101
|
Banerjee A, Narasimhulu CA, Singla DK. Immune interactions in pembrolizumab (PD-1 inhibitor) cancer therapy and cardiovascular complications. Am J Physiol Heart Circ Physiol 2023; 325:H751-H767. [PMID: 37594487 PMCID: PMC10659324 DOI: 10.1152/ajpheart.00378.2023] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/09/2023] [Accepted: 08/09/2023] [Indexed: 08/19/2023]
Abstract
The use of immunotherapies like pembrolizumab (PEM) is increasingly common for the management of numerous cancer types. The use of PEM to bolster T-cell response against tumor growth is well documented. However, the interactions PEM has on other immune cells to facilitate tumor regression and clearance is unknown and warrants further investigation. In this review, we present literature findings that have reported the interactions of PEM in stimulating innate and adaptive immune cells, which enhance cytotoxic phenotypes. This triggers secretion of cytokines and chemokines, which have both beneficial and detrimental effects. We also describe how this leads to the development of rare but underreported occurrence of PEM-induced immune-related cardiovascular complications that arise suddenly and progress rapidly to debilitating and fatal consequences. This review encourages further research and investigation of PEM-induced cardiovascular complications and other immune cell interactions in patients with cancer. As PEM therapy in treating cancer types is expanding, we expect that this review will inform health care professionals of diverse specializations of medicine like dermatology (melanoma skin cancers), ophthalmology (eye cancers), and pathology (hematological malignancies) about PEM-induced cardiac complications.
Collapse
Affiliation(s)
- Abha Banerjee
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| | - Chandrakala Aluganti Narasimhulu
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| | - Dinender K Singla
- Division of Metabolic and Cardiovascular Sciences, Burnett School of Biomedical Sciences, College of Medicine, University of Central Florida, Orlando, Florida, United States
| |
Collapse
|
102
|
Vasbinder A, Ismail A, Salem JE, Hayek SS. Role of Biomarkers in the Management of Immune-Checkpoint Inhibitor-Related Myocarditis. Curr Cardiol Rep 2023; 25:959-967. [PMID: 37436648 PMCID: PMC11729503 DOI: 10.1007/s11886-023-01915-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 06/29/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitor (ICI)-related myocarditis poses a major clinical challenge given its non-specific presentation, rapid progression, and high mortality rate. Here, we review the role of blood-based biomarkers in the clinical management of patients with ICI-related myocarditis. RECENT FINDINGS Myocardial injury, its unique pattern, and the co-occurrence with myositis are defining features of ICI-related myocarditis. Non-cardiac biomarkers, specifically creatinine phosphokinase, precedes the symptomatic presentation and is highly sensitive for diagnosing ICI-related myocarditis, making them useful screening biomarkers. Combined elevations in cardiac troponins and non-cardiac biomarkers improve the confidence of an ICI myocarditis diagnosis. High troponin and creatinine phosphokinase levels are strongly associated with severe outcomes. We propose biomarker-based algorithms for the monitoring and diagnosis of ICI-related myocarditis. Biomarkers, such as cardiac troponins and creatine phosphokinase, can be used in combination in the monitoring, diagnosis, and prognostication of patients with ICI-related myocarditis.
Collapse
Affiliation(s)
- Alexi Vasbinder
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Frankel Cardiovascular Center, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, MI, 48109, USA
| | - Anis Ismail
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Frankel Cardiovascular Center, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, MI, 48109, USA
| | - Joe-Elie Salem
- Department of Pharmacology and Clinical Investigation Centre, Pitié-Salpetriere Hospital, Sorbonne Universite, Paris, France
| | - Salim S Hayek
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan, Frankel Cardiovascular Center, 1500 E Medical Center Dr, CVC #2709, Ann Arbor, MI, 48109, USA.
| |
Collapse
|
103
|
Raschi E, Rossi S, De Giglio A, Fusaroli M, Burgazzi F, Rinaldi R, Potena L. Cardiovascular Toxicity of Immune Checkpoint Inhibitors: A Guide for Clinicians. Drug Saf 2023; 46:819-833. [PMID: 37341925 PMCID: PMC10442274 DOI: 10.1007/s40264-023-01320-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/10/2023] [Indexed: 06/22/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment and care of patients with cancer owing to unique features, including the occurrence of the so-called immune-related adverse events (irAEs). A multidisciplinary team, possibly including a cardio-oncology specialist, is warranted to achieve a favorable patient outcome. Cardiovascular toxicity, especially myocarditis, emerged as a life-threatening irAE in the real-word setting, and the European Society of Cardiology has recently published the first guideline on cardio-oncology to increase awareness and promote a standardized approach to tackle this complex multimodal issue, including diagnostic challenges, assessment, treatment, and surveillance of patients with cancer receiving ICIs. In this article, through a question & answer format made up of case vignettes, we offer a clinically oriented overview on the latest advancements of ICI-related cardiovascular toxicity, focusing on myocarditis and associated irAEs (myositis and myasthenia gravis within the so-called overlap syndrome), with the purpose of assisting clinicians and healthcare professionals in daily clinical practice.
Collapse
Affiliation(s)
- Emanuel Raschi
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy.
| | - Simone Rossi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
- Department of Biomedical and Neuromotor Sciences (DIBINEM), Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Andrea De Giglio
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
- Medical Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Michele Fusaroli
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Flavio Burgazzi
- Pharmacology Unit, Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Via Irnerio 48, 40126, Bologna, Italy
| | - Rita Rinaldi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bologna, Italy
| | - Luciano Potena
- Unit of Heart Failure and Transplantation, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| |
Collapse
|
104
|
Chitsazan M, Amin A, Ladel L, Baig A, Chitsazan M. Cardiovascular Toxicity Associated With Immune Checkpoint Inhibitor Therapy: A Comprehensive Review. Crit Pathw Cardiol 2023; 22:69-82. [PMID: 37363862 DOI: 10.1097/hpc.0000000000000327] [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: 06/28/2023]
Abstract
Immune checkpoint inhibitors (ICIs), a significant breakthrough treatment of cancer, exert their function through enhancing the immune system's ability to recognize and attack cancer cells. However, these revolutionary cancer treatments have been associated with a range of immune-related adverse effects, including cardiovascular toxicity. The most commonly reported cardiovascular toxicities associated with ICIs are myocarditis, pericarditis, arrhythmias, and vasculitis. These cardiovascular manifestations are often severe and can lead to life-threatening complications. Therefore, prompt identification and management of these toxicities is critical, and a multidisciplinary teamwork by cardiologists and oncologists are required to ensure optimal patient outcomes. In this review, we summarize the current knowledge on the mechanisms underlying ICI-associated cardiovascular toxicity, clinical presentations of these toxicities, potential risk factors, diagnosis, management, and surveillance strategies during ICI therapy. While ICIs have already transformed cancer treatment, further research is needed to better understand and manage their immune-related cardiovascular effects, and possibly, to identify biomarkers which can predict the occurrence of these cardiovascular complications.
Collapse
Affiliation(s)
| | - Ahmad Amin
- Medstar Union Memorial Hospital, Baltimore, MD
| | - Luisa Ladel
- From the Department of Medicine, Norwalk Hospital, Norwalk, CT
| | - Alyza Baig
- From the Department of Medicine, Norwalk Hospital, Norwalk, CT
| | - Mitra Chitsazan
- Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
105
|
Sanadgol G, Samimi S, Shirini D, Nakhaei P, Mohseni M, Alizadehasl A. Right ventricle toxicity in cancer treatment: a focused review on cardiac imaging. Future Cardiol 2023; 19:537-545. [PMID: 37830360 DOI: 10.2217/fca-2022-0024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023] Open
Abstract
Background: The right ventricle (RV) remains the 'forgotten chamber' in the clinical assessment of cancer therapy-related cardiac dysfunction (CTRCD). Aim: We aimed to review the role that various cardiac imaging modalities play in RV assessment as part of the integrative management of patients undergoing cancer therapy. Discussion: RV assessment remains challenging by traditional 2D echocardiography. In this review we discuss other parameters such as right atrial strain, and other echocardiographic modalities such as 3D and stress echocardiography. We also elaborate on the specific role that cardiac magnetic resonance imaging and equilibrium radionuclide angiocardiography can play in assessing the RV. Conclusion: Biventricular function should be monitored following chemotherapy for early detection of subclinical CTRCD and possible solitary RV changes.
Collapse
Affiliation(s)
- Ghazal Sanadgol
- Shahid-Beheshti University of Medical Sciences, Tehran, 1983969411, Iran
| | - Sahar Samimi
- Tehran University of Medical Sciences, Tehran, 1416634793, Iran
| | - Dorsa Shirini
- Cardiovascular Research Center, Shahid Beheshti University of Medical, Tehran, 1983969411, Iran Sciences
| | - Pooria Nakhaei
- Heart Valve Disease Research Center, Rajaie Cardiovascular Medical & Research Center, Iran University of Medical Sciences, Tehran, 1995614331, Iran
| | - Mina Mohseni
- Department of Cardio-oncology Research, Rajaie Cardiovascular Medical & Research Center, Iran University of Medical Sciences, Tehran, 1995614331, Iran
| | - Azin Alizadehasl
- Professor of Cardiology, Echocardiologist, Cardio-oncologist, Cardio-oncology Research Center, Shaheed Rajaie Cardiovascular Medical & Research Center, Iran University of Medical Science, Tehran, 1995614331, Iran
| |
Collapse
|
106
|
Toribio-García I, Olivares-Hernández A, Miramontes-González JP, Domínguez LP, Martín García A, Eiros Bachiller R, Figuero-Pérez L, Garijo Martínez M, Roldán Ruiz J, Bellido Hernández L, Fonseca-Sánchez E, Luis Sánchez P, del Barco-Morillo E. Cardiotoxicity Secondary to Immune Checkpoint Inhibitors in the Elderly: Safety in Real-World Data. Cancers (Basel) 2023; 15:4293. [PMID: 37686569 PMCID: PMC10486692 DOI: 10.3390/cancers15174293] [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: 07/02/2023] [Revised: 08/04/2023] [Accepted: 08/24/2023] [Indexed: 09/10/2023] Open
Abstract
INTRODUCTION Immunotherapy represents a key pillar of cancer treatments, with high response rates and long survival. Its use is increasing, mainly at the expense of the geriatric population due to the ageing of this population. However, despite its benefit, its safety in certain areas such as cardiotoxicity is largely unknown. The aim of this study is to assess the safety of immunotherapy in elderly patients using real-world data. METHODS This is an ambispective study of patients ≥ 70 years old with solid tumours who were treated with immunotherapy at the University Hospital of Salamanca. Cardiotoxicity was assessed using the CTCAEv5.0 criteria. RESULTS In total, 195 patients were included (76.9% male and 23.1% female), with a mean age of 75 years [70-93]. The percentage of patients with cardiotoxicity was 1.54%; 1.35% of patients with previous heart disease were diagnosed with cardiotoxicity, and 1.65% of those without previous heart disease were diagnosed with cardiotoxicity. The median time from the initiation of treatment until the cardiac event was 45 days [14-96]. The most frequent toxicity was myocarditis in 66.7% of patients, followed by arrhythmias in 33.3% of patients. CONCLUSIONS Immunotherapy is shown to be a safe treatment in elderly cancer patients in terms of cardiotoxicity. The event rate shows no difference between patients with or without cardiac comorbidity.
Collapse
Affiliation(s)
| | - Alejandro Olivares-Hernández
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
| | - José Pablo Miramontes-González
- Department of Internal Medicine, University Hospital Rio Hortega, 47012 Valladolid, Spain;
- Department of Medicine, University of Valladolid, 45005 Valladolid, Spain
| | - Luis Posado Domínguez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
| | - Ana Martín García
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
- Department of Cardiology, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Rocío Eiros Bachiller
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
- Department of Cardiology, University Hospital of Salamanca, 37007 Salamanca, Spain
| | - Luis Figuero-Pérez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
| | - María Garijo Martínez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
| | - Jonnathan Roldán Ruiz
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
| | - Lorena Bellido Hernández
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Emilio Fonseca-Sánchez
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Pedro Luis Sánchez
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
- Department of Cardiology, University Hospital of Salamanca, 37007 Salamanca, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Edel del Barco-Morillo
- Department of Medical Oncology, University Hospital of Salamanca, 37007 Salamanca, Spain; (L.P.D.); (L.F.-P.); (M.G.M.); (J.R.R.); (L.B.H.); (E.F.-S.)
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (A.M.G.); (R.E.B.); (P.L.S.)
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
107
|
Mikail N, Chequer R, Imperiale A, Meisel A, Bengs S, Portmann A, Gimelli A, Buechel RR, Gebhard C, Rossi A. Tales from the future-nuclear cardio-oncology, from prediction to diagnosis and monitoring. Eur Heart J Cardiovasc Imaging 2023; 24:1129-1145. [PMID: 37467476 PMCID: PMC10501471 DOI: 10.1093/ehjci/jead168] [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: 04/25/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
Cancer and cardiovascular diseases (CVD) often share common risk factors, and patients with CVD who develop cancer are at high risk of experiencing major adverse cardiovascular events. Additionally, cancer treatment can induce short- and long-term adverse cardiovascular events. Given the improvement in oncological patients' prognosis, the burden in this vulnerable population is slowly shifting towards increased cardiovascular mortality. Consequently, the field of cardio-oncology is steadily expanding, prompting the need for new markers to stratify and monitor the cardiovascular risk in oncological patients before, during, and after the completion of treatment. Advanced non-invasive cardiac imaging has raised great interest in the early detection of CVD and cardiotoxicity in oncological patients. Nuclear medicine has long been a pivotal exam to robustly assess and monitor the cardiac function of patients undergoing potentially cardiotoxic chemotherapies. In addition, recent radiotracers have shown great interest in the early detection of cancer-treatment-related cardiotoxicity. In this review, we summarize the current and emerging nuclear cardiology tools that can help identify cardiotoxicity and assess the cardiovascular risk in patients undergoing cancer treatments and discuss the specific role of nuclear cardiology alongside other non-invasive imaging techniques.
Collapse
Affiliation(s)
- Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Renata Chequer
- Department of Nuclear Medicine, Bichat University Hospital, AP-HP, University Diderot, 75018 Paris, France
| | - Alessio Imperiale
- Nuclear Medicine, Institut de Cancérologie de Strasbourg Europe (ICANS), University Hospitals of Strasbourg, 67093 Strasbourg, France
- Molecular Imaging-DRHIM, IPHC, UMR 7178, CNRS/Unistra, 67093 Strasbourg, France
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Kantonsspital Glarus, Burgstrasse 99, 8750 Glarus, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Alessia Gimelli
- Imaging Department, Fondazione CNR/Regione Toscana Gabriele Monasterio, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Cathérine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Cardiology, University Hospital Inselspital Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| |
Collapse
|
108
|
De Camilli A, Fischer G. Novel Cellular and Immunotherapy: Toxicities and Perioperative Implications. Curr Oncol 2023; 30:7638-7653. [PMID: 37623035 PMCID: PMC10453139 DOI: 10.3390/curroncol30080554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/19/2023] [Accepted: 08/04/2023] [Indexed: 08/26/2023] Open
Abstract
Targeted cellular and immunotherapies have welcomed a new chapter in multi-modal cancer therapy. These agents harness our innate immune system and destroy malignant cells in a precise way as compared with "legacy" chemotherapeutic agents that largely rely on abolishing cell division. New therapies can augment the T-cell recognition of tumor antigens and effectively prevent tumor cells from their historically successful ability to evade immune recognition. These novel agents cause acute and chronic toxicities to a variety of organ systems (enteritis, pneumonitis, hypophysitis, and hepatitis), and this may masquerade as other chronic illnesses or paraneoplastic effects. As the perioperative footprint of cancer patients increases, it is essential that perioperative providers-anesthesiologists, surgeons, nurse anesthetists, and inpatient hospital medicine providers-be up to date on the physiologic mechanisms that underlie these new therapies as well as their acute and subacute toxicity profiles. Immunotherapy toxicity can significantly impact perioperative morbidity as well as influence perioperative management, such as prophylaxis for adrenal insufficiency, preoperative pulmonary assessment, and screening for thyroid dysfunction, among others.
Collapse
Affiliation(s)
| | - Gregory Fischer
- Memorial Sloan Kettering, 1275 York Avenue, New York, NY 10065, USA
| |
Collapse
|
109
|
Shyam Sunder S, Sharma UC, Pokharel S. Adverse effects of tyrosine kinase inhibitors in cancer therapy: pathophysiology, mechanisms and clinical management. Signal Transduct Target Ther 2023; 8:262. [PMID: 37414756 PMCID: PMC10326056 DOI: 10.1038/s41392-023-01469-6] [Citation(s) in RCA: 141] [Impact Index Per Article: 70.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 04/06/2023] [Accepted: 04/23/2023] [Indexed: 07/08/2023] Open
Abstract
Since their invention in the early 2000s, tyrosine kinase inhibitors (TKIs) have gained prominence as the most effective pathway-directed anti-cancer agents. TKIs have shown significant utility in the treatment of multiple hematological malignancies and solid tumors, including chronic myelogenous leukemia, non-small cell lung cancers, gastrointestinal stromal tumors, and HER2-positive breast cancers. Given their widespread applications, an increasing frequency of TKI-induced adverse effects has been reported. Although TKIs are known to affect multiple organs in the body including the lungs, liver, gastrointestinal tract, kidneys, thyroid, blood, and skin, cardiac involvement accounts for some of the most serious complications. The most frequently reported cardiovascular side effects range from hypertension, atrial fibrillation, reduced cardiac function, and heart failure to sudden death. The potential mechanisms of these side effects are unclear, leading to critical knowledge gaps in the development of effective therapy and treatment guidelines. There are limited data to infer the best clinical approaches for the early detection and therapeutic modulation of TKI-induced side effects, and universal consensus regarding various management guidelines is yet to be reached. In this state-of-the-art review, we examine multiple pre-clinical and clinical studies and curate evidence on the pathophysiology, mechanisms, and clinical management of these adverse reactions. We expect that this review will provide researchers and allied healthcare providers with the most up-to-date information on the pathophysiology, natural history, risk stratification, and management of emerging TKI-induced side effects in cancer patients.
Collapse
Affiliation(s)
- Sunitha Shyam Sunder
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
| | - Umesh C Sharma
- Division of Cardiovascular Medicine, Jacob's School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, USA
| | - Saraswati Pokharel
- Cardio-Oncology Research Group, Department of Pathology and Laboratory Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA.
| |
Collapse
|
110
|
Inno A, Tarantini L, Parrini I, Spallarossa P, Maurea N, Bisceglia I, Silvestris N, Russo A, Gori S. Cardiovascular Effects of Immune Checkpoint Inhibitors: More Than Just Myocarditis. Curr Oncol Rep 2023; 25:743-751. [PMID: 37017825 DOI: 10.1007/s11912-023-01411-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/06/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors have reshaped the treatment of cancer, but they are characterized by peculiar toxicity consisting of immune-related adverse events that may potentially affect any organ or system. In this review, we summarize data on clinical presentation, diagnosis, pathogenesis, and management of the main immune-related cardiovascular toxicities of immune checkpoint inhibitors. RECENT FINDINGS The most relevant immune-related cardiovascular toxicity is myocarditis, but other non-negligible reported events include non-inflammatory heart failure, conduction abnormalities, pericardial disease, and vasculitis. More recently, growing evidence suggests a role for immune checkpoint inhibitors in accelerating atherosclerosis and promoting plaque inflammation, thus leading to myocardial infarction. Immune checkpoint inhibitors are associated with several forms of cardiovascular toxicity; thus, an accurate cardiovascular baseline evaluation and periodical monitoring are required. Furthermore, the optimization of cardiovascular risk factors before, during, and after treatment may contribute to mitigating both short-term and long-term cardiovascular toxicity of these drugs.
Collapse
Affiliation(s)
- Alessandro Inno
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, Via Don A Sempreboni 5, 37024, Negrar Di Valpolicella, VR, Italy.
| | - Luigi Tarantini
- Cardiologia Ospedaliera, AUSL - IRCCS in Tecnologie Avanzate E Modelli Assistenziali in Oncologia, Reggio Emilia, Italy
| | - Iris Parrini
- Dipartimento Di Cardiologia, Ospedale Mauriziano, Turin, Italy
| | - Paolo Spallarossa
- Clinica Di Malattie Dell'Apparato Cardiovascolare, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicola Maurea
- Struttura Complessa Cardiologia, Istituto Nazionale Tumori Di Napoli IRCCS Fondazione G. Pascale, Naples, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - Nicola Silvestris
- Oncologia Medica, Dipartimento Di Patologia Umana "G. Barresi", Università Di Messina, Messina, Italy
| | - Antonio Russo
- Dipartimento Di Discipline Chirurgiche, Oncologiche E Stomatologiche, Università Di Palermo, Palermo, Italy
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, Via Don A Sempreboni 5, 37024, Negrar Di Valpolicella, VR, Italy
| |
Collapse
|
111
|
Yousif LI, Screever EM, Versluis D, Aboumsallem JP, Nierkens S, Manintveld OC, de Boer RA, Meijers WC. Risk Factors for Immune Checkpoint Inhibitor-Mediated Cardiovascular Toxicities. Curr Oncol Rep 2023; 25:753-763. [PMID: 37079251 PMCID: PMC10256640 DOI: 10.1007/s11912-023-01414-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/21/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors (ICIs) have improved the field of cancer, especially in patients with advanced malignancies. Nevertheless, cardiovascular immune-related adverse events (irAEs) with high mortality and morbidity have been observed, including myocarditis, pericarditis, and vasculitis. To date, only a few clinical risk factors have been described and are currently being investigated. RECENT FINDINGS In this review, we address the four most prevailing risk factors for cardiovascular irAEs. ICI combination therapy is a predominant risk factor for developing ICI-mediated myocarditis. Additionally, ICI combined with other anti-cancer treatments (e.g., tyrosine kinase inhibitors, radiation, chemotherapy) seems to increase the risk of developing cardiovascular irAEs. Other risk factors include female sex, pre-existing cardiovascular disease, and specific tumors, on which we will further elaborate in this review. An a priori risk strategy to determine who is at risk to develop these cardiovascular irAEs is needed. Insights into the impact of risk factors are therefore warranted to help clinicians improve care and disease management in these patients.
Collapse
Affiliation(s)
- Laura I. Yousif
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000CA Rotterdam, The Netherlands
| | - Elles M. Screever
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000CA Rotterdam, The Netherlands
| | - Daniëlle Versluis
- Graduate School of Life Science, Utrecht University, P.O. Box 80125, 3508 TC Utrecht, The Netherlands
| | - Joseph Pierre Aboumsallem
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000CA Rotterdam, The Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, Utrecht University, University Medical Center Utrecht, P.O. Box 85500, 3508 GA Utrecht, The Netherlands
- Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584CS Utrecht, The Netherlands
| | - Olivier C. Manintveld
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000CA Rotterdam, The Netherlands
| | - Rudolf A. de Boer
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000CA Rotterdam, The Netherlands
| | - Wouter C. Meijers
- Department of Cardiology, Thorax Center, Erasmus University Medical Center, P.O. Box 2040, 3000CA Rotterdam, The Netherlands
| |
Collapse
|
112
|
Özdemir BC, Espinosa da Silva C, Arangalage D, Monney P, Guler SA, Huynh-Do U, Stirnimann G, Possamai L, Trepp R, Hoepner R, Salmen A, Gerard CL, Hruz P, Christ L, Rothschild SI. Multidisciplinary recommendations for essential baseline functional and laboratory tests to facilitate early diagnosis and management of immune-related adverse events among cancer patients. Cancer Immunol Immunother 2023; 72:1991-2001. [PMID: 37017694 PMCID: PMC10264466 DOI: 10.1007/s00262-023-03436-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 03/22/2023] [Indexed: 04/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have fundamentally changed the treatment landscape of various cancers. While ICI treatments result in improved survival, quality of life and are cost-effective, the majority of patients experience at least one immune-related adverse event (irAE). Many of these side effects cause little discomfort or are asymptomatic; however, irAEs can affect any organ and are potentially life-threatening. Consequently, early diagnosis and appropriate treatment of irAEs are critical for optimizing long-term outcomes and quality of life in affected patients. Some irAEs are diagnosed according to typical symptoms, others by abnormal findings from diagnostic tests. While there are various guidelines addressing the management of irAEs, recommendations for the early recognition of irAEs as well as the optimal extent and frequency of laboratory tests are mostly lacking. In clinical practice, blood sampling is usually performed before each ICI administration (i.e., every 2-3 weeks), often for several months, representing a burden for patients as well as health care systems. In this report, we propose essential laboratory and functional tests to improve the early detection and management of irAEs and in cancer patients treated with ICIs. These multidisciplinary expert recommendations regarding essential laboratory and functional tests can be used to identify possible irAEs at an early time point, initiate appropriate interventions to improve patient outcomes, and reduce the burden of blood sampling during ICI treatment.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Medical Oncology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| | - Cristina Espinosa da Silva
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, La Jolla, CA, USA
- Division of Epidemiology and Biostatistics, School of Public Health, San Diego State University, San Diego, USA
| | - Dimitri Arangalage
- Department of Cardiology, INSERM U1148, Bichat Hospital, University of Paris, Paris, France
| | - Pierre Monney
- Department of Cardiology, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
| | - Sabina A Guler
- Department of Pulmonary Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Uyen Huynh-Do
- Department of Nephrology and Hypertension, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Guido Stirnimann
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lucia Possamai
- Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, UK
| | - Roman Trepp
- Department of Diabetes, Endocrinology, Nutritional Medicine and Metabolism (UDEM), Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Robert Hoepner
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Anke Salmen
- Department of Neurology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Camille L Gerard
- Department of Oncology, Lausanne University Hospital (CHUV), University of Lausanne, Lausanne, Switzerland
- The Francis Crick Institute, London, UK
| | - Petr Hruz
- Department of Gastroenterology, University Hospital Basel, Basel, Switzerland
| | - Lisa Christ
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Sacha I Rothschild
- Department of Medical Oncology, University Hospital Basel, Basel, Switzerland
- Department Internal Medicine, Center for Oncology and Hematology, Cantonal Hospital Baden, Baden, Switzerland
| |
Collapse
|
113
|
Sadeq MA, Ashry MH, Ghorab RMF, Afify AY. Causes of death among patients with cutaneous melanoma: a US population-based study. Sci Rep 2023; 13:10257. [PMID: 37355743 PMCID: PMC10290704 DOI: 10.1038/s41598-023-37333-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023] Open
Abstract
Research on mortality outcomes and non-cancer-related causes of death in patients with cutaneous melanoma (CM) remains limited. This study aimed to identify the prevalence of non-cancer-related deaths following CM diagnosis. The data of 224,624 patients diagnosed with malignant CM in the United States between 2000 and 2019 were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. We stratified our cohort based on their melanoma stage at diagnosis and further calculated standardized mortality ratios (SMRs) for each cause of death, comparing their relative risk to that of the general US population. The total number of fatalities among melanoma patients was 60,110, representing 26.8% of the total cases. The percentage of deaths is directly proportional to the disease stage, reaching 80% in distant melanoma. The highest fatalities among the localized melanoma group (25,332; 60.5%) occurred from non-cancer causes, followed by melanoma-attributable deaths (10,817; 25.8%). Conversely, melanoma is the leading cause of death in regional and distant melanoma cohorts. Cardiovascular and cerebrovascular diseases were the most prevalent non-cancer causes of death among the three disease-stage cohorts. Compared to the general population, we did not observe an increased risk of death due to non-cancer causes in the localized CM cohort, while patients diagnosed with regional and distant CMs had a statistically significant higher risk of death from all the reported major causes of death.
Collapse
Affiliation(s)
- Mohammed Ahmed Sadeq
- Faculty of Medicine, Misr University for Science and Technology, 6th of October, Giza, Egypt
| | | | | | | |
Collapse
|
114
|
Chen M, Yang P, Xin Z, Chen J, Zou W, Zhou L, Yang L, Peng J, Peng H. Toxoplasma gondii gra5 deletion mutant protects hosts against Toxoplasma gondii infection and breast tumors. Front Immunol 2023; 14:1173379. [PMID: 37426671 PMCID: PMC10327641 DOI: 10.3389/fimmu.2023.1173379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 04/13/2023] [Indexed: 07/11/2023] Open
Abstract
Toxoplasma gondii is the causative agent of toxoplasmosis, a zoonotic disease that poses a threat to human health and a considerable loss to livestock farming. At present, clinical therapeutic drugs mainly target T. gondii tachyzoites and fail to eradicate bradyzoites. Developing a safe and effective vaccine against toxoplasmosis is urgent and important. Breast cancer has become a major public health problem and the therapeutic method needs to be further explored. Many similarities exist between the immune responses caused by T. gondii infection and the immunotherapy for cancers. T. gondii dense granule organelles secrete immunogenic dense granule proteins (GRAs). GRA5 is localized to the parasitophorous vacuole membrane in the tachyzoite stage and the cyst wall in the bradyzoite stage. We found that T. gondii ME49 gra5 knockout strain (ME49Δgra5) was avirulent and failed to form cysts but stimulated antibodies, inflammatory cytokines, and leukocytes infiltration in mice. We next investigated the protective efficacy of ME49Δgra5 vaccination against T. gondii infection and tumor development. All the immunized mice survived the challenge infection of either wild-type RH, ME49, VEG tachyzoites, or ME49 cysts. Moreover, ME49Δgra5 tachyzoite inoculation in situ attenuated the growth of murine breast tumor (4T1) in mice and prevented 4T1's lung metastasis. ME49Δgra5 inoculation upregulated the levels of Th1 cytokines and tumor-infiltrating T cells in the tumor microenvironment and triggered anti-tumor responses by increasing the number of natural killer, B, and T cells, macrophages, and dendritic cells in the spleen. Collectively, these results suggested that ME49Δgra5 was a potent live attenuated vaccine against T. gondii infection and breast cancer.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Hongjuan Peng
- Department of Pathogen Biology, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
115
|
Cobarro L, Valbuena-López S, Contreras C, Ugueto C, Ruiz-Gutiérrez I, Peña-López J, Ruiz-Giménez L, López-Fernández T. Immune Checkpoint Inhibitor-Related Stress Cardiomyopathy: Differential Diagnosis and Key Role of Cardiac Imaging. JACC Case Rep 2023; 16:101881. [PMID: 37396332 PMCID: PMC10313486 DOI: 10.1016/j.jaccas.2023.101881] [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: 02/01/2023] [Revised: 04/20/2023] [Accepted: 04/20/2023] [Indexed: 07/04/2023]
Abstract
A 76-year-old man with stage IV urothelial carcinoma who was receiving atezolizumab presented with dyspnea, elevated cardiac biomarkers, new negative T waves, and left ventricular apical akinesia. Coronary angiography results were normal. Immune checkpoint inhibitor-related myocarditis was suspected, and high-dose corticosteroid treatment was started. Cardiac magnetic resonance showed apical edema, suggesting stress cardiomyopathy. (Level of Difficulty: Beginner.).
Collapse
Affiliation(s)
- Lucía Cobarro
- Department of Cardiology, La Paz University Hospital, Madrid, Spain
| | | | | | - Clara Ugueto
- Department of Cardiology, La Paz University Hospital, Madrid, Spain
| | | | - Jesús Peña-López
- Department of Oncology, La Paz University Hospital, Madrid, Spain
| | | | - Teresa López-Fernández
- Department of Cardiology, La Paz University Hospital, Madrid, Spain
- Cardio-Oncology Unit, La Paz University Hospital, Madrid, Spain
| |
Collapse
|
116
|
Brociek E, Tymińska A, Giordani AS, Caforio ALP, Wojnicz R, Grabowski M, Ozierański K. Myocarditis: Etiology, Pathogenesis, and Their Implications in Clinical Practice. BIOLOGY 2023; 12:874. [PMID: 37372158 PMCID: PMC10295542 DOI: 10.3390/biology12060874] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 05/29/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023]
Abstract
Myocarditis is an inflammatory disease of the myocardium caused by infectious or non-infectious agents. It can lead to serious short-term and long-term sequalae, such as sudden cardiac death or dilated cardiomyopathy. Due to its heterogenous clinical presentation and disease course, challenging diagnosis and limited evidence for prognostic stratification, myocarditis poses a great challenge to clinicians. As it stands, the pathogenesis and etiology of myocarditis is only partially understood. Moreover, the impact of certain clinical features on risk assessment, patient outcomes and treatment options is not entirely clear. Such data, however, are essential in order to personalize patient care and implement novel therapeutic strategies. In this review, we discuss the possible etiologies of myocarditis, outline the key processes governing its pathogenesis and summarize best available evidence regarding patient outcomes and state-of-the-art therapeutic approaches.
Collapse
Affiliation(s)
- Emil Brociek
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| | - Agata Tymińska
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| | - Andrea Silvio Giordani
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy; (A.S.G.); (A.L.P.C.)
| | - Alida Linda Patrizia Caforio
- Cardiology, Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, 35-100 Padova, Italy; (A.S.G.); (A.L.P.C.)
| | - Romuald Wojnicz
- Department of Histology and Cell Pathology in Zabrze, School of Medicine with the Division of Dentistry, Medical University of Silesia, 40-055 Katowice, Poland;
| | - Marcin Grabowski
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| | - Krzysztof Ozierański
- First Department of Cardiology, Medical University of Warsaw, 02-097 Warsaw, Poland; (E.B.); (M.G.); (K.O.)
| |
Collapse
|
117
|
Bae S, Vaysblat M, Ng J, Beccarino N, Makaryus J, Sarkar K. Durvalumab-Associated Myocarditis Initially Presenting With Sinus Bradycardia Progressing Into Complete Heart Block. Cureus 2023; 15:e40171. [PMID: 37431327 PMCID: PMC10329808 DOI: 10.7759/cureus.40171] [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: 06/08/2023] [Indexed: 07/12/2023] Open
Abstract
Durvalumab is a monoclonal antibody that blocks programmed cell death ligand-1 (PD-L1). It has been recently approved for the treatment of advanced urothelial and non-small cell lung cancer (NSCLC) with a more favorable side effect profile compared to traditional chemotherapy agents. We present a case of durvalumab-induced myocarditis complicated by complete heart block (CHB). A 71-year-old male with a history of atrial flutter status post ablation, type 2 diabetes mellitus, hypertension, and non-small cell lung carcinoma (NSCLC) recently started on durvalumab, presented with new sinus bradycardia on electrocardiogram (EKG). His initial labs were notable for a troponin T of 207 ng/L (normal range ≤50). Transthoracic echo (TTE) and computed tomography angiography (CTA) of the coronaries were unremarkable. The hospital course was complicated by CHB on telemetry for 15 minutes. Given hemodynamic instability, cardiac magnetic resonance imaging (MRI) could not be obtained. The patient received transvenous pacing. Electrophysiology and cardiology-oncology were consulted to evaluate for pacemaker implantation as well as management for durvalumab-induced myocarditis. Methylprednisolone 1000 mg intravenous (IV) was started with an improvement in troponin levels but without improvement in CHB. His course was further complicated by polymorphic ventricular tachycardia prompting the placement of a permanent dual-chamber pacemaker. The patient was discharged on a prednisone taper, and durvalumab was discontinued. A diagnosis of durvalumab-induced myocarditis was made based on elevated troponin levels, with the exclusion of coronary artery disease with CTA of the coronaries. The persistence of conduction abnormalities despite treatment with steroids leads to the placement of a permanent pacemaker. Durvalumab falls under the category of immune checkpoint inhibitor (ICI) therapy which are novel agents that have more favorable side effect profiles compared to traditional chemotherapeutic agents. A review of the literature shows myocarditis with arrhythmias as a potentially rare side effect of ICI therapy. Corticosteroid therapy seems to be promising as a potential therapy.
Collapse
Affiliation(s)
- Suhwoo Bae
- Internal Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, USA
| | - Michael Vaysblat
- Internal Medicine, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, USA
| | - Jason Ng
- Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, USA
| | - Nicholas Beccarino
- Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, USA
| | - John Makaryus
- Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, USA
| | - Kumar Sarkar
- Cardiology, Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Manhasset, USA
| |
Collapse
|
118
|
Choudhary HB, Mandlik SK, Mandlik DS. Role of p53 suppression in the pathogenesis of hepatocellular carcinoma. World J Gastrointest Pathophysiol 2023; 14:46-70. [PMID: 37304923 PMCID: PMC10251250 DOI: 10.4291/wjgp.v14.i3.46] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/19/2023] [Accepted: 05/31/2023] [Indexed: 06/01/2023] Open
Abstract
In the world, hepatocellular carcinoma (HCC) is among the top 10 most prevalent malignancies. HCC formation has indeed been linked to numerous etiological factors, including alcohol usage, hepatitis viruses and liver cirrhosis. Among the most prevalent defects in a wide range of tumours, notably HCC, is the silencing of the p53 tumour suppressor gene. The control of the cell cycle and the preservation of gene function are both critically important functions of p53. In order to pinpoint the core mechanisms of HCC and find more efficient treatments, molecular research employing HCC tissues has been the main focus. Stimulated p53 triggers necessary reactions that achieve cell cycle arrest, genetic stability, DNA repair and the elimination of DNA-damaged cells’ responses to biological stressors (like oncogenes or DNA damage). To the contrary hand, the oncogene protein of the murine double minute 2 (MDM2) is a significant biological inhibitor of p53. MDM2 causes p53 protein degradation, which in turn adversely controls p53 function. Despite carrying wt-p53, the majority of HCCs show abnormalities in the p53-expressed apoptotic pathway. High p53 in-vivo expression might have two clinical impacts on HCC: (1) Increased levels of exogenous p53 protein cause tumour cells to undergo apoptosis by preventing cell growth through a number of biological pathways; and (2) Exogenous p53 makes HCC susceptible to various anticancer drugs. This review describes the functions and primary mechanisms of p53 in pathological mechanism, chemoresistance and therapeutic mechanisms of HCC.
Collapse
Affiliation(s)
- Heena B Choudhary
- Department of Pharmacology, BVDU, Poona College of Pharmacy, Pune 411038, Maharashtra, India
| | - Satish K Mandlik
- Department of Pharmaceutics, BVDU, Poona College of Pharmacy, Pune 411038, Maharashtra, India
| | - Deepa S Mandlik
- Department of Pharmacology, BVDU, Poona College of Pharmacy, Pune 411038, Maharashtra, India
| |
Collapse
|
119
|
Shi F, Huang X, Hong Z, Lu N, Huang X, Liu L, Liang T, Bai X. Improvement strategy for immune checkpoint blockade: A focus on the combination with immunogenic cell death inducers. Cancer Lett 2023; 562:216167. [PMID: 37031916 DOI: 10.1016/j.canlet.2023.216167] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 03/19/2023] [Accepted: 04/03/2023] [Indexed: 04/11/2023]
Abstract
Cancer immunotherapies have yielded promising outcomes in various malignant tumors by blocking specific immune checkpoint molecules, such as programmed cell death 1 and cytotoxic T lymphocyte antigen 4. However, only a few patients respond to immune checkpoint blockade therapy because of the poor immunogenicity of tumor cells and immune-suppressive tumor microenvironment. Accumulating evidence suggests that chemotherapeutic agents, including oxaliplatin and doxorubicin, not only mediate direct cytotoxicity in tumor cells but also induce immunogenic cancer cell death to stimulate a powerful anti-cancer immune response in the tumor microenvironment. In this review, we summarize the recent advances in cancer combination therapy based on immune checkpoint inhibitors plus immunogenic cell death inducers. Despite some clinical failures and challenges, immunogenic cell death inducers have displayed great potential when combined with immune checkpoint inhibitors for anti-cancer treatment in both preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Fukang Shi
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Xing Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Zhengtao Hong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Na Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Xin Huang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Lingyue Liu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou, 310058, Zhejiang, China.
| |
Collapse
|
120
|
Wu Y, Xu Y, Xu L. Drug therapy for myocarditis induced by immune checkpoint inhibitors. Front Pharmacol 2023; 14:1161243. [PMID: 37305530 PMCID: PMC10248045 DOI: 10.3389/fphar.2023.1161243] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 05/05/2023] [Indexed: 06/13/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs), including cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 (PD-1), and its ligand 1 (PD-L1), have improved the survival in multiple types of cancers; however, ICIs may cause cardiovascular toxicity. Although rare, ICI-mediated cardiotoxicity is an extremely serious complication with a relatively high mortality. In this review, we discuss the underlying mechanism and clinical manifestations of cardiovascular toxicity induced by ICIs. According to previous studies, multiple signaling pathways are involved in myocarditis induced by ICIs. Further, we summarize the clinical trials of drugs for the treatment of ICI-associated myocarditis. Although these drugs have shown the beneficial effects of alleviating cardiac function and reducing mortality rates, their efficacy is not optimal. Finally, we discuss the therapeutic potential of some novel compounds as well as the underlying mechanisms of their action.
Collapse
Affiliation(s)
- Yihao Wu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yizhou Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Linhao Xu
- Department of Cardiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
- Department of Laboratory Medicine, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
121
|
Uruski P, Matuszewska J, Leśniewska A, Rychlewski D, Niklas A, Mikuła-Pietrasik J, Tykarski A, Książek K. An integrative review of nonobvious puzzles of cellular and molecular cardiooncology. Cell Mol Biol Lett 2023; 28:44. [PMID: 37221467 DOI: 10.1186/s11658-023-00451-y] [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: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/25/2023] Open
Abstract
Oncologic patients are subjected to four major treatment types: surgery, radiotherapy, chemotherapy, and immunotherapy. All nonsurgical forms of cancer management are known to potentially violate the structural and functional integrity of the cardiovascular system. The prevalence and severity of cardiotoxicity and vascular abnormalities led to the emergence of a clinical subdiscipline, called cardiooncology. This relatively new, but rapidly expanding area of knowledge, primarily focuses on clinical observations linking the adverse effects of cancer therapy with deteriorated quality of life of cancer survivors and their increased morbidity and mortality. Cellular and molecular determinants of these relations are far less understood, mainly because of several unsolved paths and contradicting findings in the literature. In this article, we provide a comprehensive view of the cellular and molecular etiology of cardiooncology. We pay particular attention to various intracellular processes that arise in cardiomyocytes, vascular endothelial cells, and smooth muscle cells treated in experimentally-controlled conditions in vitro and in vivo with ionizing radiation and drugs representing diverse modes of anti-cancer activity.
Collapse
Affiliation(s)
- Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Aleksandra Leśniewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland.
| |
Collapse
|
122
|
Mallio CA, Bernetti C, Cea L, Buoso A, Stiffi M, Vertulli D, Greco F, Zobel BB. Adverse Effects of Immune-Checkpoint Inhibitors: A Comprehensive Imaging-Oriented Review. Curr Oncol 2023; 30:4700-4723. [PMID: 37232813 DOI: 10.3390/curroncol30050355] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/22/2023] [Accepted: 05/01/2023] [Indexed: 05/27/2023] Open
Abstract
Immune-checkpoint inhibitors (ICIs) are immunomodulatory monoclonal antibodies, which increase antitumor immunity of the host and facilitate T-cell-mediated actions against tumors. These medications have been used in recent years as a weapon against advanced stage malignancies, such as melanoma, renal cell carcinoma, lymphoma, small or non-small cell lung cancer, and colorectal cancer. Unfortunately, they are not free from possible adverse effects (immune-related adverse events-irAEs) that mainly affect skin, gastrointestinal, hepatic, and endocrine systems. Early diagnosis of irAEs is essential to correctly and rapidly manage patients, with ICIs suspension and therapies administration. Deep knowledge of the imaging and clinical patterns of irAEs is the key to promptly rule out other diagnoses. Here, we performed a review of the radiological signs and differential diagnosis, based on the organ involved. The aim of this review is to provide guidance to recognize the most significant radiological findings of the main irAEs, based on incidence, severity, and the role of imaging.
Collapse
Affiliation(s)
- Carlo Augusto Mallio
- Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Department of Medicine and Surgery, Research Unit of Radiology, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Caterina Bernetti
- Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Department of Medicine and Surgery, Research Unit of Radiology, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Laura Cea
- Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Department of Medicine and Surgery, Research Unit of Radiology, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Andrea Buoso
- Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Department of Medicine and Surgery, Research Unit of Radiology, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Massimo Stiffi
- Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Department of Medicine and Surgery, Research Unit of Radiology, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Daniele Vertulli
- Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Department of Medicine and Surgery, Research Unit of Radiology, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| | - Federico Greco
- Unità Operativa Complessa Diagnostica per Immagini Territoriale Aziendale, Cittadella della Salute Azienda Sanitaria Locale di Lecce, Piazza Filippo Bottazzi, 73100 Lecce, Italy
| | - Bruno Beomonte Zobel
- Department of Medicine and Surgery, Fondazione Policlinico Universitario Campus Bio-Medico, 00128 Rome, Italy
- Department of Medicine and Surgery, Research Unit of Radiology, Università Campus Bio-Medico di Roma, 00128 Roma, Italy
| |
Collapse
|
123
|
Kuhnly NM, Coviello JS. A case study of myopericarditis due to immune checkpoint inhibitor therapy: Proposed surveillance and management. J Am Assoc Nurse Pract 2023; 35:317-321. [PMID: 37000118 PMCID: PMC10568649 DOI: 10.1097/jxx.0000000000000860] [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: 12/07/2022] [Accepted: 02/21/2023] [Indexed: 04/01/2023]
Abstract
ABSTRACT Immune checkpoint inhibitor (ICI) therapy is a treatment modality used in many types of cancer. Immune-related adverse events are relatively common. Cardiovascular adverse events are uncommon, but carry a high mortality rate of 25-50%. They require cessation of therapy. There is currently no universal screening before initiation of ICI therapy to identify patients with cardiovascular risk. There is also no ongoing screening to identify myocarditis and treatment is driven by symptoms. This article provides a case study of a patient who developed myopericarditis and the patient's clinical course. Furthermore, it proposes surveillance for patients before and during ICI therapy to swiftly identify potential cases of myocarditis. There is currently no universal baseline screening for cardiovascular risk in patients planned for ICI therapy. A proposed baseline cardiac evaluation, as well as scheduled surveillance therapy, is outlined in this article. With further education and training, immune-related cardiac adverse events may be more promptly detected, leading to better patient outcomes.
Collapse
|
124
|
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
|
125
|
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
|
126
|
Xiao J, Li X, Wang X, Guan Y, Liu H, Liang J, Li Y, Wang B, Wang J. Clinical characteristics and management of immune checkpoint inhibitor-related cardiotoxicity: A single-center experience. Front Cardiovasc Med 2023; 10:1093383. [PMID: 37089888 PMCID: PMC10115988 DOI: 10.3389/fcvm.2023.1093383] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Accepted: 03/06/2023] [Indexed: 04/08/2023] Open
Abstract
BackgroundImmune checkpoint inhibitors (ICIs) have revolutionized cancer therapy in the past decade and amplify T-cell-mediated immune responses by disrupting immunoinhibitory signals. The augmented T-cell immune response has led to a range of immune-related adverse effects (irAEs). Immune-related cardiotoxicity has been reported in case series but has been underappreciated due to difficulties in diagnosis. This article describes epidemiological, clinical presentation, subtype, and treatment data and a new systematic framework for the clinical management of cardiotoxicity.MethodsData were extracted for cancer patients who received ICIs in a single center between January 1, 2020, and February 28, 2022. ICI-associated cardiotoxicity was clinically diagnosed based on clinical presentations, biochemical biomarkers, and imaging features.ResultsWe identified a total of 12 (2.46%) cases of ICI-related cardiotoxicity from 487 patients who received PD-1 or PD-L1 inhibitors. All patients were diagnosed with advanced or metastatic solid tumors. The severity of ICI-related cardiotoxicity ranged from subclinical cardiac abnormalities (subclinical type) with only asymptomatic troponin-I (TnI) elevations (25.0%) to symptomatic cardiac abnormalities (clinical type) (75.0%). Patients with symptomatic cardiac abnormalities had several manifestations, including tachyarrhythmia (16.7%), bradyarrhythmia (41.7%), or cardiac failure (8.3%). The median immunotherapy exposure time was 1.5 doses (range: 1 to 5), and the median time from the initial immunotherapy to the onset of ICI-related cardiotoxicity was 33.5 days (IQR: 20.3 to 46.8). Most patients, including those with subclinical cardiac abnormalities, were administered systemic corticosteroids (58.3%). One (8.3%) patient was put on mechanical ventilation, one (8.3%) received plasma exchange therapy, one (8.3%) was implanted with a pacemaker, and one (8.3%) was admitted to the ICU. Three patients with symptomatic cardiac abnormalities (25.0%) died, and other patients presented with significant clinical improvement with good outcomes.ConclusionICI-related cardiotoxicity is uncommon but critical with a high mortality rate and poor prognosis, especially for a small group of patients with symptomatic cardiac abnormalities. More attention should be given to cardiotoxicity associated with ICIs, and these patients should be given baseline examinations and biochemical analyses before and after the initiation of immunotherapy, intensive cardiac assessments, an accurate and rapid diagnosis, and timely multidisciplinary management with immunosuppressive agents and other necessary clinical interventions.
Collapse
|
127
|
Parashar S, Akhter N, Paplomata E, Elgendy IY, Upadhyaya D, Scherrer-Crosbie M, Okwuosa TM, Sanghani RM, Chalas E, Lindley KJ, Dent S. Cancer Treatment-Related Cardiovascular Toxicity in Gynecologic Malignancies: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2023; 5:159-173. [PMID: 37144116 PMCID: PMC10152205 DOI: 10.1016/j.jaccao.2023.02.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 02/02/2023] [Accepted: 02/07/2023] [Indexed: 05/06/2023] Open
Abstract
Improvements in early detection and treatment of gynecologic malignancies have led to an increasing number of survivors who are at risk of long-term cardiac complications from cancer treatment. Multimodality therapies for gynecologic malignancies, including conventional chemotherapy, targeted therapeutics, and hormonal agents, place patients at risk of cancer therapy-related cardiovascular toxicity during and following treatment. Although the cardiotoxicity associated with some female predominant cancers (eg, breast cancer) have been well recognized, there has been less recognition of the potential adverse cardiovascular effects of anticancer therapies used to treat gynecologic malignancies. In this review, the authors provide a comprehensive overview of the cancer therapeutic agents used in gynecologic malignancies, associated cardiovascular toxicities, risk factors for cardiotoxicity, cardiac imaging, and prevention strategies.
Collapse
Affiliation(s)
- Susmita Parashar
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, Georgia, USA
| | - Nausheen Akhter
- Division of Cardiology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | | | - Islam Y. Elgendy
- Division of Cardiology, Department of Medicine, University of Kentucky, Lexington, Kentucky, USA
| | - Deepa Upadhyaya
- Division of Cardiology, Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| | - Marielle Scherrer-Crosbie
- Cardiovascular Medicine Division, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Tochukwu M. Okwuosa
- Division of Cardio-Oncology, Department of Medicine, Rush University, Chicago, Illinois, USA
| | - Rupa M. Sanghani
- Division of Cardiology, Department of Medicine, Rush University, Chicago, Illinois, USA
| | - Eva Chalas
- Division of Obstetrics and Gynecology, New York University Long Island School of Medicine, Mineola, New York, USA
| | - Kathryn J. Lindley
- Division of Cardiology, Department of Medicine, Vanderbilt University, Nashville, Tennessee, USA
| | - Susan Dent
- Division of Medical Oncology, Duke Cancer Institute, Duke University, Durham, North Carolina, USA
| |
Collapse
|
128
|
Wang D, Bauersachs J, Berliner D. Immune Checkpoint Inhibitor Associated Myocarditis and Cardiomyopathy: A Translational Review. BIOLOGY 2023; 12:biology12030472. [PMID: 36979163 PMCID: PMC10045178 DOI: 10.3390/biology12030472] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/13/2023] [Accepted: 03/16/2023] [Indexed: 03/30/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized oncology and transformed the treatment of various malignancies. By unleashing the natural immunological brake of the immune system, ICIs were initially considered an effective, gentle therapy with few side effects. However, accumulated clinical knowledge reveals that ICIs are associated with inflammation and tissue damage in multiple organs, leading to immune-related adverse effects (irAEs). Most irAEs involve the skin and gastrointestinal tract; however, cardiovascular involvement is associated with very high mortality rates, and its underlying pathomechanisms are poorly understood. Ranging from acute myocarditis to chronic cardiomyopathies, ICI-induced cardiotoxicity can present in various forms and entities. Revealing the inciting factors, understanding the pathogenesis, and identifying effective treatment strategies are needed to improve the care of tumor patients and our understanding of the immune and cardiovascular systems.
Collapse
Affiliation(s)
- Dong Wang
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| | - Dominik Berliner
- Department of Cardiology and Angiology, Hannover Medical School, 30625 Hannover, Germany
| |
Collapse
|
129
|
Zhang Y, Huang Z, Hu S, Si J, Cheng S, Chen Z, Xue J, Lou X, Peng H, Li Z, Ouyang M, Gao X, Zeng W. The association of preoperative high-sensitivity cardiac troponin i and long-term outcomes in colorectal cancer patients received tumor resection surgery. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2023; 9:12. [PMID: 36864502 PMCID: PMC9979437 DOI: 10.1186/s40959-023-00162-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2022] [Accepted: 02/06/2023] [Indexed: 03/04/2023]
Abstract
BACKGROUND This study aimed to evaluate the association between preoperative hs-cTnI and long-term mortality and major adverse cardiovascular events (MACE) in colorectal cancer patients. METHODS This single-center retrospective cohort study included 1105 consecutive colorectal cancer patients who received tumor resection surgery between January 2018 and June 2020. Inclusion criteria were an age ≥ 18 years and had been tested for hs-cTnI on admission within 7 days prior to tumor resection surgery. Exclusion criteria were emergent surgery, failure to received tumor resection surgery, hospital death, there was clinical evidence of unstable coronary artery disease or pulmonary embolism occurred before operation according to medical record. The primary endpoint was all-cause death. Secondary endpoint was major adverse cardiovascular events (MACE). RESULTS A total of 1105 patients were enrolled: 1032 with normal hs-cTnI and 73 with elevated hs-cTnI. The mean follow-up was 24.4 ± 10.8 months, 176 patients died and 39 patients met MACE. In the elevated troponin group, 50%, 32.1% and 17.9% died from cancer, cardiovascular and other causes, while those in the normal troponin group were 75.7%, 2% and 22.3%, there was statistical difference between 2 groups (P < 0.001). Patients with elevated preoperative hs-cTnI had significantly higher mortality (P < 0.001) and more MACE (P < 0.001) compared with those with normal hs-cTnI. A propensity-matching analysis were performed, resulting in 151 patients with normal hs-cTnI and 60 patients with elevated hs-cTnI. The matched population had the similar results for all-cause death (P = 0.009) and MACE (P = 0.001). The results were consistent after further excluding 147 patients who had received chemoradiotherapy prior to surgery in subgroup analysis. The results of multivariate Cox regression analysis shown that hs-cTnI was one of the best predictors for all-cause death (hazard ratio [HR] 2.278; 95% confidence interval [CI] 1.19-4.361) and MACE (HR, 3.523; 95%CI, 1.477-8.403) in total populations, similar results were found in subgroup analysis. CONCLUSIONS Colorectal cancer patients without myocardial ischemia manifestation but with elevated hs-cTnI prior to tumor resection surgery were at increased risk for long-term all-cause death and MACE, irrespective of whether they have received chemoradiotherapy prior to surgery.
Collapse
Affiliation(s)
- Yitao Zhang
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China
- Guangdong Provincial Key Laboratory of colorectal and pelvic floor diseases, Guangzhou, 510655, China
| | - Zicheng Huang
- Guangdong Provincial Key Laboratory of colorectal and pelvic floor diseases, Guangzhou, 510655, China
- Department of Gastroenterology, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China
| | - Sutian Hu
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China
| | - Jinhong Si
- Department of Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China
- He Xian Memorial Hospital, Southern Medical University, Guangzhou, 511402, China
| | - Shiyao Cheng
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China
| | - Zhichong Chen
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China
| | - Jiaojie Xue
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China
| | - Xue Lou
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China
| | - Huajing Peng
- Department of Nephrology, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Zequan Li
- Department of Plastic Surgery, the First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Mao Ouyang
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China.
| | - Xiang Gao
- Guangdong Provincial Key Laboratory of colorectal and pelvic floor diseases, Guangzhou, 510655, China.
- Department of Gastroenterology, the Sixth Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510655, China.
| | - Weijie Zeng
- Department of Cardiovascular Internal Medicine, the Sixth Affiliated Hospital of Sun Yat-Sen University, No.26, the second crossroad of YuanCun, Tianhe District, Guangzhou, 510655, China.
- Guangdong Provincial Key Laboratory of colorectal and pelvic floor diseases, Guangzhou, 510655, China.
| |
Collapse
|
130
|
Waissengein B, Abu Ata B, Merimsky O, Shamai S, Wolf I, Arnold JH, Bar-On T, Banai S, Khoury S, Laufer-Perl M. The predictive value of high sensitivity troponin measurements in patients treated with immune checkpoint inhibitors. Clin Res Cardiol 2023; 112:409-418. [PMID: 36301334 DOI: 10.1007/s00392-022-02118-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/13/2022] [Indexed: 11/26/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of cancer; however, at the potential cost of serious adverse events including cardiac injury. OBJECTIVE To assess the baseline and longitudinal changes in high sensitivity-Troponin (hs-Tn) in patients treated with pembrolizumab as a potential predictor for the development of major adverse cardiac events (MACE) and survival. METHODS We performed a retrospective analysis of cancer patients treated with pembrolizumab at our center. All participants had baseline measurements of hs-TnI prior to initiation of pembrolizumab (T1), with half of the patients performing follow-up measurements at their second encounter for therapy introduction (T2). We first evaluated the prevalence of abnormally elevated serum hs-TnI (> 50 nanogram per liter) at T1 and T2. We then evaluated the predictive value of abnormal levels at T1 or T2 in relation to the development of MACE (composite outcomes of myocarditis, acute coronary syndrome, heart failure, venous thromboembolism, cardiovascular hospitalization and cardiovascular mortality) and all-cause mortality. RESULTS Among 135 patients, the mean age was 72 years, predominantly male (61%). Abnormally elevated hs-TnI at T1 was observed in 7 (5%) patients and emerged as a significant independent predictor for MACE (HR 8.1, 95% CI 1.67-37.4, p = 0.009) and all-cause mortality (HR 5.37, 95% CI 2.1-13.57, p < 0.001). Abnormally elevated hs-TnI at T2 was observed in 8 (11%) patients and emerged as a significant independent predictor for MACE (HR 10.49, 95% CI 1.68-65.5, p = 0.009), but not for mortality (p = 0.200). CONCLUSIONS Abnormally elevated baseline and follow-up hs-TnI served as significant independent predictors for MACE, with an increased risk of development being 8-tenfold. Furthermore, elevated baseline hs-TnI showed a predictive value for all-cause mortality. Central illustration: Novel immune checkpoint inhibitor (ICIs) therapy has been found to revolutionize cancer therapy through increased activation of host immune systems to target and reduce tumor burden, but may come at the cost of serious adverse cardiac events. Identification of early biomarkers for the prediction and detection of these events is necessary.
Collapse
Affiliation(s)
- Barliz Waissengein
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Bian Abu Ata
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ofer Merimsky
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sivan Shamai
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ido Wolf
- Division of Oncology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Joshua H Arnold
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Tali Bar-On
- Department of Internal Medicine T, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shmuel Banai
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Shafik Khoury
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Laufer-Perl
- Division of Cardiology, Tel-Aviv Sourasky Medical Center, Affiliated to the Tel Aviv University, Tel Aviv, Israel.
- Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
131
|
Gabbia D, De Martin S. Tumor Mutational Burden for Predicting Prognosis and Therapy Outcome of Hepatocellular Carcinoma. Int J Mol Sci 2023; 24:ijms24043441. [PMID: 36834851 PMCID: PMC9960420 DOI: 10.3390/ijms24043441] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/03/2023] [Accepted: 02/05/2023] [Indexed: 02/11/2023] Open
Abstract
Hepatocellular carcinoma (HCC), the primary hepatic malignancy, represents the second-highest cause of cancer-related death worldwide. Many efforts have been devoted to finding novel biomarkers for predicting both patients' survival and the outcome of pharmacological treatments, with a particular focus on immunotherapy. In this regard, recent studies have focused on unravelling the role of tumor mutational burden (TMB), i.e., the total number of mutations per coding area of a tumor genome, to ascertain whether it can be considered a reliable biomarker to be used either for the stratification of HCC patients in subgroups with different responsiveness to immunotherapy, or for the prediction of disease progression, particularly in relation to the different HCC etiologies. In this review, we summarize the recent advances on the study of TMB and TMB-related biomarkers in the HCC landscape, focusing on their feasibility as guides for therapy decisions and/or predictors of clinical outcome.
Collapse
|
132
|
Radiotherapy/Chemotherapy-Immunotherapy for Cancer Management: From Mechanisms to Clinical Implications. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:7530794. [PMID: 36778203 PMCID: PMC9911251 DOI: 10.1155/2023/7530794] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/03/2022] [Accepted: 11/24/2022] [Indexed: 02/05/2023]
Abstract
Cancer immunotherapy has drawn much attention because it can restart the recognition and killing function of the immune system to normalize the antitumor immune response. However, the role of radiotherapy and chemotherapy in cancer treatment cannot be ignored. Due to cancer heterogeneity, combined therapy has become a new trend, and its efficacy has been confirmed in many studies. This review discussed the clinical implications and the underlying mechanisms of cancer immunotherapy in combination with radiotherapy or chemotherapy, offering an outline for clinicians as well as inspiration for future research.
Collapse
|
133
|
Ritter A, Quartermaine C, Pierre-Charles J, Balasubramanian S, Raeisi-Giglou P, Addison D, Miller E. Cardiotoxicity of Anti-Cancer Radiation Therapy: a Focus on Heart Failure. Curr Heart Fail Rep 2023; 20:44-55. [PMID: 36692820 DOI: 10.1007/s11897-023-00587-0] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/06/2022] [Indexed: 01/25/2023]
Abstract
PURPOSE OF REVIEW As the percentage of patients achieving long-term survival following treatment of their cancer grows, it is increasingly important to understand the long-term toxicities of cancer-directed treatment. In this review, we highlight the recent findings regarding radiation-induced cardiotoxicity across multiple disease sites, with a particular focus on heart failure. RECENT FINDINGS Despite its relative lack of study historically, radiation-induced heart failure has now recently been implicated in several studies of breast cancer, lung cancer, esophageal cancer, and lymphoma as a non-trivial potential consequence of thoracic radiotherapy. Data regarding specific cardiac dosimetric endpoints relevant to cardiotoxicity continue to accumulate. Radiation-induced heart failure is a rare but significant toxicity of thoracic radiotherapy, that is likely underreported. Important areas for future focus include understanding the interplay between thoracic radiotherapy and concurrent cardiotoxic systemic therapy as well as development of potential mitigation strategies and novel therapeutics.
Collapse
Affiliation(s)
- Alex Ritter
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, OH, Columbus, USA
| | - Cooper Quartermaine
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Jovan Pierre-Charles
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Suryakumar Balasubramanian
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
- Velammal Medical College Hospital & Research Institute, Madurai, India
| | - Pejman Raeisi-Giglou
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
| | - Daniel Addison
- Cardio-Oncology Program, Division of Cardiology, The Ohio State University Medical Center, Columbus, OH, USA
- Division of Cancer Control, James Cancer Hospital and Solove Research Institute at The Ohio State University, Columbus, OH, USA
| | - Eric Miller
- Department of Radiation Oncology, The Ohio State University Comprehensive Cancer Center, OH, Columbus, USA.
| |
Collapse
|
134
|
Mauro AG, Mezzaroma E, Toldo S, Melendez GC, Franco RL, Lesnefsky EJ, Abbate A, Hundley WG, Salloum FN. NLRP3-mediated inflammation in cardio-oncology: sterile yet harmful. Transl Res 2023; 252:9-20. [PMID: 35948198 PMCID: PMC9839540 DOI: 10.1016/j.trsl.2022.08.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Revised: 07/28/2022] [Accepted: 08/03/2022] [Indexed: 01/17/2023]
Abstract
Despite significant advances and the continuous development of novel, effective therapies to treat a variety of malignancies, cancer therapy-induced cardiotoxicity has been identified as a prominent cause of morbidity and mortality, closely competing with secondary malignancies. This unfortunate limitation has prompted the inception of the field of cardio-oncology with its purpose to provide the necessary knowledge and key information on mechanisms that support the use of the most efficacious cancer therapy with minimal or no interruption while paying close attention to preventing cardiovascular related morbidity and mortality. Several mechanisms that contribute to cancer therapy-induced cardiotoxicity have been proposed and studied. These mainly involve mitochondrial dysfunction and reactive oxygen species-induced oxidative stress, lysosomal damage, impaired autophagy, cell senescence, DNA damage, and sterile inflammation with the formation and activation of the NLRP3 inflammasome. In this review, we focus on describing the principal mechanisms for different classes of cancer therapies that lead to cardiotoxicity involving the NLRP3 inflammasome. We also summarize current evidence of cardio-protection with inflammasome inhibitors in the context of heart disease in general, and further highlight the potential application of this evidence for clinical translation in at risk patients for the purpose of preventing cancer therapy associated cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Adolfo G Mauro
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Eleonora Mezzaroma
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Stefano Toldo
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Giselle C Melendez
- Department of Internal Medicine, Sections on Cardiovascular Medicine, Department of Pathology, Section on Comparative Medicine, Wake Forest, School of Medicine, Winston-Salem, NC
| | - R Lee Franco
- College of Humanities and Sciences, Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, VA
| | - Edward J Lesnefsky
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA; Department of the Medical Service of the McGuire Veterans Affairs Medical Center, Richmond, VA
| | - Antonio Abbate
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - W Gregory Hundley
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA
| | - Fadi N Salloum
- Pauley Heart Center, Department of Internal Medicine, Cardiology, Virginia Commonwealth University, Richmond, VA.
| |
Collapse
|
135
|
Leiva O, Bohart I, Ahuja T, Park D. Off-Target Effects of Cancer Therapy on Development of Therapy-Induced Arrhythmia: A Review. Cardiology 2023; 148:324-334. [PMID: 36702116 PMCID: PMC10614257 DOI: 10.1159/000529260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 01/17/2023] [Indexed: 01/27/2023]
Abstract
BACKGROUND Advances in cancer therapeutics have improved overall survival and prognosis in this patient population; however, this has come at the expense of cardiotoxicity including arrhythmia. SUMMARY Cancer and its therapies are associated with cardiotoxicity via several mechanisms including inflammation, cardiomyopathy, and off-target effects. Among cancer therapies, anthracyclines and tyrosine kinase inhibitors (TKIs) are particularly known for their pro-arrhythmia effects. In addition to cardiomyopathy, anthracyclines may be pro-arrhythmogenic via reactive oxygen species (ROS) generation and altered calcium handling. TKIs may mediate their cardiotoxicity via inhibition of off-target tyrosine kinases. Ibrutinib-mediated inhibition of CSK may be responsible for the increased prevalence of atrial fibrillation. Further investigation is warranted to further elucidate the mechanisms behind arrhythmias in cancer therapies. KEY MESSAGES Arrhythmias are a common cardiotoxicity of cancer therapies. Cancer therapies may induce arrhythmias via off-target effects. Understanding the mechanisms underlying arrhythmogenesis associated with cancer therapies may help design cancer therapies that can avoid these toxicities.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Isaac Bohart
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - Tania Ahuja
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| | - David Park
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York City, New York, USA
| |
Collapse
|
136
|
Zhou F, Zhu X, Liu Y, Sun Y, Zhang Y, Cheng D, Wang W. Coronary atherosclerosis and chemotherapy: From bench to bedside. Front Cardiovasc Med 2023; 10:1118002. [PMID: 36742069 PMCID: PMC9892653 DOI: 10.3389/fcvm.2023.1118002] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 01/02/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular disease, particularly coronary artery disease, is the leading cause of death in humans worldwide. Coronary heart disease caused by chemotherapy affects the prognosis and survival of patients with tumors. The most effective chemotherapeutic drugs for cancer include proteasome inhibitors, tyrosine kinase inhibitors, immune checkpoint inhibitors, 5-fluorouracil, and anthracyclines. Animal models and clinical trials have consistently shown that chemotherapy is closely associated with coronary events and can cause serious adverse cardiovascular events. Adverse cardiovascular events after chemotherapy can affect the clinical outcome, treatment, and prognosis of patients with tumors. In recent years, with the development of new chemotherapeutic drugs, new discoveries have been made about the effects of drugs used for chemotherapy on cardiovascular disease and its related mechanisms, such as inflammation. This review article summarizes the effects of chemotherapeutic drugs on coronary artery disease and its related mechanisms to guide efforts in reducing cardiovascular adverse events during tumor chemotherapy, preventing the development of coronary heart disease, and designing new prevention and treatment strategies for cardiotoxicity caused by clinical tumor chemotherapy.
Collapse
Affiliation(s)
- Fanghui Zhou
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xinxin Zhu
- Department of Endocrinology and Metabolism, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yao Liu
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yue Sun
- Department of Blood and Endocrinology, The 962nd Hospital of the PLA Joint Logistic Support Force, Harbin, Heilongjiang, China
| | - Ying Zhang
- Key Laboratory of Preservation of Human Genetic Resources and Disease Control in China (Harbin Medical University), Ministry of Education, Harbin, China
| | | | - Wei Wang
- Department of Hematology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China,*Correspondence: Wei Wang,
| |
Collapse
|
137
|
Roberto M, Panebianco M, Aschelter AM, Buccilli D, Cantisani C, Caponnetto S, Cortesi E, d’Amuri S, Fofi C, Ierinò D, Maestrini V, Marchetti P, Marignani M, Stigliano A, Vivona L, Santini D, Tomao S. The value of the multidisciplinary team in metastatic renal cell carcinoma: Paving the way for precision medicine in toxicities management. Front Oncol 2023; 12:1026978. [PMID: 36713496 PMCID: PMC9879059 DOI: 10.3389/fonc.2022.1026978] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/07/2022] [Indexed: 01/14/2023] Open
Abstract
The new landscape of treatments for metastatic clear cell renal carcinoma (mRCC) is constantly expanding, but it is associated with the emergence of novel toxicities, adding to up to those observed in the tyrosine-kinase inhibitor (TKI) era. Indeed, the introduction of immune checkpoint inhibitors (ICIs) alone or in combination has been associated with the development of immune-related adverse events (irAEs) involving multiple-organ systems which, even if rarely, had led to fatal outcomes. Moreover, due to the relatively recent addition of ICIs to the previously available treatments, the potential additive adverse effects of these combinations are still unknown. A prompt recognition and management of these toxicities currently represents a fundamental issue in oncology, since it correlates with the outcome of cancer patients. Even if clinical guidelines provide indications for the management of irAEs, no specific protocol to evaluate the individual risk of developing an adverse event during therapy is currently available. A multidisciplinary approach addressing appropriate interventions aimed at reducing the risk of any insidious, severe, and/or dose-limiting toxicity might represent the most efficacious strategy to timely prevent and manage severe irAEs, allowing indirectly to improve both patients' cancer-specific survival and quality of life. In this review, we reported a five-case series of toxicity events that occurred at our center during treatment for mRCC followed by the remarks of physicians from different specialties, pinpointing the relevant role of an integrated and extended multidisciplinary team in a modern model of mRCC patient management.
Collapse
Affiliation(s)
- Michela Roberto
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Martina Panebianco
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy,*Correspondence: Martina Panebianco,
| | - Anna Maria Aschelter
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Dorelsa Buccilli
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Carmen Cantisani
- Department of Dermatology, Complex Operative Unit (UOC) of Dermatology, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Salvatore Caponnetto
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit B, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Enrico Cortesi
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit B, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Sara d’Amuri
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Claudia Fofi
- Department of Clinical and Molecular Medicine, Nephrology and Dialysis Unit, Sant’ Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Debora Ierinò
- Department of Clinical and Molecular Medicine, Oncology Unit, Sant’ Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Viviana Maestrini
- Department of Clinical, Internal, Anesthesiological and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Paolo Marchetti
- Scientific Direction, Istituto Dermopatico dell’Immacolata (IDI-IRCCS), Rome, Italy
| | - Massimo Marignani
- Head Liver Disease Section, Digestive and Liver Diseases Department, Sant’ Andrea Hospital, Sapienza University of Rome, Rome, Italy
| | - Antonio Stigliano
- Department of Clinical and Molecular Medicine, Endocrinology Unit, Sant ‘Andrea University Hospital, Sapienza University of Rome, Rome, Italy
| | - Luca Vivona
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| | - Daniele Santini
- Complex Operative Unit (UOC) Oncologia Medica, Sapienza University, Polo Pontino, Latina, Italy
| | - Silverio Tomao
- Department of Radiological, Oncological and Anatomo-Pathological Sciences, Medical Oncology Unit A, Policlinico Umberto I Hospital, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
138
|
Jin C, Qi J, Wang Q, Pu C, Tan M. Cardiotoxicity of lung cancer-related immunotherapy versus chemotherapy: a systematic review and network meta-analysis of randomized controlled trials. Front Oncol 2023; 13:1158690. [PMID: 37124488 PMCID: PMC10141653 DOI: 10.3389/fonc.2023.1158690] [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: 02/04/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Background Previous clinical randomized controlled trials (RCTs) have demonstrated that immune checkpoint inhibitors (ICIs) cause various toxicities during cancer treatment, but the effects of different inhibitors in combination with chemotherapy for cardiotoxicity remain controversial. The aim of the present study was to assess cardiotoxicity caused by programmed cell death protein 1 (PD-1), programmed cell death-Ligand 1 (PD-L1), and cytotoxic T lymphocyte associate protein-4 (CTLA-4) in combination with chemotherapy to treat lung cancer. Methods The following ICIs were included in the present study: durvalumab, avelumab, ipilimumab, atezolizumab, pembrolizumab, cemiplimab, and nivolumab. The relevant information was extracted using a predefined data extraction table, and the risk of bias was assessed in randomized controlled trials using the Cochrane Bias Risk tool. The main outcomes were hypertension, heart failure, pericardial effusion, and other adverse cardiac events. The random effects model was used to conduct a paired meta-analysis, and a random effects network meta-analysis was then performed within a Bayesian framework. Results In total, 17 RCTs were included in the present study. There were 11,063 individuals in the experimental and control groups, with an average age greater than 60 years. Based on the evaluation of all drug classes in RCTs, CTLA-4+chemotherapy (RR, -0.69 [95% CI, 2.91-1.52] and PD-L1 (RR, -0.21 [95% CI, -1.03-0.60]) were less cardiotoxic than the control arm, which indicated they were safer options for adverse cardiac events. PD-L1 alone was less cardiotoxic than PD-1 alone (RR, -0.57 [95% CI, -1.96-0.82]). Further, the dual immunotarget inhibitor, PD-1+CTLA-4, had the lowest SUCRA value and had the highest cardiotoxicity (SUCRA=9). Conclusion When classified according to drug type, CTLA-4+chemotherapy is associated with fewer cardiac adverse events compared to other treatments. Dual immunotarget inhibitors are more likely to have adverse cardiac reactions. Therefore, clinicians should consider this evidence when developing an ICI immunotherapy regimen for lung cancer. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42023360931.
Collapse
Affiliation(s)
- Chengwei Jin
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Jia Qi
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Qilei Wang
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Chenwei Pu
- Department of Respiratory and Critical Care Medicine, Zibo Central Hospital, Shandong, China
| | - Mingming Tan
- Department of Respiratory and Critical Care Medicine, Zibo Central Hospital, Shandong, China
- *Correspondence: Mingming Tan,
| |
Collapse
|
139
|
Madanat L, Gupta R, Weber P, Kumar N, Chandra R, Ahaneku H, Bansal Y, Anderson J, Bilolikar A, Jaiyesimi I. Cardiotoxicity of Biological Therapies in Cancer Patients: An In-depth Review. Curr Cardiol Rev 2023; 19:e310522205428. [PMID: 35642110 PMCID: PMC10280990 DOI: 10.2174/1573403x18666220531094800] [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] [Received: 02/23/2022] [Revised: 04/14/2022] [Accepted: 04/14/2022] [Indexed: 11/22/2022] Open
Abstract
Cardiotoxicity from chemotherapy regimens has been long reported. However, the understanding of cardiac side effects of biological therapies is rapidly evolving. With cancer patients achieving higher life expectancy due to the use of personalized medicine and novel targeted anticancer agents, the occurrence of cardiotoxicity is becoming more significant. Novel biological therapies include anti-HER2 antibodies, tyrosine kinase inhibitors, bruton kinase inhibitors, antivascular endothelial growth factors, proteasome inhibitors, immunomodulator drugs, and immune checkpoint inhibitors. Potential cardiovascular toxicities linked to these anticancer agents include hypertension, arrhythmias, QT prolongation, myocardial ischemia and infarction, left ventricular dysfunction, congestive heart failure, and thromboembolism. Cardiac biomarkers, electrocardiography, echocardiography and magnetic resonance imaging are common diagnostic modalities used for early detection of these complications and timely intervention. This review discusses the various types of cardiotoxicities caused by novel anticancer biologic agents, their molecular and pathophysiological mechanisms, risk factors, and diagnostic and management strategies that can be used to prevent, minimize, and treat them.
Collapse
Affiliation(s)
- Luai Madanat
- Department of Internal Medicine, William Beaumont Hospital, Royal Oak, Michigan
| | - Ruby Gupta
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Paul Weber
- College of Osteopathic Medicine, Michigan State University, East Lansing, Michigan
| | - Navneet Kumar
- Department of Cardiovascular Disease, St. Joseph Mercy Oakland Hospital, Pontiac, Michigan
| | - Rohit Chandra
- Department of Internal Medicine, William Beaumont Hospital, Royal Oak, Michigan
| | - Hycienth Ahaneku
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Yatharth Bansal
- Department of Internal Medicine, University of Detroit Mercy, Detroit, Michigan
| | - Joseph Anderson
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Abhay Bilolikar
- Department of Cardiovascular Disease, William Beaumont Hospital, Royal Oak, Michigan
| | - Ishmael Jaiyesimi
- Department of Hematology and Medical Oncology, William Beaumont Hospital, Royal Oak, Michigan
| |
Collapse
|
140
|
Bhadana R, Rani V. A Patent Review on Cardiotoxicity of Anticancerous Drugs. Cardiovasc Hematol Agents Med Chem 2023; 22:28-39. [PMID: 36683367 DOI: 10.2174/1871525721666230120155734] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/16/2022] [Accepted: 11/23/2022] [Indexed: 01/24/2023]
Abstract
Chemotherapy-induced cardiotoxicity is an increasing concern and it is critical to avoid heart dysfunction induced by medications used in various cancers. Dysregulated cardiomyocyte homeostasis is a critical phenomenon of drug-induced cardiotoxicity, which hinders the cardiac tissue's natural physiological function. Drug-induced cardiotoxicity is responsible for various heart disorders such as myocardial infarction, myocardial hypertrophy, and arrhythmia, among others. Chronic cardiac stress due to drug toxicity restricts the usage of cancer medications. Anticancer medications can cause a variety of adverse effects, especially cardiovascular toxicity. This review is focused on anticancerous drugs anthracyclines, trastuzumab, nonsteroidal anti-inflammatory medications (NSAIDs), and immune checkpoint inhibitors (ICI) and associated pathways attributed to the drug-induced cardiotoxicity. Several factors responsible for enhanced cardiotoxicity are age, gender specificity, diseased conditions, and therapy are also discussed. The review also highlighted the patents assigned for different methodologies involved in the assessment and reducing cardiotoxicity. Recent advancements where the usage of trastuzumab and bevacizumab have caused cardiac dysfunction and their effects alone or in combination on cardiac cells are explained. Extensive research on patents associated with protection against cardiotoxicity has shown that chemicals like bis(dioxopiperazine)s and manganese compounds were cardioprotective when combined with other selected anticancerous drugs. Numerous patents are associated with druginduced toxicity, prevention, and diagnosis, that may aid in understanding the current issues and developing novel therapies with safer cardiovascular outcomes. Also, the advancements in technology and research going on are yet to be explored to overcome the present issue of cardiotoxicity with the development of new drug formulations.
Collapse
Affiliation(s)
- Renu Bhadana
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector- 62, Noida 201307, Uttar Pradesh, India
| | - Vibha Rani
- Center for Emerging Diseases, Department of Biotechnology, Jaypee Institute of Information Technology, A-10, Sector- 62, Noida 201307, Uttar Pradesh, India
| |
Collapse
|
141
|
Smekens L, Genoud V, Usdin N, Ben Aïssa A. Cardiovascular, Renal and Pulmonary Toxicity of Immune Checkpoint Inhibitors in Cancer: What the GP Should Know. PRAXIS 2023; 112:160-171. [PMID: 36855889 DOI: 10.1024/1661-8157/a003976] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Immunotherapy with immune checkpoint inhibitors (ICI) is administered in different cancer types and can lead to a wide range of immune-related adverse events including toxicity in vital organs such as the lungs, the kidneys, and the heart. The main hypothesis suggests an overactivation of the immune cells in the different organs. Whereas immune-related cardiotoxicity is very rare but life threatening, ICI-induced acute kidney injury and pneumonitis are more frequent but in general less severe. Renal toxicity corresponds in more than 90% to an acute tubulo-interstitial nephritis. Checkpoint inhibitors pneumonitis is diagnosed mainly on respiratory symptoms with new radiological features, especially under the form of a cryptogenic organising pneumonia. Cardiotoxicity is predominantly marked by myocarditis but also pericarditis and arrhythmias, among others. Early recognition, temporary or definitive cessation of ICI therapy and rapid initiation of high-dose corticosteroids are the cornerstones of the management, which must to be multidisciplinary in a specialised center.
Collapse
Affiliation(s)
- Laure Smekens
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
- Equal contribution
| | - Vassilis Genoud
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
- Center for Translational Research in Onco-Haematology, University of Geneva, Geneva, Switzerland
- Equal contribution
| | - Nita Usdin
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
- Equal contribution
| | - Assma Ben Aïssa
- Service d'oncologie, Hôpitaux Universitaires de Genève, Geneva, Switzerland
| |
Collapse
|
142
|
Liu Y, Zheng L, Cai X, Zhang X, Ye Y. Cardiotoxicity from neoadjuvant targeted treatment for breast cancer prior to surgery. Front Cardiovasc Med 2023; 10:1078135. [PMID: 36910540 PMCID: PMC9992214 DOI: 10.3389/fcvm.2023.1078135] [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: 10/24/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Cancer treatment has been gradually shifting from non-specific cytotoxic agents to molecularly targeted drugs. Breast cancer (BC), a malignant tumor with one of the highest incidence worldwide, has seen a rapid development in terms of targeted therapies, leading to a radical change in the treatment paradigm. However, the use of targeted drugs is accompanied by an increasing rate of deaths due to non-tumor-related causes in BC patients, with cardiovascular complications as the most common cause. Cardiovascular toxicity during antitumor therapy has become a high-risk factor for survival in BC patients. Targeted drug-induced cardiotoxicity exerts a wide range of effects on cardiac structure and function, including conduction disturbances, QT interval prolongation, impaired myocardial contractility, myocardial fibrosis, and hypertrophy, resulting in various clinical manifestations, e.g., arrhythmias, cardiomyopathy, heart failure, and even sudden death. In adult patients, the incidence of antitumor targeted drug-induced cardiotoxicity can reach 50%, and current preclinical evaluation tools are often insufficiently effective in predicting clinical cardiotoxicity. Herein, we reviewed the current status of the occurrence, causative mechanisms, monitoring methods, and progress in the prevention and treatment of cardiotoxicity associated with preoperative neoadjuvant targeted therapy for BC. It supplements the absence of relevant review on the latest research progress of preoperative neoadjuvant targeted therapy for cardiotoxicity, with a view to providing more reference for clinical treatment of BC patients.
Collapse
Affiliation(s)
- Yihua Liu
- Department of Breast Surgery, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Li Zheng
- Department of Breast Surgery, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xingjuan Cai
- Department of Breast Surgery, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaojun Zhang
- Department of Breast Surgery, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Ye
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| |
Collapse
|
143
|
Yang H, Miao Y, Yu Z, Wei M, Jiao X. Cell adhesion molecules and immunotherapy in advanced non-small cell lung cancer: Current process and potential application. Front Oncol 2023; 13:1107631. [PMID: 36895477 PMCID: PMC9989313 DOI: 10.3389/fonc.2023.1107631] [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: 11/25/2022] [Accepted: 02/07/2023] [Indexed: 02/23/2023] Open
Abstract
Advanced non-small cell lung cancer (NSCLC) is a severe disease and still has high mortality rate after conventional treatment (e.g., surgical resection, chemotherapy, radiotherapy and targeted therapy). In NSCLC patients, cancer cells can induce immunosuppression, growth and metastasis by modulating cell adhesion molecules of both cancer cells and immune cells. Therefore, immunotherapy is increasingly concerned due to its promising anti-tumor effect and broader indication, which targets cell adhesion molecules to reverse the process. Among these therapies, immune checkpoint inhibitors (mainly anti-PD-(L)1 and anti-CTLA-4) are most successful and have been adapted as first or second line therapy in advanced NSCLC. However, drug resistance and immune-related adverse reactions restrict its further application. Further understanding of mechanism, adequate biomarkers and novel therapies are necessary to improve therapeutic effect and alleviate adverse effect.
Collapse
Affiliation(s)
- Hongjian Yang
- Innovative Institute, China Medical University, Shenyang, China
| | - Yuxi Miao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China
| | - Zhaojin Yu
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Key Laboratory of Molecular Targeted Anti-Tumor Drug Development and Evaluation, Shenyang, China
| | - Minjie Wei
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Liaoning Cancer Immune Peptide Drug Engineering Technology Research Centre, Shenyang, China
| | - Xue Jiao
- Department of Pharmacology, School of Pharmacy, China Medical University, Shenyang, China.,Shenyang Kangwei Medical Laboratory Analysis Co. LTD, Shenyang, China
| |
Collapse
|
144
|
Li X, Peng W, Wu J, Yeung SCJ, Yang R. Advances in immune checkpoint inhibitors induced-cardiotoxicity. Front Immunol 2023; 14:1130438. [PMID: 36911712 PMCID: PMC9995967 DOI: 10.3389/fimmu.2023.1130438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are approved as the first-line drug for treating many cancers and has shown significant survival benefits; however, it also causes immune-related adverse events (irAEs) while activating the immune system, involving multiple organs. Among them, cardiovascular immune-related adverse events (CV-irAE) are rare, but common causes of death in ICIs treated cancer patients, which manifest as myocardial, pericardial, vascular and other cardiovascular toxicities. Therefore, it is important that irAEs, especially CV-irAE should be carefully recognized and monitored during the whole ICIs treatment because early detection and treatment of CV-irAE can significantly reduce the mortality of such patients. Consequently, it is urgent to fully understand the mechanism and management strategies of CV-irAE. The effects of ICIs are multifaceted and the exact mechanism of CV-irAE is still elusive. Generally, T cells identify tumor cell antigens as well as antigen in cardiomyocytes that are the same as or homologous to those on tumor cells, thus causing myocardial damage. In addition, ICIs promote formation of cardiac troponin I (cTnI) that induces cardiac dysfunction and myocardial dilatation; moreover, ICIs also increase the production of cytokines, which promote infiltration of inflammation-linked molecules into off-target tissues. Currently, the management and treatment of cardiovascular toxicity are largely dependent on glucocorticoids, more strategies for prevention and treatment of CV-irAE, such as predictive markers are being explored. This review discusses risk factors, potential pathophysiological mechanisms, clinical manifestations, and management and treatment of CV-irAE, guiding the development of more effective prevention, treatment and management strategies in the future.
Collapse
Affiliation(s)
- Xiang Li
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenying Peng
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiao Wu
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, TX, United States
| | - Runxiang Yang
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
145
|
Totzeck M, Aide N, Bauersachs J, Bucerius J, Georgoulias P, Herrmann K, Hyafil F, Kunikowska J, Lubberink M, Nappi C, Rassaf T, Saraste A, Sciagra R, Slart RHJA, Verberne H, Rischpler C. Nuclear medicine in the assessment and prevention of cancer therapy-related cardiotoxicity: prospects and proposal of use by the European Association of Nuclear Medicine (EANM). Eur J Nucl Med Mol Imaging 2023; 50:792-812. [PMID: 36334105 PMCID: PMC9852191 DOI: 10.1007/s00259-022-05991-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Accepted: 10/04/2022] [Indexed: 11/06/2022]
Abstract
Cardiotoxicity may present as (pulmonary) hypertension, acute and chronic coronary syndromes, venous thromboembolism, cardiomyopathies/heart failure, arrhythmia, valvular heart disease, peripheral arterial disease, and myocarditis. Many of these disease entities can be diagnosed by established cardiovascular diagnostic pathways. Nuclear medicine, however, has proven promising in the diagnosis of cardiomyopathies/heart failure, and peri- and myocarditis as well as arterial inflammation. This article first outlines the spectrum of cardiotoxic cancer therapies and the potential side effects. This will be complemented by the definition of cardiotoxicity using non-nuclear cardiovascular imaging (echocardiography, CMR) and biomarkers. Available nuclear imaging techniques are then presented and specific suggestions are made for their application and potential role in the diagnosis of cardiotoxicity.
Collapse
Affiliation(s)
- Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Nicolas Aide
- Nuclear Medicine Department, University Hospital, Caen, France
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Jan Bucerius
- Department of Nuclear Medicine, University Medicine Göttingen, Georg-August-University Göttingen, Göttingen, Germany
| | - Panagiotis Georgoulias
- Department of Nuclear Medicine, Faculty of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Ken Herrmann
- Clinic for Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Fabien Hyafil
- Department of Nuclear Medicine, DMU IMAGINA, Georges-Pompidou European Hospital, Assistance-Publique – Hôpitaux de Paris, University of Paris, Paris, France
| | - Jolanta Kunikowska
- Nuclear Medicine Department, Medical University of Warsaw, Warsaw, Poland
| | - Mark Lubberink
- Medical Physics, Uppsala University Hospital, Uppsala, Sweden
| | - Carmela Nappi
- Department of Advanced Biomedical Sciences, University of Naples “Federico II”, Naples, Italy
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center Essen, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| | - Antti Saraste
- Heart Center, Turku University Hospital and University of Turku, Turku, Finland
| | - Roberto Sciagra
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences “Mario Serio”, University of Florence, Florence, Italy
| | - Riemer H. J. A. Slart
- Medical Imaging Center, Department of Nuclear Medicine and Molecular Imaging, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands ,Department of Biomedical Photonic Imaging, Faculty of Science and Technology, Enschede, The Netherlands
| | - Hein Verberne
- Department of Radiology and Nuclear Medicine, Amsterdam UMC, Location AMC, University of Amsterdam, Amsterdam, The Netherlands
| | - Christoph Rischpler
- Clinic for Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| |
Collapse
|
146
|
Chan JSK, Lakhani I, Lee TTL, Chou OHI, Lee YHA, Cheung YM, Yeung HW, Tang P, Ng K, Dee EC, Liu T, Wong WT, Tse G, Leung FP. Cardiovascular Outcomes and Hospitalizations in Asian Patients Receiving Immune Checkpoint Inhibitors: A Population-based Study. Curr Probl Cardiol 2023; 48:101380. [PMID: 36031015 DOI: 10.1016/j.cpcardiol.2022.101380] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 08/19/2022] [Indexed: 10/15/2022]
Abstract
Immune checkpoint inhibitors (ICI) have known associations with cardiotoxicity. However, a representative quantification of the adverse cardiovascular events and cardiovascular attendances amongst Asian users of ICI has been lacking. This retrospective cohort study identified all ICI users in Hong Kong, China, between 2013 and 2021. All patients were followed up until the end of 2021 for the primary outcome of major adverse cardiovascular event (MACE; a composite of cardiovascular mortality, myocardial infarction, heart failure, and stroke). Patients with prior diagnosis of any component of MACE were excluded from all MACE analyses. In total, 4324 patients were analyzed (2905 (67.2%) males; median age 63.5 years old (interquartile range 55.4-70.7 years old); median follow-up 1.0 year (interquartile range 0.4-2.3 years)), of whom 153 were excluded from MACE analyses due to prior events. MACE occurred in 116 (2.8%) with an incidence rate (IR) of 1.7 [95% confidence interval: 1.4, 2.0] events per 100 patient-years; IR was higher within the first year of follow-up (2.9 [2.3, 3.5] events per 100 patient-years). Cardiovascular hospitalization(s) occurred in 188 (4.4%) with 254 episodes (0.5% of all episodes) and 1555 days of hospitalization (1.3% of all hospitalized days), for whom the IR of cardiovascular hospitalization was 5.6 [4.6, 6.9] episodes per 100 person-years with 52.9 [39.8, 70.3] days' stay per 100 person-years. Amongst Asian users of ICI, MACE was uncommon, and a small proportion of hospitalizations were cardiovascular in nature. Most MACE and cardiovascular hospitalizations occurred during the first year after initiating ICI.
Collapse
Affiliation(s)
- Jeffrey Shi Kai Chan
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, Hong Kong, China-United Kingdom Collaboration
| | - Ishan Lakhani
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, Hong Kong, China-United Kingdom Collaboration
| | - Teddy Tai Loy Lee
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, Hong Kong, China-United Kingdom Collaboration
| | - Oscar Hou In Chou
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, Hong Kong, China-United Kingdom Collaboration
| | - Yan Hiu Athena Lee
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, Hong Kong, China-United Kingdom Collaboration
| | - Yiu Ming Cheung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hoi Wa Yeung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Pias Tang
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, Hong Kong, China-United Kingdom Collaboration
| | - Kenrick Ng
- Department of Medical Oncology, University College London Hospitals NHS Foundation Trust, London, 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, China
| | - Wing Tak Wong
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, 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, China; Kent and Medway Medical School, University of Kent and Canterbury Christ Church University, Canterbury, UK; Epidemiology Research Unit, Cardiovascular Analytics Group, Hong Kong, China-United Kingdom Collaboration.
| | - Fung Ping Leung
- School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
147
|
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]
|
148
|
Malaty MM, Amarasekera AT, Li C, Scherrer-Crosbie M, Tan TC. Incidence of immune checkpoint inhibitor mediated cardiovascular toxicity: A systematic review and meta-analysis. Eur J Clin Invest 2022; 52:e13831. [PMID: 35788986 DOI: 10.1111/eci.13831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) are a novel class of anti-cancer therapy becoming increasingly associated with fatal cardiovascular toxicities (CVTs). The aim is to determine the incidence of CVTs in cohorts treated with ICIs as sole anti-cancer therapy. METHODS A systematic literature search of scientific and medical databases was performed using PRISMA principles to identify relevant cohorts (PROSPERO registration CRD42021272470). Data for specific CVTs (pericardial disease, myocarditis, heart failure, arrhythmia, myocardial infarction/ischaemia and angina), CVT-related death and CV risk factors were extracted. Presence of CVTs in ICI-monotherapy versus combination-ICI therapy, and programmed death 1/programmed death ligand 1- (PD1/PDL1-) versus cytotoxic T-lymphocyte-associated protein 4- (CTLA4-) inhibitor groups were dichotomised and meta-analysed using random-effect models. RESULTS Forty-eight studies (11,207 patients) were identified, from which 146 CVTs were observed (incidence 1.30%). ICI-monotherapy led to more CVTs than combination therapy (119/9009; 1.32% vs. 18/2086; 0.86%). Across monotherapies, PD1/PDL1-inhibitors had lower incidence of CVTs compared to CTLA4-inhibitors (62/6950; 0.89% vs. 57/2059; 2.77%). Based on eight studies that were meta-analysed, no significant difference was observed comparing monotherapy versus combination-ICI therapy (RR-0.69, 95% CI -1.47 to 0.09) for all CVTs, or PD1/PDL1- to CTLA4-inhibitors (RR-0.27, 95% CI -2.06 to 1.53), for all CVTs including CVT-death. CV risk factors could not be attributed to an ICI group as data was population based rather than individual based. CONCLUSION ICI-mediated CVTs are rare and potentially fatal. The role of CV risk factors in their development remains unclear.
Collapse
Affiliation(s)
- Michael M Malaty
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia
| | - Anjalee Thanuja Amarasekera
- School of Medicine, Western Sydney University, Sydney, Australia.,Department of Cardiology, Westmead Hospital, Western Sydney Local Health District, Sydney, Australia.,Westmead Applied Research Centre (WARC), Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Cindy Li
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy C Tan
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia.,School of Medicine, Western Sydney University, Sydney, Australia.,Department of Cardiology, Westmead Hospital, Western Sydney Local Health District, Sydney, Australia
| |
Collapse
|
149
|
Lee C, Drobni ZD, Zafar A, Gongora CA, Zlotoff DA, Alvi RM, Taron J, Rambarat PK, Schoenfeld S, Mosarla RC, Raghu VK, Hartmann SE, Gilman HK, Murphy SP, Sullivan RJ, Faje A, Hoffmann U, Zhang L, Mayrhofer T, Reynolds KL, Neilan TG. Pre-Existing Autoimmune Disease Increases the Risk of Cardiovascular and Noncardiovascular Events After Immunotherapy. JACC CardioOncol 2022; 4:660-669. [PMID: 36636443 PMCID: PMC9830202 DOI: 10.1016/j.jaccao.2022.11.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Revised: 11/03/2022] [Accepted: 11/08/2022] [Indexed: 12/24/2022] Open
Abstract
Background The use of immune checkpoint inhibitors (ICI) is associated with cardiovascular (CV) events, and patients with pre-existing autoimmune disease are at increased CV risk. Objectives The aim of this study was to characterize the risk for CV events in patients with pre-existing autoimmune disease post-ICI. Methods This was a retrospective study of 6,683 patients treated with ICIs within an academic network. Autoimmune disease prior to ICI was confirmed by chart review. Baseline characteristics and risk for CV and non-CV immune-related adverse events were compared with a matched control group (1:1 ratio) of ICI patients without autoimmune disease. Matching was based on age, sex, history of coronary artery disease, history of heart failure, and diabetes mellitus. CV events were a composite of myocardial infarction, percutaneous coronary intervention, coronary artery bypass graft, stroke, transient ischemic attack, deep venous thrombosis, pulmonary embolism, or myocarditis. Univariable and multivariable Cox proportional hazards models were used to determine the association between autoimmune disease and CV events. Results Among 502 patients treated with ICIs, 251 patients with and 251 patients without autoimmune disease were studied. During a median follow-up period of 205 days, there were 45 CV events among patients with autoimmune disease and 22 CV events among control subjects (adjusted HR: 1.77; 95% CI: 1.04-3.03; P = 0.0364). Of the non-CV immune-related adverse events, there were increased rates of psoriasis (11.2% vs 0.4%; P < 0.001) and colitis (24.3% vs 16.7%; P = 0.045) in patients with autoimmune disease. Conclusions Patients with autoimmune disease have an increased risk for CV and non-CV events post-ICI.
Collapse
Key Words
- CABG, coronary artery bypass graft
- CTLA-4, cytotoxic T lymphocyte–associated antigen-4
- CV, cardiovascular
- DVT, deep venous thrombosis
- ICI, immune checkpoint inhibitor
- MI, myocardial infarction
- PCI, percutaneous coronary intervention
- PD-1, programmed death-1
- PD-L1, programmed death-ligand 1
- PE, pulmonary embolism
- SMD, standardized mean difference
- TIA, transient ischemic attack
- coronary artery disease
- immunotherapy
- irAE, immune-related adverse event
- myocarditis
- thrombosis
Collapse
Affiliation(s)
- Charlotte Lee
- Division of Cardiology, Columbia University Irving Medical Center, NewYork-Presbyterian Hospital, New York, New York, USA
| | - Zsofia D. Drobni
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Amna Zafar
- Division of Cardiovascular Diseases and Hypertension, Rutgers Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Carlos A. Gongora
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Daniel A. Zlotoff
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Raza M. Alvi
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Jana Taron
- Department of Radiology, University Hospital Freiburg, Freiburg, Germany
| | - Paula K. Rambarat
- Department of Internal Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sara Schoenfeld
- Division of Rheumatology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ramya C. Mosarla
- Division of Cardiology, New York University, New York, New York, USA
| | - Vineet K. Raghu
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sarah E. Hartmann
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Hannah K. Gilman
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Sean P. Murphy
- Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Ryan J. Sullivan
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Alexander Faje
- Division of Endocrinology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Lili Zhang
- Department of Cardiology, Department of Medicine, Montefiore Medical Center, Bronx, New York, USA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Kerry L. Reynolds
- Division of Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center, Department of Radiology and Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
150
|
Haanen J, Obeid M, Spain L, Carbonnel F, Wang Y, Robert C, Lyon AR, Wick W, Kostine M, Peters S, Jordan K, Larkin J. Management of toxicities from immunotherapy: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2022; 33:1217-1238. [PMID: 36270461 DOI: 10.1016/j.annonc.2022.10.001] [Citation(s) in RCA: 473] [Impact Index Per Article: 157.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/30/2022] [Accepted: 10/02/2022] [Indexed: 11/17/2022] Open
Affiliation(s)
- J Haanen
- Division of Medical Oncology, Netherlands Cancer Institute (NKI), Amsterdam, The Netherlands
| | - M Obeid
- Immunology and Allergy Service, CHUV, Lausanne; Lausanne Center for Immuno-oncology Toxicities (LCIT), CHUV, Lausanne; Department of Oncology, CHUV, Lausanne, Switzerland
| | - L Spain
- Medical Oncology Department, Peter MacCallum Cancer Centre, Melbourne; Department of Medical Oncology, Eastern Health, Melbourne; Monash University Eastern Health Clinical School, Box Hill, Australia
| | - F Carbonnel
- Gastroenterology Department, Assistance Publique-Hôpitaux de Paris, Hôpital Universitaire Bicêtre, Le Kremlin Bicêtre, France; Université Paris Saclay 11, Le Kremlin-Bicêtre, France
| | - Y Wang
- Department of Gastroenterology, Hepatology & Nutrition, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - C Robert
- Department of Medicine, Gustave Roussy Cancer Centre, Villejuif; Paris-Saclay University, Villejuif, France
| | - A R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, London; National Heart and Lung Institute, Imperial College London, London, UK
| | - W Wick
- Neurology Clinic and National Centre for Tumour Diseases, University Hospital Heidelberg, Heidelberg; DKTK and Clinical Cooperation Unit NeuroOncology, DKFZ, Heidelberg, Germany
| | - M Kostine
- Department of Rheumatology, Hôpital Pellegrin, CHU de Bordeaux, Bordeaux, France
| | - S Peters
- Department of Oncology, CHUV, Lausanne, Switzerland
| | - K Jordan
- Department of Haematology, Oncology and Palliative Medicine, Ernst von Bergmann Hospital Potsdam, Potsdam; Department of Haematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - J Larkin
- Royal Marsden NHS Foundation Trust, London, UK
| |
Collapse
|