arm, as expected. In terms of efficacy, ICIs plus AIs achieved better ORR (OR 2.25, 95% CI: 1.70-2.97) and PFS (HR 0.49, 95% CI: 0.39-0.63) as compared to AIs alone. Conclusion The addition of ICIs to AIs significantly increased the risk of high-grade hypertension, but not that of acute vascular events.
Collapse
Affiliation(s)
- Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| | | | - Giorgio Madonia
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
- Deparment of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Alvise Berti
- Center for Medical Sciences (CISMed), Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
- Rheumatology Unit, Santa Chiara Hospital, APSS, Trento, Italy
| | | | - Lorena Incorvaia
- Deparment of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Antonio Russo
- Deparment of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Orazio Caffo
- Medical Oncology, Santa Chiara Hospital, Trento, Italy
| | - Stefania Gori
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, Negrar di Valpolicella, Italy
| |
Collapse
|
15
|
Takeuchi T, Kosugi S, Ueda Y, Ikeoka K, Yamane H, Takayasu K, Ohashi T, Fukushima T, Horiuchi K, Iehara T, Sakamoto M, Ukai K, Minami S, Mizumori Y, Muraoka N, Nakamura M, Ozaki T, Mishima T, Abe H, Inoue K, Matsumura Y. Impact of a Cancer History on Cardiovascular Events Among Patients With Myocardial Infarction Who Received Revascularization. Circ J 2024; 88:207-214. [PMID: 37045768 DOI: 10.1253/circj.cj-22-0838] [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: 04/14/2023]
Abstract
BACKGROUND It remains controversial whether a cancer history increases the risk of cardiovascular (CV) events among patients with myocardial infarction (MI) who undergo revascularization. METHODS AND RESULTS Patients who were confirmed as type 1 acute MI (AMI) by coronary angiography were retrospectively analyzed. Patients who died in hospital or those not undergoing revascularization were excluded. Patients with a cancer history were compared with those without it. A cancer history was examined in the in-hospital cancer registry. The primary outcome was a composite of cardiac death, recurrent type 1 MI, post-discharge coronary revascularization, heart failure hospitalization, and stroke. Among 551 AMI patients, 55 had a cancer history (cancer group) and 496 did not (non-cancer group). Cox proportional hazards model revealed that the risk of composite endpoint was significantly higher in the cancer group than in the non-cancer group (adjusted hazard ratio [HR]: 1.78; 95% confidence interval [CI]: 1.13-2.82). Among the cancer group, patients who were diagnosed as AMI within 6 months after the cancer diagnosis had a higher risk of the composite endpoint than those who were diagnosed as AMI 6 months or later after the cancer diagnosis (adjusted HR: 5.43; 95% CI: 1.55-19.07). CONCLUSIONS A cancer history increased the risk of CV events after discharge among AMI patients after revascularization.
Collapse
Affiliation(s)
- Taro Takeuchi
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Shumpei Kosugi
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Yasunori Ueda
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Kuniyasu Ikeoka
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Haruya Yamane
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Kohtaro Takayasu
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Takuya Ohashi
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Takashi Fukushima
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Kohei Horiuchi
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Takashi Iehara
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Mai Sakamoto
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Kazuho Ukai
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Shinya Minami
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Yuuki Mizumori
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Naoya Muraoka
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Masayuki Nakamura
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Tatsuhisa Ozaki
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Tsuyoshi Mishima
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Haruhiko Abe
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Koichi Inoue
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| | - Yasushi Matsumura
- Cardiovascular Division, National Hospital Organization Osaka National Hospital
| |
Collapse
|
16
|
Chang CH, Huang SH, Huang HY, Lin MH, Lee CS, Lee HF, Hsieh JCH, Cheng CY. Major adverse cardiovascular events in advanced-stage lung cancer: a multicenter cohort study. Ther Adv Med Oncol 2024; 16:17588359231221907. [PMID: 38249337 PMCID: PMC10798069 DOI: 10.1177/17588359231221907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Accepted: 12/04/2023] [Indexed: 01/23/2024] Open
Abstract
Background Lung cancers are common worldwide. First-line targeted therapy and chemotherapy are both standard treatments in the current guidelines. With the development of new anticancer therapy, the lifespan of patients with late-stage lung cancer has increased. Cardiovascular events can occur during cancer treatment. This observational study aimed to report the incidence of major adverse cardiovascular events (MACE) after cancer treatment using real-world data. Objectives Patients diagnosed with advanced-stage lung cancer between January 2011 and December 2017 were enrolled. Data were collected from the Chang Gung Research Database (CGRD). Design Retrospective cohort study. Methods Baseline characteristics, clinical stages, pathologies, and outcomes were retrieved from the CGRD. Results We identified 4406 patients with advanced lung cancer, of whom 2197 received first-line epidermal growth factor receptor tyrosine kinase inhibitor (EGFR-TKI) therapy and 2209 received first-line platinum-based chemotherapy. Most patients in the first-line EGFR-TKI group were never-smokers (74.9%), whereas those in the first-line chemotherapy group were ever-smokers (66.0%). The incidence of MACE was not significantly different between the two groups (12.0% versus 11.9%, p = 0.910). However, the incidence of ischemic stroke was higher in the first-line EGFR-TKI group than in the first-line chemotherapy group (3.9% versus 1.9%, p < 0.001). Conclusion MACEs are common in patients with advanced-stage lung cancer during treatment. The incidence of MACE was similar between the first-line EGFR-TKI therapy and first-line chemotherapy groups. Although more patients in the EGFR-TKI group were female and never-smokers, the risk of ischemic stroke was higher in patients who received first-line EGFR-TKI therapy than in those who received first-line chemotherapy.
Collapse
Affiliation(s)
- Chih-Hao Chang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, New Taipei Municipal Tucheng Hospital, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital Linkou Branch, 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan
| | - Shih-Hao Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, New Taipei Municipal Tucheng Hospital, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital Linkou Branch, Taoyuan City, Taiwan
| | - Hung-Yu Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, New Taipei Municipal Tucheng Hospital, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital Linkou Branch, Taoyuan City, Taiwan
| | - Meng-Hung Lin
- Health Information and Epidemiology Laboratory, Chang Gung Memorial Hospital Chiayi Branch, Chiayi County, Taiwan
| | - Chung-Shu Lee
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, New Taipei Municipal Tucheng Hospital, New Taipei City, Taiwan
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Department of Thoracic Medicine, Chang Gung Memorial Hospital Linkou Branch, Taoyuan City, Taiwan
| | - Hsin-Fu Lee
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Division of Cardiology, Department of Internal Medicine, New Taipei City Municipal Tucheng Hospital, New Taipei City, Taiwan
- The Cardiovascular Department, Chang Gung Memorial Hospital Linkou Branch, Taoyuan City, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan City, Taiwan
| | - Jason Chia-Hsun Hsieh
- College of Medicine, Chang Gung University, Taoyuan City, Taiwan
- Division of Hematology-Oncology, Department of Internal Medicine, New Taipei City Municipal Tucheng Hospital, New Taipei City, Taiwan
| | - Chun-Yu Cheng
- Department of Neurosurgery, Chang Gung Memorial Hospital Chiayi Branch, No. 8, Sec. W., Jiapu Rd., Puzi City, Chiayi County 613 , Taiwan
- Department of Biomedical Sciences and Institute of Molecular Biology, National Chung Cheng University, Chiayi County, Taiwan
| |
Collapse
|
17
|
Chan A, Torelli S, Cheng E, Batchelder R, Waliany S, Neal J, Witteles R, Nguyen P, Cheng P, Zhu H. Immunotherapy-Associated Atherosclerosis: A Comprehensive Review of Recent Findings and Implications for Future Research. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2023; 25:715-735. [PMID: 38213548 PMCID: PMC10776491 DOI: 10.1007/s11936-023-01024-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/27/2023] [Indexed: 01/13/2024]
Abstract
Purpose of the Review Even as immune checkpoint inhibitors (ICIs) have transformed the lifespan of many patients, they may also trigger acceleration of long-term cardiovascular disease. Our review aims to examine the current landscape of research on ICI-mediated atherosclerosis and address key questions regarding its pathogenesis and impact on patient management. Recent Findings Preclinical mouse models suggest that T cell dysregulation and proatherogenic cytokine production are key contributors to plaque development after checkpoint inhibition. Clinical data also highlight the significant burden of atherosclerotic cardiovascular disease (ASCVD) in patients on immunotherapy, although the value of proactively preventing and treating ASCVD in this population remains an open area of inquiry. Current treatment options include dietary/lifestyle modification and traditional medications to manage hypertension, hyperlipidemia, and diabetes risk factors; no current targeted therapies exist. Summary Early identification of high-risk patients is crucial for effective preventive strategies and timely intervention. Future research should focus on refining screening tools, elucidating targetable mechanisms driving ICI atherosclerosis, and evaluating long-term cardiovascular outcomes in cancer survivors who received immunotherapy. Moreover, close collaboration between oncologists and cardiologists is essential to optimize patient outcomes.
Collapse
Affiliation(s)
- Antonia Chan
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Stefan Torelli
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Evaline Cheng
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Ryan Batchelder
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Sarah Waliany
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Joel Neal
- Department of Medicine, Division of Oncology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA USA
| | - Ronald Witteles
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Paul Cheng
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| | - Han Zhu
- Department of Medicine, Stanford University School of Medicine, Stanford, CA USA
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA USA
- Stanford Cardiovascular Institute and Department of Medicine, Stanford University, 240 Pasteur Drive, Rm 3500, Biomedical Innovations Building, Stanford, CA 94304 USA
| |
Collapse
|
18
|
Zatarain-Nicolás E, Martín P, Márquez Rodas I, Virizuela J, Martín García A, Mitroi C, Cosín Sales J, Barrios V, Sánchez-Cabo F, Ibañez B, de Castro Carpeño J, López Fernández T. Cardiovascular toxicity of checkpoint inhibitors: review of associated toxicity and design of the Spanish Immunotherapy Registry of Cardiovascular Toxicity. Clin Transl Oncol 2023; 25:3073-3085. [PMID: 37227656 DOI: 10.1007/s12094-023-03217-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Accepted: 04/29/2023] [Indexed: 05/26/2023]
Abstract
Immune checkpoint inhibitors (ICI) have changed the prognosis of many tumors. However, concerning associated cardiotoxicity has been reported. Little is known about the real-life incidence-specific surveillance protocols or the translational correlation between the underlying mechanisms and the clinical presentation of ICI-induced cardiotoxicity. The lack of data from prospective studies led us to review the current knowledge and to present the creation of the Spanish Immunotherapy Registry of Cardiovascular Toxicity (SIR-CVT), a prospective registry of patients receiving ICI that aims to examine the role of hsa-miR-Chr8:96, (a specific serum biomarker of myocarditis) in the early diagnosis of ICI-induced myocarditis. An exhaustive prospective cardiac imaging study will be performed before and during the first 12 months of treatment. The correlation between clinical, imaging, and immunologic parameters may improve our understanding of ICI-induced cardiotoxicity and enable simpler surveillance protocols. We assess ICI-induced cardiovascular toxicity and describe the rationale of the SIR-CVT.
Collapse
Affiliation(s)
- Eduardo Zatarain-Nicolás
- Cardiology Department, Hospital General Universitario Gregorio Marañón, CIBER-CV (Instituto de Salud Carlos III), Universidad Complutense, Madrid, Spain.
| | - Pilar Martín
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBER-CV (Instituto de Salud Carlos III), Madrid, Spain
| | - Iván Márquez Rodas
- Medical Oncology Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Juan Virizuela
- Medical Oncology Department, Hospital Universitario Virgen de la Macarena, Seville, Spain
| | - Ana Martín García
- Cardiology Department, Complejo Asistencial Universitario de Salamanca, IBSAL, USAL, CIBER-CV (Instituto de Salud Carlos III), Salamanca, Spain
| | - Cristina Mitroi
- Cardiology Department, Hospital Universitario Puerta de Hierro Majadahonda, IDIPHISA, CIBER-CV (Instituto de Salud Carlos III), Madrid, Spain
| | - Juan Cosín Sales
- Agencia de Investigación de la Sociedad Española de Cardiología (AISEC), Madrid, Spain
- Cardiology Department, Hospital Arnau de Vilanova, Universidad CEU-Cardenal Herrera, Valencia, Spain
| | - Vivencio Barrios
- Agencia de Investigación de la Sociedad Española de Cardiología (AISEC), Madrid, Spain
- Cardiology Department, Hospital Universitario Ramon y Cajal, Universidad de Alcalá, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Bioinformatics Unit, Centro Nacional de Investigaciones Cardiovasculares (CNIC), CIBER-CV (Instituto de Salud Carlos III), Madrid, Spain
| | - Borja Ibañez
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, CIBER-CV (Instituto de Salud Carlos III), CNIC, Madrid, Spain
| | | | - Teresa López Fernández
- Cardiology Service, Cardio-Oncology Unit, La Paz University Hospital and IdiPAz Research Institute, Madrid, Spain.
| |
Collapse
|
19
|
Patel M, Hudson O, Han J, Kondapalli L, Arora G, Hawi R, Andrikopoulou E, Estes C, Johnson AM, Lenneman C. Update on Immunotherapy Cardiotoxicity: Checkpoint Inhibitors, CAR T, and Beyond. Curr Treat Options Oncol 2023; 24:1489-1503. [PMID: 37624557 DOI: 10.1007/s11864-023-01130-y] [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: 07/18/2023] [Indexed: 08/26/2023]
Abstract
OPINION STATEMENT Immunotherapy is an innovative approach to cancer treatment that involves using the body's immune system to fight cancer. The landscape of immunotherapy is constantly evolving, as new therapies are developed and refined. Some of the most promising approaches in immunotherapy include immune checkpoint inhibitors (ICIs): these drugs target proteins on the surface of T-cells that inhibit their ability to attack cancer cells. By blocking these proteins, checkpoint inhibitors allow T-cells to recognize and destroy cancer cells more effectively. CAR T-cell therapy: this therapy involves genetically modifying a patient's own T-cells to recognize and attack cancer cells. CAR T-cell therapy exhibits favorable response in many patients with refractory hematological cancers with growing clinical trials in solid tumors. Immune system modulators: these drugs enhance the immune system's ability to fight cancer by stimulating the production of immune cells or inhibiting the activity of immune-suppressing cells. While immunotherapy has shown great promise in the treatment of cancer, it can also pose significant cardiac side effects. Some immunotherapy drugs like ICIs can cause myocarditis, which can lead to chest pain, shortness of breath, and heart failure. Other cardiac side effects of ICIs include arrhythmias, pericarditis, vasculitis, and accelerated atherosclerosis. It is important for patients receiving immunotherapy to be monitored closely for these side effects, as prompt treatment can help prevent serious complications. Patients should also report any symptoms to their healthcare providers right away, so that appropriate action can be taken. CAR T-cell therapy can also illicit an exaggerated immune response creating cytokine release syndrome (CRS) that may precipitate cardiovascular events: arrhythmias, myocardial infarction, and heart failure. Overall, while immune modulating therapy is a promising and expanding approach to cancer treatment, it is important to weigh the potential benefits against the risks and side effects, especially in patients with high risk for cardiovascular complications.
Collapse
Affiliation(s)
- Murti Patel
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Olivia Hudson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Jingnan Han
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Lavanya Kondapalli
- University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Garima Arora
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Riem Hawi
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | | | - Courtney Estes
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Abigail M Johnson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Carrie Lenneman
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
20
|
Bisceglia I, Canale ML, Silvestris N, Gallucci G, Camerini A, Inno A, Camilli M, Turazza FM, Russo G, Paccone A, Mistrulli R, De Luca L, Di Fusco SA, Tarantini L, Lucà F, Oliva S, Moreo A, Maurea N, Quagliariello V, Ricciardi GR, Lestuzzi C, Fiscella D, Parrini I, Racanelli V, Russo A, Incorvaia L, Calabrò F, Curigliano G, Cinieri S, Gulizia MM, Gabrielli D, Oliva F, Colivicchi F. Cancer survivorship at heart: a multidisciplinary cardio-oncology roadmap for healthcare professionals. Front Cardiovasc Med 2023; 10:1223660. [PMID: 37786510 PMCID: PMC10541962 DOI: 10.3389/fcvm.2023.1223660] [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: 05/16/2023] [Accepted: 08/24/2023] [Indexed: 10/04/2023] Open
Abstract
In cancer, a patient is considered a survivor from the time of initial diagnosis until the end of life. With improvements in early diagnosis and treatment, the number of cancer survivors (CS) has grown considerably and includes: (1) Patients cured and free from cancer who may be at risk of late-onset cancer therapy-related cardiovascular toxicity (CTR-CVT); (2) Patients with long-term control of not-curable cancers in whom CTR-CVT may need to be addressed. This paper highlights the importance of the cancer care continuum, of a patient-centered approach and of a prevention-oriented policy. The ultimate goal is a personalized care of CS, achievable only through a multidisciplinary-guided survivorship care plan, one that replaces the fragmented management of current healthcare systems. Collaboration between oncologists and cardiologists is the pillar of a framework in which primary care providers and other specialists must be engaged and in which familial, social and environmental factors are also taken into account.
Collapse
Affiliation(s)
- Irma Bisceglia
- Integrated Cardiology Services, Cardio-Thoracic-Vascular Department, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - Maria Laura Canale
- Division of Cardiology, Ospedale Versilia, Azienda Usl Toscana Nord Ovest, Lido di Camaiore, Italy
| | - Nicola Silvestris
- Unit of Medical Oncology, Department of Human Pathology in Adulthood and Childhood Gaetano Barresi, University of Messina, Messina, Italy
| | - Giuseppina Gallucci
- Cardio-oncology Unit, Department of OncoHaematology, IRCCS Referral Cancer Center of Basilicata, Rionero in Vulture (PZ), Italy
| | - Andrea Camerini
- Department of Medical Oncology, Ospedale Versilia, Azienda Usl Toscana Nord Ovest, Lido di Camaiore, Italy
| | - Alessandro Inno
- Department of Oncology, Sacro Cuore Don Calabria Hospital (IRCCS), Negrar, Italy
| | - Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart and Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Fabio Maria Turazza
- Cardiology Department, National Cancer Institute Foundation (IRCCS), Milan, Italy
| | - Giulia Russo
- SC Patologie Cardiovascolari, Azienda Sanitaria Universitaria Giuliano Isontina (ASUGI), Trieste, Italy
| | - Andrea Paccone
- Department of Cardiology, G. Pascale National Cancer Institute Foundation (IRCCS), Naples, Italy
| | - Raffaella Mistrulli
- Cardiology Unit, Department of Clinical and Molecular Medicine, Faculty of Medicine and Psychology, Sapienza University of Rome, Rome, Lazio, Italy
| | - Leonardo De Luca
- Division of Cardiology, San Camillo-Forlanini Hospital, Rome, Italy
| | | | - Luigi Tarantini
- Divisione di Cardiologia, Arcispedale S. Maria Nuova, Azienda Unità Sanitaria Locale-IRCCS di Reggio-Emilia, Reggio Emilia, Italy
| | - Fabiana Lucà
- Cardiologia Interventistica, Utic, Grande Ospedale Metropolitano, Azienda Ospedaliera Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Stefano Oliva
- UOSD Cardiologia di Interesse Oncologico, IRCCS Istituto Tumori "Giovanni Paolo II", Bari, Italy
| | - Antonella Moreo
- Cardio Center De Gasperis, Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Nicola Maurea
- Department of Cardiology, G. Pascale National Cancer Institute Foundation (IRCCS), Naples, Italy
| | - Vincenzo Quagliariello
- Department of Cardiology, G. Pascale National Cancer Institute Foundation (IRCCS), Naples, Italy
| | | | | | - Damiana Fiscella
- U.O.C. Cardiologia, Ospedale Garibaldi-Nesima, Azienda di Rilievo Nazionale e Alta Specializzazione “Garibaldi”, Catania, Italy
| | - Iris Parrini
- Department of Cardiology, Hospital Mauritian Turin, Turin, Italy
| | - Vito Racanelli
- Department of Interdisciplinary Medicine, School of Medicine, University of Bari Aldo Moro, Bari, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, Palermo University Hospital, Palermo, Italy
| | - Lorena Incorvaia
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, Palermo University Hospital, Palermo, Italy
| | - Fabio Calabrò
- Department of Oncology and Specialized Medicine, San Camillo-Forlanini Hospital, Rome, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan; Division of Early Drug Development, Istituto Europeo di Oncologia, IRCCS, Milan, Italy
| | - Saverio Cinieri
- Medical Oncology Division and Breast Unit, Senatore Antonio Perrino Hospital, ASL Brindisi, Brindisi, Italy
| | - Michele Massimo Gulizia
- U.O.C. Cardiologia, Ospedale Garibaldi-Nesima, Azienda di Rilievo Nazionale e Alta Specializzazione “Garibaldi”, Catania, Italy
| | - Domenico Gabrielli
- Division of Cardiology, San Camillo-Forlanini Hospital, Rome, Italy
- Fondazione per il Tuo cuore- Heart Care Foundation, Firenze, Italy
| | - Fabrizio Oliva
- Cardiologia 1- Emodinamica, Dipartimento Cardiotoracovascolare “A. De Gasperis”, Azienda Socio Sanitaria Territoriale Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Furio Colivicchi
- Clinical and Rehabilitation Cardiology Unit, San Filippo Neri Hospital, ASL Roma 1, Rome, Italy
| |
Collapse
|
21
|
Tamura Y, Tamura Y. Usefulness of Longitudinal Strain to Assess Cancer Therapy-Related Cardiac Dysfunction and Immune Checkpoint Inhibitor-Induced Myocarditis. Pharmaceuticals (Basel) 2023; 16:1297. [PMID: 37765105 PMCID: PMC10535915 DOI: 10.3390/ph16091297] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Revised: 09/08/2023] [Accepted: 09/12/2023] [Indexed: 09/29/2023] Open
Abstract
Longitudinal strain (LS) measured by echocardiography has been reported to be useful not only for the diagnosis and risk stratification of various cardiac diseases, but also in cardio-oncology. Most previous studies have been conducted on patients undergoing treatment with anthracyclines and human epidermal growth factor receptor 2-targeted therapies. Existing guidelines recommend that global LS (GLS) should be measured before and after the administration of cancer drugs. This recommendation is based on many reports showing that a decline in GLS is indicative of early or mild cancer therapy-related cardiac dysfunction. The main purpose of this article is to provide insight into the importance of LS in patients undergoing cancer treatment and highlight the role of LS evaluation in patients undergoing immune checkpoint inhibitor (ICI) treatment, which is being used with increasing frequency. Among cancer drug therapies, immune checkpoint inhibitors (ICIs) have an important place in cancer treatment and are used for the treatment of many types of cancer. Although the efficacy of ICIs in cancer treatment has been reported, immune-related adverse events (irAEs) have also been reported. Among these irAEs, cardiovascular complications, although rare, are recognized as important adverse events that may result in ICI treatment discontinuation. Myocarditis is one severe adverse event associated with ICIs, and it is important to standardize diagnostic and therapeutic approaches to it. Several studies have reported a relationship between LS and cardiac complications associated with ICIs which may contribute to the early diagnosis of ICI-induced cardiac complications.
Collapse
Affiliation(s)
- Yudai Tamura
- Cardiovascular Center, International University of Health and Welfare Mita Hospital, Tokyo 108-8329, Japan;
- Department of Cardiology, International University of Health and Welfare School of Medicine, Narita 286-8686, Japan
| | - Yuichi Tamura
- Cardiovascular Center, International University of Health and Welfare Mita Hospital, Tokyo 108-8329, Japan;
- Department of Cardiology, International University of Health and Welfare School of Medicine, Narita 286-8686, Japan
| |
Collapse
|
22
|
Ellison JM, Nohria A. An Increased Understanding of the Association Between Atherosclerosis and Immune Checkpoint Inhibitors. Curr Cardiol Rep 2023; 25:879-887. [PMID: 37395892 DOI: 10.1007/s11886-023-01908-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/14/2023] [Indexed: 07/04/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitor (ICI) therapy activates the host immune system to promote tumor cell death. This activation of the immune system can lead to off-target immune-related adverse events (irAEs). There is an established link between inflammation and atherosclerosis. The purpose of this manuscript is to review the existing body of literature discussing the potential association between ICI treatment and atherosclerosis. RECENT FINDINGS Pre-clinical studies suggest that ICI therapy may lead to T-cell-mediated progression of atherosclerosis. Recent retrospective clinical studies have shown higher rates of myocardial infarction and stroke with ICI therapy, particularly in patients with pre-existing cardiovascular risk factors. Additionally, small observational cohort studies have used imaging modalities to demonstrate higher rates of atherosclerotic progression with ICI treatment. Early pre-clinical and clinical evidence suggests an association between ICI treatment and the progression of atherosclerosis. However, these findings are preliminary, and adequately powered prospective studies are needed to demonstrate a conclusive association. As ICI therapy is increasingly used to treat a variety of solid tumors, it is important to evaluate and mitigate the potential adverse atherosclerotic effects of ICI treatment.
Collapse
Affiliation(s)
- Judah M Ellison
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, 02115, USA
| | - Anju Nohria
- Cardiovascular Division, Brigham and Women's Hospital, Boston, MA, 02115, USA.
| |
Collapse
|
23
|
Wang Y, Cui C, Deng L, Wang L, Ren X. Cardiovascular toxicity profiles of immune checkpoint inhibitors with or without angiogenesis inhibitors: a real-world pharmacovigilance analysis based on the FAERS database from 2014 to 2022. Front Immunol 2023; 14:1127128. [PMID: 37292205 PMCID: PMC10244526 DOI: 10.3389/fimmu.2023.1127128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 05/15/2023] [Indexed: 06/10/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) combined with angiogenesis inhibitors (AGIs) have become increasingly available for multiple types of cancers, although the cardiovascular safety profiles of this combination therapy in real-world settings have not been elucidated to date. Therefore, we aimed to comprehensively investigate the cardiovascular toxicity profiles of ICIs combined with AGIs in comparison with ICIs alone. Methods The Food and Drug Administration Adverse Event Reporting System (FAERS) database from the 1st quarter of 2014 to the 1st quarter of 2022 was retrospectively queried to extract reports of cardiovascular adverse events (AEs) associated with ICIs alone, AGIs alone and combination therapy. To perform disproportionality analysis, the reporting odds ratios (RORs) and information components (ICs) were calculated with statistical shrinkage transformation formulas and a lower limit of the 95% confidence interval (CI) for ROR (ROR025) > 1 or IC (IC025) > 0 with at least 3 reports was considered statistically significant. Results A total of 18 854 cardiovascular AE cases/26 059 reports for ICIs alone, 47 168 cases/67 595 reports for AGIs alone, and 3 978 cases/5 263 reports for combination therapy were extracted. Compared to the entire database of patients without AGIs or ICIs, cardiovascular AEs were overreported in patients with combination therapy (IC025/ROR025 = 0.559/1.478), showing stronger signal strength than those taking ICIs alone (IC025/ROR025 = 0.118/1.086) or AGIs alone (IC025/ROR025 = 0.323/1.252). Importantly, compared with ICIs alone, combination therapy showed a decrease in signal strength for noninfectious myocarditis/pericarditis (IC025/ROR025 = 1.142/2.216 vs. IC025/ROR025 = 0.673/1.614), while an increase in signal value for embolic and thrombotic events (IC025/ROR025 = 0.147/1.111 vs. IC025/ROR025 = 0.591/1.519). For outcomes of cardiovascular AEs, the frequency of death and life-threatening AEs was lower for combination therapy than ICIs alone in noninfectious myocarditis/pericarditis (37.7% vs. 49.2%) as well as in embolic and thrombotic events (29.9% vs. 39.6%). Analysis among indications of cancer showed similar findings. Conclusion Overall, ICIs combined with AGIs showed a greater risk of cardiovascular AEs than ICIs alone, mainly due to an increase in embolic and thrombotic events while a decrease in noninfectious myocarditis/pericarditis. In addition, compared with ICIs alone, combination therapy presented a lower frequency of death and life-threatening in noninfectious myocarditis/pericarditis and embolic and thrombotic events.
Collapse
Affiliation(s)
- Yanfeng Wang
- Department of Comprehensive Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chanjuan Cui
- Department of Laboratory Medicine, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lei Deng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lin Wang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiayang Ren
- Department of Pharmacy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
24
|
Anastasius M, Thavendiranathan P, Kini A, Argulian E, Sharma S, Narula J, Lerakis S. Part 1: The Clinical Applications of Left Ventricular Myocardial Strain. Cardiol Rev 2023:00045415-990000000-00105. [PMID: 37126439 DOI: 10.1097/crd.0000000000000559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
There is an increasing prevalence of cardiovascular disease and heart failure. Indices of left ventricular (LV) systolic function such as LV ejection fraction used to identify those at risk of adverse cardiac events such as heart failure may not be truly representative of LV systolic function in certain cardiac diseases. Given that LV ejection fraction reduction may represent more advanced irreversible stages of disease, measures of myocardial strain have emerged as a feasible and robust instrument for the early identification of heart disease and subtle LV systolic dysfunction. The purpose of this review was to provide an overview of myocardial strain concepts and emerging clinical applications of global longitudinal strain in cardio-oncology.
Collapse
Affiliation(s)
- Malcolm Anastasius
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Annapoorna Kini
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Edgar Argulian
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Samin Sharma
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Jagat Narula
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY
| | - Stamatios Lerakis
- From the Mount Sinai Heart, Icahn School of Medicine at Mount Sinai, New York, NY
| |
Collapse
|
25
|
Delombaerde D, De Sutter J, Croes L, Vervloet D, Moerman V, Van de Veire N, Willems AM, Wouters K, Peeters M, Prenen H, Vulsteke C. Extensive CArdioVAscular Characterization and Follow-Up of Patients Receiving Immune Checkpoint Inhibitors: A Prospective Multicenter Study. Pharmaceuticals (Basel) 2023; 16:ph16040625. [PMID: 37111382 PMCID: PMC10142365 DOI: 10.3390/ph16040625] [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: 03/21/2023] [Revised: 04/13/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
BACKGROUND The increasing use of immune checkpoint inhibitors (ICIs) in the treatment of both advanced and early stages of various malignancies has resulted in a substantial increase in the incidence of cardiovascular (CV) immune-related adverse events (irAEs). The current follow-up guidelines are based on anecdotal evidence and expert opinions, due to a lack of solid data and prospective studies. As many questions remain unanswered, cardiac monitoring, in patients receiving ICIs, is not always implemented by oncologists. Hence, an urgent need to investigate the possible short- and long-term CV effects of ICIs, as ICI approval is continuing to expand to the (neo)adjuvant setting. METHODS We have initiated a prospective, multicenter study, i.e., the CAVACI trial, in which a minimum of 276 patients with a solid tumor, eligible for ICI treatment, will be enrolled. The study consists of routine investigations of blood parameters (troponin and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels, in particular) and a thorough CV follow-up (electrocardiograms, transthoracic echocardiograms, and coronary calcium scoring) at fixed time points for a total period of two years. The primary endpoint is the cumulative incidence of troponin elevation in the first three months of ICI treatment, compared to baseline levels. Furthermore, secondary endpoints include incidence above the upper limit of normal of both troponin and NT-proBNP levels, evolution in troponin and NT-proBNP levels, the incidence of CV abnormalities/major adverse cardiac events, evaluation of associations between patient characteristics/biochemical parameters and CV events, transthoracic echocardiography parameters, electrocardiography parameters, and progression of coronary atherosclerosis. Recruitment of patients started in January 2022. Enrolment is ongoing in AZ Maria Middelares, Antwerp University Hospital, AZ Sint-Vincentius Deinze, and AZ Sint-Elisabeth Zottegem. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT05699915, registered 26 January 2023.
Collapse
Affiliation(s)
- Danielle Delombaerde
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| | - Johan De Sutter
- Department of Cardiology, AZ Maria Middelares, 9000 Ghent, Belgium
- Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| | - Lieselot Croes
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| | | | | | - Nico Van de Veire
- Department of Cardiology, AZ Maria Middelares, 9000 Ghent, Belgium
- Department of Cardiology, Free University Brussels, 1000 Brussels, Belgium
| | | | - Kristien Wouters
- Antwerp University Hospital, Clinical Trial Center (CTC), CRC Antwerp, 2650 Edegem, Belgium
| | - Marc Peeters
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
- Multidisciplinary Oncologic Center Antwerp (MOCA), Antwerp University Hospital, 2650 Edegem, Belgium
| | - Hans Prenen
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
- Multidisciplinary Oncologic Center Antwerp (MOCA), Antwerp University Hospital, 2650 Edegem, Belgium
| | - Christof Vulsteke
- Integrated Cancer Center Ghent, Department of Medical Oncology, AZ Maria Middelares, 9000 Ghent, Belgium
- Center for Oncological Research (CORE), Integrated Personalized and Precision Oncology Network (IPPON), University of Antwerp, 2610 Wilrijk, Belgium
| |
Collapse
|
26
|
Ndjana Lessomo FY, Wang Z, Mukuka C. Comparative cardiotoxicity risk of pembrolizumab versus nivolumab in cancer patients undergoing immune checkpoint inhibitor therapy: A meta-analysis. Front Oncol 2023; 13:1080998. [PMID: 37064101 PMCID: PMC10090546 DOI: 10.3389/fonc.2023.1080998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
ObjectiveRecently, several researchers have reported the incidence of cardiac-related toxicities occurring with nivolumab (Opdivo) and pembrolizumab (Keytruda). There is still a need for balance between oncology treatment efficacy and reduction of cardiotoxicity burden in immune checkpoint inhibitor (ICI)-treated patients. Thus, the primary aim was to determine whether pembrolizumab or nivolumab would present with a greater risk for cardiotoxicity reports.Materials and methodsThis meta-analysis was performed with respect to the MOOSE reporting guidelines. Studies were retrieved by searching PubMed, Embase, and Google Scholar; the search terms were Keytruda or Pembrolizumab, PD1 inhibitors, anti-PD1 drugs, Nivolumab or Opdivo, and cardiotoxicities or cardiac toxicity. The study was restricted to original articles investigating ICI-induced cardiac immune-related adverse events (irAEs). The targeted population was cancer patients treated with either pembrolizumab or nivolumab monotherapy, of which those with records of any cardiac events following the therapy were labeled as events. The measures used to achieve the comparison were descriptive proportions, probabilities, and meta-analysis pooled odds ratios (ORs).ResultsFifteen studies were included in this meta-analysis. Nivolumab accounted for 55.7% cardiotoxicity and pembrolizumab, for 27.31% (P = 0.027). The meta-analysis was based on the Mantel–Haenszel method, and the random-effect model yielded a pooled OR = 0.73 (95% CI [0.43–1.23] P = 0.24), with considerable heterogeneity (I2 = 99% P = 0). Hence, the difference in cardiotoxicity odds risk between pembrolizumab and nivolumab was not statistically significant. On subgroup analysis based on cardiotoxicity type, the “myocarditis” subgroup in which there was no statistical heterogeneity was associated with a significant cardiotoxicity risk increase with pembrolizumab (OR = 1.30 [1.07;1.59], P< 0.05; I2 = 0%, Ph = 0.4).ConclusionTo our knowledge, this is the first meta-analysis to compare the cardiotoxicity potentials of nivolumab and pembrolizumab. In contrast to previous reports, the overall findings here demonstrated that nivolumab-induced cardiotoxicity was more commonly reported in the literature than pembrolizumab; however, myocarditis seemed more likely to occur with pembrolizumab therapy.
Collapse
Affiliation(s)
| | - Zhiquan Wang
- Cardiovascular Internal Medicine Department, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Zhiquan Wang, ,
| | - Chishimba Mukuka
- Internal Medicine Department, MANSA General Hospital, Mansa, Luapula, Zambia
| |
Collapse
|
27
|
Grigorescu A, Teodorescu M. Toxicitatea cardiacă a inhibitorilor punctelor de control (IPC) utilizaţi în imunoterapia cancerului. ONCOLOG-HEMATOLOG.RO 2023. [DOI: 10.26416/onhe.62.1.2023.7746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
|
28
|
Chiang CH, Chiang CH, Ma KSK, Hsia YP, Lee YW, Wu HR, Chiang CH, Peng CY, Wei JCC, Shiah HS, Peng CM, Neilan TG. The incidence and risk of cardiovascular events associated with immune checkpoint inhibitors in Asian populations. Jpn J Clin Oncol 2022; 52:1389-1398. [PMID: 36208180 PMCID: PMC9721460 DOI: 10.1093/jjco/hyac150] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Accepted: 08/30/2022] [Indexed: 12/13/2022] Open
Abstract
OBJECTIVES Immune checkpoint inhibitors are associated with adverse cardiovascular events. However, there are no data characterizing cardiovascular events among Asians on immune checkpoint inhibitors. We aim to determine the incidence and risk of cardiac events associated with immune checkpoint inhibitors in an Asian population. METHODS We performed a retrospective, propensity score-matched cohort study at two tertiary referral centers in Taiwan. Immune checkpoint inhibitor users were matched with non-immune checkpoint inhibitor users based on predetermined clinical variables. The primary outcome was major adverse cardiovascular events, defined as a composite of myocardial infarction, ischemic stroke, acute peripheral occlusive disease, pulmonary embolism, deep venous thrombosis, heart failure, pericardial disease, myocarditis, cardiac arrhythmias and conduction block. RESULTS Between January 2010 and November 2021, 868 immune checkpoint inhibitor users were matched 1:1 with non-immune checkpoint inhibitor users. Among immune checkpoint inhibitor users, 67 (7.7%) patients developed major adverse cardiovascular events. During a median follow-up period of 188 days, the incidence rate of major adverse cardiovascular events for immune checkpoint inhibitor and non-immune checkpoint inhibitor users was 94.8 and 46.2 per 1000 patient-years, respectively, resulting in an incidence rate ratio of 2.1 [95% confidence interval: 1.5-2.9]. In multivariate Cox proportional hazard models, immune checkpoint inhibitor users had a 60% increased risk for major adverse cardiovascular events [hazard ratio, 1.6 (95% confidence interval: 1.1-2.3)]. Immune checkpoint inhibitors use was independently associated with increased risk of ischemic stroke [hazard ratio, 3.0 (95% confidence interval: 1.0-9.0)] and pulmonary embolism [hazard ratio, 5.5 (95% confidence interval: 1.4-21.3)]. In multivariate logistic regression analysis, age > 65, metastatic disease, hypertension and baseline platelet-to-lymphocyte ratio < 180 were risk factors for major adverse cardiovascular events. CONCLUSIONS Among Asians, immune checkpoint inhibitors were associated with an increased risk of major adverse cardiovascular events, particularly ischemic stroke and pulmonary embolism.
Collapse
Affiliation(s)
- Cho-Han Chiang
- Department of Medicine, Mount Auburn Hospital, Harvard Medical School, Boston, MA, USA
| | - Cho-Hung Chiang
- Department of General Division, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Kevin Sheng-Kai Ma
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Center for Global Health, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Graduate Institute of Biomedical Electronics and Bioinformatics, College of Electrical Engineering and Computer Science, National Taiwan University, Taipei, Taiwan
| | - Yuan Ping Hsia
- Department of Family Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Foundation, New Taipei City, Taiwan
| | - Yu-wen Lee
- Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Han-Ru Wu
- Department of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Cho-Hsien Chiang
- Department of Medical Education, Kuang Tien General Hospital, Taichung, Taiwan
- London School of Hygiene & Tropical Medicine, London, UK
| | - Chun-Yu Peng
- Department of Medicine, Danbury Hospital, Danbury, Connecticut, USA
- Department of Medicine, National Taiwan University College of Medicine, Taipei, Taiwan
| | - James Cheng-Chung Wei
- Division of Allergy, Immunology and Rheumatology, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Her-Shyong Shiah
- Department of Hematology and Oncology, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
| | - Cheng-Ming Peng
- Department of Family Medicine, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Foundation, New Taipei City, Taiwan
- Da Vinci Minimally Invasive Surgery Center, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Tomas G Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology and Department of Radiology, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
29
|
Chen R, Zhou M, Zhu F. Immune Checkpoint Inhibitors Related to Cardiotoxicity. J Cardiovasc Dev Dis 2022; 9:jcdd9110378. [PMID: 36354777 PMCID: PMC9697232 DOI: 10.3390/jcdd9110378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/17/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have now emerged as a mainstay of treatment for various cancers. Along with development of ICIs, immune-related adverse effects (irAEs) have aroused wide attention. The cardiac irAE, one of the rare but potentially fatal effects, have been reported recently. However, the clinical comprehension of cardiac irAEs remains limited and guidelines are inadequate for cardio-oncologists to tackle the problem. In this review, we have summarized current classifications of, manifestations of, potential mechanisms of, and treatment for ICI-related myocardial injury in order to provide some clues for the understanding of cardiac irAEs in clinical work.
Collapse
Affiliation(s)
- Ru Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Zhou
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Respiratory Diseases, National Ministry of Health of the People’s Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan 430022, China
- Correspondence: (M.Z.); (F.Z.)
| | - Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence: (M.Z.); (F.Z.)
| |
Collapse
|
30
|
Shroff GS, Strange CD, Ahuja J, Altan M, Sheshadri A, Unlu E, Truong MT, Vlahos I. Imaging of Immune Checkpoint Inhibitor Immunotherapy for Non-Small Cell Lung Cancer. Radiographics 2022; 42:1956-1974. [PMID: 36240075 DOI: 10.1148/rg.220108] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The normal immune system identifies and eliminates precancerous and cancerous cells. However, tumors can develop immune resistance mechanisms, one of which involves the exploitation of pathways, termed immune checkpoints, that normally suppress T-cell function. The goal of immune checkpoint inhibitor (ICI) immunotherapy is to boost T-cell-mediated immunity to mount a more effective attack on cancer cells. ICIs have changed the treatment landscape of advanced non-small cell lung cancer (NSCLC), and numerous ICIs have now been approved as first-line treatments for NSCLC by the U.S. Food and Drug Administration. ICIs can cause atypical response patterns such as pseudoprogression, whereby the tumor burden initially increases but then decreases. Therefore, response criteria have been developed specifically for patients receiving immunotherapy. Because ICIs activate the immune system, they can lead to inflammatory side effects, termed immune-related adverse events (irAEs). Usually occurring within weeks to months after the start of therapy, irAEs range from asymptomatic abnormal laboratory results to life-threatening conditions such as encephalitis, pneumonitis, myocarditis, hepatitis, and colitis. It is important to be aware of the imaging appearances of the various irAEs to avoid misinterpreting them as metastatic disease, progressive disease, or infection. The basic principles of ICI therapy; indications for ICI therapy in the setting of NSCLC; response assessment and atypical response patterns of ICI therapy, as compared with conventional chemotherapy; and the spectrum of irAEs seen at imaging are reviewed. An invited commentary by Nishino is available online. ©RSNA, 2022.
Collapse
Affiliation(s)
- Girish S Shroff
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Chad D Strange
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Jitesh Ahuja
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Mehmet Altan
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Ajay Sheshadri
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Ebru Unlu
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Mylene T Truong
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| | - Ioannis Vlahos
- From the Departments of Thoracic Imaging (G.S.S., C.D.S., J.A., E.U., M.T.T., I.V.), Thoracic/Head and Neck Medical Oncology (M.A.), and Pulmonary Medicine (A.S.), The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1478, Houston, TX 77030
| |
Collapse
|
31
|
Irabor OC, Nelson N, Shah Y, Niazi MK, Poiset S, Storozynsky E, Singla DK, Hooper DC, Lu B. Overcoming the cardiac toxicities of cancer therapy immune checkpoint inhibitors. Front Oncol 2022; 12:940127. [PMID: 36185227 PMCID: PMC9523689 DOI: 10.3389/fonc.2022.940127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Accepted: 08/29/2022] [Indexed: 11/13/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have led recent advances in the field of cancer immunotherapy improving overall survival in multiple malignancies with abysmal prognoses prior to their introduction. The remarkable efficacy of ICIs is however limited by their potential for systemic and organ specific immune-related adverse events (irAEs), most of which present with mild to moderate symptoms that can resolve spontaneously, with discontinuation of therapy or glucocorticoid therapy. Cardiac irAEs however are potentially fatal. The understanding of autoimmune cardiotoxicity remains limited due to its rareness. In this paper, we provide an updated review of the literature on the pathologic mechanisms, diagnosis, and management of autoimmune cardiotoxicity resulting from ICIs and their combinations and provide perspective on potential strategies and ongoing research developments to prevent and mitigate their occurrence.
Collapse
Affiliation(s)
- Omoruyi Credit Irabor
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Philadelphia, PA, United States
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, United States
- *Correspondence: Omoruyi Credit Irabor,
| | - Nicolas Nelson
- Sidney Kimmel Medical College (SKMC), Philadelphia, PA, United States
| | - Yash Shah
- Sidney Kimmel Medical College (SKMC), Philadelphia, PA, United States
| | - Muneeb Khan Niazi
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Philadelphia, PA, United States
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Spencer Poiset
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Philadelphia, PA, United States
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Eugene Storozynsky
- Division of Cardiology, Thomas Jefferson University Hospital, Philadelphia, PA, United States
| | - Dinender K. Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Douglas Craig Hooper
- Sidney Kimmel Medical College (SKMC), Philadelphia, PA, United States
- Department of Pharmacology, Physiology, and Cancer Biology, Thomas Jefferson University, Philadelphia, PA, United States
| | - Bo Lu
- Department of Radiation Oncology, Sidney Kimmel Cancer Center, Philadelphia, PA, United States
- Department of Radiation Oncology, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
32
|
Wu S, Bai H, Zhang L, He J, Luo X, Wang S, Fan G, Sun N. Cardiovascular adverse events induced by immune checkpoint inhibitors: A real world study from 2018 to 2022. Front Cardiovasc Med 2022; 9:969942. [PMID: 36035942 PMCID: PMC9399397 DOI: 10.3389/fcvm.2022.969942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/13/2022] [Indexed: 12/19/2022] Open
Abstract
Background The reported rate of cardiovascular adverse events (CAE) caused by immune checkpoint inhibitors (ICI) is low but potentially fatal. Assess the risk of CAE in cancer patients and compare the incidence of CAE between Chinese developed ICIs and imported ICIs. Methods A retrospective analysis was performed on cancer patients treated with ICI for at least four cycles in the Second Affiliated Hospital of Dalian Medical University from January 2018 to March 2022. Baseline characteristics, physiological and biochemical values, electrocardiographic and echocardiographic findings were compared between patients with and without CAE. Results Among 495 patients treated with ICIs, CAEs occurred in 64 patients (12.93%). The median time to the event was 105 days (61–202). The patients with low neutrophil-to-lymphocyte ratio (L-NLR) were significantly associated with the risk of developing CAE (hazard ratio HR 3.64, 95% confidence ratio CI 1.86–7.15, P = 0.000). Patients with higher comorbidity burden significantly increased the risk of developing CAE (HR 1.30, 95% CI 1.05–1.61, P = 0.014). Those who received a combination of ICI and vascular endothelial growth factor receptor (VEGFR) inhibitors (HR 2.57, 95% CI 1.37–4.84, P = 0.003) or thoracic radiation therapy (HR 32.93, 95% CI 8.81–123.14, P = 0.000) were at a significantly increased risk of developing CAE. Compared to baseline values, creatine kinase is -oenzymes (CK-MB) (95% CI -9.73 to -2.20, P = 0.003) and cardiac troponin I (cTnI) (95% CI -1.06 to -0.06, P = 0.028) were elevated, and the QTc interval prolonged (95% CI -27.07 to -6.49, P = 0.002). Using nivolumab as a control, there was no difference in CAE risk among the eight ICIs investigated. However, the results of the propensity matching showed that programmed death-ligand 1 (PD-L1) inhibitors had lower CAE occurrence compared with programmed cell death protein 1 (PD-1) inhibitors (adjusted HR = 0.38, P = 0.045). Conclusion Patients who received concurrent VEGFR inhibitors and ICIs had a history of thoracic radiation therapy, L-NLR, and higher comorbidity burden had an increased risk of CAEs. Elevated cTnI, CK-MB, and QTc, can be used to monitor CAEs. There was no significant difference in CAE risks between Chinese domestic and imported ICIs. PD-L1 inhibitors had lower CAE occurrence than PD-1 inhibitors.
Collapse
|
33
|
Sorodoc V, Sirbu O, Lionte C, Haliga RE, Stoica A, Ceasovschih A, Petris OR, Constantin M, Costache II, Petris AO, Morariu PC, Sorodoc L. The Value of Troponin as a Biomarker of Chemotherapy-Induced Cardiotoxicity. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081183. [PMID: 36013362 PMCID: PMC9410123 DOI: 10.3390/life12081183] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
In cancer survivors, cardiac dysfunction is the main cause of mortality. Cardiotoxicity represents a decline in cardiac function associated with cancer therapy, and the risk factors include smoking, dyslipidemia, an age of over 60 years, obesity, and a history of coronary artery disease, diabetes, atrial fibrillation, or heart failure. Troponin is a biomarker that is widely used in the detection of acute coronary syndromes. It has a high specificity, although it is not exclusively associated with myocardial ischemia. The aim of this paper is to summarize published studies and to establish the role of troponin assays in the diagnosis of cardiotoxicity associated with various chemotherapeutic agents. Troponin has been shown to be a significant biomarker in the diagnosis of the cardiac dysfunction associated with several types of chemotherapeutic drugs: anthracyclines, anti-human epidermal growth factor receptor 2 treatment, and anti-vascular endothelial growth factor therapy. Based on the data available at this moment, troponin is useful for baseline risk assessment, the diagnosis of cardiotoxicity, and as a guide for the initiation of cardioprotective treatment. There are currently clear regulations regarding the timing of troponin surveillance depending on the patient’s risk of cardiotoxicity and the type of medication administered, but data on the cut-off values of this biomarker are still under investigation.
Collapse
Affiliation(s)
- Victorita Sorodoc
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Oana Sirbu
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Correspondence: (O.S.); (C.L.)
| | - Catalina Lionte
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Correspondence: (O.S.); (C.L.)
| | - Raluca Ecaterina Haliga
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Alexandra Stoica
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Alexandr Ceasovschih
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Ovidiu Rusalim Petris
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Mihai Constantin
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Irina Iuliana Costache
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Department of Cardiology, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania
| | - Antoniu Octavian Petris
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Department of Cardiology, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania
| | - Paula Cristina Morariu
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
| | - Laurentiu Sorodoc
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| |
Collapse
|
34
|
Dong M, Yu T, Zhang Z, Zhang J, Wang R, Tse G, Liu T, Zhong L. ICIs-Related Cardiotoxicity in Different Types of Cancer. J Cardiovasc Dev Dis 2022; 9:203. [PMID: 35877565 PMCID: PMC9324462 DOI: 10.3390/jcdd9070203] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/17/2022] [Accepted: 06/20/2022] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) are rapidly developing immunotherapy cancer drugs that have prolonged patient survival. However, ICIs-related cardiotoxicity has been recognized as a rare, but fatal, consequence. Although there has been extensive research based on different types of ICIs, these studies have not indicated whether cardiotoxicity is specific to a type of cancer. Therefore, we conducted a systematic review to analyze a variety of ICIs-related cardiotoxicity, focusing on different types of cancer. We found that the incidence of ICIs-related cardiac adverse events (CAEs) and common cardiotoxic manifestations vary with cancer type. This inspired us to explore the underlying mechanisms to formulate targeted clinical strategies for maintaining the cardiovascular health of cancer patients.
Collapse
Affiliation(s)
- Mei Dong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| | - Ting Yu
- Medical College, Qingdao University, Qingdao 266003, China;
| | - Zhenzhen Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| | - Jing Zhang
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| | - Rujian Wang
- Department of Oncology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China;
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300070, China;
- Kent and Medway Medical School, Canterbury CT2 7FS, UK
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300070, China;
| | - Lin Zhong
- Department of Cardiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai 264000, China; (M.D.); (Z.Z.); (J.Z.)
| |
Collapse
|
35
|
Tonry C, Russel-Hallinan A, McCune C, Collier P, Harbinson M, Dixon L, Watson CJ. Circulating biomarkers for management of cancer therapeutics related cardiac dysfunction. Cardiovasc Res 2022; 119:710-728. [PMID: 35640873 PMCID: PMC10153425 DOI: 10.1093/cvr/cvac087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 05/09/2022] [Accepted: 05/15/2022] [Indexed: 11/14/2022] Open
Abstract
Cancer therapeutics related cardiac dysfunction (CTRCD) has emerged as a major cause of morbidity and mortality in cancer survivors. Effective clinical management of CTRCD is impeded by a lack of sensitive diagnostic and prognostic strategies. Circulating molecular markers could potentially address this need as they are often indicative of cardiac stress before cardiac damage can be detected clinically. A growing understanding of the underlying physiological mechanisms for CTRCD has inspired research efforts to identify novel pathophysiologically-relevant biomarkers that may also guide development of cardio-protective therapeutic approaches. The purpose of this review is to evaluate current circulating biomarkers of cardiac stress and their potential role in diagnosis and management of CTRCD. We also discuss some emerging avenues for CTRCD-focused biomarker investigations.
Collapse
Affiliation(s)
- Claire Tonry
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Adam Russel-Hallinan
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | - Claire McCune
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| | | | | | | | - Chris J Watson
- Wellcome-Wolfson Institute for Experimental Medicine, School of Medicine, Dentistry and Biomedical Sciences, Queens University Belfast, Northern Ireland, United Kingdom
| |
Collapse
|
36
|
Immune Checkpoint Inhibitors-Associated Cardiotoxicity. Cancers (Basel) 2022; 14:cancers14051145. [PMID: 35267453 PMCID: PMC8909315 DOI: 10.3390/cancers14051145] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 02/17/2022] [Accepted: 02/19/2022] [Indexed: 12/19/2022] Open
Abstract
Large population-based studies examining differences in ICI-associated cardiotoxicity across cancer types and agents are limited. Data of 5518 cancer patients who received at least one cycle of ICIs were extracted from a large network of health care organizations. ICI treatment groups were classified by the first ICI agent(s) (ipilimumab, nivolumab, pembrolizumab, cemiplimab, avelumab, atezolizumab, or durvalumab) or its class (PD-1 inhibitors, PD-L1 inhibitors, CTLA4-inhibitors, or their combination (ipilimumab + nivolumab)). Time to first cardiac adverse event (CAE) (arrhythmia, acute myocardial infarction, myocarditis, cardiomyopathy, or pericarditis) developed within one year after ICI initiation was analyzed using a competing-risks regression model adjusting for ICI treatment groups, patient demographic and clinical characteristics, and cancer sites. By month 12, 12.5% developed cardiotoxicity. The most common cardiotoxicity was arrhythmia (9.3%) and 2.1% developed myocarditis. After adjusting for patient characteristics and cancer sites, patients who initiated on monotherapy with ipilimumab (adjusted Hazard Ratio (aHR): 2.00; 95% CI: 1.49−2.70; p < 0.001) or pembrolizumab (aHR: 1.21; 95% CI: 1.01−1.46; p = 0.040) had a higher risk of developing CAEs within one year compared to nivolumab monotherapy. Ipilimumab and pembrolizumab use may increase the risk of cardiotoxicity compared to other agents. Avelumab also estimated a highly elevated risk (aHR: 1.92; 95% CI: 0.85−4.34; p = 0.117) compared to nivolumab and other PD-L1 agents, although the estimate did not reach statistical significance, warranting future studies.
Collapse
|
37
|
Chitturi KR, Burns EA, Muhsen IN, Anand K, Trachtenberg BH. Cardiovascular Risks with Epidermal Growth Factor Receptor (EGFR) Tyrosine Kinase Inhibitors and Monoclonal Antibody Therapy. Curr Oncol Rep 2022; 24:475-491. [PMID: 35192115 DOI: 10.1007/s11912-022-01215-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2021] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Tyrosine kinase inhibitors (TKI) and monoclonal antibodies (mAbs) that target the epidermal growth factor receptor (EGFR) have changed the therapeutic landscape across a range of solid malignancies. However, there is little data regarding the cardiovascular (CV) impact of these agents. The purpose of this review is to discuss reported CV effects, pathophysiology, pre-treatment screening, diagnostic workup, and treatment recommendations in this patient population. RECENT FINDINGS It is apparent that CV events are not class dependent, and while infrequently reported in clinical trials, unique CV toxicity may occur with EGFR inhibitors, including structural, electrical, and vascular events. There remains an unmet need to fully elucidate the spectrum of CV events associated with EGFR inhibitors. Early CV screening, close clinical monitoring, coupled with a multidisciplinary approach between medical and cardio-oncology is needed to minimize the potentially detrimental impact of cardiotoxicity in this patient population.
Collapse
Affiliation(s)
- Kalyan R Chitturi
- Division of Cardiovascular Medicine, Department of Medicine, University of Missouri, 1 Hospital Drive, Columbia, MO, CE30665201, USA
| | - Ethan A Burns
- Houston Methodist Cancer Center, 6445 Main St. Outpatient Center, Floor 24, Houston, TX, 77030, USA
| | - Ibrahim N Muhsen
- Department of Medicine, Houston Methodist Hospital, Houston, TX, 77030, USA
| | - Kartik Anand
- Great Plains Health Callahan Cancer Center, 601 W Leota St, North Platte, NE, 69101, USA
| | - Barry H Trachtenberg
- Methodist DeBakey Heart and Vascular Center, 6400 Fannin St. Suite 3000, Houston, TX, 77030, USA.
| |
Collapse
|
38
|
Nardi Agmon I, Itzhaki Ben Zadok O, Kornowski R. The Potential Cardiotoxicity of Immune Checkpoint Inhibitors. J Clin Med 2022; 11:jcm11030865. [PMID: 35160316 PMCID: PMC8836470 DOI: 10.3390/jcm11030865] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/26/2022] [Accepted: 02/01/2022] [Indexed: 12/13/2022] Open
Abstract
The use of immune checkpoint inhibitors (ICIs) as a mono- or adjuvant oncologic treatment is rapidly expanding to most fields of cancer. Alongside their efficacy, ICIs carry the risk of immune-related adverse events (irAEs) arising from misguided immune-mediated response to normal tissues. In the cardiovascular system, the cardiac toxicity of ICIs has been primarily related to the development of an acute, immune-mediated myocarditis; beyond this potentially fatal complication, evidence of an increased risk of cardiovascular events and accelerated atherosclerosis is emerging, as well as reports of other cardiovascular adverse events such as arrythmias, Takotsubo-like syndrome and vascular events. The absence of identified risk factors for cardiotoxic complications, specific monitoring strategies or diagnostic tests, pose challenges to the timely recognition and optimal management of such events. The rising numbers of patients being treated with ICIs make this potential cardiotoxic effect one of paramount importance for further investigation and understanding. This review will discuss the most recent data on different cardiotoxic effects of ICIs treatment.
Collapse
Affiliation(s)
- Inbar Nardi Agmon
- Department of Cardiology, Rabin Medical Center–Beilinson Hospital, Petach Tikva 4941492, Israel; (O.I.B.Z.); (R.K.)
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
- Correspondence: ; Tel.: +972-54-2661422
| | - Osnat Itzhaki Ben Zadok
- Department of Cardiology, Rabin Medical Center–Beilinson Hospital, Petach Tikva 4941492, Israel; (O.I.B.Z.); (R.K.)
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center–Beilinson Hospital, Petach Tikva 4941492, Israel; (O.I.B.Z.); (R.K.)
- Sackler School of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| |
Collapse
|
39
|
Kushnareva E, Kushnarev V, Artemyeva A, Mitrofanova L, Moiseeva O. Myocardial PD-L1 Expression in Patients With Ischemic and Non-ischemic Heart Failure. Front Cardiovasc Med 2022; 8:759972. [PMID: 35096992 PMCID: PMC8792535 DOI: 10.3389/fcvm.2021.759972] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 12/17/2021] [Indexed: 12/19/2022] Open
Abstract
Objective: Immune checkpoints inhibitors are promising and wide-spread agents in anti-cancer therapy. However, despite their efficacy, these agents could cause cardiotoxicity, a rare but life-threatening event. In addition, there are still no well-described predictive factors for the development of immune-related adverse events and information on high risk groups. According to known experimental studies we hypothesized that cardiovascular diseases may increase myocardial PD-L1 expression, which could be an extra target for Checkpoint inhibitors and a potential basis for complications development. Methods: We studied patterns of myocardial PD-L1 expression in non-cancer-related cardiovascular diseases, particularly ischemic heart disease (n = 12) and dilated cardiomyopathy (n = 7), compared to patients without known cardiovascular diseases (n = 10) using mouse monoclonal anti-PD-L1 antibody (clone 22C3, 1:50, Dako). Correlation between immunohistochemical data and echocardiographic parameters was assessed. Statistical analyses were performed using R Statistical Software—R studio version 1.3.1093. Results: In the myocardium of cardiac patients, we found membranous, cytoplasmic, and endothelial expression of PD-L1 compared to control group. In samples from patients with a history of myocardial infarction, PD-L1 membrane and endothelial expression was more prominent and frequent, and cytoplasmic and intercalated discs staining was more localized. In contrast, samples from patients with dilated cardiomyopathy displayed very faint endothelial staining, negative membrane staining, and more diffuse PD-L1 expression in the cytoplasm and intercalated discs. In samples from the non-cardiac patients, no convincing PD-L1 expression was observed. Moreover, we discovered a significant negative correlation between PD-L1 expression level and left ventricular ejection fraction and a positive correlation between PD-L1 expression level and left ventricular end-diastolic volume. Conclusions: The present findings lay the groundwork for future experimental and clinical studies of the role of the PD-1/PD-L1 pathway in cardiovascular diseases. Further studies are required to find patients at potentially high risk of cardiovascular adverse events associated with immune checkpoint inhibitors therapy.
Collapse
Affiliation(s)
- Ekaterina Kushnareva
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Vladimir Kushnarev
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia.,N. N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - Anna Artemyeva
- N. N. Petrov National Medical Research Center of Oncology, Saint Petersburg, Russia
| | - Lubov Mitrofanova
- Personalized Medicine Centre, Almazov National Medical Research Centre, Saint Petersburg, Russia
| | - Olga Moiseeva
- Non-coronary Heart Disease Department, Almazov National Medical Research Centre, Saint Petersburg, Russia
| |
Collapse
|
40
|
Isawa T, Toi Y, Sugawara S, Taguri M, Toyoda S. OUP accepted manuscript. Oncologist 2022; 27:e410-e419. [PMID: 35348766 PMCID: PMC9074992 DOI: 10.1093/oncolo/oyac056] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 02/04/2022] [Indexed: 11/25/2022] Open
Abstract
Background Cardiovascular immune-related adverse events (CV–irAEs) associated with immune checkpoint inhibitors (ICIs) may have been underreported given that most previous reports were retrospective. We aimed to evaluate the incidence, clinical characteristics, and predictors of CV–irAEs and determine the feasibility of serial cardiac monitoring using a combination of B-type natriuretic peptide, cardiac troponin T, and electrocardiogram for the prediction of future symptomatic (grade ≥2) CV–irAEs. Materials and Methods This was a prospective observational study that included 129 consecutive patients with non–small-cell lung cancer who received ICI monotherapy at a single center. Serial cardiac monitoring was performed during ICI monotherapy. Results A total of 35 (27%) patients developed any grade ≥1 CV–irAEs with a median time of onset of 72 (interquartile range 44-216) days after ICI treatment initiation. Multivariate Fine–Gray regression analysis showed that prior acute coronary syndrome (adjusted hazard ratio [HR] 3.15 (95% [CI], 2.03-4.91), prior heart failure hospitalization (adjusted HR 1.65 [95% CI, 1.17-2.33]), and achievement of disease control (adjusted HR 1.91, [95% CI, 1.16-3.14]) were significantly associated with grade ≥1 CV–irAEs. Serial cardiac monitoring revealed that patients with preceding grade 1 CV–irAEs were associated with a significantly higher risk of onset of grade ≥2 CV–irAEs compared with those without preceding grade 1 CV–irAEs (HR: 6.17 [95% CI, 2.97-12.83]). Conclusion CV–irAEs were more common than previously recognized and have several predictors. Moreover, serial cardiac monitoring may be feasible for the prediction of future grade ≥2 CV–irAEs.
Collapse
Affiliation(s)
- Tsuyoshi Isawa
- Department of Cardiology, Sendai Kousei Hospital, Sendai, Japan
- Corresponding author: Tsuyoshi Isawa, MD, Department of Cardiology, Sendai Kousei Hospital, 4-15, Hirose-machi, Sendai, Miyagi 980-0873, Japan. Tel: 81 22 222 6181; Fax: 81 22 223 8422; e-mail:
| | - Yukihiro Toi
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Shunichi Sugawara
- Department of Pulmonary Medicine, Sendai Kousei Hospital, Sendai, Japan
| | - Masataka Taguri
- Department of Data Science, Yokohama City University School of Data Science, Yokohama, Japan
| | - Shigeru Toyoda
- Department of Cardiovascular Medicine, Dokkyo Medical University, Mibu, Japan
| |
Collapse
|
41
|
Zaha VG, Hayek SS, Alexander KM, Beckie TM, Hundley WG, Kondapalli L, Ky B, Leger KJ, Meijers WC, Moslehi JJ, Shah SH. Future Perspectives of Cardiovascular Biomarker Utilization in Cancer Survivors: A Scientific Statement From the American Heart Association. Circulation 2021; 144:e551-e563. [PMID: 34753300 DOI: 10.1161/cir.0000000000001032] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Improving cancer survival represents the most significant effect of precision medicine and personalized molecular and immunologic therapeutics. Cardiovascular health becomes henceforth a key determinant for the direction of overall outcomes after cancer. Comprehensive tissue diagnostic studies undoubtedly have been and continue to be at the core of the fight against cancer. Will a systemic approach integrating circulating blood-derived biomarkers, multimodality imaging technologies, strategic panomics, and real-time streams of digitized physiological data overcome the elusive cardiovascular tissue diagnosis in cardio-oncology? How can such a systemic approach be personalized for application in day-to-day clinical work, with diverse patient populations, cancer diagnoses, and therapies? To address such questions, this scientific statement approaches a broad definition of the biomarker concept. It summarizes the current literature on the utilization of a multitude of established cardiovascular biomarkers at the intersection with cancer. It identifies limitations and gaps of knowledge in the application of biomarkers to stratify the cardiovascular risk before cancer treatment, monitor cardiovascular health during cancer therapy, and detect latent cardiovascular damage in cancer survivors. Last, it highlights areas in biomarker discovery, validation, and clinical application for concerted efforts from funding agencies, scientists, and clinicians at the cardio-oncology nexus.
Collapse
|
42
|
Eftekhar SP, Yazdanpanah N, Rezaei N. Immune checkpoint inhibitors and cardiotoxicity: possible mechanisms, manifestations, diagnosis and management. Expert Rev Anticancer Ther 2021; 21:1211-1228. [PMID: 34511008 DOI: 10.1080/14737140.2021.1979396] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
INTRODUCTION Immune checkpoint inhibitors (ICIs) are a new class of anticancer drugs that enhance the immune system function and activate T cells against cancerous cells. Although cardiac complications are not common, they could be accompanied with high morbidity and mortality. AREAS COVERED Regarding the importance of cardiac complications and their subsequent burden on individuals and the healthcare system, this review attempts to discuss the mechanism, diagnosis, and management of myocarditis, besides recapitulating the possible mechanism of other cardiac adverse events. Moreover, we briefly discuss the concurrent administration of other chemotherapeutic agents. EXPERT OPINION Due to insufficient knowledge concerning the physiopathology of immune-related adverse events (irAEs) and their potential further complications, cardiovascular complications in particular and in the context of this paper's focus, cooperation of oncologists, immunologists, and cardiologists is necessary for the management of patients. Experimental approaches such as using corticosteroids are becoming a part of guidelines for managing cardiac irAEs. However, a unique algorithm for diagnosis and management is necessary, especially in myocarditis cases. Furthermore, more studies are required to resolve current challenges, including prevention of myocarditis, concurrent administration of other chemotherapeutic agents, and re-introducing patients with ICIs.
Collapse
Affiliation(s)
- Seyed Parsa Eftekhar
- School of Medicine, Babol University of Medical Sciences, Babol, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Babol, Iran
| | - Niloufar Yazdanpanah
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.,Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Network of Immunity in Infection, Malignancy and Autoimmunity (Niima), Universal Scientific Education and Research Network (Usern), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Cardiovascular toxicity of angiogenesis inhibitors and immune checkpoint inhibitors: synergistic anti-tumour effects at the cost of increased cardiovascular risk? Clin Sci (Lond) 2021; 135:1649-1668. [PMID: 34283204 DOI: 10.1042/cs20200300] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022]
Abstract
In the past two decades, treatment outcomes for a wide range of malignancies have improved remarkably due to the development of novel anti-cancer therapies, including vascular endothelial growth factor inhibitors (VEGFIs) and immune checkpoint inhibitors (ICIs). Despite their unprecedented anti-tumour effects, it is becoming increasingly clear that both types of agents are associated with specific cardiovascular toxicity, including hypertension, congestive heart failure, myocarditis and acceleration of atherosclerosis. Currently, VEGFI and ICI combination therapy is recommended for the treatment of advanced renal cell carcinoma (RCC) and has shown promising treatment efficacy in other tumour types as well. Consequently, VEGFI and ICI combination therapy will most likely become an important therapeutic strategy for various malignancies. However, this combinatory approach is expected to be accompanied by a substantial increase in cardiovascular risk, as both types of agents could act synergistically to induce cardiovascular sequelae. Therefore, a comprehensive baseline assessment and adequate monitoring by specialised cardio-oncology teams is essential in case these agents are used in combination, particularly in high-risk patients. This review summarises the mechanisms of action and treatment indications for currently registered VEGFIs and ICIs, and discusses their main vascular and cardiac toxicity. Subsequently, we provide the biological rationales for the observed promising synergistic anti-tumour effects of combined VEGFI/ICI administration. Lastly, we speculate on the increased risk for cardiovascular toxicity in case these agents are used in combination and its implications and future directions for the clinical situation.
Collapse
|
44
|
Fradley MG, Beckie TM, Brown SA, Cheng RK, Dent SF, Nohria A, Patton KK, Singh JP, Olshansky B. Recognition, Prevention, and Management of Arrhythmias and Autonomic Disorders in Cardio-Oncology: A Scientific Statement From the American Heart Association. Circulation 2021; 144:e41-e55. [PMID: 34134525 DOI: 10.1161/cir.0000000000000986] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
With the advent of novel cancer therapeutics and improved screening, more patients are surviving a cancer diagnosis or living longer with advanced disease. Many of these treatments have associated cardiovascular toxicities that can manifest in both an acute and a delayed fashion. Arrhythmias are an increasingly identified complication with unique management challenges in the cancer population. The purpose of this scientific statement is to summarize the current state of knowledge regarding arrhythmia identification and treatment in patients with cancer. Atrial tachyarrhythmias, particularly atrial fibrillation, are most common, but ventricular arrhythmias, including those related to treatment-induced QT prolongation, and bradyarrhythmias can also occur. Despite increased recognition, dedicated prospective studies evaluating true incidence are lacking. Moreover, few studies have addressed appropriate prevention and treatment strategies. As such, this scientific statement serves to mobilize the cardio-oncology, electrophysiology, and oncology communities to develop clinical and scientific collaborations that will improve the care of patients with cancer who have arrhythmias.
Collapse
|
45
|
Chen C, Chen T, Liang J, Guo X, Xu J, Zheng Y, Guo Z, Chi L, Wei L, Chen X, Ye X, He J. Cardiotoxicity Induced by Immune Checkpoint Inhibitors: A Pharmacovigilance Study From 2014 to 2019 Based on FAERS. Front Pharmacol 2021; 12:616505. [PMID: 33643048 PMCID: PMC7907652 DOI: 10.3389/fphar.2021.616505] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/05/2021] [Indexed: 12/19/2022] Open
Abstract
This study was to scientifically and systematically explore the association between cardiotoxicity and immune checkpoint inhibitors (ICIs) and also to characterize the spectrum of ICI-related cardiac complications. From the first quarter of 2014 to the fourth quarter of 2019, data from the FDA Adverse Event Reporting System database were selected to conduct the disproportionality analysis. Reporting odds ratios and information components were used to evaluate the signal after statistical shrinkage transformation. In total, 7,443,137 cases and 36,326,611 drug-adverse event pairs were collected, among which 9,271 cases were identified to be related to ICI-induced cardiotoxicities. The number of male patients was much higher than that of females (5,579 vs. 3,031) and males presented a slightly higher reporting frequency than females in general, which was statistically significant (ROR = 1.04, 95%CI: 0.99-1.09, p < 0.001). Simultaneously, the proportion of serious or life-threatening outcomes in males was significantly higher than in females (ROR = 1.05, 95%CI: 0.96-1.15, p < 0.001). Importantly, ICIs were associated with over-reporting frequencies of cardiotoxicities in general (ROR025 = 1.06, IC025 = 0.08). PD-1 and PD-L1 were found to be related to cardiac adverse events, corresponding to ROR025 = 1.06, IC025 = 0.08, and ROR025 = 1.06, IC025 = 0.08, respectively, while anti-CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) was significantly associated with some specific adverse events rather than common adverse events. The spectrum of cardiotoxicities induced by ICIs mostly differed among individual agents, but also demonstrated some common features. Dyspnea (N = 2,527, 21.25%), myocarditis (N = 614, 5.16%), atrial fibrillation (N = 576, 4.84%), cardiac failure (N = 476, 4.00%), and pericardial effusion (N = 423, 3.56%) were the top five cardiac adverse events reported in the database. Among them, myocarditis was the only one caused by all ICIs with strong signal value and high risk, warranting further attention. Overall, this investigation mainly showed the profile of cardiotoxicities caused by ICIs, which varied between different ICI therapies, but also shared some similarities in specific symptoms such as myocarditis. Therefore, it is vital and urgent to recognize and manage ICI-related cardiotoxicities, known to frequently occur in clinical practice, at the earliest point.
Collapse
Affiliation(s)
- Chenxin Chen
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Ting Chen
- Department of Health Statistics, Second Military Medical University, Shanghai, China
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai, China
| | - Jizhou Liang
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Xiaojing Guo
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Jinfang Xu
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Yi Zheng
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Zhijian Guo
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Lijie Chi
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Lianhui Wei
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Xiao Chen
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Xiaofei Ye
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| | - Jia He
- Department of Health Statistics, Second Military Medical University, Shanghai, China
| |
Collapse
|
46
|
Drobni ZD, Alvi RM, Taron J, Zafar A, Murphy SP, Rambarat PK, Mosarla RC, Lee C, Zlotoff DA, Raghu VK, Hartmann SE, Gilman HK, Gong J, Zubiri L, Sullivan RJ, Reynolds KL, Mayrhofer T, Zhang L, Hoffmann U, Neilan TG. Association Between Immune Checkpoint Inhibitors With Cardiovascular Events and Atherosclerotic Plaque. Circulation 2020; 142:2299-2311. [PMID: 33003973 PMCID: PMC7736526 DOI: 10.1161/circulationaha.120.049981] [Citation(s) in RCA: 353] [Impact Index Per Article: 70.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) treat an expanding range of cancers. Consistent basic data suggest that these same checkpoints are critical negative regulators of atherosclerosis. Therefore, our objectives were to test whether ICIs were associated with accelerated atherosclerosis and a higher risk of atherosclerosis-related cardiovascular events. METHODS The study was situated in a single academic medical center. The primary analysis evaluated whether exposure to an ICI was associated with atherosclerotic cardiovascular events in 2842 patients and 2842 controls matched by age, a history of cardiovascular events, and cancer type. In a second design, a case-crossover analysis was performed with an at-risk period defined as the 2-year period after and the control period as the 2-year period before treatment. The primary outcome was a composite of atherosclerotic cardiovascular events (myocardial infarction, coronary revascularization, and ischemic stroke). Secondary outcomes included the individual components of the primary outcome. In addition, in an imaging substudy (n=40), the rate of atherosclerotic plaque progression was compared from before to after the ICI was started. All study measures and outcomes were blindly adjudicated. RESULTS In the matched cohort study, there was a 3-fold higher risk for cardiovascular events after starting an ICI (hazard ratio, 3.3 [95% CI, 2.0-5.5]; P<0.001). There was a similar increase in each of the individual components of the primary outcome. In the case-crossover, there was also an increase in cardiovascular events from 1.37 to 6.55 per 100 person-years at 2 years (adjusted hazard ratio, 4.8 [95% CI, 3.5-6.5]; P<0.001). In the imaging study, the rate of progression of total aortic plaque volume was >3-fold higher with ICIs (from 2.1%/y before 6.7%/y after). This association between ICI use and increased atherosclerotic plaque progression was attenuated with concomitant use of statins or corticosteroids. CONCLUSIONS Cardiovascular events were higher after initiation of ICIs, potentially mediated by accelerated progression of atherosclerosis. Optimization of cardiovascular risk factors and increased awareness of cardiovascular risk before, during, and after treatment should be considered among patients on an ICI.
Collapse
Affiliation(s)
- Zsofia D. Drobni
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Raza M. Alvi
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jana Taron
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amna Zafar
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean P. Murphy
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paula K. Rambarat
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rayma C. Mosarla
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Charlotte Lee
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel A. Zlotoff
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vineet K. Raghu
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah E. Hartmann
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hannah K. Gilman
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jingyi Gong
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leyre Zubiri
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan J. Sullivan
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lili Zhang
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
47
|
Park SH, Kim T, Cha WC, Yoon H, Hwang SY, Shin TG, Sim MS, Jo I, Lee S, Park H, Choi J. Cardiac troponin I predicts clinical outcome of patients with cancer at emergency department. Clin Cardiol 2020; 43:1585-1591. [PMID: 33085130 PMCID: PMC7724208 DOI: 10.1002/clc.23486] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 10/01/2020] [Accepted: 10/12/2020] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND The prognostic ability of cardiac troponin I (TnI) has been demonstrated in general populations and among cardiovascular disease patients, but it has not been evaluated in cancer patients. HYPOTHESIS This study assumes to have the prognostic ability of cardiac troponin in cancer patients visiting the emergency department. METHODS Cancer patients visiting the emergency department were enrolled in this retrospective cohort study. Patients with previously known coronary artery disease or clinically indicated coronary angiography were not included. The maximal value from Siemens ADVIA Centaur troponin I Ultra assay within 24 hours was assessed. The primary endpoint was 180-day all-cause death, including cardiovascular and noncardiovascular death. RESULTS A total of 9135 cancer patients (mean age: 63 years, male gender: 60%) were enrolled. Lowest (0.006 ng/mL), assay-specific <99th % (0.007-0.039 ng/mL), below median ≥ 99th % (0.040-0.129 ng/mL), and above median ≥ 99th % (≥0.130 ng/mL) TnI were found in 4487 (49.1%), 3158 (34.6%), 852 (9.3%), and 638 (7.0%) patients, respectively. There was 3192 (34.9%) all-cause deaths including 137 (1.5%) cardiovascular and 3047 (33.4%) noncardiovascular deaths in the 180-day follow-up period. The risks of all-cause, cardiovascular, and noncardiovascular death increased across higher TnI strata (hazard ratio [HR] = 1.3-2.9; 2.1-9.3; 1.3-1.8; P < .001, all). These findings were consistent within clinical subgroups including solid and hematologic cancers. CONCLUSIONS Cancer patients visiting the emergency department with elevated troponin I were at increased risk of 180-day death. Cancer patients with elevated TnI may need additional evaluation or careful follow-up even without cardiovascular disease diagnosis.
Collapse
Affiliation(s)
- Soo Hyun Park
- Department of Medicine, Graduate SchoolKyung Hee UniversitySeoulSouth Korea
| | - Taerim Kim
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - Won Cul Cha
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - Hee Yoon
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - Sung Yeon Hwang
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - Tae Gun Shin
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - Min Seob Sim
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - IkJoon Jo
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| | - Seung‐Hwa Lee
- Department of MedicineSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulSouth Korea
| | - Hyung‐Doo Park
- Department of Laboratory Medicine and GeneticsSamsung Medical Center, Sungkyunkwan University School of MedicineSeoulSouth Korea
| | - Jin‐Ho Choi
- Department of Emergency Medicine, Samsung Medical CenterSungkyunkwan University School of MedicineSeoulSouth Korea
| |
Collapse
|
48
|
Sławiński G, Wrona A, Dąbrowska-Kugacka A, Raczak G, Lewicka E. Immune Checkpoint Inhibitors and Cardiac Toxicity in Patients Treated for Non-Small Lung Cancer: A Review. Int J Mol Sci 2020; 21:E7195. [PMID: 33003425 PMCID: PMC7582741 DOI: 10.3390/ijms21197195] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is a major cause of cancer-related mortality worldwide, both in men and women. The vast majority of patients are diagnosed with non-small-cell lung cancer (NSCLC, 80-85% of lung cancer cases). Therapeutics named immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment in the last decade. They are monoclonal antibodies, and those directed against PD-1 (programmed cell death protein 1) or PD-L1 (programmed cell death-ligand 1) have been used in the treatment of lung cancer and significantly improved the prognosis of NSCLC patients. However, during treatment with ICIs, immune-related adverse events (irAEs) can occur in any organ and any tissue. At the same time, although cardiac irAEs are relatively rare compared to irAEs in other organs, they have a high mortality rate. The two most common clinical manifestations of immunotherapy-related cardiotoxicity are myocarditis and pericarditis. Various types of arrhythmias have been reported in patients treated with ICIs, including the occurrence of life-threatening complete atrioventricular block or ventricular tachyarrhythmias. Here, we aim to summarize the incidence, clinical manifestations, underlying mechanisms, diagnosis, and treatment strategies for ICI-associated cardiotoxicity as these issues become very important in view of the increasing use of ICI in the treatment of lung cancer.
Collapse
Affiliation(s)
- Grzegorz Sławiński
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| | - Anna Wrona
- Department of Oncology & Radiotherapy, Medical University of Gdańsk, 80-952 Gdańsk, Poland;
| | - Alicja Dąbrowska-Kugacka
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| | - Grzegorz Raczak
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| | - Ewa Lewicka
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| |
Collapse
|
49
|
Fournel L, Boudou-Rouquette P, Prieto M, Hervochon R, Guinet C, Arrondeau J, Alexandre J, Damotte D, Wislez M, Batteux F, Icard P, Goldwasser F, Alifano M. Nivolumab increases pulmonary artery pressure in patients treated for non-small cell lung cancer. Cancer Chemother Pharmacol 2020; 86:497-505. [PMID: 32936316 DOI: 10.1007/s00280-020-04142-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 09/06/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE The widespread use of Nivolumab results in an increasing number of side effects and adverse events. Herein, we evaluated the impact of Nivolumab on crude and normalized pulmonary artery diameter (PAD). METHODS We analyzed clinical, morphometric, pathological and radiological data of lung cancer patients treated by Nivolumab in an 18-month period. Blinded radiological evaluation was performed, by three observers measuring axial PAD and Aorta diameter (AoD) in secondarily matched pre- and post-Nivolumab CT-scans. Correlation between ΔPAD and clinicopathological data was investigated. RESULTS 59 patients receiving Nivolumab for treatment of advanced lung carcinoma were identified. Pre-and post-Nivolumab comparison of CT-scan measures revealed that mean PAD was 26.3 ± 2.8 mm versus 28.0 ± 3.0 mm (p < 0.001), and mean PAD/AoD ratio was 0.82 ± 0.09 versus 0.87 ± 0.11 (p < 0.001), respectively. Median ΔPAD was 0.05 [0.01-0.122] was significantly higher in hypometabolic patients exhibiting low Rest Energy Expenditure (p = 0.03). Patients exhibiting ΔPAD > 1% had significantly lower serum albumin level (p = 0.03), and higher nutritional risk (p = 0.02), compared to others. Unlike Nivolumab therapy, there was no increase of PAD after chemotherapy in the same cohort of patients with available scans (n = 45, 25.9 ± 2.9 mm pre-chemotherapy versus 25.7 ± 2.4 mm post-chemotherapy, p = 0.51). Anti-PD-1 treatment was associated with immune-related adverse events in 11 (18.6%) cases including 2 cases of life-threatening acute pulmonary hypertension, both exhibiting post-treatment PAD/AoD ratio > 1. CONCLUSION Nivolumab is associated to PAD enlargement, a potential marker of pulmonary hypertension, sometimes leading to lethal adverse events. Careful CT-scan and echocardiographic evaluation of PAD should be part of the therapeutic work-up of patients receiving Nivolumab, especially those suffering cancer-associated malnutrition.
Collapse
Affiliation(s)
- Ludovic Fournel
- Thoracic Surgery Department, Cochin Hospital, AP-HP.Center-University of Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France. .,Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.
| | - Pascaline Boudou-Rouquette
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.,Oncology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Mathilde Prieto
- Thoracic Surgery Department, Cochin Hospital, AP-HP.Center-University of Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France
| | - Remi Hervochon
- Radiology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Claude Guinet
- Radiology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Jennifer Arrondeau
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.,Oncology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Jérôme Alexandre
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.,Oncology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Diane Damotte
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.,Pathology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Marie Wislez
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.,Respiratory Medicine and Thoracic Oncology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Frédéric Batteux
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.,Biology and Immunology Department, AP-HP.Center, University of Paris, Paris, France
| | - Philippe Icard
- Thoracic Surgery Department, Cochin Hospital, AP-HP.Center-University of Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.,INSERM U-119, UNICAEN, University of Caen-Normandy, Caen, France
| | - François Goldwasser
- Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France.,Oncology Department, Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| | - Marco Alifano
- Thoracic Surgery Department, Cochin Hospital, AP-HP.Center-University of Paris, 27 rue du Faubourg Saint-Jacques, 75014, Paris, France.,Immunomodulatory Therapies Multidisciplinary Study Group (CERTIM), Cochin Hospital, AP-HP.Center-University of Paris, Paris, France
| |
Collapse
|
50
|
Schiffer WB, Deych E, Lenihan DJ, Zhang KW. Coronary and aortic calcification are associated with cardiovascular events on immune checkpoint inhibitor therapy. Int J Cardiol 2020; 322:177-182. [PMID: 32800916 DOI: 10.1016/j.ijcard.2020.08.024] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 07/30/2020] [Accepted: 08/07/2020] [Indexed: 01/09/2023]
Abstract
BACKGROUND Although the incidence of immune checkpoint inhibitor (ICI)-related cardiovascular (CV) toxicity is low, the overall burden of CV events after ICI is unknown. Risk factors for CV events after ICI have yet to be identified. OBJECTIVES We sought to evaluate the association between vascular calcification on routine baseline computed tomography (CT) imaging and CV events following ICI. METHOD This was a single-center, retrospective cohort study of 76 patients referred to Cardio-Oncology with prior ICI treatment. Coronary and aortic calcification on non-gated chest and abdominal CT imaging were qualitatively assessed. The association of baseline clinical parameters and vascular calcification with symptomatic heart failure (HF), acute coronary syndrome, myocarditis, symptomatic arrhythmia, or pericardial effusion after ICI was evaluated. RESULTS Over 11 months of follow-up, there were 80 CV events that occurred in 49 patients. Worse coronary and aortic calcification on pre-treatment CT imaging was seen in patients with a CV event (p = .018 and p = .014, respectively). There were no differences in traditional CV risk factors between those with and without a CV event. Eighteen patients (37%) were restarted on ICI therapy after a non- myocarditis or symptomatic systolic HF CV event without recurrent events or mortality over 13 months of follow-up. CONCLUSIONS Symptomatic HF was the most common CV event seen after ICI therapy. Worse coronary and aortic calcification on baseline CT imaging was associated with CV events following ICI. With careful clinical evaluation, selected patients may be re-treated with ICI following a non- myocarditis or symptomatic systolic HF CV event.
Collapse
Affiliation(s)
- Walter B Schiffer
- Department of Internal Medicine, Barnes-Jewish Hospital, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Elena Deych
- Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Daniel J Lenihan
- Cardio-Oncology Center of Excellence, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, United States of America
| | - Kathleen W Zhang
- Cardio-Oncology Center of Excellence, Cardiovascular Division, Washington University School of Medicine, St. Louis, MO, United States of America.
| |
Collapse
|