351
|
Hu JR, Florido R, Lipson EJ, Naidoo J, Ardehali R, Tocchetti CG, Lyon AR, Padera RF, Johnson DB, Moslehi J. Cardiovascular toxicities associated with immune checkpoint inhibitors. Cardiovasc Res 2020; 115:854-868. [PMID: 30715219 DOI: 10.1093/cvr/cvz026] [Citation(s) in RCA: 337] [Impact Index Per Article: 67.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 01/11/2019] [Accepted: 01/24/2019] [Indexed: 12/24/2022] Open
Abstract
Cardiovascular toxicities associated with immune checkpoint inhibitors (ICIs) have been reported in case series but have been underappreciated due to their recent emergence, difficulties in diagnosis and non-specific clinical manifestations. ICIs are antibodies that block negative regulators of the T cell immune response, including cytotoxic T lymphocyte-associated protein-4 (CTLA-4), programmed cell death protein-1 (PD-1), and PD-1 ligand (PD-L1). While ICIs have introduced a significant mortality benefit in several cancer types, the augmented immune response has led to a range of immune-related toxicities, including cardiovascular toxicity. ICI-associated myocarditis often presents with arrhythmias, may co-exist with myositis and myasthenia gravis, can be severe, and portends a poor prognosis. In addition, pericardial disease, vasculitis, including temporal arteritis, and non-inflammatory heart failure, have been recently described as immune-related toxicities from ICI. This narrative review describes the epidemiology, diagnosis, pathophysiology, and treatment of cardiovascular toxicities of ICI therapy, highlighting recent developments in the field in the past year.
Collapse
Affiliation(s)
- Jiun-Ruey Hu
- Division of Cardiology, Cardio-Oncology Program, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, USA
| | - Roberta Florido
- Division of Cardiology, Johns Hopkins University School of Medicine, 600 N Wolfe St, Baltimore, MD, USA
| | - Evan J Lipson
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, USA.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, USA
| | - Jarushka Naidoo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, USA.,Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, 1550 Orleans Street, Baltimore, MD, USA
| | - Reza Ardehali
- Division of Cardiology, David Geffen School of Medicine, University of California Los Angeles, 675 Charles E Young Dr S, Los Angeles, CA, USA
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Interdepartmental Center for Clinical and Translational Research (CIRCET), Federico II University, Via Pansini 5, Naples, NA, Italy
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Dovehouse St, London, UK.,National Heart and Lung Institute, Imperial College London, Sydney Street, London, UK
| | - Robert F Padera
- Department of Pathology, Brigham and Women's Hospital, 75 Francis St, Boston, MA, USA
| | - Douglas B Johnson
- Division of Hematology-Oncology, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, TN, USA
| | - Javid Moslehi
- Division of Cardiology, Cardio-Oncology Program, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, USA
| |
Collapse
|
352
|
Hosoda H. [The mechanism of anticancer drug-induced DNA damage response and repair in cardiomyocytes]. Nihon Yakurigaku Zasshi 2020; 155:175-178. [PMID: 32378639 DOI: 10.1254/fpj.19128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Genomic DNA, which contains all of the genetic information, is damaged by a variety of endogenous and environmental factors such as genotoxic chemicals, ionizing radiation and UV light. Consequently, the DNA repair process is constantly active as it responds to damage in the DNA structure. Not only cardiotoxicity of anticancer drug treatment but also ischemic heart disease and heart failure associated with overloaded pressure interfere with DNA damage response and DNA repair regulation in cardiomyocytes. DNA methylation, catalyzed by the DNMTs, plays an important role in maintaining genome stability, but the molecular mechanism is not clear. In this study, we examine and outline the links between DNA methylation and the DNA damage repair systems and discuss the possible mechanisms of how they are orchestrated, with a focus on cardiotoxicity of anticancer drugs.
Collapse
Affiliation(s)
- Hiroshi Hosoda
- National Cerebral and Cardiovascular Center Research Institute
| |
Collapse
|
353
|
Zhang L, Awadalla M, Mahmood SS, Nohria A, Hassan MZO, Thuny F, Zlotoff DA, Murphy SP, Stone JR, Golden DLA, Alvi RM, Rokicki A, Jones-O’Connor M, Cohen JV, Heinzerling LM, Mulligan C, Armanious M, Barac A, Forrestal BJ, Sullivan RJ, Kwong RY, Yang EH, Damrongwatanasuk R, Chen CL, Gupta D, Kirchberger MC, Moslehi JJ, Coelho-Filho OR, Ganatra S, Rizvi MA, Sahni G, Tocchetti CG, Mercurio V, Mahmoudi M, Lawrence DP, Reynolds KL, Weinsaft JW, Baksi AJ, Ederhy S, Groarke JD, Lyon AR, Fradley MG, Thavendiranathan P, Neilan TG. Cardiovascular magnetic resonance in immune checkpoint inhibitor-associated myocarditis. Eur Heart J 2020; 41:1733-1743. [PMID: 32112560 PMCID: PMC7205467 DOI: 10.1093/eurheartj/ehaa051] [Citation(s) in RCA: 244] [Impact Index Per Article: 48.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 10/30/2019] [Accepted: 01/21/2020] [Indexed: 12/27/2022] Open
Abstract
AIMS Myocarditis is a potentially fatal complication of immune checkpoint inhibitors (ICI). Sparse data exist on the use of cardiovascular magnetic resonance (CMR) in ICI-associated myocarditis. In this study, the CMR characteristics and the association between CMR features and cardiovascular events among patients with ICI-associated myocarditis are presented. METHODS AND RESULTS From an international registry of patients with ICI-associated myocarditis, clinical, CMR, and histopathological findings were collected. Major adverse cardiovascular events (MACE) were a composite of cardiovascular death, cardiogenic shock, cardiac arrest, and complete heart block. In 103 patients diagnosed with ICI-associated myocarditis who had a CMR, the mean left ventricular ejection fraction (LVEF) was 50%, and 61% of patients had an LVEF ≥50%. Late gadolinium enhancement (LGE) was present in 48% overall, 55% of the reduced EF, and 43% of the preserved EF cohort. Elevated T2-weighted short tau inversion recovery (STIR) was present in 28% overall, 30% of the reduced EF, and 26% of the preserved EF cohort. The presence of LGE increased from 21.6%, when CMR was performed within 4 days of admission to 72.0% when CMR was performed on Day 4 of admission or later. Fifty-six patients had cardiac pathology. Late gadolinium enhancement was present in 35% of patients with pathological fibrosis and elevated T2-weighted STIR signal was present in 26% with a lymphocytic infiltration. Forty-one patients (40%) had MACE over a follow-up time of 5 months. The presence of LGE, LGE pattern, or elevated T2-weighted STIR were not associated with MACE. CONCLUSION These data suggest caution in reliance on LGE or a qualitative T2-STIR-only approach for the exclusion of ICI-associated myocarditis.
Collapse
Affiliation(s)
- Lili Zhang
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Magid Awadalla
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Syed S Mahmood
- Cardiology Division, Department of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical Center, 1300 York Avenue, New York, NY 10065, USA
| | - Anju Nohria
- Cardio-Oncology Program, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, 75 Francis St, Boston, MA 02115, USA
| | - Malek Z O Hassan
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Franck Thuny
- Department of Cardiology, Aix-Marseille University, Assistance Publique–Hôpitaux de Marseille, Mediterranean university, Cardio-Oncology center (MEDI-CO center), Unit of Heart Failure and Valvular Heart Diseases, Hôpital Nord, Jardin du Pharo, 58 Boulevard Charles Livon 13007, Marseille, France
- Groupe Méditerranéen de Cardio-Oncologie (gMEDICO), AP-HM, Chemin des Bourrely, 13015, Marseille, France
- Aix-Marseille University, Center for CardioVascular and Nutrition research (C2VN), Inserm 1263, Inra 1260, 13385 Marseille, France
| | - Daniel A Zlotoff
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Sean P Murphy
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - James R Stone
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
| | - Doll Lauren Alexandra Golden
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Raza M Alvi
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Adam Rokicki
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Maeve Jones-O’Connor
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Justine V Cohen
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA
| | - Lucie M Heinzerling
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Schloßplatz 4, 91054 Erlangen, Germany
| | - Connor Mulligan
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| | - Merna Armanious
- Cardio-Oncology Program, Division of Cardiovascular Medicine, H. Lee Moffitt Cancer Center & Research Institute and University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Ana Barac
- Cardio-Oncology program, MedStar Heart and Vascular Institute, Georgetown University, 110 Irving St NW, Washington, DC 20010, USA
| | - Brian J Forrestal
- Cardio-Oncology program, MedStar Heart and Vascular Institute, Georgetown University, 110 Irving St NW, Washington, DC 20010, USA
| | - Ryan J Sullivan
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA
| | - Raymond Y Kwong
- Noninvasive Cardiovascular Imaging Section, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, 75 Francis St, Boston, MA 02115, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, 757 Westwood Plaza, Los Angeles, CA 90095, USA
| | - Rongras Damrongwatanasuk
- Cardio-Oncology Program, Division of Cardiovascular Medicine, H. Lee Moffitt Cancer Center & Research Institute and University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Carol L Chen
- Cardiology Division, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Avenue, New York, NY 10065, USA
| | - Dipti Gupta
- Cardiology Division, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Avenue, New York, NY 10065, USA
| | - Michael C Kirchberger
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Schloßplatz 4, 91054 Erlangen, Germany
| | - Javid J Moslehi
- Cardio-Oncology Program, Vanderbilt University Medical Center, 1211 Medical Center Dr, Nashville, TN 37232, USA
| | - Otavio R Coelho-Filho
- Cardiology Division, State University of Campinas, Cidade Universitária Zeferino Vaz - Barão Geraldo, Campinas, São Paulo 13083-970, Brazil
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Lahey Hospital and Medical Center, 41 Burlington Mall Road, Burlington, MA 01805, USA
| | - Muhammad A Rizvi
- Division of Oncology and Hematology, Department of Medicine, Lehigh Valley Hospital, 1200 S Cedar Crest Blvd, Allentown, PA 18103, USA
| | - Gagan Sahni
- Cardiology Division, The Mount Sinai Hospital, 1468 Madison Ave, New York, NY 10029, USA
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, via S. Pansini 5, 80131 Naples, NA, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, via S. Pansini 5, 80131 Naples, NA, Italy
| | - Michael Mahmoudi
- Faculty of Medicine, University of Southampton, University Road Southampton SO17 1BJ, UK
| | - Donald P Lawrence
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA
| | - Kerry L Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, 55 Fruit St, Boston, MA 02114, USA
| | - Jonathan W Weinsaft
- Cardiology Division, Department of Medicine, New York-Presbyterian Hospital, Weill Cornell Medical Center, 1300 York Avenue, New York, NY 10065, USA
- Cardiology Division, Memorial Sloan Kettering Cancer Center, Weill Cornell Medical College, 1275 York Avenue, New York, NY 10065, USA
| | - A John Baksi
- Cardiovascular Research Centre and Cardiovascular Magnetic Resonance Unit, Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
- National Heart and Lung Institute, Imperial College London, Kensington, London SW7 2DD, UK
| | - Stephane Ederhy
- UNICO-GRECO cardio-oncology program, sorbonne universite, Hopital Saint Antoine, 27 Rue de Chaligny, 75012 Paris, France
| | - John D Groarke
- Cardio-Oncology Program, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, 75 Francis St, Boston, MA 02115, USA
| | - Alexander R Lyon
- Cardio-Oncology Program, Royal Brompton Hospital, Sydney St, Chelsea, London SW3 6NP, UK
- National Heart and Lung Institute, Imperial College London, Cale Street, Chelsea, London, SW3 6LY, United Kingdom
| | - Michael G Fradley
- Cardio-Oncology Program, Division of Cardiovascular Medicine, H. Lee Moffitt Cancer Center & Research Institute and University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| | - Paaladinesh Thavendiranathan
- Ted Rogers Program in Cardiotoxicity Prevention, Division of Cardiology, Toronto General Hospital, Peter Munk Cardiac Center, University of Toronto, Toronto, ON, Canada
| | - Tomas G Neilan
- Cardiovascular Imaging Research Center (CIRC), Division of Cardiology, Department of Radiology, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, 165 Cambridge Street, Suite 400, Boston, MA 02114, USA
| |
Collapse
|
354
|
Avila MS, Siqueira SRR, Ferreira SMA, Bocchi EA. Prevention and Treatment of Chemotherapy-Induced Cardiotoxicity. Methodist Debakey Cardiovasc J 2020; 15:267-273. [PMID: 31988687 DOI: 10.14797/mdcj-15-4-267] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent decades have seen an increase in survival rates for cancer patients, partially explained by earlier diagnoses and new chemotherapeutic agents. However, chemotherapy may be associated with adverse cardiovascular events, including hypertension and pulmonary hypertension, supraventricular and ventricular arrhythmias, cardiomyopathy, and other forms of cardiovascular disease. For patients, the benefits of chemotherapy may be partially obfuscated by deleterious effects on the cardiovascular system, resulting in a significant increase in morbidity and mortality. In this article, we review strategies for prevention and treatment of chemotherapy-related cardiotoxicity.
Collapse
Affiliation(s)
- Monica Samuel Avila
- HOSPITAL DAS CLINICAS DA FACULDADE DE MEDICINA DA UNIVERSIDADE DE SAO PAULO, SAO PAULO, BRAZIL
| | | | | | - Edimar Alcides Bocchi
- HOSPITAL DAS CLINICAS DA FACULDADE DE MEDICINA DA UNIVERSIDADE DE SAO PAULO, SAO PAULO, BRAZIL
| |
Collapse
|
355
|
Jagielska B, Ozdowska P, Gepner K, Kubala S, Siedlecki JA, Sarnowski TJ, Sarnowska E. Cardiotoxicity danger in immunotherapy. IUBMB Life 2020; 72:1160-1167. [PMID: 32359132 DOI: 10.1002/iub.2299] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Revised: 04/15/2020] [Accepted: 04/16/2020] [Indexed: 12/12/2022]
Abstract
Immunotherapy based on immune checkpoint inhibitors (ICIs) is currently broadly used in the treatment of different types of cancer. The treatment targeting programmed cell death protein 1/programmed death-ligand 1 axis is already approved by Food and Drug Administration for numerous cancers. These kinds of therapy brought spectacular results in the treatment of non-small cell lung cancer where systemic therapy was ineffective. However, a wide range of applied therapies based on ICIs in the clinic have led to unexpected side effects, such as severe cardiotoxicity. It needs to be underlined that the molecular mechanism of myocarditis in response to ICIs is still not fully understood. Lack of sufficient knowledge, especially concerning the kind of risk factors increasing probability of myocarditis, poses currently a large clinical problem. Continuous cardiac monitoring of patients who undergo ICI treatment presents another problem as it is cost-ineffective for the healthcare system. Herein, we highlight the risks of use of anticancer therapy based on ICIs. We also stress that detailed monitoring of any event of cardiotoxicity following ICIs treatment should be carefully investigated and registered to give a global overview of the frequency of myocarditis occurrence. Moreover, we propose that the extension of molecular and systemic knowledge of etiology of myocarditis as a side effect, including the role of protein kinases, will be highly beneficial for the medical field. Last but not least, better understanding of mechanisms of cardiotoxicity induction will improve the safety of cancer patients and will help clinicians in prediction of unexpected side effect occurrence.
Collapse
Affiliation(s)
- Beata Jagielska
- Department of Oncology and Internal Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Patrycja Ozdowska
- Department of Oncology and Internal Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Katarzyna Gepner
- Department of Oncology and Internal Medicine, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Szymon Kubala
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Janusz A Siedlecki
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| | - Tomasz J Sarnowski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, Poland
| | - Elzbieta Sarnowska
- Department of Molecular and Translational Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Warsaw, Poland
| |
Collapse
|
356
|
Zaborowska-Szmit M, Krzakowski M, Kowalski DM, Szmit S. Cardiovascular Complications of Systemic Therapy in Non-Small-Cell Lung Cancer. J Clin Med 2020; 9:E1268. [PMID: 32349387 PMCID: PMC7287714 DOI: 10.3390/jcm9051268] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular diseases may determine therapy outcomes of non-small-cell lung cancer (NSCLC). The evidence for how iatrogenic cardiovascular complications contribute to ceasing anticancer treatment, decreasing the quality of life or even premature death, is unclear. Older patients and smokers are at risk of atherosclerosis and arterial thromboembolic events (TE), such as myocardial infarction or stroke. Venous TE can be observed in up to 15% of NSCLC patients, but the risk increases three to five times in ALK (anaplastic lymphoma kinase)-rearranged NSCLC. ALK inhibitors are associated with electrophysiological disorders. Cytotoxic agents and anti-VEGF inhibitors mainly cause vascular complications, including venous or arterial TE. Cardiac dysfunction and arrhythmias seem to be less frequent. Chemotherapy is often administered in two-drug regimens. Clinical events can be triggered by different mechanisms. Among epidermal growth factor inhibitors, erlotinib and gefitinib can lead to coronary artery events; however, afatinib and osimertinib can be associated with the development of heart failure. During anti-PD1/anti-PDL1 therapy, myocarditis is possible, which must be differentiated from acute coronary syndrome and heart failure. Awareness of all possible cardiovascular complications in NSCLC encourages vigilance in early diagnostics and treatment.
Collapse
Affiliation(s)
- Magdalena Zaborowska-Szmit
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.Z.-S.); (M.K.); (D.M.K.)
| | - Maciej Krzakowski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.Z.-S.); (M.K.); (D.M.K.)
| | - Dariusz M. Kowalski
- Department of Lung Cancer and Thoracic Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland; (M.Z.-S.); (M.K.); (D.M.K.)
| | - Sebastian Szmit
- Department of Pulmonary Circulation, Thromboembolic Diseases and Cardiology, Centre of Postgraduate Medical Education, European Health Centre, 05-400 Otwock, Poland
| |
Collapse
|
357
|
Re-introducing immunotherapy in patients surviving immune checkpoint inhibitors-mediated myocarditis. Clin Res Cardiol 2020; 110:50-60. [DOI: 10.1007/s00392-020-01648-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 04/07/2020] [Indexed: 12/12/2022]
|
358
|
Imaging of Adverse Events Related to Checkpoint Inhibitor Therapy. Diagnostics (Basel) 2020; 10:diagnostics10040216. [PMID: 32294888 PMCID: PMC7235714 DOI: 10.3390/diagnostics10040216] [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: 03/18/2020] [Revised: 04/10/2020] [Accepted: 04/11/2020] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy with checkpoint inhibitors (ICIs) is becoming standard of practice for an increasing number of cancer types. ICIs enhance T-cell action against the cancer cells. By unbalancing the immune system ICIs may cause dysimmune toxicities, a series of disorders broadly defined immune-related adverse events (irAEs). IrAEs may affect any organ or apparatus and most frequently involve skin, colon, endocrine organs, liver, and lungs. Early identification and appropriate treatment of irAEs can improve patient outcome. The paper aims at reviewing mechanisms of the occurrence of irAEs, the importance of a proper diagnosis and the main pillars of therapy. To provide effective guidance to the comprehension of major irAEs imaging findings will be reviewed.
Collapse
|
359
|
Tu L, Liu J, Li Z, Liu Y, Luo F. Early detection and management of immune-related myocarditis: Experience from a case with advanced squamous cell lung carcinoma. Eur J Cancer 2020; 131:5-8. [PMID: 32248072 DOI: 10.1016/j.ejca.2020.02.046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/13/2020] [Indexed: 02/08/2023]
Affiliation(s)
- Li Tu
- Department of Medical Oncology, Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Jiewei Liu
- Department of Medical Oncology, Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Zhixi Li
- Department of Medical Oncology, Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Yanyang Liu
- Department of Medical Oncology, Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China
| | - Feng Luo
- Department of Medical Oncology, Lung Cancer Center, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, People's Republic of China.
| |
Collapse
|
360
|
Totzeck M, Rassaf T. Neue onkologische Therapien und ihre kardiovaskulären Risiken. Herz 2020; 45:129-133. [DOI: 10.1007/s00059-020-04902-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
361
|
Alhumaid W, Paterson DI. Drug-Induced Acute Coronary Syndrome: A New Cardiovascular Concern With Immune Checkpoint Inhibitors and the Need for a Prospective Registry. Can J Cardiol 2020; 36:455-456. [DOI: 10.1016/j.cjca.2020.02.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 02/03/2020] [Accepted: 02/03/2020] [Indexed: 12/25/2022] Open
|
362
|
Wang A, Xu Y, Fei Y, Wang M. The role of immunosuppressive agents in the management of severe and refractory immune-related adverse events. Asia Pac J Clin Oncol 2020; 16:201-210. [PMID: 32212243 DOI: 10.1111/ajco.13332] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2019] [Accepted: 02/27/2020] [Indexed: 12/15/2022]
Abstract
The advent of immune checkpoint inhibitors has improved survival in some types of cancer and brought promising prospects to cancer immunotherapy. Despite their clinical benefits, significant off-target toxicities resulting from the immune system activation have been observed, namely immune-related adverse events (irAEs), which pose to clinicians a new challenge of optimal management. With steroids being the mainstay of current management of irAEs, immunosuppressive agents are especially indicated for severe or steroid-refractory cases, based on current immunopathophysiological knowledge and on extrapolations of treatment options for primary autoimmune disorders. This review focuses on the status and recent clinical progress of immunosuppressive agents in the management of severe and steroid-refractory irAEs.
Collapse
Affiliation(s)
- Anqi Wang
- Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan Xu
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yunyun Fei
- Department of Rheumatology and Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengzhao Wang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
363
|
State-of-the-art Review: Interventional Onco-Cardiology. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2020. [DOI: 10.1007/s11936-020-00809-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
364
|
Moriyama S, Fukata M, Kusaba H, Maruyama T, Akashi K. Acute and Chronic Effects of Cancer Drugs on the Cardiovascular System. Heart Fail Clin 2020; 16:231-241. [PMID: 32143767 DOI: 10.1016/j.hfc.2019.11.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Several cancer treatments cause cardiotoxicity that can lead to heart failure, coronary artery disease, arrhythmia, and pericardial disease. In this review, representative cases of heart failure following cardiotoxicity caused by trastuzumab, anthracycline, and hematopoietic stem cell transplantation are described with case notes. Additionally, other important points regarding cardiotoxicity related to heart failure are reported. During and after potentially cardiotoxic therapy, periodic cardiac examinations are recommended to detect any cardiovascular disorders; these are ameliorated if appropriately diagnosed at an earlier stage. It is important for cardiologists and oncologists to understand the pathophysiology of representative cardiovascular disease cases following cancer treatment.
Collapse
Affiliation(s)
- Shohei Moriyama
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan.
| | - Mitsuhiro Fukata
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Hitoshi Kusaba
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Toru Maruyama
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| | - Koichi Akashi
- Department of Hematology, Oncology and Cardiovascular Medicine, Kyushu University Hospital, Maidashi 3-1-1, Higashi-ku, Fukuoka 812-8582, Japan
| |
Collapse
|
365
|
Liu Y, Wu W. Cardiovascular immune-related adverse events: Evaluation, diagnosis and management. Asia Pac J Clin Oncol 2020; 16:232-240. [PMID: 32129935 PMCID: PMC7496884 DOI: 10.1111/ajco.13326] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 01/28/2020] [Indexed: 12/15/2022]
Abstract
Cardiotoxicities are associated with immune checkpoint inhibitor (ICI) therapy. Recent case series and retrospective studies have shown that cardiac immune‐related adverse events (irAEs) are rare but potentially fatal complications of immunotherapy, with various underlying risk factors such as combinations of different ICIs. High mortality rates and overreactive inflammation have been observed with ICI‐associated cardiotoxicities, highlighting the necessity of baseline and serial evaluations and the identification and management of cardiac irAEs as early as possible. The clinical presentations of irAEs range from asymptomatic cardiac biomarker elevation, myocarditis and pericardial diseases to heart failure and mild to fatal arrhythmia. Troponin measurement and electrocardiogram are sensitive initial tests, whereas cardiac magnetic resonance imaging and endomyocardial biopsy are both gold standard components of the diagnostic criteria. Close monitoring and timely consultation with a cardiologist are important for the diagnosis of ICI‐related cardiotoxicities, with decisions of stopping or rechallenging ICIs and strategies to manage heart injuries. Treatment principles are made according to risk stratifications. The first‐line medication is glucocorticoids of various doses, and the second‐line immunosuppression includes intravenous immunoglobin, antithymocyte globulin and other immunosuppressants, which are recommended in life‐threatening cases or in cases of resistance/no response to steroids.
Collapse
Affiliation(s)
- Yingxian Liu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| | - Wei Wu
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Science, Beijing, China
| |
Collapse
|
366
|
Hayek SS, Ganatra S, Lenneman C, Scherrer-Crosbie M, Leja M, Lenihan DJ, Yang E, Ryan TD, Liu J, Carver J, Mousavi N, O'Quinn R, Arnold A, Banchs J, Barac A, Ky B. Preparing the Cardiovascular Workforce to Care for Oncology Patients: JACC Review Topic of the Week. J Am Coll Cardiol 2020; 73:2226-2235. [PMID: 31047011 DOI: 10.1016/j.jacc.2019.02.041] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 02/04/2019] [Indexed: 10/26/2022]
Abstract
Cardiovascular disease and cancer are the 2 main causes of death in the United States. They intersect on multiple levels, sharing common causal mechanisms and epidemiological risk factors. The growing prevalence and complexity of cardiovascular disease and cancer have resulted in the development of the discipline of cardio-oncology. Preparing the cardiovascular workforce for the care of a growing population of cancer patients is necessary to enhance the delivery of high-quality cardiovascular care for patients with cancer. The goal of this review is to present the dedicated efforts of the cardio-oncology community to meet the growing need for education and training of cardiovascular practitioners providing care to cancer patients and survivors. Integration in general cardiology training programs and the efforts of the stakeholder organizations serve as an example of how a multidimensional, innovative approach can address provider education and training needs in a relatively new discipline.
Collapse
Affiliation(s)
- Salim S Hayek
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Sarju Ganatra
- Lahey Hospital and Medical Center, Burlington, Massachusetts
| | - Carrie Lenneman
- Division of Cardiovascular Disease, University of Alabama at Birmingham, Birmingham, Alabama
| | | | - Monika Leja
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Daniel J Lenihan
- Division of Cardiology, Cardio-Oncology Center of Excellence, Washington University in St. Louis, St. Louis, Missouri
| | - Eric Yang
- Division of Cardiology, University of California at Los Angeles, Los Angeles, California
| | - Thomas D Ryan
- Department of Pediatrics, University College of Medicine and Heart Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Jennifer Liu
- Memorial Sloan Kettering Cancer Center and Weill Cornell Medical Center, New York, New York
| | - Joseph Carver
- Division of Cardiology, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Negareh Mousavi
- Division of Cardiology, McGill University, Montreal, Quebec, Canada
| | - Rupal O'Quinn
- Division of Cardiology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Anita Arnold
- Division of Cardiology, Florida State University, Tallahassee, Florida
| | - Jose Banchs
- Division of Cardiology, MD Anderson Cancer Center, Houston, Texas
| | - Ana Barac
- MedStar Heart and Vascular Institute, Washington, DC.
| | - Bonnie Ky
- Department of Epidemiology, Biostatistics and Informatics, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
367
|
Li CH, Viladés D, López-Fernández T, Barreiro-Pérez M, Pizzi MN, Vidal-Pérez R, Martínez-Monzonís MA, Jiménez-Borreguero LJ. Selección de lo mejor del año 2019 en imagen cardiovascular. REC: CARDIOCLINICS 2020; 55:10-17. [DOI: 10.1016/j.rccl.2019.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/17/2025]
|
368
|
Passos LSA, Lupieri A, Becker-Greene D, Aikawa E. Innate and adaptive immunity in cardiovascular calcification. Atherosclerosis 2020; 306:59-67. [PMID: 32222287 DOI: 10.1016/j.atherosclerosis.2020.02.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 01/10/2020] [Accepted: 02/20/2020] [Indexed: 12/19/2022]
Abstract
Despite the focus placed on cardiovascular research, the prevalence of vascular and valvular calcification is increasing and remains a leading contributor of cardiovascular morbidity and mortality. Accumulating studies provide evidence that cardiovascular calcification is an inflammatory disease in which innate immune signaling becomes sustained and/or excessive, shaping a deleterious adaptive response. The triggering immune factors and subsequent inflammatory events surrounding cardiovascular calcification remain poorly understood, despite sustained significant research interest and support in the field. Most studies on cardiovascular calcification focus on innate cells, particularly macrophages' ability to release pro-osteogenic cytokines and calcification-prone extracellular vesicles and apoptotic bodies. Even though substantial evidence demonstrates that macrophages are key components in triggering cardiovascular calcification, the crosstalk between innate and adaptive immune cell components has not been adequately addressed. The only therapeutic options currently used are invasive procedures by surgery or transcatheter intervention. However, no approved drug has shown prophylactic or therapeutic effectiveness. Conventional diagnostic imaging is currently the best method for detecting, measuring, and assisting in the treatment of calcification. However, these common imaging modalities are unable to detect early subclinical stages of disease at the level of microcalcifications; therefore, the vast majority of patients are diagnosed when macrocalcifications are already established. In this review, we unravel the current knowledge of how innate and adaptive immunity regulate cardiovascular calcification; and put forward differences and similarities between vascular and valvular disease. Additionally, we highlight potential immunomodulatory drugs with the potential to target calcification and propose avenues in need of further translational inquiry.
Collapse
Affiliation(s)
- Livia S A Passos
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Adrien Lupieri
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Dakota Becker-Greene
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Elena Aikawa
- Center for Excellence in Vascular Biology, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Center for Interdisciplinary Cardiovascular Sciences, Division of Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA; Department of Pathology, Sechenov First Moscow State Medical University, Moscow, 119992, Russia.
| |
Collapse
|
369
|
Lee MS, Liu DW, Hung SK, Yu CC, Chi CL, Chiou WY, Chen LC, Lin RI, Huang LW, Chew CH, Hsu FC, Chan MWY, Lin HY. Emerging Challenges of Radiation-Associated Cardiovascular Dysfunction (RACVD) in Modern Radiation Oncology: Clinical Practice, Bench Investigation, and Multidisciplinary Care. Front Cardiovasc Med 2020; 7:16. [PMID: 32154267 PMCID: PMC7047711 DOI: 10.3389/fcvm.2020.00016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2019] [Accepted: 01/31/2020] [Indexed: 02/06/2023] Open
Abstract
Radiotherapy (RT) is a crucial treatment modality in managing cancer patients. However, irradiation dose sprinkling to tumor-adjacent normal tissues is unavoidable, generating treatment toxicities, such as radiation-associated cardiovascular dysfunction (RACVD), particularly for those patients with combined therapies or pre-existing adverse features/comorbidities. Radiation oncologists implement several efforts to decrease heart dose for reducing the risk of RACVD. Even applying the deep-inspiration breath-hold (DIBH) technique, the risk of RACVD is though reduced but still substantial. Besides, available clinical methods are limited for early detecting and managing RACVD. The present study reviewed emerging challenges of RACVD in modern radiation oncology, in terms of clinical practice, bench investigation, and multidisciplinary care. Several molecules are potential for serving as biomarkers and therapeutic targets. Of these, miRNAs, endogenous small non-coding RNAs that function in regulating gene expression, are of particular interest because low-dose irradiation, i.e., 200 mGy (one-tenth of conventional RT daily dose) induces early changes of pro-RACVD miRNA expression. Moreover, several miRNAs, e.g., miR-15b and miR21, involve in the development of RACVD, further demonstrating the potential bio-application in RACVD. Remarkably, many RACVDs are late RT sequelae, characterizing highly irreversible and progressively worse. Thus, multidisciplinary care from oncologists and cardiologists is crucial. Combined managements with commodities control (such as hypertension, hypercholesterolemia, and diabetes), smoking cessation, and close monitoring are recommended. Some agents show abilities for preventing and managing RACVD, such as statins and angiotensin-converting enzyme inhibitors (ACEIs); however, their real roles should be confirmed by further prospective trials.
Collapse
Affiliation(s)
- Moon-Sing Lee
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Dai-Wei Liu
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
| | - Shih-Kai Hung
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan
| | - Chih-Chia Yu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, Taiwan
| | - Chen-Lin Chi
- School of Medicine, Tzu Chi University, Hualien, Taiwan.,Department of Anatomic Pathology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan
| | - Wen-Yen Chiou
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan
| | - Liang-Cheng Chen
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan
| | - Ru-Inn Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, Taiwan
| | - Li-Wen Huang
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan
| | - Chia-Hui Chew
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan
| | - Feng-Chun Hsu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan
| | - Michael W Y Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, Taiwan
| | - Hon-Yi Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,School of Medicine, Tzu Chi University, Hualien, Taiwan.,Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Dalin, Taiwan.,Department of Biomedical Sciences, National Chung Cheng University, Chia-Yi, Taiwan
| |
Collapse
|
370
|
Bishopric NH, Lippman ME. Adverse Cardiovascular Events in Cancer Trials: Missing in Action? J Am Coll Cardiol 2020; 75:629-631. [PMID: 32057378 DOI: 10.1016/j.jacc.2019.12.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 12/22/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Nanette H Bishopric
- MedStar Heart and Vascular Institute, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC.
| | - Marc E Lippman
- Georgetown University Medical Center, Georgetown Lombardi Comprehensive Cancer Center, Washington, DC
| |
Collapse
|
371
|
Choi J, Lee SY. Clinical Characteristics and Treatment of Immune-Related Adverse Events of Immune Checkpoint Inhibitors. Immune Netw 2020; 20:e9. [PMID: 32158597 PMCID: PMC7049586 DOI: 10.4110/in.2020.20.e9] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2019] [Revised: 02/05/2020] [Accepted: 02/06/2020] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have been changing the paradigm of cancer treatment. However, immune-related adverse effects (irAEs) have also increased with the exponential increase in the use of ICIs. ICIs can break up the immunologic homeostasis and reduce T-cell tolerance. Therefore, inhibition of immune checkpoint can lead to the activation of autoreactive T-cells, resulting in various irAEs similar to autoimmune diseases. Gastrointestinal toxicity, endocrine toxicity, and dermatologic toxicity are common side effects. Neurotoxicity, cardiotoxicity, and pulmonary toxicity are relatively rare but can be fatal. ICI-related gastrointestinal toxicity, dermatologic toxicity, and hypophysitis are more common with anti- CTLA-4 agents. ICI-related pulmonary toxicity, thyroid dysfunction, and myasthenia gravis are more common with PD-1/PD-L1 inhibitors. Treatment with systemic steroids is the principal strategy against irAEs. The use of immune-modulatory agents should be considered in case of no response to the steroid therapy. Treatment under the supervision of multidisciplinary specialists is also essential, because the symptoms and treatments of irAEs could involve many organs. Thus, this review focuses on the mechanism, clinical presentation, incidence, and treatment of various irAEs.
Collapse
Affiliation(s)
- Juwhan Choi
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Sung Yong Lee
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| |
Collapse
|
372
|
Donisan T, Balanescu DV, Palaskas N, Lopez-Mattei J, Karimzad K, Kim P, Charitakis K, Cilingiroglu M, Marmagkiolis K, Iliescu C. Cardiac Interventional Procedures in Cardio-Oncology Patients. Cardiol Clin 2020; 37:469-486. [PMID: 31587788 DOI: 10.1016/j.ccl.2019.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Comorbidities specific to the cardio-oncology population contribute to the challenges in the interventional management of patients with cancer and cardiovascular disease (CVD). Patients with cancer have generally been excluded from cardiovascular randomized clinical trials. Endovascular procedures may represent a valid option in patients with cancer with a range of CVDs because of their minimally invasive nature. Patients with cancer are less likely to be treated according to societal guidelines because of perceived high risk. This article presents the specific challenges that interventional cardiologists face when caring for patients with cancer and the modern tools to optimize care.
Collapse
Affiliation(s)
- Teodora Donisan
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77030, USA. https://twitter.com/TDonisan
| | - Dinu Valentin Balanescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77030, USA. https://twitter.com/dinubalanescu
| | - Nicolas Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77030, USA
| | - Juan Lopez-Mattei
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77030, USA
| | - Kaveh Karimzad
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77030, USA
| | - Peter Kim
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77030, USA
| | - Konstantinos Charitakis
- Department of Cardiology, McGovern Medical School at The University of Texas Health Science Center at Houston, 6431 Fannin Street, Houston, TX 77030, USA
| | - Mehmet Cilingiroglu
- Department of Cardiology, Arkansas Heart Hospital, 1701 South Shackleford Road, Little Rock, AR 72211, USA
| | | | - Cezar Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77030, USA.
| |
Collapse
|
373
|
Pembrolizumab-Induced Mobitz Type 2 Second-Degree Atrioventricular Block. Case Rep Cardiol 2020; 2020:8428210. [PMID: 32047674 PMCID: PMC7007942 DOI: 10.1155/2020/8428210] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Accepted: 12/14/2019] [Indexed: 01/22/2023] Open
Abstract
Pembrolizumab is a monoclonal antibody directed towards programmed cell death protein 1 (PD-1) and is an antineoplastic drug which has a growing variety of oncologic uses. Pembrolizumab is commonly associated with immune-related adverse events (IRAEs) but is infrequently noted to cause cardiotoxicities such as myocarditis, arrhythmias, and heart failure. The following case report illustrates the clinical course of a 67-year-old female patient with stage IV non-small-cell lung cancer who developed Mobitz type 2 second-degree atrioventricular block three weeks after receiving her first infusion of pembrolizumab. Within a few hours of presentation, she progressed to symptomatic complete heart block requiring emergent placement of a temporary transvenous pacemaker. The article further discusses proposed mechanisms to explain IRAEs and management of IRAEs. We conclude by recommending a higher degree of caution and awareness among all physicians when treating patients on immunotherapy and a multidisciplinary approach when considering resumption of immune checkpoint inhibitor therapy.
Collapse
|
374
|
Oncolytic Virus Encoding a Master Pro-Inflammatory Cytokine Interleukin 12 in Cancer Immunotherapy. Cells 2020; 9:cells9020400. [PMID: 32050597 PMCID: PMC7072539 DOI: 10.3390/cells9020400] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2019] [Revised: 01/25/2020] [Accepted: 01/29/2020] [Indexed: 02/07/2023] Open
Abstract
Oncolytic viruses (OVs) are genetically modified or naturally occurring viruses, which preferentially replicate in and kill cancer cells while sparing healthy cells, and induce anti-tumor immunity. OV-induced tumor immunity can be enhanced through viral expression of anti-tumor cytokines such as interleukin 12 (IL-12). IL-12 is a potent anti-cancer agent that promotes T-helper 1 (Th1) differentiation, facilitates T-cell-mediated killing of cancer cells, and inhibits tumor angiogenesis. Despite success in preclinical models, systemic IL-12 therapy is associated with significant toxicity in humans. Therefore, to utilize the therapeutic potential of IL-12 in OV-based cancer therapy, 25 different IL-12 expressing OVs (OV-IL12s) have been genetically engineered for local IL-12 production and tested preclinically in various cancer models. Among OV-IL12s, oncolytic herpes simplex virus encoding IL-12 (OHSV-IL12) is the furthest along in the clinic. IL-12 expression locally in the tumors avoids systemic toxicity while inducing an efficient anti-tumor immunity and synergizes with anti-angiogenic drugs or immunomodulators without compromising safety. Despite the rapidly rising interest, there are no current reviews on OV-IL12s that exploit their potential efficacy and safety to translate into human subjects. In this article, we will discuss safety, tumor-specificity, and anti-tumor immune/anti-angiogenic effects of OHSV-IL12 as mono- and combination-therapies. In addition to OHSV-IL12 viruses, we will also review other IL-12-expressing OVs and their application in cancer therapy.
Collapse
|
375
|
Affiliation(s)
- Dorothy M. Gujral
- Department of Clinical Oncology, Imperial College Healthcare NHS Trust, London, United Kingdom
- Department of Surgery & Cancer, Imperial College, London, United Kingdom
| | - Elie N. Mouhayar
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Sanjeev Bhattacharyya
- Echocardiography Laboratory, St. Bartholomew's Hospital, London, United Kingdom
- William Harvey Research Institute, Queen Mary University of London, London, United Kingdom
- Institute of Cardiovascular Sciences, UCL, London, United Kingdom
| |
Collapse
|
376
|
Curigliano G, Lenihan D, Fradley M, Ganatra S, Barac A, Blaes A, Herrmann J, Porter C, Lyon AR, Lancellotti P, Patel A, DeCara J, Mitchell J, Harrison E, Moslehi J, Witteles R, Calabro MG, Orecchia R, de Azambuja E, Zamorano JL, Krone R, Iakobishvili Z, Carver J, Armenian S, Ky B, Cardinale D, Cipolla CM, Dent S, Jordan K, ESMO Guidelines Committee. Management of cardiac disease in cancer patients throughout oncological treatment: ESMO consensus recommendations. Ann Oncol 2020; 31:171-190. [PMID: 31959335 PMCID: PMC8019325 DOI: 10.1016/j.annonc.2019.10.023] [Citation(s) in RCA: 626] [Impact Index Per Article: 125.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/13/2022] Open
Abstract
Cancer and cardiovascular (CV) disease are the most prevalent diseases in the developed world. Evidence increasingly shows that these conditions are interlinked through common risk factors, coincident in an ageing population, and are connected biologically through some deleterious effects of anticancer treatment on CV health. Anticancer therapies can cause a wide spectrum of short- and long-term cardiotoxic effects. An explosion of novel cancer therapies has revolutionised this field and dramatically altered cancer prognosis. Nevertheless, these new therapies have introduced unexpected CV complications beyond heart failure. Common CV toxicities related to cancer therapy are defined, along with suggested strategies for prevention, detection and treatment. This ESMO consensus article proposes to define CV toxicities related to cancer or its therapies and provide guidance regarding prevention, screening, monitoring and treatment of CV toxicity. The majority of anticancer therapies are associated with some CV toxicity, ranging from asymptomatic and transient to more clinically significant and long-lasting cardiac events. It is critical however, that concerns about potential CV damage resulting from anticancer therapies should be weighed against the potential benefits of cancer therapy, including benefits in overall survival. CV disease in patients with cancer is complex and treatment needs to be individualised. The scope of cardio-oncology is wide and includes prevention, detection, monitoring and treatment of CV toxicity related to cancer therapy, and also ensuring the safe development of future novel cancer treatments that minimise the impact on CV health. It is anticipated that the management strategies discussed herein will be suitable for the majority of patients. Nonetheless, the clinical judgment of physicians remains extremely important; hence, when using these best clinical practices to inform treatment options and decisions, practitioners should also consider the individual circumstances of their patients on a case-by-case basis.
Collapse
Affiliation(s)
- G. Curigliano
- European Institute of Oncology IRCCS, Milan
- Department of Oncology and Haematology (DIPO), University of Milan, Milan, Italy
| | - D. Lenihan
- Cardiovascular Division, Cardio-Oncology Center of Excellence, Washington University Medical Center, St. Louis
| | - M. Fradley
- Cardio-oncology Program, Division of Cardiovascular Medicine, Morsani College of Medicine and H. Lee Moffitt Cancer Center and Research Institute, University of South Florida, Tampa
| | - S. Ganatra
- Cardio-Oncology Program, Lahey Medical Center, Burlington
| | - A. Barac
- Cardio-Oncology Program, Medstar Heart and Vascular Institute and MedStar Georgetown Cancer Institute, Georgetown University Hospital, Washington DC
| | - A. Blaes
- Division of Hematology, Oncology and Transplantation, University of Minnesota, Minneapolis
| | | | - C. Porter
- University of Kansas Medical Center, Lawrence, USA
| | - A. R. Lyon
- Royal Brompton Hospital and Imperial College, London, UK
| | - P. Lancellotti
- GIGA Cardiovascular Sciences, Acute Care Unit, Heart Failure Clinic, CHU Sart Tilman, University Hospital of Liège, Liège, Belgium
| | - A. Patel
- Morsani College of Medicine, University of South Florida, Tampa
| | - J. DeCara
- Medicine Section of Cardiology, University of Chicago, Chicago
| | - J. Mitchell
- Washington University Medical Center, St. Louis
| | - E. Harrison
- HCA Memorial Hospital and University of South Florida, Tampa
| | - J. Moslehi
- Vanderbilt University School of Medicine, Nashville
| | - R. Witteles
- Division of Cardiovascular Medicine, Falk CVRC, Stanford University School of Medicine, Stanford, USA
| | - M. G. Calabro
- Department of Anesthesia and Intensive Care, IRCCS, San Raffaele Scientific Institute, Milan, Italy
| | | | - E. de Azambuja
- Institut Jules Bordet and L’Université Libre de Bruxelles, Brussels, Belgium
| | | | - R. Krone
- Division of Cardiology, Washington University, St. Louis, USA
| | - Z. Iakobishvili
- Clalit Health Services, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - J. Carver
- Division of Cardiology, Abramson Cancer Center, Hospital of the University of Pennsylvania, Philadelphia
| | - S. Armenian
- Department of Population Sciences, City of Hope Comprehensive Cancer Center, Duarte
| | - B. Ky
- University of Pennsylvania School of Medicine, Philadelphia, USA
| | - D. Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Milan
| | - C. M. Cipolla
- Cardiology Department, European Institute of Oncology, IRCCS, Milan, Italy
| | - S. Dent
- Duke Cancer Institute, Duke University, Durham, USA
| | - K. Jordan
- Department of Medicine V, Hematology, Oncology and Rheumatology, University of Heidelberg, Heidelberg, Germany
| | - ESMO Guidelines Committee
- Correspondence to: ESMO Guidelines Committee, ESMO Head Office, Via Ginevra 4, CH-6900 Lugano, Switzerland, (ESMO Guidelines Committee)
| |
Collapse
|
377
|
Georgiou A, Yousaf N. CV complications of anti-cancer ICI therapy and their combinations: are we ready for challenges ahead? THE BRITISH JOURNAL OF CARDIOLOGY 2020; 27:01. [PMID: 35747422 PMCID: PMC9205254 DOI: 10.5837/bjc.2020.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Affiliation(s)
- Alexandros Georgiou
- Clinical Research Fellow in Medical Oncology The Royal Marsden NHS Foundation Trust, Downs Road, Sutton, SM2 5PT
| | - Nadia Yousaf
- Consultant Medical Oncologist The Royal Marsden NHS Foundation Trust, 203 Fulham Road, Chelsea, London, SW3 6JJ
| |
Collapse
|
378
|
Affiliation(s)
- Michael A. Biersmith
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
| | - Matthew S. Tong
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
| | - Avirup Guha
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
- Harrington Heart and Vascular InstituteCase Western Reserve UniversityClevelandOH
| | - Orlando P. Simonetti
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
| | - Daniel Addison
- Cardio‐Oncology ProgramDivision of Cardiovascular MedicineDepartment of MedicineThe Ohio State UniversityColumbusOH
- Division of Cancer Prevention and ControlDepartment of MedicineCollege of MedicineThe Ohio State UniversityColumbusOH
| |
Collapse
|
379
|
Baker JK, Shank-Coviello J, Zhou B, Dixon J, McCorkle R, Sarpong D, Medoff E, Cooper D, Seropian S, Dai F. Cardiotoxicity in Hematopoietic Stem Cell Transplant: Keeping the Beat. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 20:244-251.e4. [PMID: 32067953 DOI: 10.1016/j.clml.2019.12.027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 12/21/2019] [Accepted: 12/28/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION The number of hematopoietic stem cell transplants (HSCTs) performed in the United States and worldwide is increasing. Cardiac events have been well described in HSCT, and the incidence and type of cardiac events have not changed over recent decades. PATIENTS AND METHODS This study adds to the body of evidence in describing the incidence and type of cardiac events experienced by an allogeneic and autologous HSCT population at a single institution from 2012 to 2017. RESULTS Sixty-five (9.8%) patients experienced cardiac events, including atrial arrhythmia (N = 39), acute heart failure (N = 9), acute coronary syndrome (N = 7), and new onset hypertension (N = 9), with a few instances of bradycardia, ventricular arrhythmia, pericardial effusion, and pericarditis. Our multivariable regression analysis identified age (older), creatinine (higher), and history of coronary artery disease to significantly correlate with risk of cardiac event (P = .005, P = .039, and P = .038, respectively). A subgroup analysis of those patients experiencing a cardiac event found pre-transplant atrial dilation by trans-thoracic echocardiogram to correlate with increased risk of atrial arrhythmia (33.8% vs. 9.7%; P = .03). Patients developing a CE had an increased risk of death within 1 year (11% vs. 32%; P < .001). CONCLUSION We review our results in context of other important HSCT cardiac studies to illuminate the most relevant factors of medical history, laboratory data, and cardiac measurements that will identify patients at higher risk, allowing for intervention to improve HSCT outcomes.
Collapse
Affiliation(s)
| | | | - Bin Zhou
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT
| | - Jane Dixon
- School of Nursing, Yale University, Orange, CT
| | | | - Daniel Sarpong
- Center for Minority Health and Health Disparities Research and Education, Xavier University, New Orleans, LA
| | - Erin Medoff
- Department of Medicine, Yale University, New Haven, CT
| | - Dennis Cooper
- Department of Medicine, Stem Cell Transplantation, Rutgers Cancer Institute of New Jersey, New Brunswick, NJ
| | - Stuart Seropian
- Department of Medicine, Blood and Marrow Transplantation, Smilow Cancer Institute at Yale University, New Haven, CT
| | - Feng Dai
- Department of Biostatistics, Yale University School of Public Health, New Haven, CT
| |
Collapse
|
380
|
Abstract
BACKGROUND Immunotherapy is a significant breakthrough in cancer therapy in the last decade. Immunotherapy is better tolerated compared with chemotherapy. However, it does have side effects, and one of the rare and serious side effects of immunotherapy is cardiotoxicity. Cardiotoxicity has been described with other cancer-related treatments such as chemotherapy and targeted therapy. A high index of suspicion is required, and prompt management with immunosuppression needs to be instituted as soon as possible to prevent fatal outcomes. AREAS OF UNCERTAINTY Research is still ongoing to identify biomarkers that will help us to choose the patients, who will respond well to immunotherapy. Tumor-infiltrating lymphocytes, tumor PD-L1 expression, and tumor mutational burden explored as potential biomarkers. There are no predictive biomarkers to identify patients who are at higher risk of severe cardiotoxicity. Both cardiologists and oncologists should be aware of cardiac toxicity from immune checkpoint inhibitors. CONCLUSION All patients who are starting immune checkpoint inhibitors should undergo baseline cardiac risk factor assessment with referral to a cardiologist in a patient with multiple risk factors or previous history of cardiovascular disease. Cardiac immune-related adverse events are higher in patients taking combination therapy with anti-CTLA-4/anti-PD-1 agents compared with monotherapy. Patients with known cardiac comorbidities require a higher level of vigilance to monitor for cardiac toxicity because nonspecific symptoms can lead to rapid clinical deterioration and a higher rate of mortality when treated with checkpoint inhibitors.
Collapse
Affiliation(s)
- Venu Madhav Konala
- Division of Medical Oncology, Department of Internal Medicine, Ashland Bellefonte Cancer Center, Ashland, KY
| | - Sreedhar Adapa
- Division of Nephrology, The Nephrology Group, Fresno, CA; and
| | - Wilbert S Aronow
- Division of Cardiology, Westchester Medical Center, New York Medical College, Valhalla, NY
| |
Collapse
|
381
|
Abstract
A growing number of effective cancer therapies is associated with cardiovascular (CV) toxicities including myocardial injury or dysfunction, leading to reduced ventricular function, and increased risk of heart failure. As the timing of administration of cancer treatment is known, the potential for risk stratification pre-treatment, and appropriate surveillance and monitoring during treatment, and intervention with cardio-protective treatment strategies in patients exhibiting early evidence of CV toxicity is an appealing clinical strategy. The field of cardio-oncology has developed, and the application of monitoring strategies using CV biomarkers and CV imaging has been to focus of many studies and is now implemented in dedicated cardio-oncology services supporting oncology centres. In this article, we review the background and rationale for monitoring, the different options and their strengths, weaknesses and where they are helpful in specific cardiotoxic cancer therapies, and the impact in cardio-oncology care.
Collapse
Affiliation(s)
- Mohsen Habibian
- The Prince Charles Hospital and University of Queensland, Rode Road, Chermside, QLD 4032, Australia.,Cardio-Oncology Service, Royal Brompton Hospital, Sydney Street, Chelsea, London, SW3 6NP, UK
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Sydney Street, Chelsea, London, SW3 6NP, UK.,National Heart and Lung Institute, Imperial College London, Cale Street, Chelsea, London, SW3 6LY, UK
| |
Collapse
|
382
|
Nichetti F, Ligorio F, Zattarin E, Signorelli D, Prelaj A, Proto C, Galli G, Marra A, Apollonio G, Porcu L, de Braud F, Lo Russo G, Ferrara R, Garassino MC. Is There an Interplay between Immune Checkpoint Inhibitors, Thromboprophylactic Treatments and Thromboembolic Events? Mechanisms and Impact in Non-Small Cell Lung Cancer Patients. Cancers (Basel) 2019; 12:cancers12010067. [PMID: 31881699 PMCID: PMC7016680 DOI: 10.3390/cancers12010067] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 12/19/2019] [Accepted: 12/20/2019] [Indexed: 12/14/2022] Open
Abstract
PD-1 pathway blockade has been shown to promote proatherogenic T-cell responses and destabilization of atherosclerotic plaques. Moreover, preclinical evidence suggests a potential synergy of antiplatelet drugs with immune checkpoint inhibitors (ICIs). We conducted an analysis within a prospective observational protocol (APOLLO study) to investigate the rates, predictors, and prognostic significance of thromboembolic events (TE) and thromboprophylaxis in patients with advanced NSCLC treated with ICIs. Among 217 patients treated between April 2014 and September 2018, 13.8% developed TE events. Current smoking status (HR 3.61 (95% CI 1.52–8.60), p = 0.004) and high (>50%) PD-L1 (HR 2.55 (95% CI 1.05–6.19), p = 0.038) resulted in being independent TE predictors. An increased risk of death following a diagnosis of TE (HR 2.93; 95% CI 1.59–5.42; p = 0.0006) was observed. Patients receiving antiplatelet treatment experienced longer progression-free survival (PFS) (6.4 vs. 3.4 months, HR 0.67 (95% CI 0.48–0.92), p = 0.015) and a trend toward better OS (11.2 vs. 9.6 months, HR 0.78 (95% CI 0.55–1.09), p = 0.14), which were not confirmed in a multivariate model. No impact of anticoagulant treatment on patients’ outcomes was observed. NSCLC patients treated with ICIs bear a consistent risk for thrombotic complications, with a detrimental effect on survival. The impact of antiplatelet drugs on ICIs efficacy deserves further investigation in prospective trials.
Collapse
Affiliation(s)
- Federico Nichetti
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
- Correspondence: ; Tel.: +39-022-390-3066
| | - Francesca Ligorio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Emma Zattarin
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Diego Signorelli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Arsela Prelaj
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Claudia Proto
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Giulia Galli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Antonio Marra
- Department of Medical Oncology, Istituto Europeo di Oncologia, 20141 Milan, Italy;
| | - Giulia Apollonio
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Luca Porcu
- Department of Oncology, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, 20156 Milan, Italy;
| | - Filippo de Braud
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Giuseppe Lo Russo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Roberto Ferrara
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| | - Marina Chiara Garassino
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, 20133 Milan, Italy; (F.L.); (E.Z.); (D.S.); (A.P.); (C.P.); (G.G.); (G.A.); (F.d.B.); (G.L.R.); (R.F.); (M.C.G.)
| |
Collapse
|
383
|
Guo X, Wang H, Zhou J, Li Y, Duan L, Si X, Zhang L, Fang L, Zhang L. Clinical manifestation and management of immune checkpoint inhibitor-associated cardiotoxicity. Thorac Cancer 2019; 11:475-480. [PMID: 31849171 PMCID: PMC6996973 DOI: 10.1111/1759-7714.13250] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 10/25/2019] [Indexed: 01/02/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) targeting programmed death-1 (PD-1), its ligand (PD-L1), and cytotoxic T-lymphocyte-associated antigen 4 (CTLA4) have revolutionized cancer treatment by recovering the attack of T lymphocytes on the malignant cells. They have improved clinical outcomes dramatically in multiple types of advanced-stage malignancies. Even though the tolerance and safety profiles are generally good, it has been widely reported that ICIs can cause severe or fatal immune-related adverse events (irAEs), since the activated T lymphocytes are not specific for tumor cells. Cardiac irAEs appear to occur less frequently than irAEs in other organ systems but are notorious for high mortality. Here, we aim to identify and characterize the ICI-associated cardiotoxicity and summarize the optional diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Hanping Wang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Jiaxin Zhou
- Department of Rheumatism and Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Yue Li
- Department of Gastroenterology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Lian Duan
- Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Xiaoyan Si
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Ligang Fang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| | - Li Zhang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
384
|
Cautela J, Rouby F, Salem JE, Alexandre J, Scemama U, Dolladille C, Cohen A, Paganelli F, Ederhy S, Thuny F. Acute Coronary Syndrome With Immune Checkpoint Inhibitors: A Proof-of-Concept Case and Pharmacovigilance Analysis of a Life-Threatening Adverse Event. Can J Cardiol 2019; 36:476-481. [PMID: 32144037 DOI: 10.1016/j.cjca.2019.11.035] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 11/28/2019] [Accepted: 11/30/2019] [Indexed: 12/20/2022] Open
Abstract
Isolated cases of acute coronary syndrome (ACS) associated with immune checkpoint inhibitors (ICIs) have been described without the establishment of a formal cause-and-effect relationship between treatment and adverse event. We reported a case of ACS after the first administration of an ICI and with a fatal recurrence in another coronary area immediately after readministration. According to guidelines, causality was considered to be certain. Subsequently, we queried the French pharmacovigilance database and found 4 cases of ACS with coronary artery thrombosis. Causality was probable in those patients. These data suggest that ACS may be another life-threatening cardiac adverse event occurring with ICI exposure.
Collapse
Affiliation(s)
- Jennifer Cautela
- University Mediterranean Centre of Cardio-Oncology, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, and Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Hôpital Nord, Marseille, France; Groupe Méditerranéen de Cardio-Oncologie, Marseille, France; Centre for Cardiovascular and Nutrition Research, Aix-Marseille University, INSERM 1263, INRA 1260, Marseille, France
| | - Franck Rouby
- Department of Clinical Pharmacology and Pharmacovigilance, Regional Pharmacovigilance Centre of Marseille Provence Corse, Marseille, France
| | - Joe-Elie Salem
- UNICO-GRECO Cardio-Oncology Program, Sorbonne Université, INSERM, CIC Paris-Est, AP-HP, and Department of Pharmacology, Pitié-Salpêtrière Hospital, Paris, France
| | - Joachim Alexandre
- PICARO Cardio-Oncology Program, Department of Pharmacology, CHU de Caen, Caen, France; Department of Cardiology, CHU de Caen, Caen, France; Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, Medical School, Université de Caen Normandie, EA 4650, Caen, France
| | - Ugo Scemama
- Department of Imaging, Hôpital Nord, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, Marseille, France
| | - Charles Dolladille
- PICARO Cardio-Oncology Program, Department of Pharmacology, CHU de Caen, Caen, France; Department of Cardiology, CHU de Caen, Caen, France; Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, Medical School, Université de Caen Normandie, EA 4650, Caen, France
| | - Ariel Cohen
- Sorbonne Université, Hôpitaux Universitaires Est Parisien, Hôpital Saint Antoine, and Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, INSERM 856, Paris, France
| | - Franck Paganelli
- University Mediterranean Centre of Cardio-Oncology, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, and Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Hôpital Nord, Marseille, France; Centre for Cardiovascular and Nutrition Research, Aix-Marseille University, INSERM 1263, INRA 1260, Marseille, France
| | - Stéphane Ederhy
- Sorbonne Université, Hôpitaux Universitaires Est Parisien, Hôpital Saint Antoine, and Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, INSERM 856, Paris, France
| | - Franck Thuny
- University Mediterranean Centre of Cardio-Oncology, Assistance Publique-Hôpitaux de Marseille, Aix-Marseille University, and Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Hôpital Nord, Marseille, France; Groupe Méditerranéen de Cardio-Oncologie, Marseille, France; Centre for Cardiovascular and Nutrition Research, Aix-Marseille University, INSERM 1263, INRA 1260, Marseille, France.
| |
Collapse
|
385
|
Edmonds KP, Saunders IM, Willeford A, Ajayi TA, Atayee RS. Emerging Challenges to the Safe and Effective Use of Methadone for Cancer-Related Pain in Paediatric and Adult Patient Populations. Drugs 2019; 80:115-130. [PMID: 31820362 DOI: 10.1007/s40265-019-01234-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Methadone continues to be an important medication for the treatment of paediatric and adult cancer-related pain. Appropriate patient selection to ensure safe and effective treatment by a team of clinicians who appreciate and are familiar with methadone and its unique pharmacology is crucial. Unlike morphine and other more common opioids, methadone is purported to have involvement with delta-opioid receptor and higher affinity as an N-methyl-D-aspartate-receptor antagonist. Clinically this gives it the advantage of being effective for both nociceptive and neuropathic pain, but also may be useful in the setting of tolerance to other opioids. Methadone also comes in multiple available formulations that can be administrated through a variety of routes beyond the oral route. Challenges with methadone in treating cancer-related pain include drug interactions specifically as it relates to new targeted cancer therapies. Recent guidelines recommend electrocardiogram monitoring with methadone and there is potential for additive cardiac toxicity in the oncology setting. Appropriate dosing of methadone for pain management given age, organ dysfunction, and patients who are on methadone maintenance therapy are also key factors. This article aims to provide clinicians with evidence and clinical practice guidelines for safe and appropriate use of methadone including indication, initiation, and monitoring given its complexity for management of pain in the dynamic oncology setting.
Collapse
Affiliation(s)
- Kyle P Edmonds
- Doris A. Howell Palliative Care Teams, University of California San Diego, La Jolla, CA, 92093, USA
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
| | - Ila M Saunders
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Pharmacy, University of California San Diego Health, La Jolla, CA, USA
| | - Andrew Willeford
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA
- Department of Pharmacy, University of California San Diego Health, La Jolla, CA, USA
| | - Toluwalase A Ajayi
- Digital Medicine, Scripps Research Translational Institute, La Jolla, CA, 92037, USA
| | - Rabia S Atayee
- Doris A. Howell Palliative Care Teams, University of California San Diego, La Jolla, CA, 92093, USA.
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, 92093, USA.
- Department of Pharmacy, University of California San Diego Health, La Jolla, CA, USA.
| |
Collapse
|
386
|
Edahiro R, Shiroyama T, Hijiki S, Nojima S, Shirai Y, Koyama S, Kumanogoh A. Severe myocarditis with slight lymphocytic infiltration after nivolumab treatment. Lung Cancer 2019; 140:116-117. [PMID: 31848027 DOI: 10.1016/j.lungcan.2019.12.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2019] [Accepted: 12/08/2019] [Indexed: 11/15/2022]
Affiliation(s)
- Ryuya Edahiro
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Takayuki Shiroyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan.
| | - Sachiko Hijiki
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Satoshi Nojima
- Department of Pathology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yuya Shirai
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shohei Koyama
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Osaka, Japan
| |
Collapse
|
387
|
Chitturi KR, Xu J, Araujo-Gutierrez R, Bhimaraj A, Guha A, Hussain I, Kassi M, Bernicker EH, Trachtenberg BH. Immune Checkpoint Inhibitor-Related Adverse Cardiovascular Events in Patients With Lung Cancer. JACC CardioOncol 2019; 1:182-192. [PMID: 34396181 PMCID: PMC8352266 DOI: 10.1016/j.jaccao.2019.11.013] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2019] [Revised: 11/10/2019] [Accepted: 11/11/2019] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES The purpose of this study was to evaluate whether immune checkpoint inhibitors (ICIs) are associated with an increased risk of major adverse cardiovascular events (MACE) compared with non-ICI therapies in patients with lung cancer. BACKGROUND ICIs activate the host immune system to target cancer cells. Though uncommon, cardiovascular immune-related adverse events can be life-threatening. METHODS A retrospective single-institution cohort study of 252 patients with pathologically confirmed lung cancer who received ICI or non-ICI therapy was analyzed. The primary endpoint was MACE, defined as a composite of cardiovascular death, nonfatal myocardial infarction, nonfatal stroke, and hospitalization for heart failure. RESULTS During a median follow-up of 6 months, MACE occurred in 13.3% of ICI-treated patients, with a median time to event of 51 days, compared with 10.3% and 64 days in non-ICI patients. ICIs were not associated with MACE (hazard ratio [HR]: 1.18; 95% confidence interval [CI]: 0.57 to 2.43; p = 0.66) in a univariable Fine-Gray regression analysis incorporating noncardiovascular death as a competing risk. Multivariable regression analyses determined that patients treated with ICIs with elevated serum troponin I >0.01 ng/ml (HR: 7.27; 95% CI: 2.72 to 19.43; p < 0.001) and B-type natriuretic peptide (BNP) >100 pg/ml (HR: 2.65; 95% CI: 1.01 to 6.92; p = 0.047) had an increased risk of MACE. Patients pre-treated or receiving combined immunotherapy with ICIs and vascular endothelial growth factor inhibitors (VEGFIs) or tyrosine kinase inhibitors (TKIs) had an increased risk of MACE (HR: 2.15; 95% CI: 1.05 to 4.37; p = 0.04). CONCLUSIONS ICIs were not independently associated with an increased risk of MACE in patients with lung cancer, although power is an important limitation in these analyses. ICI-associated cardiotoxicity was associated with elevations in serum troponin and BNP, and combined immunotherapy with VEGFIs or TKIs. Future studies are needed to further define the role of cardiac biomarkers as a monitoring strategy with ICI therapy.
Collapse
Key Words
- BNP
- BNP, B-type natriuretic peptide
- CI, confidence interval
- HR, hazard ratio
- ICI, immune checkpoint inhibitor
- IQR, interquartile range
- LVEF, left ventricular ejection fraction
- MACE
- MACE, major adverse cardiovascular events
- PD, programmed cell death protein
- PD-L1, programmed cell death-ligand 1
- TKI, tyrosine kinase inhibitor
- TnI, troponin I
- VEGFI, vascular endothelial growth factor inhibitor
- cardiotoxicity
- immune checkpoint inhibitors
- lung cancer
- troponin
Collapse
Affiliation(s)
| | - Jiaqiong Xu
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
- Center for Outcomes Research, Houston Methodist Research Institute, Houston, Texas, USA
| | | | - Arvind Bhimaraj
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
| | - Ashrith Guha
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
| | - Imad Hussain
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
| | - Mahwash Kassi
- Houston Methodist DeBakey Heart & Vascular Center, Houston, Texas, USA
| | - Eric H. Bernicker
- Department of Medical Oncology, Houston Methodist Cancer Center, Houston, Texas, USA
| | | |
Collapse
|
388
|
Liu Y, Wang H, Deng J, Sun C, He Y, Zhou C. Toxicity of tumor immune checkpoint inhibitors-more attention should be paid. Transl Lung Cancer Res 2019; 8:1125-1133. [PMID: 32010590 PMCID: PMC6976385 DOI: 10.21037/tlcr.2019.11.26] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 10/21/2019] [Indexed: 12/17/2022]
Abstract
In recent years, immunotherapy, especially immune checkpoint inhibitors (ICIs), has achieved amazing results in the treatment of lung cancer, melanoma, renal clear cell carcinoma and other malignant tumors. Although ICIs have achieved significant efficacy in tumor treatment, the immune-related adverse events (irAEs) caused by non-specific immune activation of ICIs can directly affect the result of treatment, even threaten the life of patients. The most common form of irAEs involve the skin, lung, colon, liver and endocrine organs. However, it is noticeable that although irAEs of some organs are more common, actually any organ and tissue are likely to be affected, because of non-specific activation of the immune system. Other tissues and organs, though rare, can be more severe and even fatal, such as neurological disorders and myocarditis. Therefore, effective management of irAEs is of great importance for the efficacy of immunotherapy. This review is focused on the morbidity, clinical manifestations, diagnosis and treatment of tumor immune toxic effects.
Collapse
Affiliation(s)
- Yu Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
- Tongji University, Shanghai 200433, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
- Tongji University, Shanghai 200433, China
| | - Juan Deng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
- Tongji University, Shanghai 200433, China
| | - Chenglong Sun
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
- Anhui No. 2 Provincial People’s Hospital, Hefei 230041, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, Shanghai 200433, China
| |
Collapse
|
389
|
Anker MS, Hadzibegovic S, Lena A, Belenkov Y, Bergler‐Klein J, de Boer RA, Farmakis D, von Haehling S, Iakobishvili Z, Maack C, Pudil R, Skouri H, Cohen‐Solal A, Tocchetti CG, Coats AJ, Seferović PM, Lyon AR, for the Heart Failure Association Cardio‐Oncology Study Group of the European Society of Cardiology. Recent advances in cardio-oncology: a report from the 'Heart Failure Association 2019 and World Congress on Acute Heart Failure 2019'. ESC Heart Fail 2019; 6:1140-1148. [PMID: 31884717 PMCID: PMC6989292 DOI: 10.1002/ehf2.12551] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
While anti-cancer therapies, including chemotherapy, immunotherapy, radiotherapy, and targeted therapy, are constantly advancing, cardiovascular toxicity has become a major challenge for cardiologists and oncologists. This has led to an increasing demand of cardio-oncology units in Europe and a growing interest of clinicians and researchers. The Heart Failure 2019 meeting of the Heart Failure Association of the European Society of Cardiology in Athens has therefore created a scientific programme that included four dedicated sessions on the topic along with several additional lectures. The major points that were discussed at the congress included the implementation and delivery of a cardio-oncology service, the collaboration among cardio-oncology experts, and the risk stratification, prevention, and early recognition of cardiotoxicity. Furthermore, sessions addressed the numerous different anti-cancer therapies associated with cardiotoxic effects and provided guidance on how to treat cancer patients who develop cardiovascular disease before, during, and after treatment.
Collapse
Affiliation(s)
- Markus S. Anker
- Division of Cardiology and Metabolism, Department of CardiologyCharité 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 FranklinBerlinGermany
| | - Sara Hadzibegovic
- Division of Cardiology and Metabolism, Department of CardiologyCharité 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 FranklinBerlinGermany
| | - Alessia Lena
- Division of Cardiology and Metabolism, Department of CardiologyCharité 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 FranklinBerlinGermany
| | | | | | - Rudolf A. de Boer
- Department of CardiologyUniversity of GroningenUniversity Medical Center Groningen, GroningenThe Netherlands
| | - Dimitrios Farmakis
- University of Cyprus Medical SchoolNicosiaCyprus
- Department of Cardiology, Cardio‐Oncology Clinic, Heart Failure UnitAthens University Hospital ‘Attikon’, National and Kapodistrian University of AthensAthensGreece
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart Center Göttingen, German Center for Cardiovascular Medicine (DZHK)University of Göttingen Medical Center, Georg‐August‐UniversityGöttingenGermany
| | - Zaza Iakobishvili
- Department of Community Cardiology, Clalit Health Fund, and Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC)University Clinic WürzburgWürzburgGermany
| | - Radek Pudil
- 1st Department of Medicine–Cardioangiology, Faculty of MedicineUniversity HospitalHradec KrálovéCzech Republic
| | - Hadi Skouri
- Department of Internal MedicineAmerican University of Beirut Medical CenterBeirutLebanon
| | - Alain Cohen‐Solal
- Department of Cardiology, Lariboisière Hospital and U942 INSERM, BIOCANVAS (Biomarqueurs Cardiovasculaires)Paris UniversityParisFrance
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center for Clinical and Translational Sciences (CIRCET)Federico II UniversityNaplesItaly
| | | | - Petar M. Seferović
- Faculty of Medicine and Heart Failure CenterBelgrade University Medical Center, University of BelgradeBelgradeSerbia
| | | | | |
Collapse
|
390
|
Lancellotti P, Moonen M, Galderisi M. Chimeric Antigen Receptor T-Cells and Cardiovascular Toxicity. J Am Coll Cardiol 2019; 74:3109-3111. [DOI: 10.1016/j.jacc.2019.10.028] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 10/22/2019] [Indexed: 10/25/2022]
|
391
|
Zhou YW, Zhu YJ, Wang MN, Xie Y, Chen CY, Zhang T, Xia F, Ding ZY, Liu JY. Immune Checkpoint Inhibitor-Associated Cardiotoxicity: Current Understanding on Its Mechanism, Diagnosis and Management. Front Pharmacol 2019; 10:1350. [PMID: 31849640 PMCID: PMC6897286 DOI: 10.3389/fphar.2019.01350] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 10/24/2019] [Indexed: 02/05/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) that target cytotoxic T lymphocyte antigen 4, programmed cell death-1, and PD-ligand 1 have revolutionized cancer treatment, achieving unprecedented efficacy in multiple malignancies. ICIs are increasingly being used in early cancer settings and in combination with various other types of therapies, including targeted therapy, radiotherapy, and chemotherapy. However, despite the excellent therapeutic effect of ICIs, these medications typically result in a broad spectrum of toxicity reactions, termed immune-related adverse events (irAEs). Of all irAEs, cardiotoxicity, uncommon but with high mortality, has not been well recognized. Herein, based on previous published reports and current evidence, we summarize the incidence, diagnosis, clinical manifestations, underlying mechanisms, treatments, and outcomes of ICI-associated cardiotoxicity and discuss possible management strategies. A better understanding of these characteristics is critical to managing patients with ICI-associated cardiotoxicity.
Collapse
Affiliation(s)
- Yu-Wen Zhou
- Department of Biotherapy, Cancer Center, and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Ya-Juan Zhu
- Department of Biotherapy, Cancer Center, and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Man-Ni Wang
- Department of Biotherapy, Cancer Center, and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Yao Xie
- Department of Dermatovenerology, West China Hospital, Sichuan University, Chengdu, China
| | - Chao-Yue Chen
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tao Zhang
- West China School of Medicine, Sichuan University, Chengdu, China
| | - Fan Xia
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, China
| | - Zhen-Yu Ding
- Department of Biotherapy, Cancer Center, and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| | - Ji-Yan Liu
- Department of Biotherapy, Cancer Center, and National Clinical Research Center for Geriatrics, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
392
|
Guo X, Wang H, Zhou J, Duan L, Li Y, Si X, Zhang L, Fang L, Zhang L. [Clinical Diagnosis and Treatment Recommendations for Cardiac Adverse Reactions Related to Immune Checkpoint Inhibitor]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2019; 22:627-632. [PMID: 31650944 PMCID: PMC6817436 DOI: 10.3779/j.issn.1009-3419.2019.10.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Immunotherapy of malignant tumors has become a hot spot in the field of cancer research and treatment, bringing new hope to patients with advanced cancer. Activation of molecular programmer death protein-1 and T lymphocyte-associated antigen 4-related signaling pathways at the immunological checkpoint can inhibit T lymphocyte activation and thereby block the inflammatory response. Tumor cells achieve immune escape by activating the molecular pathways associated with immune checkpoints. The immune checkpoint inhibitor can wake up T lymphocytes and enhance the body's clearance of tumor cells. However, the role of immune checkpoint inhibitors is not specific to tumor cells, and it can cause side effects of multiple systems including the cardiovascular system while killing tumor cells. We will summarize the relevant cardiac side effects and give advice on how to manage it.
Collapse
Affiliation(s)
- Xiaoxiao Guo
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Hanping Wang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Jiaxin Zhou
- Department of Rheumatism and Immunology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Lian Duan
- Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Yue Li
- Department of Digestive Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Xiaoyan Si
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Li Zhang
- Department of Clinical Laboratory, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Ligang Fang
- Department of Cardiology, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Li Zhang
- Department of Respiratory Medicine, Peking Union Medical College Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Beijing 100730, China
| |
Collapse
|
393
|
Agunbiade TA, Zaghlol RY, Barac A. Heart Failure in Relation to Tumor-Targeted Therapies and Immunotherapies. Methodist Debakey Cardiovasc J 2019; 15:250-257. [PMID: 31988685 PMCID: PMC6977568 DOI: 10.14797/mdcj-15-4-250] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Tumor-targeted therapies such as trastuzumab have led to significant improvements in survival of human epidermal growth factor receptor 2 (HER2)-positive breast cancer. However, these therapies have also been associated with significant left ventricular dysfunction. The incidence of trastuzumab-induced heart failure has decreased significantly since the initial reports, in large part due to improved screening, closer monitoring for early changes in left ventricular function, and a significant decrease in the concurrent administration of anthracyclines. The mechanism of trastuzumab cardiotoxicity is still not well understood, but current knowledge suggests that ErbB2 inhibition in cardiac myocytes plays a key role. In addition to trastuzumab and other HER2-targeted agents, vascular endothelial growth factor inhibitors, proteasome inhibitors, and immune checkpoint inhibitors are all additional classes of drugs used with great success in the treatment of solid tumors and hematologic malignancies. Yet these, too, have been associated with cardiac toxicity that ranges from a mild asymptomatic decrease in ejection fraction to fulminant myocarditis. In this review, we summarize the cardiotoxic effects of tumor-targeted and immunotherapies with a focus on HER2 antagonists.
Collapse
Affiliation(s)
| | - Raja Y Zaghlol
- MEDSTAR WASHINGTON HOSPITAL CENTER, GEORGETOWN UNIVERSITY, WASHINGTON, DC
| | - Ana Barac
- MEDSTAR WASHINGTON HOSPITAL CENTER, GEORGETOWN UNIVERSITY, WASHINGTON, DC
| |
Collapse
|
394
|
Itzhaki Ben Zadok O, Ben-Avraham B, Nohria A, Orvin K, Nassar M, Iakobishvili Z, Neiman V, Goldvaser H, Kornowski R, Ben Gal T. Immune-Checkpoint Inhibitor-Induced Fulminant Myocarditis and Cardiogenic Shock. JACC: CARDIOONCOLOGY 2019; 1:141-144. [PMID: 34396174 PMCID: PMC8352193 DOI: 10.1016/j.jaccao.2019.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 07/29/2019] [Accepted: 07/30/2019] [Indexed: 11/25/2022]
Affiliation(s)
- Osnat Itzhaki Ben Zadok
- Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Ben Ben-Avraham
- Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Anju Nohria
- Cardiovascular Division, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Katia Orvin
- Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Mithal Nassar
- Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Zaza Iakobishvili
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Tel-Aviv-Jaffa District, Clalit Health Services, Tel-Aviv, Israel
| | - Victoria Neiman
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Hadar Goldvaser
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Davidoff Cancer Center, Rabin Medical Center, Petah Tikva, Israel
| | - Ran Kornowski
- Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tuvia Ben Gal
- Department of Cardiology, Rabin Medical Center, Petah Tikva, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
395
|
Michel L, Rassaf T, Totzeck M. Cardiotoxicity from immune checkpoint inhibitors. IJC HEART & VASCULATURE 2019; 25:100420. [PMID: 31517036 PMCID: PMC6736791 DOI: 10.1016/j.ijcha.2019.100420] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Revised: 08/22/2019] [Accepted: 08/31/2019] [Indexed: 12/20/2022]
Abstract
Immune checkpoint inhibitor (ICI) therapy has greatly improved treatment of various advanced cancers but increasing use of ICI therapy has exposed the risk of ICI-related cardiovascular side effects. Immune checkpoints are inhibitory regulators of T cell activation and mediate T cell effector functions during physiological responses to shield from autoimmune reactions. ICI therapy for advanced cancers promotes immune activity against tumors and is applied within a broad collective of cancer patients. Widespread use of ICI therapy has revealed the burden of immune related adverse events with various organ manifestations and characteristics. Since immune checkpoints are highly relevant for maintaining myocardial homeostasis as emerging evidence implicates, inhibition of immune checkpoint pathways has been associated with various forms of cardiotoxicity in preclinical models and patients. Although ICI-related cardiotoxicity is rare, it has significant relevance due to high mortality rates. This review focuses on current knowledge about cardiac ICI-related toxicity. We summarize the most common forms and delineate incidence, presentation, and treatment. Clinical characteristics are correlated to potential underlying pathomechanisms. We outline epidemiology, risk factors, and course of disease. Recommendations for monitoring and critical diagnostic measures are specified within the context of different forms of cardiac involvement. Different therapeutic implications for suspected ICI-related cardiotoxicity and their limitations are critically summarized. We highlight current gaps of knowledge concerning the underlying pathomechanisms and clinical characteristics of ICI-related cardiotoxicity. Future challenges are depicted for optimum cardio-oncology care of patients receiving ICI therapy.
Collapse
Affiliation(s)
- Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Hufelandstraße 55, 45147 Essen, Germany
| |
Collapse
|
396
|
Is PD-1 blockade a potential therapy for HBV? JHEP Rep 2019; 1:142-144. [PMID: 32040093 PMCID: PMC7001582 DOI: 10.1016/j.jhepr.2019.07.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 07/24/2019] [Accepted: 07/26/2019] [Indexed: 01/05/2023] Open
|
397
|
Tsuruda T, Sato Y, Kajihara K, Kawabata T, Kubuki Y, Komaki S, Kikuchi M, Ishikawa T, Tono T, Kitamura K. Non-canonical Expression of Cardiac Troponin-T in Neuroendocrine Ethmoid Sinus Carcinoma Following Immune Checkpoint Blockade. Front Cardiovasc Med 2019; 6:124. [PMID: 31508427 PMCID: PMC6716019 DOI: 10.3389/fcvm.2019.00124] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 08/12/2019] [Indexed: 01/08/2023] Open
Abstract
We describe the case of a patient with neuroendocrine ethmoid sinus carcinoma, who exhibited markedly elevated levels of serum cardiac troponin-T and creatine kinase (CK)-MB isoenzyme without any symptom after the administration of nivolumab, immune checkpoint inhibitor. The repeated 12-leads-electrocardiogram did not show any changes in the ST-T segments or arrhythmias. The echocardiogram showed normal ranges of left ventricular contraction in the clinical course. Cardiac magnetic resonance imaging showed minimal myocardial edema and inflammation. Blood clots in the metastatic lesion of bone marrow aspirates exhibited positive staining for cardiac troponin-T and CK-MB in the cytoplasm and nucleoplasm of neoplastic cells. Although we did not perform a second cardiac magnetic resonance imaging and autopsy, we postulate that the attack of the neoplastic cells by the immune checkpoint inhibitor or the secretion from neoplastic cell-derived extracellular vesicles may have exacerbated the increase in concentrations of these molecules in the blood. Our case should warrant consideration a false-positive value of cardiac troponin-T and CK-MB can be obtained in cases with malignancy.
Collapse
Affiliation(s)
- Toshihiro Tsuruda
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yuichiro Sato
- Department of Diagnostic Pathology, Faculty of Medicine, University of Miyazaki, Miyazaki University Hospital, Miyazaki, Japan
| | - Kei Kajihara
- Department of Otolaryngology, Head & Neck Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Takayuki Kawabata
- Department of Otolaryngology, Head & Neck Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Yoko Kubuki
- Department of Gastroenterology and Hematology, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Soichi Komaki
- Department of Cardiovascular Medicine, Miyazaki Prefectural Nobeoka Hospital, Miyazaki, Japan
| | - Masao Kikuchi
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tetsunori Ishikawa
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Tetsuya Tono
- Department of Otolaryngology, Head & Neck Surgery, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| | - Kazuo Kitamura
- Department of Internal Medicine, Circulatory and Body Fluid Regulation, Faculty of Medicine, University of Miyazaki, Miyazaki, Japan
| |
Collapse
|
398
|
Cai H, Wang C, Shukla S, Steinmetz NF. Cowpea Mosaic Virus Immunotherapy Combined with Cyclophosphamide Reduces Breast Cancer Tumor Burden and Inhibits Lung Metastasis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2019; 6:1802281. [PMID: 31453050 PMCID: PMC6702650 DOI: 10.1002/advs.201802281] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 04/14/2019] [Indexed: 05/10/2023]
Abstract
Patients with metastatic triple-negative breast cancer (TNBC) have a poor prognosis, so new therapies or drug combinations that achieve more effective and durable responses are urgently needed. Here, a combination therapy using cowpea mosaic virus (CPMV) and low doses of cyclophosphamide (CPA) is developed with remarkable synergistic efficacy against 4T1 mouse tumors in vivo. The combination therapy not only attenuates the growth of primary tumor and increases survival, but also suppresses distant tumor growth and reduces lung metastasis. Mechanistic analysis indicates that the combination of CPMV and CPA increases the secretion of several cytokines, activates antigen-presenting cells, increases the abundance of tumor infiltrating T cells, and systematically reverses the immunosuppression. These results show that the combination of CPMV in situ vaccination with chemotherapy may become a potent new strategy for the treatment of TNBC.
Collapse
Affiliation(s)
- Hui Cai
- Department of NanoEngineering/Department of Radiology/Moores Cancer Center/Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Department of Biomedical EngineeringCase Western Reserve University10900 Euclid AvenueClevelandOH44106USA
| | - Chao Wang
- Department of NanoEngineering/Department of Radiology/Moores Cancer Center/Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Department of Biomedical EngineeringCase Western Reserve University10900 Euclid AvenueClevelandOH44106USA
| | - Sourabh Shukla
- Department of NanoEngineering/Department of Radiology/Moores Cancer Center/Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Department of Biomedical EngineeringCase Western Reserve University10900 Euclid AvenueClevelandOH44106USA
| | - Nicole F. Steinmetz
- Department of NanoEngineering/Department of Radiology/Moores Cancer Center/Department of BioengineeringUniversity of California, San DiegoLa JollaCA92093USA
- Department of Biomedical EngineeringCase Western Reserve University10900 Euclid AvenueClevelandOH44106USA
| |
Collapse
|
399
|
Ji C, Roy MD, Golas J, Vitsky A, Ram S, Kumpf SW, Martin M, Barletta F, Meier WA, Hooper AT, Sapra P, Khan NK, Finkelstein M, Guffroy M, Buetow BS. Myocarditis in Cynomolgus Monkeys Following Treatment with Immune Checkpoint Inhibitors. Clin Cancer Res 2019; 25:4735-4748. [PMID: 31085720 DOI: 10.1158/1078-0432.ccr-18-4083] [Citation(s) in RCA: 92] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 03/28/2019] [Accepted: 05/08/2019] [Indexed: 11/16/2022]
Abstract
PURPOSE Immune checkpoint inhibitors (ICI) targeting PD1, PDL1, or CTLA4 are associated with immune-related adverse events (irAE) in multiple organ systems including myocarditis. The pathogenesis and early diagnostic markers for ICI-induced myocarditis are poorly understood, and there is currently a lack of laboratory animal model to enhance our understanding. We aimed to develop such a model using cynomolgus monkeys. EXPERIMENTAL DESIGN Chinese-origin cynomolgus monkeys were dosed intravenously with vehicle or nivolumab 20 mg/kg plus ipilimumab 15 mg/kg once weekly and euthanized on day 29. RESULTS Multiple organ toxicities were observed in cynomolgus monkeys, and were characterized by loose feces, lymphadenopathy, and mononuclear cell infiltrations of varying severity in heart, colon, kidneys, liver, salivary glands, and endocrine organs. Increased proliferation of CD4+ and CD8+ T lymphocytes as well as an increase in activated T cells and central memory T cells in the blood, spleen, and lymph nodes, were observed. Transcriptomic analysis suggested increased migration and activation of T cells and increased phagocytosis and antigen presentation in the heart. Mononuclear cell infiltration in myocardium was comprised primarily of T cells, with lower numbers of macrophages and occasional B cells, and was associated with minimal cardiomyocyte degeneration as well as increases in cardiac troponin-I and NT-pro-BNP. Morphologically, cardiac lesions in our monkey model are similar to the reported ICI myocarditis in humans. CONCLUSIONS We have developed a monkey model characterized by multiple organ toxicities including myocarditis. This model may provide insight into the immune mechanisms and facilitate biomarker identification for ICI-associated irAEs.
Collapse
Affiliation(s)
- Changhua Ji
- Immunotoxicology, DSRD, Pfizer, La Jolla, California.
| | - Marc D Roy
- Investigative Toxicology, DSRD, Pfizer, Groton, Connecticut
| | - Jonathan Golas
- Oncology Targeted Therapeutics Discovery, Pearl River, New York
| | | | - Sripad Ram
- Global Pathology, DSRD, Pfizer, La Jolla, California
| | - Steven W Kumpf
- Investigative Toxicology, DSRD, Pfizer, Groton, Connecticut
| | - Matthew Martin
- Investigative Toxicology, DSRD, Pfizer, Groton, Connecticut
| | | | | | - Andrea T Hooper
- Oncology Targeted Therapeutics Discovery, Pearl River, New York
| | - Puja Sapra
- Oncology Targeted Therapeutics Discovery, Pearl River, New York
| | | | | | | | | |
Collapse
|
400
|
Connolly C, Bambhania K, Naidoo J. Immune-Related Adverse Events: A Case-Based Approach. Front Oncol 2019; 9:530. [PMID: 31293970 PMCID: PMC6598598 DOI: 10.3389/fonc.2019.00530] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 05/31/2019] [Indexed: 12/23/2022] Open
Abstract
Immunotherapy has heralded the advent of a new era in oncology. Immune checkpoint inhibitors (ICIs) enhance anti-tumor immunity, thereby reinvigorating a patient's immune system to fight cancer. While therapy with this class of agents has resulted in improved clinical outcomes for patients with multiple tumor types, a broad spectrum of immune-related adverse events (irAEs) may affect any organ system, with variable clinical presentations. Prompt recognition and management of irAEs are associated with improved irAE outcomes, and represents an important new clinical challenge for practicing oncologists. Herein, we provide a comprehensive case-based review of the most common and clinically-important irAEs, focussing on epidemiology, clinical manifestations, and management. We also examine future strategies that may provide meaningful insights into the prevention and management of irAEs.
Collapse
Affiliation(s)
- Caoilfhionn Connolly
- Department of Internal Medicine, Johns Hopkins Hospital, Baltimore, MD, United States
| | - Kalindi Bambhania
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, United States
| | - Jarushka Naidoo
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins University, Baltimore, MD, United States
- Department of Oncology, Bloomberg-Kimmel Institute for Cancer Immunotherapy, Baltimore, MD, United States
| |
Collapse
|