301
|
Drobni ZD, Alvi RM, Taron J, Zafar A, Murphy SP, Rambarat PK, Mosarla RC, Lee C, Zlotoff DA, Raghu VK, Hartmann SE, Gilman HK, Gong J, Zubiri L, Sullivan RJ, Reynolds KL, Mayrhofer T, Zhang L, Hoffmann U, Neilan TG. Association Between Immune Checkpoint Inhibitors With Cardiovascular Events and Atherosclerotic Plaque. Circulation 2020; 142:2299-2311. [PMID: 33003973 PMCID: PMC7736526 DOI: 10.1161/circulationaha.120.049981] [Citation(s) in RCA: 343] [Impact Index Per Article: 68.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) treat an expanding range of cancers. Consistent basic data suggest that these same checkpoints are critical negative regulators of atherosclerosis. Therefore, our objectives were to test whether ICIs were associated with accelerated atherosclerosis and a higher risk of atherosclerosis-related cardiovascular events. METHODS The study was situated in a single academic medical center. The primary analysis evaluated whether exposure to an ICI was associated with atherosclerotic cardiovascular events in 2842 patients and 2842 controls matched by age, a history of cardiovascular events, and cancer type. In a second design, a case-crossover analysis was performed with an at-risk period defined as the 2-year period after and the control period as the 2-year period before treatment. The primary outcome was a composite of atherosclerotic cardiovascular events (myocardial infarction, coronary revascularization, and ischemic stroke). Secondary outcomes included the individual components of the primary outcome. In addition, in an imaging substudy (n=40), the rate of atherosclerotic plaque progression was compared from before to after the ICI was started. All study measures and outcomes were blindly adjudicated. RESULTS In the matched cohort study, there was a 3-fold higher risk for cardiovascular events after starting an ICI (hazard ratio, 3.3 [95% CI, 2.0-5.5]; P<0.001). There was a similar increase in each of the individual components of the primary outcome. In the case-crossover, there was also an increase in cardiovascular events from 1.37 to 6.55 per 100 person-years at 2 years (adjusted hazard ratio, 4.8 [95% CI, 3.5-6.5]; P<0.001). In the imaging study, the rate of progression of total aortic plaque volume was >3-fold higher with ICIs (from 2.1%/y before 6.7%/y after). This association between ICI use and increased atherosclerotic plaque progression was attenuated with concomitant use of statins or corticosteroids. CONCLUSIONS Cardiovascular events were higher after initiation of ICIs, potentially mediated by accelerated progression of atherosclerosis. Optimization of cardiovascular risk factors and increased awareness of cardiovascular risk before, during, and after treatment should be considered among patients on an ICI.
Collapse
Affiliation(s)
- Zsofia D. Drobni
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Cardiovascular Imaging Research Group, Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Raza M. Alvi
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jana Taron
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Amna Zafar
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sean P. Murphy
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Paula K. Rambarat
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Rayma C. Mosarla
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Charlotte Lee
- Division of Internal Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Daniel A. Zlotoff
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Vineet K. Raghu
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Sarah E. Hartmann
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Hannah K. Gilman
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jingyi Gong
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Leyre Zubiri
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Ryan J. Sullivan
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Thomas Mayrhofer
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Lili Zhang
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Udo Hoffmann
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Division of Cardiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
302
|
Cautela J, Zeriouh S, Gaubert M, Bonello L, Laine M, Peyrol M, Paganelli F, Lalevee N, Barlesi F, Thuny F. Intensified immunosuppressive therapy in patients with immune checkpoint inhibitor-induced myocarditis. J Immunother Cancer 2020; 8:jitc-2020-001887. [PMID: 33298621 PMCID: PMC7725077 DOI: 10.1136/jitc-2020-001887] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/20/2020] [Indexed: 12/14/2022] Open
Abstract
Background Myocarditis is a rare but life-threatening adverse event of cancer treatments with immune checkpoint inhibitors (ICIs). Recent guidelines recommend the use of high doses of corticosteroids as a first-line treatment, followed by intensified immunosuppressive therapy (IIST) in the case of unfavorable evolution. However, this strategy is empirical, and no studies have specifically addressed this issue. Therefore, we aimed to investigate and compare the clinical course, management and outcome of ICI-induced myocarditis patients requiring or not requiring IIST. Methods This case–control study included all patients consecutively admitted to The Mediterranean University Center of Cardio-Oncology (Aix-Marseille University, France) for the diagnosis of ICI-induced myocarditis according to Bonaca’s criteria and treated with or without IIST. In addition, we searched PubMed and included patients from previously published case reports treated with IIST in the analysis. The clinical, biological, imaging, treatment, all-cause death and cardiovascular death data of patients who required IIST were compared with those of patients who did not. Results A total of 60 patients (69±12 years) were included (36 were treated with IIST and 24 were not). Patients requiring IIST were more likely to have received a combination of ICIs (39% vs 8%, p=0.01), and developed the first symptoms/signs of myocarditis earlier after the onset of ICI therapy (median, 18 days vs 60 days, p=0.002). They had a significantly higher prevalence of sustained ventricular arrhythmia, complete atrioventricular block, cardiogenic shock and troponin elevation. Moreover, they were more likely to have other immune-related adverse events simultaneously (p<0.0001), especially myositis (p=0.0002) and myasthenia gravis (p=0.009). Patients who required IIST were more likely to die from any cause (50% vs 21%, p=0.02). Among them, patients who received infliximab were more likely to die from cardiovascular causes (OR, 12.0; 95% CI 2.1 to 67.1; p=0.005). Conclusion The need for IIST was more common in patients who developed myocarditis very early after the start of ICI therapy, as well as when hemodynamic/electrical instability or neuromuscular adverse events occurred. Treatment with infliximab might be associated with an increased risk of cardiovascular death.
Collapse
Affiliation(s)
- Jennifer Cautela
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France.,Mediterranean Group of Cardio-oncology (gMEDICO), Aix-Marseille-University, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Sarah Zeriouh
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Melanie Gaubert
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Laurent Bonello
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Marc Laine
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Michael Peyrol
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Franck Paganelli
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France
| | - Nathalie Lalevee
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France.,Technological Advances for Genomics and Clinics (TAGC), UMR/INSERM 1090, Aix-Marseille-University, Marseille, France
| | - Fabrice Barlesi
- Mediterranean Group of Cardio-oncology (gMEDICO), Aix-Marseille-University, Marseille, Provence-Alpes-Côte d'Azur, France.,Gustave Roussy, Drug Development Department (DITEP), Paris-Saclay University, Villejuif, France
| | - Franck Thuny
- University Mediterranean Centre of Cardio-Oncology (MEDI-CO centre), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Nord Hospital, Centre for CardioVascular and Nutrition research (C2VN), INSERM 1263, INRAE 1260, Aix-Marseille University, Assistance Publique - Hôpitaux de Marseille, Marseille, Provence-Alpes-Côte d'Azur, France .,Mediterranean Group of Cardio-oncology (gMEDICO), Aix-Marseille-University, Marseille, Provence-Alpes-Côte d'Azur, France
| |
Collapse
|
303
|
Behravesh S, Shomali N, Danbaran GR, Aslani S, Hemmatzadeh M, Hosseinzadeh R, Gowhari-Shabgah A, Mohammadi H. Cardiotoxicity of immune checkpoint inhibitors: An updated review. Biotechnol Appl Biochem 2020; 69:61-69. [PMID: 33289168 DOI: 10.1002/bab.2081] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 11/25/2020] [Indexed: 12/11/2022]
Abstract
The immune checkpoint molecules are involved in the regulation of T cells in order to prevent them from attacking to sell tissues and play a role in the immune response homeostasis. Application of the immune checkpoint inhibitors (ICIs) has provided a promising therapeutic approach in pathologies where the immune system is suppressed. The extended utilization of ICIs in several cancers has caused immune-related side effects in the cardiovascular system like cardiomyopathy and myocarditis. Cardiac toxicity, one of the main side effects of the ICIs based therapeutic approach has less been concerned; however, during the last years, many cases of fatal heart failure and myocarditis have been reported in patients treated with ICIs. In this review article, we attempted to discuss the cardiac adverse effects of inhibiting different immune checkpoint molecules. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Soheil Behravesh
- Student Research Committee, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Saeed Aslani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hamed Mohammadi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran.,Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
304
|
Broberg AM, Geisler J, Tuohinen S, Skytta T, Hrafnkelsdóttir ÞJ, Nielsen KM, Hedayati E, Omland T, Offersen BV, Lyon AR, Gulati G. Prevention, Detection, and Management of Heart Failure in Patients Treated for Breast Cancer. Curr Heart Fail Rep 2020; 17:397-408. [PMID: 32979150 PMCID: PMC7683437 DOI: 10.1007/s11897-020-00486-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Long-term survival has increased significantly in breast cancer patients, and cardiovascular side effects are surpassing cancer-related mortality. We summarize risk factors, prevention strategies, detection, and management of cardiotoxicity, with focus on left ventricular dysfunction and heart failure, during breast cancer treatment. RECENT FINDINGS Baseline treatment of cardiovascular risk factors is recommended. Anthracycline and trastuzumab treatment constitute a substantial risk of developing cardiotoxicity. There is growing evidence that this can be treated with beta blockers and angiotensin antagonists. Early detection of cardiotoxicity with cardiac imaging and circulating cardiovascular biomarkers is currently evaluated in clinical trials. Chest wall irradiation accelerates atherosclerotic processes and induces fibrosis. Immune checkpoint inhibitors require consideration for surveillance due to a small risk of severe myocarditis. Cyclin-dependent kinases4/6 inhibitors, cyclophosphamide, taxanes, tyrosine kinase inhibitors, and endocrine therapy have a lower-risk profile for cardiotoxicity. Preventive and management strategies to counteract cancer treatment-related left ventricular dysfunction or heart failure in breast cancer patients should include a comprehensive cardiovascular risk assessment and individual clinical evaluation. This should include both patient and treatment-related factors. Further clinical trials especially on early detection, cardioprevention, and management are urgently needed.
Collapse
Affiliation(s)
- Agneta Månsson Broberg
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog & Institute of Clinical Medicine, University of Oslo, Campus AHUS, Lørenskog, Norway
| | - Suvi Tuohinen
- Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Tanja Skytta
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | - Þórdís Jóna Hrafnkelsdóttir
- Department of Cardiology, Landspitali University Hospital, Reykjavík, Iceland and Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | - Elham Hedayati
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Breast Cancer, Sarcoma and Endocrine Tumors, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte V. Offersen
- Department of Experimental Clinical Oncology & Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Alexander R. Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and Imperial College London, London, UK
| | - Geeta Gulati
- Department of Cardiology, Oslo University Hospital, Postbox 4950, Ullevål, Nydalen, 0424 Oslo, Norway
- Department of Research, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
305
|
Yeung SJ, Qdaisat A, Chaftari P, Lipe D, Merlin J, Rajha E, Wechsler A, Sandoval M, Viets J, Al‐Breiki A, Shah M, Pandey R, Kamal M, Khattab O, Toale K, Wattana M, Elsayem A, Gaeta S, Brock P, Reyes‐Gibby C, Alagappan K. Diagnosis and management of immune-related adverse effects of immune checkpoint therapy in the emergency department. J Am Coll Emerg Physicians Open 2020; 1:1637-1659. [PMID: 33392573 PMCID: PMC7771833 DOI: 10.1002/emp2.12209] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 07/02/2020] [Accepted: 07/13/2020] [Indexed: 12/14/2022] Open
Abstract
Rapid advances in cancer immunotherapy using immune checkpoint inhibitors have led to significantly improved survival. Rapid identification of the toxicity syndromes associated with these therapeutic agents is very important for emergency physicians because the population of patients diagnosed with cancer is increasing and cancer therapies including immune checkpoint inhibitors have become the first-line treatment for more and more types of cancer. The emergency medicine literature lags behind rapid advances in oncology, and oncology guidelines for rapid recognition and management of these emerging toxicity syndromes are not familiar to emergency physicians. In this review article, we discuss the clinical presentation and management of immune-related adverse effects during the critical first hours of emergency care. We also suggest a workflow for the recognition and treatment of emergencies arising from serious immune-related adverse effects, including but not limited to colitis, adrenal crisis, myocarditis, pneumonitis, myasthenic crisis, diabetic ketoacidosis, bullous pemphigus, and hemophagocytic lymphohistiocytosis. Rapid advances in cancer therapy are bringing new diagnostic and therapeutic challenges to emergency providers, and therefore it is crucial to raise awareness and provide guidelines for the management of new treatment-related toxicities.
Collapse
Affiliation(s)
- Sai‐Ching Jim Yeung
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Aiham Qdaisat
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Patrick Chaftari
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Demis Lipe
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Jeffrey Merlin
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Eva Rajha
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Adriana Wechsler
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Marcelo Sandoval
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Jayne Viets
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Aisha Al‐Breiki
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Mohsin Shah
- Center for Clinical Epidemiology and BiostatisticsPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPennsylvaniaUSA
| | - Ramesh Pandey
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Mona Kamal
- Department of Symptom ResearchThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
- Department of Clinical Oncology and Nuclear MedicineFaculty of MedicineAin Shams UniversityCairoEgypt
| | - Osama Khattab
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Katy Toale
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Monica Wattana
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Ahmed Elsayem
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Susan Gaeta
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Patricia Brock
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Cielito Reyes‐Gibby
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | - Kumar Alagappan
- Department of Emergency MedicineThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
306
|
Zamorano JL, Gottfridsson C, Asteggiano R, Atar D, Badimon L, Bax JJ, Cardinale D, Cardone A, Feijen EA, Ferdinandy P, López-Fernández T, Gale CP, Maduro JH, Moslehi J, Omland T, Plana Gomez JC, Scott J, Suter TM, Minotti G. The cancer patient and cardiology. Eur J Heart Fail 2020; 22:2290-2309. [PMID: 32809231 PMCID: PMC8278961 DOI: 10.1002/ejhf.1985] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 08/12/2020] [Accepted: 08/14/2020] [Indexed: 02/06/2023] Open
Abstract
Advances in cancer treatments have improved clinical outcomes, leading to an increasing population of cancer survivors. However, this success is associated with high rates of short- and long-term cardiovascular (CV) toxicities. The number and variety of cancer drugs and CV toxicity types make long-term care a complex undertaking. This requires a multidisciplinary approach that includes expertise in oncology, cardiology and other related specialties, and has led to the development of the cardio-oncology subspecialty. This paper aims to provide an overview of the main adverse events, risk assessment and risk mitigation strategies, early diagnosis, medical and complementary strategies for prevention and management, and long-term follow-up strategies for patients at risk of cancer therapy-related cardiotoxicities. Research to better define strategies for early identification, follow-up and management is highly necessary. Although the academic cardio-oncology community may be the best vehicle to foster awareness and research in this field, additional stakeholders (industry, government agencies and patient organizations) must be involved to facilitate cross-discipline interactions and help in the design and funding of cardio-oncology trials. The overarching goals of cardio-oncology are to assist clinicians in providing optimal care for patients with cancer and cancer survivors, to provide insight into future areas of research and to search for collaborations with industry, funding bodies and patient advocates. However, many unmet needs remain. This document is the product of brainstorming presentations and active discussions held at the Cardiovascular Round Table workshop organized in January 2020 by the European Society of Cardiology.
Collapse
Affiliation(s)
- José Luis Zamorano
- Department of Cardiology, University Hospital Ramón y Cajal, CiberCV, Madrid, Spain
| | - Christer Gottfridsson
- Cardiovascular Safety Centre of Excellence, Patient Safety, CMO Organization, AstraZeneca, Gothenburg, Sweden
| | - Riccardo Asteggiano
- ESC Council of Cardio-Oncology, Insubria University of Medicine, Varese, Italy
- LARC (Laboratorio Analisi Ricerca Clinica), Turin, Italy
| | - Dan Atar
- Department of Cardiology, Oslo University Hospital Ulleval, Oslo, Norway
- Institute of Clinical Sciences, University of Oslo, Oslo, Norway
| | - Lina Badimon
- ESC Advocacy Committee 2018–2020, Director Cardiovascular Programme (ICCC)-IR Hospital de la Santa Creu I Sant Pau, CiberCV, Barcelona, Spain
| | - Jeroen J. Bax
- Department of Cardiology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Daniela Cardinale
- Cardio-Oncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | | | | | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | | | - Chris P. Gale
- Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - John H. Maduro
- Department of Radiation Oncology, University of Groningen, University Medical Centre Groningen, Groningen, the Netherlands
| | - Javid Moslehi
- Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital, University of Oslo, Oslo, Norway
| | - Juan Carlos Plana Gomez
- Department of Cardiology, Texas Heart Institute and Baylor College of Medicine, Houston, TX, USA
| | - Jessica Scott
- Exercise Oncology Research Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Thomas M. Suter
- Department of Cardiology, Inselspital, Bern University Hospital, Bern, Switzerland
| | - Giorgio Minotti
- Campus Bio-Medico University School of Medicine, Rome, Italy
| |
Collapse
|
307
|
Nso N, Antwi-Amoabeng D, Beutler BD, Ulanja MB, Ghuman J, Hanfy A, Nimo-Boampong J, Atanga S, Doshi R, Enoru S, Gullapalli N. Cardiac adverse events of immune checkpoint inhibitors in oncology patients: A systematic review and meta-analysis. World J Cardiol 2020; 12:584-598. [PMID: 33312443 PMCID: PMC7701899 DOI: 10.4330/wjc.v12.i11.584] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are novel therapeutic agents used for various types of cancer. ICIs have revolutionized cancer treatment and improved clinical outcomes among cancer patients. However, immune-related adverse effects of ICI therapy are common. Cardiovascular immune-related adverse events (irAEs) are rare but potentially life-threatening complications.
AIM To estimate the incidence of cardiovascular irAEs among patients undergoing ICI therapy for various malignancies.
METHODS We conducted this systematic review and meta-analysis by searching PubMed, Cochrane CENTRAL, Web of Science, and SCOPUS databases for relevant interventional trials reporting cardiovascular irAEs. We performed a single-arm meta-analysis using OpenMeta [Analyst] software of the following outcomes: Myocarditis, pericardial effusion, heart failure, cardiomyopathy, atrial fibrillation, myocardial infarction, and cardiac arrest. We assessed the heterogeneity using the I2 test and managed to solve it with Cochrane’s leave-one-out method. The risk of bias was performed with the Cochrane’s risk of bias tool.
RESULTS A total of 26 studies were included. The incidence of irAEs follows: Myocarditis: 0.5% [95% confidence interval (CI): 0.1%-0.9%]; Pericardial effusion: 0.5% (95%CI: 0.1%-1.0%); Heart failure: 0.3% (95%CI: 0.0%-0.5%); Cardiomyopathy: 0.3% (95%CI: -0.1%-0.6%); atrial fibrillation: 4.6% (95%CI: 1.0%-14.1%); Myocardial infarction: 0.4% (95%CI: 0.0%-0.7%); and Cardiac arrest: 0.4% (95%CI: 0.1%-0.8%).
CONCLUSION The most common cardiovascular irAEs were atrial fibrillation, myocarditis, and pericardial effusion. Although rare, data from post market surveillance will provide estimates of the long-term prevalence and prognosis in patients with ICI-associated cardiovascular complications.
Collapse
Affiliation(s)
- Nso Nso
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens, NY 10029, United States
| | - Daniel Antwi-Amoabeng
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Bryce D Beutler
- Department of Radiology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, United States
| | - Mark B Ulanja
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Jasmine Ghuman
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Ahmed Hanfy
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Joyce Nimo-Boampong
- Department of Internal Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Sirri Atanga
- Department of Medicine, United Health Services Wilson Medical Center, Johnson City, NY 13790, United States
| | - Rajkumar Doshi
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Sostanie Enoru
- Department of Cardiovascular Disease, SUNY Downstate Health Science University, Brooklyn, NY 11203, United States
| | - Nageshwara Gullapalli
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| |
Collapse
|
308
|
Farmakis D. Is cardio‐oncology a rapidly growing field of precision medicine? Eur J Heart Fail 2020; 22:2310-2313. [DOI: 10.1002/ejhf.2051] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 01/01/2023] Open
|
309
|
Iwata H, Akita K, Yamaba Y, Kunii E, Takakuwa O, Yoshihara M, Hattori Y, Nakajima K, Hayashi K, Toshito T, Ogino H, Shibamoto Y. Concurrent Chemo-Proton Therapy Using Adaptive Planning for Unresectable Stage 3 Non-Small Cell Lung Cancer: A Phase 2 Study. Int J Radiat Oncol Biol Phys 2020; 109:1359-1367. [PMID: 33227444 DOI: 10.1016/j.ijrobp.2020.11.035] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 11/05/2020] [Accepted: 11/12/2020] [Indexed: 02/07/2023]
Abstract
PURPOSE This study prospectively evaluated the efficacy and safety of concurrent chemo-proton therapy (CCPT) using adaptive planning for unresectable stage III non-small cell lung cancer (NSCLC). METHODS AND MATERIALS The primary endpoint was overall survival (OS). Secondary endpoints were local control rate (LCR), progression-free survival (PFS), incidence of grade 3 or higher adverse events, and changes in quality of life (QOL). Patients received cisplatin (60 mg/m2) on day 1 and S-1 (∼40 mg/m2 twice daily) on days 1 to 14, q4w, for up to 4 cycles, plus concurrent proton therapy at a total dose of 70 GyRBE for the primary lesion and 66 GyRBE for lymph node metastasis with 2 GyRBE per day. Proton therapy was performed using respiratory-gated and image guided techniques, and adaptive plans were implemented. RESULTS Forty-seven patients were enrolled between August 2013 and August 2018. Four cycles of cisplatin plus S-1 were completed in 34 patients. The mean number of cycles was 4 (range, 1-4). The median follow-up of all and surviving patients was 37 (range, 4-84) and 52 months (range, 26-84), respectively. The mean number of replanning sessions was 2.5 (range, 1-4). The 2- and 5-year OS, LCR, and PFS were 77% (95% confidence interval 64%-89%) and 59% (43%-76%), 84% (73%-95%) and 61% (44%-78%), and 43% (28%-57%) and 37% (22%-51%), respectively. The median OS was not reached. No grade 3 or higher radiation pneumonitis was observed. There was no significant deterioration in the QOL scores after 24 months except for alopecia. CONCLUSIONS CCPT with adaptive planning was well tolerated and yielded remarkable OS for unresectable stage III NSCLC.
Collapse
Affiliation(s)
- Hiromitsu Iwata
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City West Medical Center, Nagoya, Japan; Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan.
| | - Kenji Akita
- Department of Respiratory Tract Oncology Center, Nagoya City West Medical Center, Nagoya, Japan
| | - Yusuke Yamaba
- Department of Respiratory Tract Oncology Center, Nagoya City West Medical Center, Nagoya, Japan
| | - Eiji Kunii
- Department of Respiratory Tract Oncology Center, Nagoya City West Medical Center, Nagoya, Japan
| | - Osamu Takakuwa
- Department of Respiratory Tract Oncology Center, Nagoya City West Medical Center, Nagoya, Japan
| | - Misuzu Yoshihara
- Department of Respiratory Tract Oncology Center, Nagoya City West Medical Center, Nagoya, Japan
| | - Yukiko Hattori
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City West Medical Center, Nagoya, Japan
| | - Koichiro Nakajima
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City West Medical Center, Nagoya, Japan; Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Kensuke Hayashi
- Department of Proton Therapy Technology, Nagoya Proton Therapy Center, Nagoya, Japan
| | - Toshiyuki Toshito
- Department of Proton Therapy Physics, Nagoya Proton Therapy Center, Nagoya, Japan
| | - Hiroyuki Ogino
- Department of Radiation Oncology, Nagoya Proton Therapy Center, Nagoya City West Medical Center, Nagoya, Japan; Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| | - Yuta Shibamoto
- Department of Radiology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
310
|
Abstract
In recent decades, major advances in the treatment of malignant diseases have significantly improved long-term survival. However, this has increased the spectrum of side effects of these treatment methods, particularly for the cardiovascular system. Cardiotoxicity can be acute and chronic, including hypertension, heart failure, arrhythmias, acute myocardial infarction, venous thromboembolism, stroke, and valvular heart disease. While the occurrence of cardiotoxicity is known for many older cancer therapies, it needs to be largely evaluated for newer forms of therapy. Diagnosing possible cardiotoxic side effects is essential for optimal treatment, but remains a challenge. Troponin and the natriuretic peptides play an essential role as cardiac biomarkers in the diagnosis of conventional heart diseases. However, they also appear to play an important role in the detection of cardiotoxicity, as well as in the treatment of cardio-oncology patients. Elevated troponin or B-type natriuretic peptide (BNP)/N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels are associated with increased overall mortality and were associated with the development of heart failure in selected cohorts. Troponin can also be used to identify myocarditis associated with immune checkpoint inhibitor therapy. This overview summarizes the current knowledge about biomarkers for the detection of cardiotoxicity due to tumor therapy. Possible clinical recommendations for the detection of cardiotoxic effects using biomarkers are also outlined.
Collapse
Affiliation(s)
- M Totzeck
- Westdeutsches Herz- und Gefäßzentrum, Klinik für Kardiologie und Angiologie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| | - M Glas
- Abteilung für Klinische Neuroonkologie, Klinik für Neurologie, Universitätsklinikum Essen, Essen, Deutschland
| | - T Rassaf
- Westdeutsches Herz- und Gefäßzentrum, Klinik für Kardiologie und Angiologie, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland
| |
Collapse
|
311
|
Salido Iniesta M, López López L, Carreras Costa F, Sionis A. A different type of acute myocarditis: a case report of acute autoimmune myocarditis mediated by anti-PD-1 T lymphocyte receptor (pembrolizumab). EUROPEAN HEART JOURNAL-CASE REPORTS 2020; 4:1-6. [PMID: 33204987 PMCID: PMC7649507 DOI: 10.1093/ehjcr/ytaa214] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 12/30/2019] [Accepted: 06/17/2020] [Indexed: 01/22/2023]
Abstract
Background Pembrolizumab is an immune check-point inhibitor (ICI), which acts by blocking the T lymphocyte PD-1 inhibitor receptor. It has been increasingly used in advanced or non-responsive tumours with promising results. However, acute myocarditis is an infrequent but potentially life-threatening autoimmune adverse effect related to ICIs. Case summary This case deals with a 69-year-old gentleman on second-line therapy with pembrolizumab for advanced non-small cell lung cancer. Three weeks after first dose, the patient was diagnosed with an autoimmune hepatitis, treated with decreasing corticoid dosage, followed by acute heart failure. On admission, his electrocardiogram (ECG) showed diffuse repolarization changes and a transthoracic echocardiography revealed severe left ventricle impairment (left ventricular ejection fraction 32%). High-sensitivity cardiac troponin was elevated and a coronary angiogram was performed showing non-significant obstructive disease. An autoimmune myocarditis was suspected, and high-dose intravenous corticoid, intravenous vasodilators, and loop diuretics were started with favourable response. Cardiac magnetic resonance (CMR) imaging, performed 2 weeks after clinical onset, revealed extracellular oedema in the anteroseptal-apical left ventricle segments. A new transthoracic echocardiography, performed after 3 months, showed preserved left ventricle ejection fraction. Finally, the patient was readmitted due to an autoimmune myasthenia-like syndrome. Discussion Acute autoimmune myocarditis related to ICIs is a challenging diagnosis and its incidence has been underestimated in early studies. Endomyocardial biopsy (EMB) is the gold standard test for its diagnosis. Nevertheless, a definite myocarditis diagnosis is possible without EMB when characteristic clinical syndrome, elevated myonecrosis markers, and electrocardiographic, echocardiographic, and CMR changes are present together.
Collapse
Affiliation(s)
- Mario Salido Iniesta
- Hospital de la Santa Creu i Sant Pau IIB Sant Pau, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| | - Laura López López
- Hospital de la Santa Creu i Sant Pau IIB Sant Pau, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| | - Francesc Carreras Costa
- Hospital de la Santa Creu i Sant Pau IIB Sant Pau, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| | - Alessandro Sionis
- Hospital de la Santa Creu i Sant Pau IIB Sant Pau, Carrer de Sant Quintí, 89, 08041 Barcelona, Spain
| |
Collapse
|
312
|
Londoño MC, Reig M, on behalf of the RETOINMUNO Multidisciplinary Group. Multidisciplinary Clinical Approach to Cancer Patients with Immune-Related Adverse Events Induced by Checkpoint Inhibitors. Cancers (Basel) 2020; 12:E3446. [PMID: 33228219 PMCID: PMC7699587 DOI: 10.3390/cancers12113446] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Revised: 11/12/2020] [Accepted: 11/16/2020] [Indexed: 02/07/2023] Open
Abstract
Immune-oncology is a major breakthrough in cancer treatment and has become the standard of care for a wide variety of solid organ malignancies. Unfortunately, manipulation of the immune system with checkpoint inhibitors may result in an immune-based attack of normal tissues which can lead to treatment discontinuation. These immune-related adverse events (irAEs) are diverse and affect several organs, constituting a new clinical challenge in the management of cancer patients. The complexity of this scenario requires a multidisciplinary approach that allows the early identification, diagnosis and treatment of specific irAE, ruling out other non-related adverse events. Hospital Clinic has a multidisciplinary team seeking to develop a coordinated strategy to facilitate the access of patients with suspected irAEs to specialised care resulting in harmonised management that guarantees the best patient care. The aim of the manuscript was to describe the current evidence on the management of irAEs reflecting a coordinated multidisciplinary approach to face this clinical challenge regardless of the immunotherapy indication.
Collapse
Affiliation(s)
- Maria-Carlota Londoño
- Liver Unit, Hospital Clinic Barcelona, IDIBAPS, University of Barcelona, CIBERehd, 08036 Barcelona, Spain
| | - Maria Reig
- Liver Liver Cancer Group (BCLC), Liver Unit, Hospital Clínic Barcelona, IDIBAPS, University of Barcelona, CIBERehd, 08036 Barcelona, Spain
| | | |
Collapse
|
313
|
Milazzo V, Cosentino N, Campodonico J, Lucci C, Cardinale D, Cipolla CM, Marenzi G. Characteristics, Management, and Outcomes of Acute Coronary Syndrome Patients with Cancer. J Clin Med 2020; 9:E3642. [PMID: 33198355 PMCID: PMC7696544 DOI: 10.3390/jcm9113642] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/09/2020] [Indexed: 02/06/2023] Open
Abstract
Patients with cancer are at increased risk of cardiovascular disease, with a reported prevalence of acute coronary syndrome (ACS) ranging from 3% to 17%. The increased risk of ACS in these patients seems to be due to the complex interaction of shared cardiovascular risk factors, cancer type and stage, and chemotherapeutic and radiotherapy regimens. The management of ACS in patients with cancer is a clinical challenge, particularly due to cancer's unique pathophysiology, which makes it difficult to balance thrombotic and bleeding risks in this specific patient population. In addition, patients with cancer have largely been excluded from ACS trials. Hence, an evidence-based treatment for ACS in this group of patients is unknown and only a limited proportion of them is treated with antiplatelets or invasive revascularization, despite initial reports suggesting their beneficial prognostic effects in cancer patients. Finally, cancer patients experiencing ACS are also at higher risk of in-hospital and long-term mortality as compared to non-cancer patients. In this review, we will provide an overview on the available evidence of the relationship between ACS and cancer, in terms of clinical manifestations, possible underlying mechanisms, and therapeutic and prognostic implications.
Collapse
Affiliation(s)
- Valentina Milazzo
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (V.M.); (N.C.); (J.C.); (C.L.)
| | - Nicola Cosentino
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (V.M.); (N.C.); (J.C.); (C.L.)
| | - Jeness Campodonico
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (V.M.); (N.C.); (J.C.); (C.L.)
| | - Claudia Lucci
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (V.M.); (N.C.); (J.C.); (C.L.)
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, 20141 Milan, Italy;
| | - Carlo M. Cipolla
- Cardiology Division, European Institute of Oncology, IRCCS, 20141 Milan, Italy;
| | - Giancarlo Marenzi
- Centro Cardiologico Monzino IRCCS, 20138 Milan, Italy; (V.M.); (N.C.); (J.C.); (C.L.)
| |
Collapse
|
314
|
|
315
|
Romitan DM, Rădulescu D, Berindan-Neagoe I, Stoicescu L, Grosu A, Rădulescu L, Gulei D, Ciuleanu TE. Cardiomyopathies and Arrhythmias Induced by Cancer Therapies. Biomedicines 2020; 8:biomedicines8110496. [PMID: 33198152 PMCID: PMC7696637 DOI: 10.3390/biomedicines8110496] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 12/17/2022] Open
Abstract
Cardiology and oncology are two fields dedicated to the study of various types of oncological and cardiac diseases, but when they collide, a new specialty is born, i.e., cardio-oncology. Continuous research on cancer therapy has brought into the clinic novel therapeutics that have significantly improved patient survival. However, these therapies have also been associated with adverse effects that can impede the proper management of oncological patients through the necessity of drug discontinuation due to life-threatening or long-term morbidity risks. Cardiovascular toxicity from oncological therapies is the main issue that needs to be solved. Proper knowledge, interpretation, and management of new drugs are key elements for developing the best therapeutic strategies for oncological patients. Upon continuous investigations, the profile of cardiotoxicity events has been enlarged with the inclusion of myocarditis upon administration of immune checkpoint inhibitors and cardiac dysfunction in the context of cytokine release syndrome with chimeric antigen receptor T cell therapy. Affinity enhanced and chimeric antigen receptor T cells have both been associated with hypotension, arrhythmia, and left ventricular dysfunction, typically in the setting of cytokine release syndrome. Therefore, the cardiologist must adhere to the progressing field of cancer therapy and become familiar with the adverse effects of novel drugs, and not only the ones of standard care, such as anthracycline, trastuzumab, and radiation therapy. The present review provides essential information summarized from the latest studies from cardiology, oncology, and hematology to bring together the three specialties and offers proper management options for oncological patients.
Collapse
Affiliation(s)
- Dragoș-Mihai Romitan
- Department of Cardiology, Municipal Clinical Hospital of Cluj-Napoca, 400139 Cluj-Napoca, Romania; (D.R.); (L.S.); (A.G.); (L.R.)
- Correspondence:
| | - Dan Rădulescu
- Department of Cardiology, Municipal Clinical Hospital of Cluj-Napoca, 400139 Cluj-Napoca, Romania; (D.R.); (L.S.); (A.G.); (L.R.)
| | - Ioana Berindan-Neagoe
- Research Center for Functional Genomic, Biomedicine and Translational Medicine, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400139 Cluj-Napoca, Romania;
| | - Laurențiu Stoicescu
- Department of Cardiology, Municipal Clinical Hospital of Cluj-Napoca, 400139 Cluj-Napoca, Romania; (D.R.); (L.S.); (A.G.); (L.R.)
| | - Alin Grosu
- Department of Cardiology, Municipal Clinical Hospital of Cluj-Napoca, 400139 Cluj-Napoca, Romania; (D.R.); (L.S.); (A.G.); (L.R.)
| | - Liliana Rădulescu
- Department of Cardiology, Municipal Clinical Hospital of Cluj-Napoca, 400139 Cluj-Napoca, Romania; (D.R.); (L.S.); (A.G.); (L.R.)
| | - Diana Gulei
- Research Center for Advanced Medicine-Medfuture, “Iuliu Hațieganu” University of Medicine and Pharmacy Cluj-Napoca, 400139 Cluj-Napoca, Romania;
| | - Tudor-Eliade Ciuleanu
- Department of Chemotherapy, Ion Chiricuta Clinical Cancer Center, 400139 Cluj Napoca, Romania;
| |
Collapse
|
316
|
Onishi T, Fukuda Y, Miyazaki S, Yamada H, Tanaka H, Sakamoto J, Daimon M, Izumi C, Nonaka A, Nakatani S, Akaishi M. Practical guidance for echocardiography for cancer therapeutics-related cardiac dysfunction. J Echocardiogr 2020; 19:1-20. [PMID: 33159650 PMCID: PMC7932955 DOI: 10.1007/s12574-020-00502-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2020] [Revised: 10/15/2020] [Accepted: 10/21/2020] [Indexed: 01/06/2023]
Abstract
The prognosis of patients with cancer has improved due to an early diagnosis of cancer and advances in cancer treatment. There are emerging reports on cardiotoxicity in cancer treatment and on cardiovascular disease in cancer patients, from which cardiovascular disease has been recognized as a common cause of death among cancer survivors. This situation has led to the need for a medical system in which oncologists and cardiologists work together to treat patients. With the growing importance of onco-cardiology, the role of echocardiography in cancer care is rapidly expanding, but at present, the practice of echocardiography in clinical settings varies from institution to institution, and is empirical with no established systematic guidance. In view of these circumstances, we thought that brief guidance for clinical application was necessary and have therefore developed this guidance, although evidence in this field is still insufficient.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Chisato Izumi
- Department of Cardiovascular Medicine, National Cerebral and Cardiovascular Center, 6-1 Kishibeshimmachi, Suita, Osaka, Japan.
| | | | | | | | | |
Collapse
|
317
|
Dolladille C, Ederhy S, Allouche S, Dupas Q, Gervais R, Madelaine J, Sassier M, Plane AF, Comoz F, Cohen AA, Thuny FR, Cautela J, Alexandre J. Late cardiac adverse events in patients with cancer treated with immune checkpoint inhibitors. J Immunother Cancer 2020; 8:jitc-2019-000261. [PMID: 31988143 PMCID: PMC7057417 DOI: 10.1136/jitc-2019-000261] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/18/2019] [Indexed: 12/17/2022] Open
Abstract
Background Immune checkpoint inhibitor (ICI)-associated early cardiac adverse events (CAEs), mostly acute and fulminant myocarditis, have been well characterized and mainly occur during the first 90 days after ICI therapy initiation. ICI-associated late CAEs (occurring after the first 90 days of treatment) have not yet been described. Methods First, we compared characteristics of a cohort involving early (defined as a CAE time to onset (TTO) of <90 days after ICI therapy initiation) and late (defined as a CAE TTO of ≥90 days after ICI therapy initiation) ICI-associated CAE consecutive cases who were referred to three French cardio-oncology units. Second, ICI-associated CAE cases were searched in VigiBase, the WHO global individual case safety report database, and early and late ICI-associated CAEs were compared. Results In the cohort study, compared with early CAE cases (n=19, median TTO of 14 days), late ICI-associated CAE cases (n=19, median TTO of 304 days) exhibited significantly more left ventricular systolic dysfunction (LVSD) and heart failure (HF) and less frequent supraventricular arrhythmias. In VigiBase, compared with early cases (n=437, 73.3%, median TTO 21 days), the late ICI-associated CAE reports (n=159, 26.7%, median TTO 178 days) had significantly more frequent HF (21.1% vs 31.4%, respectively, p=0.01). Early and late ICI-associated CAE cases had similarly high mortality rates (40.0% vs 44.4% in the cohort and 30.0% vs 27.0% in VigiBase, respectively). Conclusions Late CAEs could occur with ICI therapy and were mainly revealed to be HF with LVSD. Trial registration numbers NCT03678337, NCT03882580, and NCT03492528.
Collapse
Affiliation(s)
- Charles Dolladille
- CHU de Caen, PICARO Cardio-oncology Program, Department of Pharmacology, CHU de Caen, Caen, France .,CHU de Caen, Department of Cardiology, CHU de Caen, Caen F-14000, France
| | - Stephane Ederhy
- Department of Cardiology, Cardio-oncology Unit, Cardio-oncology Research G, INSERM U 856, Thrombose, Athérothrombose et Pharmacologie Appliquée, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Stéphane Allouche
- EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-reperfusion Myocardique, Université de Caen Normandie, Caen, Normandie, France
| | - Querntin Dupas
- EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-reperfusion Myocardique, Université de Caen Normandie, Caen, Normandie, France
| | - Radj Gervais
- Onco-pneumology, Centre François Baclesse Centre de Lutte Contre le Cancer, Caen, France
| | | | - Marion Sassier
- CHU de Caen, PICARO Cardio-oncology Program, Department of Pharmacology, CHU de Caen, Caen, France
| | - Anne-Flore Plane
- CHU de Caen, Department of Cardiology, CHU de Caen, Caen F-14000, France
| | | | - Ariel Aron Cohen
- Department of Cardiology, Cardio-oncology Unit, Cardio-oncology Research G, INSERM U 856, Thrombose, Athérothrombose et Pharmacologie Appliquée, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Franck Roland Thuny
- Mediterranean University Cardio-oncology Center (MEDI-CO Center), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Hôpital Nord, Marseille, APHM, Marseille, Provence-Alpes-Côte d'Azu, France
| | - Jennifer Cautela
- Mediterranean University Cardio-oncology Center (MEDI-CO Center), Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Hôpital Nord, Marseille, APHM, Marseille, Provence-Alpes-Côte d'Azu, France
| | - Joachim Alexandre
- CHU de Caen, PICARO Cardio-oncology Program, Department of Pharmacology, CHU de Caen, Caen, France.,EA4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-reperfusion Myocardique, Université de Caen Normandie, Caen, Normandie, France
| |
Collapse
|
318
|
Lyon AR, Dent S, Stanway S, Earl H, Brezden-Masley C, Cohen-Solal A, Tocchetti CG, Moslehi JJ, Groarke JD, Bergler-Klein J, Khoo V, Tan LL, Anker MS, von Haehling S, Maack C, Pudil R, Barac A, Thavendiranathan P, Ky B, Neilan TG, Belenkov Y, Rosen SD, Iakobishvili Z, Sverdlov AL, Hajjar LA, Macedo AV, Manisty C, Ciardiello F, Farmakis D, de Boer RA, Skouri H, Suter TM, Cardinale D, Witteles RM, Fradley MG, Herrmann J, Cornell RF, Wechelaker A, Mauro MJ, Milojkovic D, de Lavallade H, Ruschitzka F, Coats AJ, Seferovic PM, Chioncel O, Thum T, Bauersachs J, Andres MS, Wright DJ, López-Fernández T, Plummer C, Lenihan D. Baseline cardiovascular risk assessment in cancer patients scheduled to receive cardiotoxic cancer therapies: a position statement and new risk assessment tools from the Cardio-Oncology Study Group of the Heart Failure Association of the European Society of Cardiology in collaboration with the International Cardio-Oncology Society. Eur J Heart Fail 2020; 22:1945-1960. [PMID: 32463967 PMCID: PMC8019326 DOI: 10.1002/ejhf.1920] [Citation(s) in RCA: 453] [Impact Index Per Article: 90.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2020] [Revised: 05/13/2020] [Accepted: 05/25/2020] [Indexed: 12/11/2022] Open
Abstract
This position statement from the Heart Failure Association of the European Society of Cardiology Cardio-Oncology Study Group in collaboration with the International Cardio-Oncology Society presents practical, easy-to-use and evidence-based risk stratification tools for oncologists, haemato-oncologists and cardiologists to use in their clinical practice to risk stratify oncology patients prior to receiving cancer therapies known to cause heart failure or other serious cardiovascular toxicities. Baseline risk stratification proformas are presented for oncology patients prior to receiving the following cancer therapies: anthracycline chemotherapy, HER2-targeted therapies such as trastuzumab, vascular endothelial growth factor inhibitors, second and third generation multi-targeted kinase inhibitors for chronic myeloid leukaemia targeting BCR-ABL, multiple myeloma therapies (proteasome inhibitors and immunomodulatory drugs), RAF and MEK inhibitors or androgen deprivation therapies. Applying these risk stratification proformas will allow clinicians to stratify cancer patients into low, medium, high and very high risk of cardiovascular complications prior to starting treatment, with the aim of improving personalised approaches to minimise the risk of cardiovascular toxicity from cancer therapies.
Collapse
Affiliation(s)
- Alexander R. Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and Imperial College, London, UK
- Corresponding author. Cardio-Oncology Service, Royal Brompton Hospital, Sydney Street, London SW3 6NP, UK. Tel: +44 20 7352 8121,
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | | | - Helena Earl
- Department of Oncology, University of Cambridge and NIHR Cambridge Biomedical Research Centre, Cambridge, UK
| | | | - Alain Cohen-Solal
- UMR-S 942, Paris University, Cardiology Department, Lariboisiere Hospital, AP-HP, Paris, France
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center for Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Javid J. Moslehi
- Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - John D. Groarke
- Cardio-Oncology Program, Brigham & Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Vincent Khoo
- Department of Clinical Oncology, Royal Marsden Hospital and Institute of Cancer Research, London, UK
- Department of Medical Imaging and Radiation Sciences, Monash University and Department of Medicine, Melbourne University, Melbourne, Australia
| | - Li Ling Tan
- Department of Cardiology, National University Heart Centre, Singapore, National University Health System, Singapore, Singapore
| | - Markus S. Anker
- Division of Cardiology and Metabolism, Department of Cardiology, Charité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research), partner site Berlin and Department of Cardiology, Charité Campus Benjamin Franklin, Berlin, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, University of Goettingen Medical Center, Goettingen, Germany
- German Center for Cardiovascular Research (DZHK), partner site Goettingen, Goettingen, Germany
| | - Christoph Maack
- Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Radek Pudil
- First Department of Medicine – Cardioangiology, Charles University Prague, Medical Faculty and University Hospital Hradec Kralove, Prague, Czech Republic
| | - Ana Barac
- MedStar Heart and Vascular Institute, Georgetown University, Washington, DC, USA
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention and Joint Division of Medical Imaging, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Bonnie Ky
- University of Pennsylvania, Philadelphia, PA, USA
| | - Tomas G. Neilan
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Stuart D. Rosen
- Cardio-Oncology Service, Royal Brompton Hospital and Imperial College, London, UK
| | - Zaza Iakobishvili
- Department of Community Cardiology, Tel Aviv Jaffa District, Clalit Health Fund and Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Aaron L. Sverdlov
- School of Medicine and Public Health, University of Newcastle and “Cancer and the Heart” Program, Hunter New England LHD, Newcastle, Australia
| | - Ludhmila A. Hajjar
- Cardio-Oncology, Department of Cardio-Pneumology, University of São Paulo, São Paulo, Brazil
| | - Ariane V.S. Macedo
- Santa Cardio-Oncology, Santa Casa de São Paulo and Rede Dor São Luiz, São Paulo, Brazil
| | | | - Fortunato Ciardiello
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Dimitrios Farmakis
- University of Cyprus Medical School, Nicosia, Cyprus
- Cardio-Oncology Clinic, Heart Failure Unit, “Attikon” University Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Rudolf A. de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Hadi Skouri
- Cardiology Division, Internal Medicine Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Thomas M. Suter
- Department of Cardiology, Bern University Hospital, Inselspital, University of Bern, Bern, Switzerland
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan, Italy
| | | | | | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Dragana Milojkovic
- Department of Haematology, Hammersmith Hospital, Imperial College, London, UK
| | | | - Frank Ruschitzka
- University Heart Center, Department of Cardiology, University Hospital Zurich, Zurich, Switzerland
| | - Andrew J.S. Coats
- University of Warwick, Warwick, UK
- Pharmacology, Centre of Clinical and Experimental Medicine, IRCCS San Raffaele Pisana, Rome, Italy
| | - Petar M. Seferovic
- Faculty of Medicine and Serbian Academy of Sciences and Arts, University of Belgrade, Belgrade, Serbia
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases ‘Prof. C.C. Iliescu’, Bucharest, Romania
- University of Medicine Carol Davila, Bucharest, Romania
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - M. Sol Andres
- Cardio-Oncology Service, Royal Brompton Hospital and Imperial College, London, UK
| | - David J. Wright
- Liverpool Centre for Cardiovascular Science, Liverpool Heart and Chest Hospital, Liverpool, UK
| | - Teresa López-Fernández
- Cardiology Service, Cardio-Oncology Unit, La Paz University Hospital and IdiPAz Research Institute, Ciber CV, Madrid, Spain
| | - Chris Plummer
- Department of Cardiology, The Newcastle upon Tyne Hospitals NHS Foundation Trust and Newcastle University, Newcastle, UK
| | - Daniel Lenihan
- Cardio-Oncology Center of Excellence, Washington University in St Louis, St Louis, MO, USA
| |
Collapse
|
319
|
Gutierrez C, Rajendram P, Pastores SM. Toxicities Associated with Immunotherapy and Approach to Cardiotoxicity with Novel Cancer Therapies. Crit Care Clin 2020; 37:47-67. [PMID: 33190775 DOI: 10.1016/j.ccc.2020.08.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In recent years, major advances in oncology especially the advent of targeted agents and immunotherapies (immune checkpoint inhibitors [ICIs] and chimeric antigen receptor [CAR] T-cell therapy) have led to improved quality of life and survival rates in patients with cancer. This article focuses on the clinical features, and grading and management of toxicities associated with ICIs and CAR T-cell therapy. In addition, because cardiotoxicity is one of the most harmful effects of anticancer therapeutics, we describe the risk factors and mechanisms of cardiovascular injury associated with newer agents, screening technologies for at-risk patients, and preventive and treatment strategies.
Collapse
Affiliation(s)
- Cristina Gutierrez
- Department of Critical Care Medicine, University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX 77030, USA
| | - Prabalini Rajendram
- Department of Critical Care, Respiratory Institute, Cleveland Clinic, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | - Stephen M Pastores
- Department of Anesthesiology and Critical Care Medicine, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Avenue C-1179, New York, NY 10065, USA.
| |
Collapse
|
320
|
Pudil R, Mueller C, Čelutkienė J, Henriksen PA, Lenihan D, Dent S, Barac A, Stanway S, Moslehi J, Suter TM, Ky B, Štěrba M, Cardinale D, Cohen‐Solal A, Tocchetti CG, Farmakis D, Bergler‐Klein J, Anker MS, Von Haehling S, Belenkov Y, Iakobishvili Z, Maack C, Ciardiello F, Ruschitzka F, Coats AJ, Seferovic P, Lainscak M, Piepoli MF, Chioncel O, Bax J, Hulot J, Skouri H, Hägler‐Laube ES, Asteggiano R, Fernandez TL, Boer RA, Lyon AR. Role of serum biomarkers in cancer patients receiving cardiotoxic cancer therapies: a position statement from the
Cardio‐Oncology Study Group
of the
Heart Failure Association
and the
Cardio‐Oncology Council of the European Society of Cardiology. Eur J Heart Fail 2020; 22:1966-1983. [DOI: 10.1002/ejhf.2017] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/14/2020] [Accepted: 09/25/2020] [Indexed: 12/19/2022] Open
Affiliation(s)
- Radek Pudil
- 1st Department Medicine – Cardioangiology Charles University Prague, Medical Faculty and University Hospital Hradec Kralove Prague Czech Republic
| | - Christian Mueller
- Cardiovascular Research Institute Basel (CRIB) and Department of Cardiology University Hospital Basel, University of Basel Basel Switzerland
| | - Jelena Čelutkienė
- Clinic of Cardiac and Vascular Diseases Institute of Clinical Medicine, Faculty of Medicine, Vilnius University Vilnius Lithuania
- State Research Institute Centre For Innovative Medicine Vilnius Lithuania
| | | | - Dan Lenihan
- Cardio‐Oncology Center of Excellence Washington University in St Louis St Louis MO USA
| | - Susan Dent
- Duke Cancer Institute Duke University Durham NC USA
| | - Ana Barac
- MedStar Heart and Vascular Institute Georgetown University Washington DC USA
| | | | - Javid Moslehi
- Cardio‐Oncology Program, Department of Medicine Vanderbilt University Medical Center Nashville TN USA
| | - Thomas M. Suter
- Department of Cardiology Bern University Hospital, Inselspital, University of Bern Bern Switzerland
| | - Bonnie Ky
- University of Pennsylvania Philadelphia PA USA
| | - Martin Štěrba
- Department of Pharmacology Faculty of Medicine in Hradec Kralove, Charles University Hradec Kralove Czech Republic
| | - Daniela Cardinale
- Cardioncology Unit European Institute of Oncology, IRCCS Milan Italy
| | - Alain Cohen‐Solal
- UMR‐S 942, Paris University, Cardiology Department, Lariboisiere Hospital, AP‐HP Paris France
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center for Clinical and Translational Research (CIRCET) ‘Federico II’ University Naples Italy
| | - Dimitrios Farmakis
- University of Cyprus Medical School Nicosia Cyprus
- Cardio‐Oncology Clinic, Heart Failure Unit, ‘Attikon’ University Hospital Athens Greece
- National and Kapodistrian University of Athens Medical School Athens Greece
| | | | - Markus S. Anker
- Division of Cardiology and Metabolism, Department of Cardiology Charité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research), partner site Berlin and Department of Cardiology, Charité Campus Benjamin Franklin Berlin Germany
| | - Stephan Von Haehling
- Department of Cardiology and Pneumology University of Goettingen Medical Center Goettingen Germany
- German Center for Cardiovascular Research (DZHK), partner site Goettingen Goettingen Germany
| | | | - Zaza Iakobishvili
- Department of Community Cardiology Tel Aviv Jaffa District, Clalit Health Fund and Sackler Faculty of Medicine, Tel Aviv University Tel Aviv Israel
| | - Christoph Maack
- Comprehensive Heart Failure Center University Clinic Würzburg Würzburg Germany
| | - Fortunato Ciardiello
- Department of Precision Medicine ‘Luigi Vanvitelli’ University of Campania Naples Italy
| | - Frank Ruschitzka
- University Heart Center, Department of Cardiology University Hospital Zurich Zurich Switzerland
| | - Andrew J.S. Coats
- University of Warwick Warwick UK
- Pharmacology Centre of Clinical and Experimental Medicine, IRCCS San Raffaele Pisana Rome Italy
| | - Petar Seferovic
- Faculty of Medicine and Serbian Academy of Sciences and Arts University of Belgrade Belgrade Serbia
| | | | - Massimo F. Piepoli
- Cardiac Department ‘Guglielmo da Saliceto’ Polichirurgico Hospital AUSL Piacenza Piacenza Italy
| | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases ‘Prof. C.C. Iliescu’ Bucharest Romania
- University of Medicine Carol Davila Bucharest Romania
| | - Jereon Bax
- Department of Cardiology Leiden University Medical Centre Leiden The Netherlands
| | - Jean‐Sebastien Hulot
- Université de Paris CIC1418, Paris Cardiovascular Research Center, INSERM Paris France
| | - Hadi Skouri
- Cardiology Division, Internal Medicine Department at American University of Beirut Medical Center Beirut Lebanon
| | | | | | - Teresa Lopez Fernandez
- Cardiology Service Cardio‐Oncology Unit, La Paz University Hospital and IdiPAz Research Institute, Ciber CV Madrid Spain
| | - Rudolf A. Boer
- Department of Cardiology University of Groningen, University Medical Center Groningen Groningen The Netherlands
| | - Alexander R. Lyon
- Cardio‐Oncology Service Royal Brompton Hospital and Imperial College London London UK
| |
Collapse
|
321
|
Quagliariello V, Passariello M, Rea D, Barbieri A, Iovine M, Bonelli A, Caronna A, Botti G, De Lorenzo C, Maurea N. Evidences of CTLA-4 and PD-1 Blocking Agents-Induced Cardiotoxicity in Cellular and Preclinical Models. J Pers Med 2020; 10:jpm10040179. [PMID: 33086484 PMCID: PMC7711520 DOI: 10.3390/jpm10040179] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/29/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Several strategies based on immune checkpoint inhibitors (ICIs) have been developed for cancer therapy, opening to advantages in cancer outcomes. However, several ICI-induced side effects have emerged in these patients, especially a rare but clinically significant cardiotoxicity with high rate of mortality. We studied the cytotoxic and pro-inflammatory properties of Ipilimumab and Nivolumab, the underlying pathways and cytokine storm involved. Methods: Co-cultures of human cardiomyocytes and lymphocytes were exposed to Ipilimumab or Nivolumab; cell viability and expression of leukotrienes, NLRP3, MyD88, and p65/NF-kB were performed. C57 mice were treated with Ipilimumab (15 mg/kg); analysis of fractional shortening, ejection fraction, radial and longitudinal strain were made before and after treatments through 2D-echocardiography. Expression of NLRP3, MyD88, p65/NF-kB, and 12 cytokines were analyzed in murine myocardium. Results: Nivolumab and Ipilimumab exert effective anticancer, but also significant cardiotoxic effects in co-cultures of lymphocytes and tumor or cardiac cells. Both ICIs increased NLRP3, MyD88, and p65/NF-kB expression compared to untreated cells, however, the most pro-inflammatory and cardiotoxic effects were seen after exposure to Ipilimumab. Mice treated with Ipilimumab showed a significant decrease in fractional shortening and radial strain with respect to untreated mice, coupled with a significant increase in myocardial expression of NLRP3, MyD88, and several interleukins. Conclusions: Nivolumab and Ipilimumab exert cytotoxic effects mediated by the NLRP3/IL-1β and MyD88 pathways, leading to pro-inflammatory cytokine storm in heart tissue.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | | | - Domenica Rea
- Animal Facility, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (D.R.); (A.B.)
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (D.R.); (A.B.)
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Annamaria Bonelli
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Antonietta Caronna
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Claudia De Lorenzo
- CEINGE—Biotecnologie Avanzate s.c.a.r.l., 80131 Naples, Italy;
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Napoli, Italy
- Correspondence: (C.D.L.); (N.M.)
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
- Correspondence: (C.D.L.); (N.M.)
| |
Collapse
|
322
|
Lewis LD. Medicamenta ad sentinam, et defectum vitae: Drugs and failure of the pump of life-cardiotoxicity of oncotherapeutics. Br J Clin Pharmacol 2020; 87:735-737. [PMID: 33073394 DOI: 10.1111/bcp.14558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 07/29/2020] [Accepted: 09/11/2020] [Indexed: 11/30/2022] Open
Affiliation(s)
- Lionel D Lewis
- Section of Clinical Pharmacology, Department of Medicine, The Geisel School of Medicine at Dartmouth and The Dartmouth-Hitchcock Medical Center, Lebanon, New Hampshire, USA
| |
Collapse
|
323
|
Safi M, Ahmed H, Al-Azab M, Xia YL, Shan X, Al-radhi M, Al-danakh A, Shopit A, Liu J. PD-1/PDL-1 Inhibitors and Cardiotoxicity; Molecular, Etiological and Management Outlines. J Adv Res 2020; 29:45-54. [PMID: 33842004 PMCID: PMC8020146 DOI: 10.1016/j.jare.2020.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022] Open
Abstract
Background The US Food and Drug Administration (FDA) has approved several immunotherapeutic drugs for cancer since 2010, and many more are still being evaluated in other clinical studies. These inhibitors significantly increase response rates and result in the treatment of patients with advanced cancer. However, cancer immunotherapy leads to essential cardiac toxicity properties that have become distinct from other cancer patients' care and are mostly related to their etiology. Aim of review As potential implications, the occurrence of cardiovascular adverse events is particularly challenging and needs a comprehensive understanding of overall cancer-related etiology, clinical outcomes with different variable severity, and management. Key scientific concepts of review In terms of improving the overall survival of patients with cancer, clinicians should be careful in selecting either programmed cell death-1 (PD-1) or its programmed cell death ligand (PDL-1) inhibitors by evaluating their risk and clinical benefit for early intervention and decrease the level of morbidity and mortality of their patients. This review focuses on the effectiveness of PD-1/PL-1 antibodies and associated cardiotoxicity adverse events, including etiological mechanisms, diagnosis, and treatment.
Collapse
Affiliation(s)
- Mohammed Safi
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Hyat Ahmed
- Department of Stomatology, Oral Pathology, Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Mahmoud Al-Azab
- Department of Immunology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yun-long Xia
- Head of Department of Cardiology, Vice president of the First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian 116021, Liaoning, China
| | - Xiu Shan
- First Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Mohammed Al-radhi
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Abdullah Al-danakh
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Abdullah Shopit
- Department of Pharmacology, Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Jiwei Liu
- Head of Department of Oncology First Affiliated Hospital of Dalian Medical University, Zhongshan Road Dalian, Dalian Liaoning Province 116044, China
- Corresponding author.
| |
Collapse
|
324
|
Rassaf T, Totzeck M, Backs J, Bokemeyer C, Hallek M, Hilfiker-Kleiner D, Hochhaus A, Lüftner D, Müller OJ, Neudorf U, Pfister R, von Haehling S, Lehmann LH, Bauersachs J. Onco-Cardiology: Consensus Paper of the German Cardiac Society, the German Society for Pediatric Cardiology and Congenital Heart Defects and the German Society for Hematology and Medical Oncology. Clin Res Cardiol 2020; 109:1197-1222. [PMID: 32405737 PMCID: PMC7515958 DOI: 10.1007/s00392-020-01636-7] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/11/2022]
Abstract
The acute and long-lasting side effects of modern multimodal tumour therapy significantly impair quality of life and survival of patients afflicted with malignancies. The key components of this therapy include radiotherapy, conventional chemotherapy, immunotherapy and targeted therapies. In addition to established tumour therapy strategies, up to 30 new therapies are approved each year with only incompletely characterised side effects. This consensus paper discusses the risk factors that contribute to the development of a potentially adverse reaction to tumour therapy and, in addition, defines specific side effect profiles for different treatment groups. The focus is on novel therapeutics and recommendations for the surveillance and treatment of specific patient groups.
Collapse
Affiliation(s)
- Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre Essen, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Centre Essen, University Hospital Essen, Hufelandstrasse 55, 45147, Essen, Germany
| | - Johannes Backs
- Institute for Experimental Cardiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Carsten Bokemeyer
- Department of Oncology, Hematology and Bone Marrow Transplantation with the Section Pneumology, Centre for Oncology, University Hospital Hamburg-Eppendorf, Hamburg, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Center for Integrated Oncology ABCD, University Hospital of Cologne, Cologne, Germany
| | | | - Andreas Hochhaus
- Department of Hematology and Medical Oncology, University Hospital Jena, Jena, Germany
| | - Diana Lüftner
- Department of Haematology, Oncology and Tumour Immunology, Charité, Humboldt University Berlin, Berlin, Germany
| | - Oliver J Müller
- Department of Internal Medicine III (Cardiology, Angiology and Internal Intensive Care Medicine), University Hospital Schleswig-Holstein, University of Kiel, Kiel, Germany
| | - Ulrich Neudorf
- Department of Pediatrics III, West German Heart and Vascular Centre Essen, University Hospital Essen, Essen, Germany
| | - Roman Pfister
- Clinic III for Internal Medicine, General and Interventional Cardiology, Electrophysiology, Angiology, Pneumology and Internal Intensive Care Medicine, University Hospital Cologne, Cologne, Germany
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart Center Göttingen, University of Göttingen Medical Center and German Center for Cardiovascular Research (DZHK), partner site Göttingen, Göttingen, Germany
| | - Lorenz H Lehmann
- Department of Cardiology, Angiology, Pneumology, University Hospital Heidelberg, Heidelberg, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
325
|
Chen DY, Huang WK, Chien-Chia Wu V, Chang WC, Chen JS, Chuang CK, Chu PH. Cardiovascular toxicity of immune checkpoint inhibitors in cancer patients: A review when cardiology meets immuno-oncology. J Formos Med Assoc 2020; 119:1461-1475. [DOI: 10.1016/j.jfma.2019.07.025] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/23/2019] [Accepted: 07/29/2019] [Indexed: 12/28/2022] Open
|
326
|
Neves KB, Montezano AC, Lang NN, Touyz RM. Vascular toxicity associated with anti-angiogenic drugs. Clin Sci (Lond) 2020; 134:2503-2520. [PMID: 32990313 DOI: 10.1042/cs20200308] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Over the past two decades, the treatment of cancer has been revolutionised by the highly successful introduction of novel molecular targeted therapies and immunotherapies, including small-molecule kinase inhibitors and monoclonal antibodies that target angiogenesis by inhibiting vascular endothelial growth factor (VEGF) signaling pathways. Despite their anti-angiogenic and anti-cancer benefits, the use of VEGF inhibitors (VEGFi) and other tyrosine kinase inhibitors (TKIs) has been hampered by potent vascular toxicities especially hypertension and thromboembolism. Molecular processes underlying VEGFi-induced vascular toxicities still remain unclear but inhibition of endothelial NO synthase (eNOS), reduced nitric oxide (NO) production, oxidative stress, activation of the endothelin system, and rarefaction have been implicated. However, the pathophysiological mechanisms still remain elusive and there is an urgent need to better understand exactly how anti-angiogenic drugs cause hypertension and other cardiovascular diseases (CVDs). This is especially important because VEGFi are increasingly being used in combination with other anti-cancer dugs, such as immunotherapies (immune checkpoint inhibitors (ICIs)), other TKIs, drugs that inhibit epigenetic processes (histone deacetylase (HDAC) inhibitor) and poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors, which may themselves induce cardiovascular injury. Here, we discuss vascular toxicities associated with TKIs, especially VEGFi, and provide an up-to-date overview on molecular mechanisms underlying VEGFi-induced vascular toxicity and cardiovascular sequelae. We also review the vascular effects of VEGFi when used in combination with other modern anti-cancer drugs.
Collapse
Affiliation(s)
- Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Ninian N Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| |
Collapse
|
327
|
Sławiński G, Wrona A, Dąbrowska-Kugacka A, Raczak G, Lewicka E. Immune Checkpoint Inhibitors and Cardiac Toxicity in Patients Treated for Non-Small Lung Cancer: A Review. Int J Mol Sci 2020; 21:E7195. [PMID: 33003425 PMCID: PMC7582741 DOI: 10.3390/ijms21197195] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/25/2020] [Accepted: 09/27/2020] [Indexed: 12/24/2022] Open
Abstract
Lung cancer is a major cause of cancer-related mortality worldwide, both in men and women. The vast majority of patients are diagnosed with non-small-cell lung cancer (NSCLC, 80-85% of lung cancer cases). Therapeutics named immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment in the last decade. They are monoclonal antibodies, and those directed against PD-1 (programmed cell death protein 1) or PD-L1 (programmed cell death-ligand 1) have been used in the treatment of lung cancer and significantly improved the prognosis of NSCLC patients. However, during treatment with ICIs, immune-related adverse events (irAEs) can occur in any organ and any tissue. At the same time, although cardiac irAEs are relatively rare compared to irAEs in other organs, they have a high mortality rate. The two most common clinical manifestations of immunotherapy-related cardiotoxicity are myocarditis and pericarditis. Various types of arrhythmias have been reported in patients treated with ICIs, including the occurrence of life-threatening complete atrioventricular block or ventricular tachyarrhythmias. Here, we aim to summarize the incidence, clinical manifestations, underlying mechanisms, diagnosis, and treatment strategies for ICI-associated cardiotoxicity as these issues become very important in view of the increasing use of ICI in the treatment of lung cancer.
Collapse
Affiliation(s)
- Grzegorz Sławiński
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| | - Anna Wrona
- Department of Oncology & Radiotherapy, Medical University of Gdańsk, 80-952 Gdańsk, Poland;
| | - Alicja Dąbrowska-Kugacka
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| | - Grzegorz Raczak
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| | - Ewa Lewicka
- Department of Cardiology & Electrotherapy, Medical University of Gdańsk, Debinki 7 Street, 80-952 Gdańsk, Poland; (G.S.); (A.D.-K.); (G.R.)
| |
Collapse
|
328
|
Awadalla M, Mahmood SS, Groarke JD, Hassan MZO, Nohria A, Rokicki A, Murphy SP, Mercaldo ND, Zhang L, Zlotoff DA, Reynolds KL, Alvi RM, Banerji D, Liu S, Heinzerling LM, Jones-O'Connor M, Bakar RB, Cohen JV, Kirchberger MC, Sullivan RJ, Gupta D, Mulligan CP, Shah SP, Ganatra S, Rizvi MA, Sahni G, Tocchetti CG, Lawrence DP, Mahmoudi M, Devereux RB, Forrestal BJ, Mandawat A, Lyon AR, Chen CL, Barac A, Hung J, Thavendiranathan P, Picard MH, Thuny F, Ederhy S, Fradley MG, Neilan TG. Global Longitudinal Strain and Cardiac Events in Patients With Immune Checkpoint Inhibitor-Related Myocarditis. J Am Coll Cardiol 2020; 75:467-478. [PMID: 32029128 DOI: 10.1016/j.jacc.2019.11.049] [Citation(s) in RCA: 189] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/03/2019] [Accepted: 11/22/2019] [Indexed: 01/04/2023]
Abstract
BACKGROUND There is a need for improved methods for detection and risk stratification of myocarditis associated with immune checkpoint inhibitors (ICIs). Global longitudinal strain (GLS) is a sensitive marker of cardiac toxicity among patients receiving standard chemotherapy. There are no data on the use of GLS in ICI myocarditis. OBJECTIVES This study sought to evaluate the role of GLS and assess its association with cardiac events among patients with ICI myocarditis. METHODS This study retrospectively compared echocardiographic GLS by speckle tracking at presentation with ICI myocarditis (cases, n = 101) to that from patients receiving an ICI who did not develop myocarditis (control subjects, n = 92). Where available, GLS was also measured pre-ICI in both groups. Major adverse cardiac events (MACE) were defined as a composite of cardiogenic shock, arrest, complete heart block, and cardiac death. RESULTS Cases and control subjects were similar in age, sex, and cancer type. At presentation with myocarditis, 61 cases (60%) had a normal ejection fraction (EF). Pre-ICI, GLS was similar between cases and control subjects (20.3 ± 2.6% vs. 20.6 ± 2.0%; p = 0.60). There was no change in GLS among control subjects on an ICI without myocarditis (pre-ICI vs. on ICI, 20.6 ± 2.0% vs. 20.5 ± 1.9%; p = 0.41); in contrast, among cases, GLS decreased to 14.1 ± 2.8% (p < 0.001). The GLS at presentation with myocarditis was lower among cases presenting with either a reduced (12.3 ± 2.7%) or preserved EF (15.3 ± 2.0%; p < 0.001). Over a median follow-up of 162 days, 51 (51%) experienced MACE. The risk of MACE was higher with a lower GLS among patients with either a reduced or preserved EF. After adjustment for EF, each percent reduction in GLS was associated with a 1.5-fold increase in MACE among patients with a reduced EF (hazard ratio: 1.5; 95% confidence interval: 1.2 to 1.8) and a 4.4-fold increase with a preserved EF (hazard ratio: 4.4; 95% confidence interval: 2.4 to 7.8). CONCLUSIONS GLS decreases with ICI myocarditis and, compared with control subjects, was lower among cases presenting with either a preserved or reduced EF. Lower GLS was strongly associated with MACE in ICI myocarditis presenting with either a preserved or reduced EF.
Collapse
Affiliation(s)
- Magid Awadalla
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts; Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Syed S Mahmood
- Cardiology Division, New York-Presbyterian Hospital, Weill Cornell Medical Center, New York, New York
| | - John D Groarke
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Malek Z O Hassan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Anju Nohria
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Adam Rokicki
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Sean P Murphy
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Nathaniel D Mercaldo
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Lili Zhang
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts; Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Daniel A Zlotoff
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Kerry L Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Raza M Alvi
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Dahlia Banerji
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Shiying Liu
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Lucie M Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nurnberg (FAU), Erlangen and Nurnberg, Germany
| | - Maeve Jones-O'Connor
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Rula B Bakar
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Justine V Cohen
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Michael C Kirchberger
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nurnberg (FAU), Erlangen and Nurnberg, Germany
| | - Ryan J Sullivan
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Dipti Gupta
- Cardiology Division, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | - Connor P Mulligan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Sachin P Shah
- Cardiology Division, Lahey Hospital & Medical Center, Burlington, Massachusetts
| | - Sarju Ganatra
- Cardiology Division, Lahey Hospital & Medical Center, Burlington, Massachusetts
| | - Muhammad A Rizvi
- Division of Oncology and Hematology, Department of Medicine, Lehigh Valley Hospital, Allentown, Pennsylvania
| | - Gagan Sahni
- Cardiovascular Institute, School of Medicine, The Mount Sinai Hospital, New York, New York
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center for Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Donald P Lawrence
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Michael Mahmoudi
- Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Richard B Devereux
- Cardiology Division, New York-Presbyterian Hospital, Weill Cornell Medical Center, New York, New York
| | - Brian J Forrestal
- Cardio-Oncology Program, Department of Cardiology, Medstar Washington Hospital Center, Medstar Heart and Vascular institute, Washington, DC
| | - Anant Mandawat
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - Alexander R Lyon
- Cardio-Oncology Program, Royal Brompton Hospital and Imperial College, London, United Kingdom
| | - Carol L Chen
- Cardiology Division, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, New York, New York
| | - Ana Barac
- Cardio-Oncology Program, Department of Cardiology, Medstar Washington Hospital Center, Medstar Heart and Vascular institute, Washington, DC
| | - Judy Hung
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Center, Division of Cardiology, Toronto General Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Michael H Picard
- Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts
| | - Franck Thuny
- Cardiology Division, Cardiovascular Division, Department of Medicine, Aix-Marseille Universite, Marseille, France
| | - Stephane Ederhy
- UNICO-GRECO, Cardio-Oncology Program, Department of Cardiology, Assistance Publique-Hopitaux de Paris, Saint-Antoine Hospital, Paris, France
| | - Michael G Fradley
- Cardio-Oncology Program, H. Lee Moffitt Cancer Center & Research Institute and University of South Florida Division of Cardiovascular Medicine, Tampa, Florida
| | - Tomas G Neilan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology, Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts; Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts.
| |
Collapse
|
329
|
Beasley GS, Towbin JA. Acquired and modifiable cardiovascular risk factors in patients treated for cancer. J Thromb Thrombolysis 2020; 51:846-853. [PMID: 32918669 DOI: 10.1007/s11239-020-02273-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/02/2020] [Indexed: 11/28/2022]
Abstract
Cardiac mortality is the leading cause of death secondary to malignancy in survivors of cancer. The field of cardio-oncology is dedicated to identifying and, if possible, modifying risk factors that contribute to significant cardiac morbidity and mortality. Many risk factors for the development of cancer-related cardiotoxicity overlap with risk factors in cardiovascular disease such as hypertension, obesity, dyslipidemia, and diabetes among others. These risk factors are usually modifiable while others such as genetics, type of malignancy, and need for chemotherapy are less modifiable. This article summarizes acquired and modifiable risk factors in both pediatric and adult patients treated for cancer.
Collapse
Affiliation(s)
- Gary S Beasley
- Department of Pediatrics, Division of Cardiology Le Bonheur Children's Hospital and The Heart Institute, University of Tennessee Health Science Center, College of Medicine, 49 N. Dunlap Street, 3rd Floor, Faculty Office Building, Memphis, TN, 38105, USA. .,Cardio-Hema-Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Jeffrey A Towbin
- Department of Pediatrics, Division of Cardiology Le Bonheur Children's Hospital and The Heart Institute, University of Tennessee Health Science Center, College of Medicine, 49 N. Dunlap Street, 3rd Floor, Faculty Office Building, Memphis, TN, 38105, USA.,Cardio-Hema-Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
330
|
Okamatsu Y, Tsubouchi K, Ibusuki R, Maehara-Keshino E, Shimauchi A, Kayukawa T, Eto D, Inoue K, Harada T. Rapid Onset of Takotsubo Cardiomyopathy Induced by an Infusion Reaction to Pembrolizumab in a Patient with NSCLC. JTO Clin Res Rep 2020; 1:100055. [PMID: 34589948 PMCID: PMC8474362 DOI: 10.1016/j.jtocrr.2020.100055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 05/02/2020] [Accepted: 05/07/2020] [Indexed: 11/20/2022] Open
Affiliation(s)
- Yuki Okamatsu
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| | - Kazuya Tsubouchi
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
- Corresponding author. Address for correspondence: Kazuya Tsubouchi, MD, PhD, Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, 1-8-1 Kishinoura, Yahata-nishi-ku, Kitakyushu City, Fukuoka 806-8501, Japan.
| | - Ritsu Ibusuki
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| | - Eri Maehara-Keshino
- Department of Cardiovascular Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| | - Atsushi Shimauchi
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| | - Takafumi Kayukawa
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| | - Daisuke Eto
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| | - Katsuhiro Inoue
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| | - Taishi Harada
- Department of Respiratory Medicine, Japan Community Health Care Organization Kyushu Hospital, Fukuoka, Japan
| |
Collapse
|
331
|
Chung WB, Youn JC, Youn HJ. Cardiovascular Complications of Novel Anti-Cancer Immunotherapy: Old Problems from New Agents? Korean Circ J 2020; 50:743-753. [PMID: 32725983 PMCID: PMC7440999 DOI: 10.4070/kcj.2020.0158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022] Open
Abstract
Many novel anti-cancer therapies have dramatically improved outcomes of various cancer patients. However, it also poses a risk for cardiovascular complications as well. For the novel anti-cancer agent with which physicians does not have enough clinical experiences to determine the characteristics of cardiovascular complications, it is important to assess risk factors for cardiotoxicity before starting anti-cancer therapy. High-risk patient should be consulted to cardiologist before initiating anti-cancer therapy and pre-emptive cardiac function monitoring plan might be prepared in advance. The biomarkers, electrocardiography and echocardiography are useful tools for the detection of subclinical cardiotoxicity during anti-cancer therapy. This review article tried to suggest the cardiac function monitoring strategies for newly encountered potential cardiotoxic anti-cancer agents and to summarize the cardiovascular complications of novel anti-cancer immunotherapies including immune checkpoint inhibitor (ICI) and chimeric antigen receptor (CAR) T-cell therapy. ICIs can cause fatal myocarditis, which usually occurs early after initiation, and prompt treatment with high-dose corticosteroid is necessary. CAR T-cell therapy can cause cytokine release syndrome, which may result in circulatory collapse. Supportive treatment as well as tocilizumab, an anti-interleukin-6 receptor antibody are cornerstones of treatment.
Collapse
Affiliation(s)
- Woo Baek Chung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Chan Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Joong Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
332
|
Čelutkienė J, Pudil R, López‐Fernández T, Grapsa J, Nihoyannopoulos P, Bergler‐Klein J, Cohen‐Solal A, Farmakis D, Tocchetti CG, Haehling S, Barberis V, Flachskampf FA, Čeponienė I, Haegler‐Laube E, Suter T, Lapinskas T, Prasad S, Boer RA, Wechalekar K, Anker MS, Iakobishvili Z, Bucciarelli‐Ducci C, Schulz‐Menger J, Cosyns B, Gaemperli O, Belenkov Y, Hulot J, Galderisi M, Lancellotti P, Bax J, Marwick TH, Chioncel O, Jaarsma T, Mullens W, Piepoli M, Thum T, Heymans S, Mueller C, Moura B, Ruschitzka F, Zamorano JL, Rosano G, Coats AJ, Asteggiano R, Seferovic P, Edvardsen T, Lyon AR. Role of cardiovascular imaging in cancer patients receiving cardiotoxic therapies: a position statement on behalf of the
H
eart
F
ailure
A
ssociation (
HFA
), the
E
uropean
A
ssociation of
C
ardiovascular
I
maging (
EACVI
) and the
Cardio‐Oncology C
ouncil of the
E
uropean
S
ociety of
C
ardiology (
ESC
). Eur J Heart Fail 2020; 22:1504-1524. [DOI: 10.1002/ejhf.1957] [Citation(s) in RCA: 125] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 06/08/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023] Open
Affiliation(s)
- Jelena Čelutkienė
- Clinic of Cardiac and Vascular Diseases, Institute of Clinical Medicine, Faculty of Medicine, Vilnius University Vilnius Lithuania
- State Research Institute Centre For Innovative Medicine Vilnius Lithuania
| | - Radek Pudil
- First Department of Medicine ‐ Cardioangiology Charles University Prague, Medical Faculty and University Hospital Hradec Králové Hradec Kralove Czech Republic
| | | | - Julia Grapsa
- Department of Cardiology St Bartholomew Hospital, Barts Health Trust London UK
| | - Petros Nihoyannopoulos
- Unit of Inherited Cardiovascular Diseases/Heart Center of the Young and Athletes, First Department of Cardiology Hippokrateion General Hospital, National and Kapodistrian University of Athens Athens Greece
- National Heart and Lung Institute Imperial College London, Hammersmith Hospital London UK
| | | | - Alain Cohen‐Solal
- UMR‐S 942, Cardiology Department Hôpital Lariboisière, AP‐HP, Université de Paris Paris France
| | - Dimitrios Farmakis
- University of Cyprus Medical School Nicosia Cyprus
- Cardio‐Oncology Clinic, Heart Failure Unit, Department of Cardiology Athens University Hospital Attikon, National and Kapodistrian University of Athens Athens Greece
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, and Interdepartmental Center for Clinical and Translational Research (CIRCET) Federico II University Naples Italy
| | - Stephan Haehling
- Department of Cardiology and Pneumology University of Göttingen Medical Centre Göttingen Germany
| | | | - Frank A. Flachskampf
- Department of Medical Sciences Uppsala University, Clinical Physiology and Cardiology, Akademiska Hospital Uppsala Sweden
| | - Indrė Čeponienė
- Department of Cardiology, Medical Academy Lithuanian University of Health Sciences Kaunas Lithuania
| | - Eva Haegler‐Laube
- Department of Cardiology, Inselspital University of Bern Bern Switzerland
| | - Thomas Suter
- Department of Cardiology, Inselspital University of Bern Bern Switzerland
| | - Tomas Lapinskas
- Department of Cardiology, Medical Academy Lithuanian University of Health Sciences Kaunas Lithuania
| | - Sanjay Prasad
- Department of Cardiac Magnetic Resonance Royal Brompton Hospital London UK
- National Heart and Lung Institute, Imperial College London UK
| | - Rudolf A. Boer
- Department of Cardiology University Medical Center Groningen, University of Groningen Groningen The Netherlands
| | | | - Markus S. Anker
- Division of Cardiology and Metabolism, Department of Cardiology, Charité; and Berlin Institute of Health Center for Regenerative Therapies (BCRT); and DZHK (German Centre for Cardiovascular Research), partner site Berlin; and Department of Cardiology, Charité Campus Benjamin Franklin Berlin Germany
| | - Zaza Iakobishvili
- Sackler Faculty of Medicine Tel Aviv University Tel Aviv Israel
- Tel Aviv‐Jaffa District, Clalit Health Services Tel Aviv Israel
| | - Chiara Bucciarelli‐Ducci
- Bristol Heart Institute, Bristol NIHR Biomedical Research Centre and Clinical Research and Imaging Centre (CRIC) Bristol University Hospitals Bristol NHS Trust and University of Bristol Bristol UK
| | - Jeanette Schulz‐Menger
- Working Group on Cardiovascular Magnetic Resonance, Experimental and Clinical Research Center a joint cooperation between the Charité ‐ Universitätsmedizin Berlin, Department of Internal Medicine and Cardiology and the Max‐Delbrueck Center for Molecular Medicine, and HELIOS Klinikum Berlin Buch, Department of Cardiology and Nephrology Berlin Germany
- DZHK (German Centre for Cardiovascular Research), partner site Berlin Berlin Germany
| | - Bernard Cosyns
- Department of Cardiology CHVZ (Centrum voor Hart en Vaatziekten), ICMI (In Vivo Cellular and Molecular Imaging) Laboratory, Universitair Ziekenhuis Brussel Brussels Belgium
| | | | - Yury Belenkov
- I.M. Sechenov's First Moscow State Medical University of Ministry of Health (Sechenov University) Moscow Russia
| | - Jean‐Sébastien Hulot
- Université de Paris, CIC1418, Paris Cardiovascular Research Center, INSERM Paris France
| | - Maurizio Galderisi
- Department of Advanced Biomedical Sciences Federico II University Hospital Naples Italy
| | - Patrizio Lancellotti
- University of Liège Hospital, GIGA Cardiovascular Sciences, Department of Cardiology, CHU SartTilman Liège Belgium
| | - Jeroen Bax
- Department of Cardiology Leiden University Medical Centre Leiden The Netherlands
| | | | - Ovidiu Chioncel
- Emergency Institute for Cardiovascular Diseases C.C. Iliescu Bucuresti Romania
- University of Medicine Carol Davila Bucuresti Romania
| | - Tiny Jaarsma
- Department of Health, Medicine and Caring Sciences Linköping University Linköping Sweden
- Julius Center for Health Sciences and Primary Care University Medical Center Utrecht and Utrecht University Utrecht The Netherlands
| | | | - Massimo Piepoli
- Heart Failure Unit, Cardiology Guglielmo da Saliceto Hospital Piacenza Italy
- University of Parma Parma Italy
| | - Thomas Thum
- Hannover Medical School, Institute of Molecular and Translational Therapeutic Strategies (IMTTS) Hannover Germany
| | - Stephane Heymans
- Department of Cardiology, CARIM School for Cardiovascular Diseases Faculty of Health, Medicine and Life Sciences Maastricht University Maastricht The Netherlands
- William Harvey Research Institute, Barts Heart Centre, Queen Mary University of London, Charterhouse Square London UK
- Department of Cardiovascular Sciences Centre for Molecular and Vascular Biology, KU Leuven Leuven Belgium
| | - Christian Mueller
- Department of Cardiology and Cardiovascular Research Institute Basel (CRIB) University Hospital Basel, University of Basel Basel Switzerland
| | - Brenda Moura
- Cardiology Department, Military Hospital, and CINTESIS, CardioCare, Faculty of Medicine Porto University Porto Portugal
| | - Frank Ruschitzka
- University Heart Center, Department of Cardiology University Hospital Zurich Zurich Switzerland
| | - Jose Luis Zamorano
- Cardiology Department University Hospital Ramón y Cajal Madrid Spain
- University Alcala Madrid Spain
- CIBERCV, Instituto de Salud Carlos III (ISCIII) Madrid Spain
| | - Giuseppe Rosano
- Centre for Clinical and Basic Research, Department of Medical Sciences IRCCS San Raffaele Pisana Rome Italy
| | | | | | - Petar Seferovic
- University of Belgrade Faculty of Medicine and Serbian Academy of Sciences and Arts Belgrade Serbia
| | - Thor Edvardsen
- Department of Cardiology Oslo University Hospital, Rikshospitalet Oslo Norway
- Faculty of Medicine University of Oslo Oslo Norway
| | - Alexander R. Lyon
- National Heart and Lung Institute, Imperial College London UK
- Cardio‐Oncology Service, Royal Brompton Hospital London UK
| |
Collapse
|
333
|
Samejima Y, Iuchi A, Kanai T, Noda Y, Nasu S, Tanaka A, Morishita N, Suzuki H, Okamoto N, Harada H, Ezumi A, Ueda K, Kawahara K, Hirashima T. Development of Severe Heart Failure in a Patient with Squamous Non-small-cell Lung Cancer During Nivolumab Treatment. Intern Med 2020; 59:2003-2008. [PMID: 32448839 PMCID: PMC7492128 DOI: 10.2169/internalmedicine.4550-20] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Cardiac side effects associated with immune checkpoint inhibitors (ICIs) are an uncommon but serious complication with a relatively high mortality. We experienced a case of cardiomyopathy induced by nivolumab. Echocardiography showed diffuse hypo-kinesis of the left ventricular cardiac wall and a significant decrease in the ejection fraction, like dilated cardiomyopathy. The myocardial biopsy showed non-inflammatory change; cardiac function gradually improved after treatment of acute heart failure without a corticosteroid. Although non-inflammatory left ventricular dysfunction induced by ICIs is rare, it is a reported cardiovascular toxicity. Physicians should consider this complication when treating patients with ICIs for malignant diseases.
Collapse
Affiliation(s)
- Yumiko Samejima
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Atsuhiko Iuchi
- Department of Cardiovascular Medicine, Osaka Habikino Medical Center, Japan
| | - Tomohiro Kanai
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Yoshimi Noda
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Shingo Nasu
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Ayako Tanaka
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Naoko Morishita
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Hidekazu Suzuki
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Norio Okamoto
- Department of Thoracic Oncology, Osaka Habikino Medical Center, Japan
| | - Hiroshi Harada
- Department of Cardiovascular Medicine, Osaka Habikino Medical Center, Japan
| | - Akira Ezumi
- Department of Cardiovascular Medicine, Osaka Habikino Medical Center, Japan
| | - Kayo Ueda
- Department of Clinical Pathology, Osaka Habikino Medical Center, Japan
| | | | | |
Collapse
|
334
|
Waliany S, Lee D, Witteles RM, Neal JW, Nguyen P, Davis MM, Salem JE, Wu SM, Moslehi JJ, Zhu H. Immune Checkpoint Inhibitor Cardiotoxicity: Understanding Basic Mechanisms and Clinical Characteristics and Finding a Cure. Annu Rev Pharmacol Toxicol 2020; 61:113-134. [PMID: 32776859 DOI: 10.1146/annurev-pharmtox-010919-023451] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Immune checkpoint inhibitors (ICIs) attenuate mechanisms of self-tolerance in the immune system, enabling T cell responses to cancerous tissues and revolutionizing care for cancer patients. However, by loweringbarriers against self-reactivity, ICIs often result in varying degrees of autoimmunity. Cardiovascular complications, particularly myocarditis but also arrhythmias, pericarditis, and vasculitis, have emerged as significant complications associated with ICIs. In this review, we examine the clinical aspects and basic science principles that underlie ICI-associated myocarditis and other cardiovascular toxicities. In addition, we discuss current therapeutic approaches. We believe a better mechanistic understanding of ICI-associated toxicities can lead to improved patient outcomes by reducing treatment-related morbidity.
Collapse
Affiliation(s)
- Sarah Waliany
- Department of Medicine, Stanford University, Stanford, California 94305, USA;
| | - Daniel Lee
- Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA
| | - Ronald M Witteles
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joel W Neal
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Division of Oncology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Patricia Nguyen
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Mark M Davis
- Department of Microbiology and Immunology and Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, California 94305, USA.,Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Joe-Elie Salem
- Sorbonne Université, INSERM, CIC-1901 Paris-Est, CLIP² Galilée, UNICO-GRECO Cardio-Oncology Program, and Department of Pharmacology, Pitié-Salpêtrière Hospital, Assistance Publique-Hôpitaux de Paris, F-75013 Paris, France.,Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA; .,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Sean M Wu
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Javid J Moslehi
- Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA; .,Division of Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, Tennessee 37203, USA
| | - Han Zhu
- Department of Medicine, Stanford University, Stanford, California 94305, USA; .,Stanford Cardiovascular Institute, Stanford University, Stanford, California 94305, USA.,Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, California 94305, USA
| |
Collapse
|
335
|
Gürdoğan M, Yalta K. Myocarditis associated with immune checkpoint inhibitors: Practical considerations in diagnosis and management. Anatol J Cardiol 2020; 24:68-75. [PMID: 32749254 PMCID: PMC7460679 DOI: 10.14744/anatoljcardiol.2020.79584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) have caused radical changes in the treatment scheme of many types of cancer in the past 10 years. ICIs are specific monoclonal antibodies that increase T-cell mediated immune response against cancer cells. Despite important advances in cancer treatment, uncontrolled activation of cytotoxic T cells has brought along many autoimmune clinical side effects, especially acute myocarditis. Although the incidence of ICI-related myocarditis is about 1%, it is remarkable in terms of mortality rate reaching 46% and demonstrating the necessity of rapid diagnosis and multidisciplinary approach. The present review aimed to summarize the heterogeneous symptomatology of ICI-associated myocarditis, clinical presentation ranging from elevated asymptomatic cardiac enzyme levels to cardiogenic shock, prominent diagnostic value of cardiac magnetic resonance imaging, and current information on the effectiveness of immunosuppressants in therapy.
Collapse
Affiliation(s)
- Muhammet Gürdoğan
- Department of Cardiology, Faculty of Medicine, Trakya University; Edirne-Turkey
| | - Kenan Yalta
- Department of Cardiology, Faculty of Medicine, Trakya University; Edirne-Turkey
| |
Collapse
|
336
|
Abstract
Remarkable progress has been made in the development of new therapies for cancer, dramatically changing the landscape of treatment approaches for several malignancies and continuing to increase patient survival. Accordingly, adverse effects of cancer therapies that interfere with the continuation of best-possible care, induce life-threatening risks or lead to long-term morbidity are gaining increasing importance. Cardiovascular toxic effects of cancer therapeutics and radiation therapy are the epitome of such concerns, and proper knowledge, interpretation and management are needed and have to be placed within the context of the overall care of individual patients with cancer. Furthermore, the cardiotoxicity spectrum has broadened to include myocarditis with immune checkpoint inhibitors and cardiac dysfunction in the setting of cytokine release syndrome with chimeric antigen receptor T cell therapy. An increase in the incidence of arrhythmias related to inflammation such as atrial fibrillation can also be expected, in addition to the broadening set of cancer therapeutics that can induce prolongation of the corrected QT interval. Therefore, cardiologists of today have to be familiar not only with the cardiotoxicity associated with traditional cancer therapies, such as anthracycline, trastuzumab or radiation therapy, but even more so with an ever-increasing repertoire of therapeutics. This Review provides this information, summarizing the latest developments at the juncture of cardiology, oncology and haematology.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA.
| |
Collapse
|
337
|
Tocchetti CG, Ameri P, de Boer RA, D’Alessandra Y, Russo M, Sorriento D, Ciccarelli M, Kiss B, Bertrand L, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Linke WA, Mayr M, van der Velden J, Zacchigna S, Ghigo A, Hirsch E, Lyon AR, Görbe A, Ferdinandy P, Madonna R, Heymans S, Thum T. Cardiac dysfunction in cancer patients: beyond direct cardiomyocyte damage of anticancer drugs: novel cardio-oncology insights from the joint 2019 meeting of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc Res 2020; 116:1820-1834. [DOI: 10.1093/cvr/cvaa222] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/17/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
In western countries, cardiovascular (CV) disease and cancer are the leading causes of death in the ageing population. Recent epidemiological data suggest that cancer is more frequent in patients with prevalent or incident CV disease, in particular, heart failure (HF). Indeed, there is a tight link in terms of shared risk factors and mechanisms between HF and cancer. HF induced by anticancer therapies has been extensively studied, primarily focusing on the toxic effects that anti-tumour treatments exert on cardiomyocytes. In this Cardio-Oncology update, members of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart discuss novel evidence interconnecting cardiac dysfunction and cancer via pathways in which cardiomyocytes may be involved but are not central. In particular, the multiple roles of cardiac stromal cells (endothelial cells and fibroblasts) and inflammatory cells are highlighted. Also, the gut microbiota is depicted as a new player at the crossroads between HF and cancer. Finally, the role of non-coding RNAs in Cardio-Oncology is also addressed. All these insights are expected to fuel additional research efforts in the field of Cardio-Oncology.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, AB31, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Yuri D’Alessandra
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michele Russo
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine Surgery and Odontology, University of Salerno, Salerno, Italy
| | - Bernadett Kiss
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
| | - Luc Bertrand
- IREC Institute, Pole of Cardiovascular Research, Université Catholique de Louvain, Brussels, Belgium
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Ines Falcao-Pires
- Unidade de Investigação e Desenvolvimento Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Portugal
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr Universität Bochum, Bochum, Germany
- Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Witten, Germany
| | | | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Imperial College London, London, UK
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy
- Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| |
Collapse
|
338
|
Abstract
The world of cardio-oncology is an evolving field involving the assessment of cardiovascular disease in patients suffering from cancer. Cancer and cardiovascular diseases are the two leading causes of morbidity and mortality in the developed world. Globally, cancer is diagnosed in 12.7 million patients annually, and cancer incidence is projected to increase by 40% in high-income countries from 2008 to 2030. Chemotherapy is the main treatment for most cancers and improves survival, but is associated with significant cardiotoxicity. In recent years, the introduction of new biological anti-cancer treatments in addition to "classic" chemotherapy has further improved survival, but has also introduced new cardiovascular side effects beyond "pure" myocardial damage. The increasing number of patients with cancer and cancer survivors, and the growing complexity of cancer treatment and cardiovascular side effects, call for teamwork including cardiologists with specific training and expertise working in teams with oncologists, hematologists, and others. The purpose of this review was to describe the clinical background and importance of cardio-oncology, with an emphasis on the use of imaging in this clinical setting.
Collapse
|
339
|
Abstract
The era of modern oncology incorporates an ever-evolving personalized approach to hematological malignancies and solid tumors. As a result, patient survival rates have, in part, substantially improved, depending on the specific type of underlying malignancy. However, systemic therapies may come along with potential cardiotoxic effects resulting in heart failure with increased morbidity and mortality. Ultimately, patients may survive their malignancy but die as a result of cancer treatment. Cardiovascular magnetic resonance imaging has long been in use for the assessment of function and tissue characteristics in patients with various nonischemic cardiac diseases. Besides an introductory overview on the general definition of cardiotoxicity including potential underlying mechanisms, this review provides insight into the application of various cardiovascular magnetic resonance imaging techniques in the setting of cancer therapy-related cardiac and vascular toxicity. Early identification of cardiotoxic effects may allow for on-time therapy adjustment and/or cardioprotective measures to avoid subsequent long-term heart failure with increased mortality.
Collapse
|
340
|
Targeted anti-inflammatory therapy is a new insight for reducing cardiovascular events: A review from physiology to the clinic. Life Sci 2020; 253:117720. [PMID: 32360620 DOI: 10.1016/j.lfs.2020.117720] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 04/13/2020] [Accepted: 04/14/2020] [Indexed: 12/30/2022]
Abstract
Despite considerable progressions, cardiovascular disease (CVD) is still one of the major causes of mortality around the world, indicates an important and unmet clinical need. Recently, extensive studies have been performed on the role of inflammatory factors as either a major or surrogate factor in the pathophysiology of CVD. Epidemiological observations suggest the theory of the role of inflammatory mediators in the development of cardiovascular events. This may support the idea that targeted anti-inflammatory therapies, on the background of traditional validated medical therapies, can play a significant role in prevention and even reduction of cardiovascular disorders. Many randomized controlled trials have shown that drugs commonly useful for primary and secondary prevention of CVD have an anti-inflammatory mechanism. Further, many anti-inflammatory drugs are being examined because of their potential to reduce the risk of cardiovascular problems. In this study, we review the process of inflammation in the development of cardiovascular events, both in vivo and clinical evidence in immunotherapy for CVD.
Collapse
|
341
|
Cutroneo PM, Isgrò V, Ientile V, Santarpia M, Ferlazzo G, Fontana A, Carrega P, Matarangolo E, Barnaba S, Spina E, Trifirò G. Safety profile of immune checkpoint inhibitors: An analysis of the Italian spontaneous reporting system database. Br J Clin Pharmacol 2020; 87:527-541. [PMID: 32495965 DOI: 10.1111/bcp.14413] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Revised: 05/08/2020] [Accepted: 05/19/2020] [Indexed: 12/13/2022] Open
Abstract
AIMS To provide an overview of immune checkpoint inhibitors (ICIs) safety profile using the Italian spontaneous adverse drug reaction (ADR) reporting system. METHODS We selected all ADR reports attributed to ipilimumab (CTLA-4 inhibitor), nivolumab, pembrolizumab, atezolizumab (PD-1/PD-L1 inhibitors) from the Italian spontaneous reporting system (2011-2018). Descriptive analyses of reports for ICIs have been conducted. Time to onset of adverse effects was stratified by system organ class. Reporting odds ratio was used as measure of ADR reporting disproportionality. ICI-related ADR reports were compared with 2 reference groups, i.e. all other suspected drugs or all other antineoplastic agents. RESULTS Overall, 2217 (0.7%) reports were related to ICIs (nivolumab: 72.2% of those reports; ipilimumab: 14.3%; pembrolizumab: 10.3%; and atezolizumab: 3.5%). ICI-related ADR reports mostly involved males (65%) and median age was 67 (interquartile range 59-73) years. Serious reports accounted for 48.8%. Frequencies of endocrine, general, hepatobiliary, metabolism, musculoskeletal, respiratory disorders, infections and neoplasms were significantly higher for ICIs than for all other drugs (P < .001). Except for infections, similar results emerged through comparison with other anticancer drugs. Colitis, hypophysitis and skin disorders were more frequently reported for anti-CTLA-4 drugs than PD-1/PD-L1 ICIs, and the opposite for musculoskeletal effects, pneumonia, and thyroid dysfunctions. ICIs were disproportionally associated also with less known risks, e.g. ischaemic heart disease, cardiac failure and optic nerve disorders. CONCLUSION The most frequently reported safety issues were probably immune-related adverse events including general, gastrointestinal and respiratory disorders. Potentially emerging safety signals, such as ischaemic heart disease and cardiac failure, requiring further investigation were detected.
Collapse
Affiliation(s)
- Paola Maria Cutroneo
- Sicilian Regional Pharmacovigilance Center, University Hospital of Messina, Messina, Italy
| | - Valentina Isgrò
- Dept. of Biomedical and Dental Sciences and Morpho-functional Imaging, University of Messina, Messina, Italy
| | - Valentina Ientile
- Dept. of Biomedical and Dental Sciences and Morpho-functional Imaging, University of Messina, Messina, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, A.O.U. Policlinico "G. Martino", Dept. of Human Pathology, University of Messina, Messina, Italy
| | - Guido Ferlazzo
- Research Center Cell Factory UniMe, Dept. of Human Pathology, University of Messina and Cell Therapy Program, A.O.U. Policlinico "G. Martino", Messina, Italy
| | - Andrea Fontana
- Unit of Biostatistics, Fondazione IRCCS "Casa Sollievo della Sofferenza", San Giovanni Rotondo, Italy
| | - Paolo Carrega
- Research Center Cell Factory UniMe, Dept. of Human Pathology, University of Messina and Cell Therapy Program, A.O.U. Policlinico "G. Martino", Messina, Italy
| | | | - Simona Barnaba
- Italian Medicines Agency, Pharmacovigilance Office, Rome, Italy
| | - Edoardo Spina
- Sicilian Regional Pharmacovigilance Center, University Hospital of Messina, Messina, Italy.,Dept. of Clinical and Experimental medicine, University of Messina, Messina, Italy
| | - Gianluca Trifirò
- Dept. of Biomedical and Dental Sciences and Morpho-functional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
342
|
Abstract
In the field of cardio-oncology, it is well recognised that despite the benefits of chemotherapy in treating and possibly curing cancer, it can cause catastrophic damage to bystander tissues resulting in a range of potentially of life-threatening cardiovascular toxicities, and leading to a number of damaging side effects including heart failure and myocardial infarction. Cardiotoxicity is responsible for significant morbidity and mortality in the long-term in oncology patients, specifically due to left ventricular dysfunction. There is increasing emphasis on the early use of biomarkers in order to detect the cardiotoxicity at a stage before it becomes irreversible. The most important markers of cardiac injury are cardiac troponin and natriuretic peptides, whilst markers of inflammation such as interleukin-6, C-reactive protein, myeloperoxidase, Galectin-3, growth differentiation factor-15 are under investigation for their use in detecting cardiotoxicity early. In addition, microRNAs, genome-wide association studies and proteomics are being studied as novel markers of cardiovascular injury or inflammation. The aim of this literature review is to discuss the evidence base behind the use of these biomarkers for the detection of cardiotoxicity.
Collapse
|
343
|
De Luca G, Campochiaro C, Sartorelli S, Peretto G, Dagna L. Therapeutic strategies for virus-negative myocarditis: a comprehensive review. Eur J Intern Med 2020; 77:9-17. [PMID: 32402564 DOI: 10.1016/j.ejim.2020.04.050] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Revised: 04/16/2020] [Accepted: 04/20/2020] [Indexed: 12/19/2022]
Abstract
Virus-negative or autoimmune myocarditis(VNM) is an inflammatory disease affecting the myocardium that may occur as a distinct disease with exclusive cardiac involvement, or in the context of systemic autoimmune or inflammatory disorders. The pathogenesis of VNM involves both innate and acquired immunity and is not completely elucidated: an early immune-mediated pathogenic process lead to subacute and chronic stages and eventually results in tissue remodeling, fibrosis, contractile dysfunction, dilated cardiomyopathy and arrhythmic burden, accounting for a dismal prognosis. Treatment interventions effectively curbing the acute inflammatory process at an early stage can prevent late cardiac remodeling and improve patient's outcome. The mainstay of treatment of VNM remains symptomatic therapy of heart failure and arrhythmia, while the use of immunosuppressive treatments has long been considered controversial until recently, and strategies effectively targeting the inflammatory and immune-mediated substrate of the disease remain elusive. Only steroids and azathioprine have been tested in clinical trials, and nowadays represent the therapy of choice. A substantial proportion of patients are resistant to first line strategies, suggesting that some critical inflammatory mechanisms are not responsive to conventional immunosuppression with steroids and azathioprine, or experience drug-related adverse events. Thus, second-line targeted therapeutic strategies to treat VNM are eagerly awaited. Recent data on the pathogenic mechanisms underlying myocardial inflammation are paving the way to novel, promising treatment strategies for myocarditis, which could reformulate future treatment strategies for VNM. In this review, we summarize the current therapeutic opportunities, beyond corticosteroids, to treat VNM, including conventional and biologic immunosuppressive drugs and cytokine blocking agents.
Collapse
Affiliation(s)
- Giacomo De Luca
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy.
| | - Corrado Campochiaro
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| | - Silvia Sartorelli
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy
| | - Giovanni Peretto
- Department of Cardiac Electrophysiology and Arrhythmology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Unit of Immunology, Rheumatology, Allergy and Rare Diseases (UnIRAR), IRCCS San Raffaele Scientific Institute, Via Olgettina, 60-20132, Milan, Italy; Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
344
|
ECG Changes in Melanoma Patients Undergoing Cancer Therapy-Data From the ECoR Registry. J Clin Med 2020; 9:jcm9072060. [PMID: 32630003 PMCID: PMC7408861 DOI: 10.3390/jcm9072060] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 06/18/2020] [Accepted: 06/25/2020] [Indexed: 12/16/2022] Open
Abstract
We aimed to evaluate whether therapy with immune checkpoint inhibitors (ICI) leads to changes in electrocardiogram (ECG) parameters in melanoma patients. We retrospectively examined 41 patients (46% women, age 61 ± 12years) with advanced melanoma (stage III/IV) before and during ICI treatment from our “Essen Cardio-oncology Registry” (ECoR). ECGs were analyzed before and 4–12 weeks after therapy started (follow-up, 90 ± 51 days). Heart rate, PR time, QRS duration and duration of the corrected QT (QTc) interval were recorded. QT dispersion (QTd) was calculated. Heart rate, PR time, QRS and QTc did not differ when comparing values before and after therapy started. QTd was prolonged after therapy started (32 ± 16 ms vs. 47 ± 19 ms, n = 41, p < 0.0001). Subgroup analyses revealed prolonged QTd in patients that received a combination immunotherapy with ipilimumab and nivolumab (31 ± 14 ms vs. 50 ± 14 ms, n = 21, p < 0.0001), while QTd in patients with anti–programmed death 1 (PD-1) inhibitor monotherapy did not change after therapy started. QTd is prolonged in patients under ICI combination therapy, potentially signaling an increased susceptibility to ventricular arrhythmias.
Collapse
|
345
|
Harinstein ME. Avaliação da Gravidade da Doença Arterial Coronariana em Pacientes Tratados com Quimioterapia: A Necessidade Adicional da Cardio-Oncologia. Arq Bras Cardiol 2020; 114:1013-1014. [PMID: 32638898 PMCID: PMC8416125 DOI: 10.36660/abc.20200408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
|
346
|
Kooshkaki O, Derakhshani A, Hosseinkhani N, Torabi M, Safaei S, Brunetti O, Racanelli V, Silvestris N, Baradaran B. Combination of Ipilimumab and Nivolumab in Cancers: From Clinical Practice to Ongoing Clinical Trials. Int J Mol Sci 2020; 21:ijms21124427. [PMID: 32580338 PMCID: PMC7352976 DOI: 10.3390/ijms21124427] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 06/15/2020] [Accepted: 06/18/2020] [Indexed: 12/13/2022] Open
Abstract
Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1) are inhibitory checkpoints that are commonly seen on activated T cells and have been offered as promising targets for the treatment of cancers. Immune checkpoint inhibitors (ICIs)targeting PD-1, including pembrolizumab and nivolumab, and those targeting its ligand PD-L1, including avelumab, atezolizumab, and durvalumab, and two drugs targeting CTLA-4, including ipilimumab and tremelimumab have been approved for the treatment of several cancers and many others are under investigating in advanced trial phases. ICIs increased antitumor T cells’ responses and showed a key role in reducing the acquired immune system tolerance which is overexpressed by cancer and tumor microenvironment. However, 50% of patients could not benefit from ICIs monotherapy. To overcome this, a combination of ipilimumab and nivolumab is frequently investigated as an approach to improve oncological outcomes. Despite promising results for the combination of ipilimumab and nivolumab, safety concerns slowed down the development of such strategies. Herein, we review data concerning the clinical activity and the adverse events of ipilimumab and nivolumab combination therapy, assessing ongoing clinical trials to identify clinical outlines that may support combination therapy as an effective treatment. To the best of our knowledge, this paper is one of the first studies to evaluate the efficacy and safety of ipilimumab and nivolumab combination therapy in several cancers.
Collapse
Affiliation(s)
- Omid Kooshkaki
- Student research committee, Birjand University of Medical Sciences, Birjand 9717853577, Iran;
- Department of Immunology, Birjand University of Medical Sciences, Birjand 9717853577, Iran
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (S.S.)
| | - Negar Hosseinkhani
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
| | - Mitra Torabi
- Student research committee, Tabriz University of medical sciences, Tabriz 5165665811, Iran;
| | - Sahar Safaei
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (S.S.)
| | - Oronzo Brunetti
- Medical Oncology Unit, IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
| | - Nicola Silvestris
- Medical Oncology Unit, IRCCS IstitutoTumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology, University of Bari “Aldo Moro”, 70124 Bari, Italy;
- Correspondence: (N.S.); (B.B.)
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 5165665811, Iran; (A.D.); (S.S.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 5166614766, Iran;
- Correspondence: (N.S.); (B.B.)
| |
Collapse
|
347
|
Michel L, Totzeck M, Lehmann L, Finke D. Emerging role of immune checkpoint inhibitors and their relevance for the cardiovascular system. Herz 2020; 45:645-651. [DOI: 10.1007/s00059-020-04954-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
348
|
Abstract
Patients with cancer are at a higher risk of cardiovascular disease, which contributes to significant morbidity and mortality. The rapid progress in the field of oncological treatments has led to a steady increase in long-term cancer survivors. Care for cardiovascular complications is therefore becoming increasingly important. In addition, the establishment of new oncological therapies has resulted in the identification of previously unknown cardiovascular side effects. Oncocardiology aims to detect and treat cardiovascular diseases associated with cancer and cancer therapy. Continuous scientific, clinical, and structural developments are necessary as the basis for the best care of the growing number of affected patients. This review summarizes current developments in the field of oncocardiology with regard to advances in cancer therapy and challenges in clinical oncocardiology work. Cardiovascular side effects by targeted cancer therapies are characterized and recent advances in the field of cardiovascular diagnostics are outlined. Developments to better integrate oncocardiology into the medical care system and perspectives for modern, patient-oriented care are shown. In light of the coronavirus disease 2019 (COVID-19) pandemic, current challenges and opportunities are highlighted. The relevance of profitable further advances in oncocardiology including standardized guidelines and educational programs is delineated as a mandatory requirement for the successful development of oncocardiology.
Collapse
|
349
|
Khunger A, Battel L, Wadhawan A, More A, Kapoor A, Agrawal N. New Insights into Mechanisms of Immune Checkpoint Inhibitor-Induced Cardiovascular Toxicity. Curr Oncol Rep 2020; 22:65. [DOI: 10.1007/s11912-020-00925-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
350
|
de La Rochefoucauld J, Noël N, Lambotte O. Management of immune-related adverse events associated with immune checkpoint inhibitors in cancer patients: a patient-centred approach. Intern Emerg Med 2020; 15:587-598. [PMID: 32144552 DOI: 10.1007/s11739-020-02295-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2019] [Accepted: 02/13/2020] [Indexed: 02/06/2023]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment. The number of indications is increasing and antibodies targeting the CTLA-4 and PD-1/PD-L1 pathways are now also prescribed in adjuvant settings and for metastatic cancer. However, ICIs reactivate autoreactive immune cells as well as tumour-specific T cells, which lead to immune-related adverse events (irAEs) in around 70% of treated patients. Although all organs can potentially be involved, the skin, gut, thyroid, lungs, liver, and joints are most frequently affected. Most irAEs occur in the first few months of treatment but late-onset toxicity-even after the ICI has been discontinued-is also possible. In terms of severity, most irAEs are grade 1-2. Some irAEs (especially myocarditis, pneumonitis, and encephalitis) are potentially fatal; in patients with highly suggestive clinical signs, treatment should be initiated before the diagnostic work-up has been completed. When confronted with an unexpected clinical sign, the physician must differentiate rapidly between an irAE, cancer progression, and another (unrelated) cause. The management of irAEs is based on the temporary or permanent discontinuation of the ICI and (for grade ≥ 2 events) the administration of steroids.
Collapse
Affiliation(s)
- Jeanne de La Rochefoucauld
- Department of Internal Medicine and Clinical Immunology, APHP-Université-Paris-Saclay, Hôpital Universitaire Bicêtre, CHU Bicêtre, APHP, 78 rue du Général Leclerc, 94275, Le Kremlin Bicêtre, France
| | - Nicolas Noël
- Department of Internal Medicine and Clinical Immunology, APHP-Université-Paris-Saclay, Hôpital Universitaire Bicêtre, CHU Bicêtre, APHP, 78 rue du Général Leclerc, 94275, Le Kremlin Bicêtre, France
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, 94276, Le Kremlin Bicêtre, France
- Université Paris Saclay, UMR 1184, 94276, Le Kremlin Bicêtre, France
- CEA, DSV/iMETI, IDMIT, 92265, Fontenay-aux-Roses, France
| | - Olivier Lambotte
- Department of Internal Medicine and Clinical Immunology, APHP-Université-Paris-Saclay, Hôpital Universitaire Bicêtre, CHU Bicêtre, APHP, 78 rue du Général Leclerc, 94275, Le Kremlin Bicêtre, France.
- INSERM U1184, Immunology of Viral Infections and Autoimmune Diseases, 94276, Le Kremlin Bicêtre, France.
- Université Paris Saclay, UMR 1184, 94276, Le Kremlin Bicêtre, France.
- CEA, DSV/iMETI, IDMIT, 92265, Fontenay-aux-Roses, France.
| |
Collapse
|