251
|
Zhang RS, Padegimas A, Murphy KM, Evans PT, Peters CJ, Domenico CM, Vidula MK, Mather PJ, Cevasco M, Cohen RB, Carver JR, O'Quinn RP. Treatment of corticosteroid refractory immune checkpoint inhibitor myocarditis with Infliximab: a case series. CARDIO-ONCOLOGY 2021; 7:13. [PMID: 33785062 PMCID: PMC8008661 DOI: 10.1186/s40959-021-00095-x] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 02/07/2021] [Indexed: 12/22/2022]
Abstract
Background Glucocorticoid treatment remains the cornerstone of therapy for immune checkpoint inhibitor (ICI) myocarditis, but data supporting the use of additional immunotherapy for steroid refractory cases remains limited. We investigate the safety and efficacy of infliximab in patients with ICI myocarditis who are refractory to corticosteroids. Additionally, we highlight the importance of a multi-disciplinary approach in the care for these complex patients. Methods We retrospectively identified consecutive patients who developed ICI myocarditis at our institution between January 2017 and January 2020. Baseline characteristics, laboratory data and clinical outcomes were compared between patients who received infliximab and those who did not. Results Of a total of 11 patients who developed ICI myocarditis, 4 were treated with infliximab. Aside from age, there were no significant differences in baseline patient characteristics between the two groups including total number of ICI doses received and duration from initial ICI dose to onset of symptoms. The time to troponin normalization was 58 vs. 151.5 days (p = 0.25). The duration of prednisone taper was longer in the infliximab group (90 vs. 150 days p = 0.32). All patients survived initial hospital admission. Over a median follow-up period of 287 days, two of the 4 patients died from sepsis 2 and 3 months after initial treatment of their myocarditis; one of these patients was on a steroid taper and the other patient had just completed a steroid taper. Conclusions Infliximab, despite its black box warning in patients with heart failure, may be a safe and effective treatment for ICI myocarditis.
Collapse
Affiliation(s)
- Robert S Zhang
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA
| | - Allison Padegimas
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA.,Division of Cardiovascular Medicine, University of Pennsylvania, PA, Philadelphia, USA
| | - Kathleen M Murphy
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA.,Division of Infectious Disease, University of Pennsylvania, PA, Philadelphia, USA
| | - Peter T Evans
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA
| | - Carli J Peters
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA
| | | | - Mahesh K Vidula
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA.,Division of Cardiovascular Medicine, University of Pennsylvania, PA, Philadelphia, USA
| | - Paul J Mather
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA.,Division of Cardiovascular Medicine, University of Pennsylvania, PA, Philadelphia, USA
| | - Marisa Cevasco
- Department of Cardiothoracic Surgery, University of Pennsylvania, PA, Philadelphia, USA
| | - Roger B Cohen
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA.,Division of Hematology-Oncology, University of Pennsylvania, PA, Philadelphia, USA
| | - Joseph R Carver
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA.,Division of Cardiovascular Medicine, University of Pennsylvania, PA, Philadelphia, USA.,Division of Hematology-Oncology, University of Pennsylvania, PA, Philadelphia, USA
| | - Rupal P O'Quinn
- Department of Medicine, University of Pennsylvania, PA, Philadelphia, USA. rupal.o'.,Division of Cardiovascular Medicine, University of Pennsylvania, PA, Philadelphia, USA. rupal.o'
| |
Collapse
|
252
|
Baik AH, Tsai KK, Oh DY, Aras MA. Mechanisms and clinical manifestations of cardiovascular toxicities associated with immune checkpoint inhibitors. Clin Sci (Lond) 2021; 135:703-724. [PMID: 33686402 PMCID: PMC8647663 DOI: 10.1042/cs20200331] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 02/22/2021] [Accepted: 02/24/2021] [Indexed: 12/17/2022]
Abstract
Immunotherapies have greatly expanded the armamentarium of cancer-directed therapies in the past decade, allowing the immune system to recognize and fight cancer. Immune checkpoint inhibitors (ICIs), in particular, have revolutionized cancer treatment and have demonstrated survival benefit in numerous types of cancer. These monoclonal antibodies increase anti-cancer immunity by blocking down-regulators of adaptive immunity, including cytotoxic T lymphocyte-associated protein 4 (CTLA-4), programmed cell death protein 1 (PD-1), and its ligand (PD-L1), resulting in anti-tumor activity. As ICIs increase immune system activation, they can cause a wide range of inflammatory side effects, termed immune-released adverse events. Though these toxicities can affect nearly any organ, the most fatal toxicity is myocarditis. Here, we discuss the diverse spectrum of cardiovascular toxicities associated with ICI use. In addition, we provide insight and future directions on mechanisms and treatments for immune-related adverse events (irAEs) involving the myocardium, pericardium, vasculature, and conduction system.
Collapse
Affiliation(s)
- Alan H. Baik
- UCSF Department of Medicine, Division of Cardiology, San Francisco, CA 94143
| | - Katy K. Tsai
- UCSF Department of Medicine, Division of Hematology/Oncology, San Francisco, CA 94143
| | - David Y. Oh
- UCSF Department of Medicine, Division of Hematology/Oncology, San Francisco, CA 94143
| | - Mandar A. Aras
- UCSF Department of Medicine, Division of Cardiology, San Francisco, CA 94143
| |
Collapse
|
253
|
Combination of echocardiography and emergency endomyocardial biopsy for suspected myocarditis in the cardiovascular emergency medical care. J Echocardiogr 2021; 19:86-94. [PMID: 33661474 DOI: 10.1007/s12574-021-00521-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 02/10/2021] [Accepted: 02/22/2021] [Indexed: 10/22/2022]
Abstract
Myocarditis is a fatal inflammatory disease of myocardium, diagnosed with clinical and histopathological findings by endomyocardial biopsy (EMB). Myocarditis has a variety of clinical presentations and a dynamic and sometimes rapid process of severity. Echocardiography plays an important role in the management of myocarditis because it has noninvasiveness and portability. Once acute myocarditis is suspected by an echocardiography, pathological information should be required as early as possible. In our cardiovascular center, emergency EMB suspecting myocarditis was performed in 19 cases (1.3%) among consecutive 1469 cases (70.1 ± 12.6 years old, male 67.5%) undergoing emergency coronary angiograms from April 2014 to September 2017. Hematoxylin-eosin stain of the biopsy specimens were prepared with microwave-accelerated histoprocessing within 3-5 hours after EMB for rapid pathological diagnosis of myocarditis. We reviewed the value of emergency echo-EMB combination leading to the early decision making of intensive care, corticosteroids and proper mechanical circulatory support prior to the possible sudden collapse in patients with myocarditis.
Collapse
|
254
|
Zhang L, Reynolds KL, Lyon AR, Palaskas N, Neilan TG. The Evolving Immunotherapy Landscape and the Epidemiology, Diagnosis, and Management of Cardiotoxicity: JACC: CardioOncology Primer. JACC CardioOncol 2021; 3:35-47. [PMID: 33842895 PMCID: PMC8034586 DOI: 10.1016/j.jaccao.2020.11.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 11/08/2020] [Accepted: 11/11/2020] [Indexed: 02/07/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are newer therapies being applied to an increasing number of patients with cancer. Data suggest that up to 36% of cancer patients may be eligible for immunotherapy and, in late 2019, there were more than 3,362 clinical trials initiated to evaluate the effectiveness of immunotherapy, either as single agents or in combination with other immunotherapy, targeted therapies, or traditional cytotoxic or radiation therapy. With the combination of both immune and non-immune treatment approaches, the complexity in making the diagnosis of cardiotoxicity related to an ICI will increase substantially. Here, we summarize the published data on the epidemiology, diagnosis, and management of cardiotoxicity of ICIs. This is a rapidly evolving field, and as our understanding continues to evolve, previously considered hypotheses may not prove to be entirely correct. Research and continued collaborations are urgently needed to provide evidence-based cardiovascular care for this rapidly expanding and vulnerable cohort of patients. (J Am Coll Cardiol CardioOnc 2021;3:35-47) © 2021 The Authors. Published by Elsevier on behalf of the American College of Cardiology Foundation. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).
Collapse
Affiliation(s)
- Lili Zhang
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, New York, USA
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Massachusetts, USA
| | - Alexander R. Lyon
- Cardio-Oncology Program, Royal Brompton Hospital, London, United Kingdom
- Imperial College London, London, United Kingdom
| | - Nicolas Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center (CIRC), Department of Radiology and Division of Cardiology, Massachusetts General Hospital, Boston, Massachusetts, USA
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
255
|
Munn LL, Stylianopoulos T, Jain NK, Hardin CC, Khandekar MJ, Jain RK. Vascular Normalization to Improve Treatment of COVID-19: Lessons from Treatment of Cancer. Clin Cancer Res 2021; 27:2706-2711. [PMID: 33648989 DOI: 10.1158/1078-0432.ccr-20-4750] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/17/2021] [Accepted: 02/25/2021] [Indexed: 12/21/2022]
Abstract
The dramatic impact of the COVID-19 pandemic has resulted in an "all hands on deck" approach to find new therapies to improve outcomes in this disease. In addition to causing significant respiratory pathology, infection with SARS-CoV-2 (like infection with other respiratory viruses) directly or indirectly results in abnormal vasculature, which may contribute to hypoxemia. These vascular effects cause significant morbidity and may contribute to mortality from the disease. Given that abnormal vasculature and poor oxygenation are also hallmarks of solid tumors, lessons from the treatment of cancer may help identify drugs that can be repurposed to treat COVID-19. Although the mechanisms that result in vascular abnormalities in COVID-19 are not fully understood, it is possible that there is dysregulation of many of the same angiogenic and thrombotic pathways as seen in patients with cancer. Many anticancer therapeutics, including androgen deprivation therapy (ADT) and immune checkpoint blockers (ICB), result in vascular normalization in addition to their direct effects on tumor cells. Therefore, these therapies, which have been extensively explored in clinical trials of patients with cancer, may have beneficial effects on the vasculature of patients with COVID-19. Furthermore, these drugs may have additional effects on the disease course, as some ADTs may impact viral entry, and ICBs may accelerate T-cell-mediated viral clearance. These insights from the treatment of cancer may be leveraged to abrogate the vascular pathologies found in COVID-19 and other forms of hypoxemic respiratory failure.
Collapse
Affiliation(s)
- Lance L Munn
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Natalie K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - C Corey Hardin
- Department of Pulmonary and Critical Care Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Melin J Khandekar
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
256
|
Wei SC, Meijers WC, Axelrod ML, Anang NAAS, Screever EM, Wescott EC, Johnson DB, Whitley E, Lehmann L, Courand PY, Mancuso JJ, Himmel LE, Lebrun-Vignes B, Wleklinski MJ, Knollmann BC, Srinivasan J, Li Y, Atolagbe OT, Rao X, Zhao Y, Wang J, Ehrlich LIR, Sharma P, Salem JE, Balko JM, Moslehi JJ, Allison JP. A Genetic Mouse Model Recapitulates Immune Checkpoint Inhibitor-Associated Myocarditis and Supports a Mechanism-Based Therapeutic Intervention. Cancer Discov 2021; 11:614-625. [PMID: 33257470 PMCID: PMC8041233 DOI: 10.1158/2159-8290.cd-20-0856] [Citation(s) in RCA: 175] [Impact Index Per Article: 43.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 10/08/2020] [Accepted: 11/23/2020] [Indexed: 11/16/2022]
Abstract
Immune checkpoint inhibitors (ICI) targeting CTLA4 or PD-1/PD-L1 have transformed cancer therapy but are associated with immune-related adverse events, including myocarditis. Here, we report a robust preclinical mouse model of ICI-associated myocarditis in which monoallelic loss of Ctla4 in the context of complete genetic absence of Pdcd1 leads to premature death in approximately half of mice. Premature death results from myocardial infiltration by T cells and macrophages and severe ECG abnormalities, closely recapitulating the clinical and pathologic hallmarks of ICI-associated myocarditis observed in patients. Using this model, we show that Ctla4 and Pdcd1 functionally interact in a gene dosage-dependent manner, providing a mechanism by which myocarditis arises with increased frequency in the setting of combination ICI therapy. We demonstrate that intervention with CTLA4-Ig (abatacept) is sufficient to ameliorate disease progression and additionally provide a case series of patients in which abatacept mitigates the fulminant course of ICI myocarditis. SIGNIFICANCE: We provide a preclinical model of ICI-associated myocarditis which recapitulates this clinical syndrome. Using this model, we demonstrate that CTLA4 and PD-1 (ICI targets) functionally interact for myocarditis development and that intervention with CTLA4-Ig (abatacept) attenuates myocarditis, providing mechanistic rationale and preclinical support for therapeutic clinical studies.See related commentary by Young and Bluestone, p. 537.This article is highlighted in the In This Issue feature, p. 521.
Collapse
Affiliation(s)
- Spencer C Wei
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
| | - Wouter C Meijers
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Margaret L Axelrod
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Nana-Ama A S Anang
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Elles M Screever
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elizabeth C Wescott
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Douglas B Johnson
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Elizabeth Whitley
- Department of Veterinary Medicine and Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lorenz Lehmann
- Department of Cardiology, University Hospital of Heidelberg, Heidelberg, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Heidelberg/Mannheim, German Research Center (DKFZ), Heidelberg, Germany
| | - Pierre-Yves Courand
- Hospices Civils de Lyon, Service de cardiologie, IMMUCARE, Hôpital de la Croix-Rousse et Hôpital Lyon Sud, Lyon, France; Université de Lyon, CREATIS UMR INSERM U1044, INSA, Lyon France
| | - James J Mancuso
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren E Himmel
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Benedicte Lebrun-Vignes
- Department of Pharmacology, APHP. Sorbonne Université, INSERM, CIC-1901, UNICO-GRECO Cardiooncology Program, Paris, France
| | - Matthew J Wleklinski
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Bjorn C Knollmann
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Jayashree Srinivasan
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas
| | - Yu Li
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas
| | | | - Xiayu Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Yang Zhao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Jing Wang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Lauren I R Ehrlich
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, Texas
- Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, Texas
| | - Padmanee Sharma
- Department of Genitourinary Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Joe-Elie Salem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, APHP. Sorbonne Université, INSERM, CIC-1901, UNICO-GRECO Cardiooncology Program, Paris, France
| | - Justin M Balko
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Javid J Moslehi
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| | - James P Allison
- Department of Immunology, The University of Texas MD Anderson Cancer Center, Houston, Texas.
- Parker Institute for Cancer Immunotherapy, The University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
257
|
Lee C, Maan A, Singh JP, Fradley MG. Arrhythmias and device therapies in patients with cancer therapy-induced cardiomyopathy. Heart Rhythm 2021; 18:1223-1229. [PMID: 33640446 DOI: 10.1016/j.hrthm.2021.02.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 02/08/2021] [Accepted: 02/22/2021] [Indexed: 11/30/2022]
Abstract
Our knowledge of associated cardiotoxicities from novel therapeutics in oncology continues to expand. These include arrhythmias from cancer-therapy induced cardiomyopathy resulting from both direct and indirect effects on cardiomyocytes and other mechanisms that can adversely impact cardiovascular outcomes and overall mortality. In this review, we focus on both the arrhythmias of various classes of oncologic agents as well as the use of cardiac implantable electronic devices (cardioverter-defibrillators, permanent pacemakers, and cardiac resynchronization therapy) in cardio-oncology patients.
Collapse
Affiliation(s)
- Charlotte Lee
- Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Abhishek Maan
- Department of Cardiac Electrophysiology, The Mount Sinai Hospital, New York, New York
| | - Jagmeet P Singh
- Cardiac Arrhythmia Service, Massachusetts General Hospital, Boston, Massachusetts
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
258
|
Bermas BL, Zaha VG. Mending Broken Hearts: A New Treatment Paradigm for Immune Checkpoint Inhibitor-Induced Myocarditis. Circulation 2021; 143:767-769. [PMID: 33617309 DOI: 10.1161/circulationaha.120.052307] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Bonnie L Bermas
- Rheumatic Diseases Division (B.L.B.), University of Texas Southwestern Medical Center, Dallas.,Parkland Health and Hospital System, Dallas, TX (B.L.B., V.G.Z.)
| | - Vlad G Zaha
- Cardiology Division (V.G.Z.), University of Texas Southwestern Medical Center, Dallas.,Department of Internal Medicine, Cardio-Oncology Program, Harold C. Simmons Comprehensive Cancer Center (V.G.Z.), University of Texas Southwestern Medical Center, Dallas.,Advanced Imaging Research Center (V.G.Z.), University of Texas Southwestern Medical Center, Dallas.,Parkland Health and Hospital System, Dallas, TX (B.L.B., V.G.Z.)
| |
Collapse
|
259
|
Xu S, Sharma UC, Tuttle C, Pokharel S. Immune Checkpoint Inhibitors: Cardiotoxicity in Pre-clinical Models and Clinical Studies. Front Cardiovasc Med 2021; 8:619650. [PMID: 33614750 PMCID: PMC7887301 DOI: 10.3389/fcvm.2021.619650] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 01/06/2021] [Indexed: 12/12/2022] Open
Abstract
Since the approval of the first immune checkpoint inhibitor (ICI) 9 years ago, ICI-therapy have revolutionized cancer treatment. Lately, antibodies blocking the interaction of programmed cell death protein (PD-1) and ligand (PD-L1) are gaining momentum as a cancer treatment, with multiple agents and cancer types being recently approved for treatment by the US Food and Drug Administration (FDA). Unfortunately, immunotherapy often leads to a wide range of immune related adverse events (IRAEs), including several severe cardiac effects and most notably myocarditis. While increased attention has been drawn to these side effects, including publication of multiple clinical observational data, the underlying mechanisms are unknown. In the event of IRAEs, the most widely utilized clinical solution is administration of high dose corticosteroids and in severe cases, discontinuation of these ICIs. This is detrimental as these therapies are often the last line of treatment options for many types of advanced cancer. In this review, we have systematically described the pathophysiology of the PD-1/PD-L1 axis (including a historical perspective) and cardiac effects in pre-clinical models, clinical trials, autoimmune mechanisms, and immunotherapy in combination with other cancer treatments. We have also reviewed the current challenges in the diagnosis of cardiac events and future directions in the field. In conclusion, this review will delve into this expanding field of cancer immunotherapy and the emerging adverse effects that should be quickly detected and prevented.
Collapse
Affiliation(s)
- Shirley Xu
- Division of Thoracic Pathology and Oncology, Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States.,Department of Medicine, Jacob's School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Umesh C Sharma
- Department of Medicine, Jacob's School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| | - Cheyanna Tuttle
- Division of Thoracic Pathology and Oncology, Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| | - Saraswati Pokharel
- Division of Thoracic Pathology and Oncology, Department of Pathology, Roswell Park Comprehensive Cancer Center, Buffalo, NY, United States
| |
Collapse
|
260
|
Affiliation(s)
- Siqi Li
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Japan
| | - Kazuko Tajiri
- Department of Cardiology, Faculty of Medicine, University of Tsukuba, Japan
| |
Collapse
|
261
|
Chen DH, Tyebally S, Malloupas M, Roylance R, Spurrell E, Raja F, Ghosh AK. Cardiovascular Disease Amongst Women Treated for Breast Cancer: Traditional Cytotoxic Chemotherapy, Targeted Therapy, and Radiation Therapy. Curr Cardiol Rep 2021; 23:16. [PMID: 33501515 DOI: 10.1007/s11886-021-01446-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 12/13/2022]
Abstract
PURPOSE OF REVIEW Cardiotoxicity can occur acutely during breast cancer treatment and impact the potential for the intended cancer treatment regime to be completed, or as a late effect affecting cancer survivorship. Indeed, the most common cause of mortality in females with early breast cancer is cardiovascular disease, especially in those over the age of 65. Optimal cancer care therefore needs to be delivered without jeopardising cardiovascular health. Understanding the different cardiotoxicities associated with breast cancer treatment is vital to this approach, and therefore, this article seeks to provide an overview of this. RECENT FINDINGS Tyrosine kinase inhibitors targeting human epidermal growth factor receptor (HER)-2, immune checkpoint inhibitors (ICI), and cyclin-dependent kinase (CDK) inhibitors are new targeted breast cancer treatments. In particular, ICI are associated with myocarditis that carries a significant mortality, whilst the CDK inhibitor ribociclib causes QT prolongation that requires cardiac surveillance and appropriate dose adjustment to prevent ventricular arrhythmias. The need has always been for strategies to mitigate the risks of cardiovascular toxicities, and new data is promising for the use of dexrazoxane in anthracyclines, and the role of beta blockers and angiotensin converting enzymes inhibitors in anthracyclines and HER-2 monoclonal antibodies such as trastuzumab. Significant headways in breast cancer treatment have resulted in reductions in disease recurrence and mortality, but cardiovascular complications continue to impact the ability to deliver some of these cancer treatments, and the period of cancer survivorship.
Collapse
Affiliation(s)
- Daniel H Chen
- Cardio-Oncology Service, Bart's Heart Centre, St Bartholomew's Hospital, London, EC1A 7BE, UK.,University College London Hospital, 235 Euston Road, London, NW1 2BU, UK.,The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Sara Tyebally
- Cardio-Oncology Service, Bart's Heart Centre, St Bartholomew's Hospital, London, EC1A 7BE, UK.,University College London Hospital, 235 Euston Road, London, NW1 2BU, UK
| | - Michael Malloupas
- University College London Hospital, 235 Euston Road, London, NW1 2BU, UK.,The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK
| | - Rebecca Roylance
- University College London Hospital, 235 Euston Road, London, NW1 2BU, UK.,NIHR University College London Hospitals Biomedical Research Centre, Maple House 1st Floor, 149 Tottenham Court Road, London, W1T 7DN, UK
| | - Emma Spurrell
- University College London Hospital, 235 Euston Road, London, NW1 2BU, UK.,Whittington Health, Magdala Avenue, London, N19 5NF, UK
| | - Fharat Raja
- University College London Hospital, 235 Euston Road, London, NW1 2BU, UK.,North Middlesex University Hospital, Sterling Way, London, N18 1QX, UK
| | - Arjun K Ghosh
- Cardio-Oncology Service, Bart's Heart Centre, St Bartholomew's Hospital, London, EC1A 7BE, UK. .,University College London Hospital, 235 Euston Road, London, NW1 2BU, UK. .,The Hatter Cardiovascular Institute, 67 Chenies Mews, London, WC1E 6HX, UK.
| |
Collapse
|
262
|
Immune Checkpoint Inhibitor (ICI)-Related Cardiotoxicity. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1342:377-387. [DOI: 10.1007/978-3-030-79308-1_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
263
|
Kumar M, Thangavel C, Becker RC, Sadayappan S. Monoclonal Antibody-Based Immunotherapy and Its Role in the Development of Cardiac Toxicity. Cancers (Basel) 2020; 13:E86. [PMID: 33396766 PMCID: PMC7795565 DOI: 10.3390/cancers13010086] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/22/2020] [Accepted: 12/26/2020] [Indexed: 12/14/2022] Open
Abstract
Immunotherapy is one of the most effective therapeutic options for cancer patients. Five specific classes of immunotherapies, which includes cell-based chimeric antigenic receptor T-cells, checkpoint inhibitors, cancer vaccines, antibody-based targeted therapies, and oncolytic viruses. Immunotherapies can improve survival rates among cancer patients. At the same time, however, they can cause inflammation and promote adverse cardiac immune modulation and cardiac failure among some cancer patients as late as five to ten years following immunotherapy. In this review, we discuss cardiotoxicity associated with immunotherapy. We also propose using human-induced pluripotent stem cell-derived cardiomyocytes/ cardiac-stromal progenitor cells and cardiac organoid cultures as innovative experimental model systems to (1) mimic clinical treatment, resulting in reproducible data, and (2) promote the identification of immunotherapy-induced biomarkers of both early and late cardiotoxicity. Finally, we introduce the integration of omics-derived high-volume data and cardiac biology as a pathway toward the discovery of new and efficient non-toxic immunotherapy.
Collapse
Affiliation(s)
- Mohit Kumar
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA; (R.C.B.); (S.S.)
| | - Chellappagounder Thangavel
- Department of Radiation Oncology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA;
- Department of Dermatology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Richard C. Becker
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA; (R.C.B.); (S.S.)
| | - Sakthivel Sadayappan
- Heart, Lung and Vascular Institute, Department of Internal Medicine, Division of Cardiovascular Health and Disease, University of Cincinnati, Cincinnati, OH 45267, USA; (R.C.B.); (S.S.)
| |
Collapse
|
264
|
Shroff GS, Shroff S, Ahuja J, Truong MT, Vlahos I. Imaging spectrum of adverse events of immune checkpoint inhibitors. Clin Radiol 2020; 76:262-272. [PMID: 33375984 DOI: 10.1016/j.crad.2020.11.117] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 11/20/2020] [Indexed: 12/16/2022]
Abstract
Immune checkpoint inhibitors (ICIs), a form of immunotherapy, are increasingly used for a variety of malignancies and have been linked to numerous treatment-related side effects known as immune-related adverse events (irAEs). IrAEs can affect multiple organ systems and are important to recognise in order to avoid misinterpretation as progressive tumour and to ensure appropriate management. In this pictorial review, we will briefly discuss radiological response criteria of immunotherapy and describe the imaging appearances of the wide spectrum of these ICI-associated toxicities.
Collapse
Affiliation(s)
- G S Shroff
- MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 1478, Houston, TX, 77030, USA.
| | - S Shroff
- Houston Methodist, 6560 Fannin St. Ste 802, Houston, TX, 77030, USA
| | - J Ahuja
- MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 1478, Houston, TX, 77030, USA
| | - M T Truong
- MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 1478, Houston, TX, 77030, USA
| | - I Vlahos
- MD Anderson Cancer Center, 1515 Holcombe Blvd Unit 1478, Houston, TX, 77030, USA
| |
Collapse
|
265
|
Ruddy KJ, Patel SR, Higgins AS, Armenian SH, Herrmann J. Cardiovascular Health during and after Cancer Therapy. Cancers (Basel) 2020; 12:E3737. [PMID: 33322622 PMCID: PMC7763346 DOI: 10.3390/cancers12123737] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 01/22/2023] Open
Abstract
Certain cancer treatments have been linked to specific cardiovascular toxicities, including (but not limited to) cardiomyopathy, atrial fibrillation, arterial hypertension, and myocarditis. Radiation, anthracyclines, human epidermal growth factor receptor 2 (Her2)-directed therapies, fluoropyrimidines, platinums, tyrosine kinase inhibitors and proteasome inhibitors, immune checkpoint inhibitors, and chimeric antigen-presenting (CAR)-T cell therapy can all cause cardiovascular side effects. Management of cardiovascular dysfunction that occurs during cancer therapy often requires temporary or permanent cessation of the risk-potentiating anti-neoplastic drug as well as optimization of medical management from a cardiovascular standpoint. Stem cell or bone marrow transplant recipients face unique cardiovascular challenges, as do patients at extremes of age.
Collapse
Affiliation(s)
| | - Shruti R. Patel
- Department of Internal Medicine, Mayo Clinic, Rochester, MN 55905, USA;
| | | | - Saro H. Armenian
- Department of Population Sciences, City of Hope, Duarte, CA 91010, USA;
| | - Joerg Herrmann
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, MN 55905, USA;
| |
Collapse
|
266
|
Sessums M, Yarrarapu S, Guru PK, Sanghavi DK. Atezolizumab-induced myositis and myocarditis in a patient with metastatic urothelial carcinoma. BMJ Case Rep 2020; 13:13/12/e236357. [PMID: 33298477 DOI: 10.1136/bcr-2020-236357] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Immune checkpoint inhibitors have revolutionised cancer therapy in the past decade. Although they have been indicated to treat a diverse range of malignant neoplasms, they are also associated with various immune-related adverse effects. We report the case of a 74-year-old man with a history of urothelial carcinoma who had atezolizumab-induced myocarditis and myositis resulting in acute hypercapnic respiratory failure, despite the discontinuation of atezolizumab and aggressive treatment with corticosteroids. This case highlights the importance of a multidisciplinary approach for early diagnosis and treatment of immune-related adverse events. Physicians must be aware of the risks associated with immune checkpoint inhibitors and have a basic knowledge regarding their management.
Collapse
Affiliation(s)
- Mary Sessums
- Internal Medicine, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Siva Yarrarapu
- Critical Care Medicine, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Pramod K Guru
- Critical Care Medicine, Mayo Clinic Florida, Jacksonville, Florida, USA
| | - Devang K Sanghavi
- Critical Care Medicine, Mayo Clinic Florida, Jacksonville, Florida, USA
| |
Collapse
|
267
|
Broberg AM, Geisler J, Tuohinen S, Skytta T, Hrafnkelsdóttir ÞJ, Nielsen KM, Hedayati E, Omland T, Offersen BV, Lyon AR, Gulati G. Prevention, Detection, and Management of Heart Failure in Patients Treated for Breast Cancer. Curr Heart Fail Rep 2020; 17:397-408. [PMID: 32979150 PMCID: PMC7683437 DOI: 10.1007/s11897-020-00486-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Long-term survival has increased significantly in breast cancer patients, and cardiovascular side effects are surpassing cancer-related mortality. We summarize risk factors, prevention strategies, detection, and management of cardiotoxicity, with focus on left ventricular dysfunction and heart failure, during breast cancer treatment. RECENT FINDINGS Baseline treatment of cardiovascular risk factors is recommended. Anthracycline and trastuzumab treatment constitute a substantial risk of developing cardiotoxicity. There is growing evidence that this can be treated with beta blockers and angiotensin antagonists. Early detection of cardiotoxicity with cardiac imaging and circulating cardiovascular biomarkers is currently evaluated in clinical trials. Chest wall irradiation accelerates atherosclerotic processes and induces fibrosis. Immune checkpoint inhibitors require consideration for surveillance due to a small risk of severe myocarditis. Cyclin-dependent kinases4/6 inhibitors, cyclophosphamide, taxanes, tyrosine kinase inhibitors, and endocrine therapy have a lower-risk profile for cardiotoxicity. Preventive and management strategies to counteract cancer treatment-related left ventricular dysfunction or heart failure in breast cancer patients should include a comprehensive cardiovascular risk assessment and individual clinical evaluation. This should include both patient and treatment-related factors. Further clinical trials especially on early detection, cardioprevention, and management are urgently needed.
Collapse
Affiliation(s)
- Agneta Månsson Broberg
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog & Institute of Clinical Medicine, University of Oslo, Campus AHUS, Lørenskog, Norway
| | - Suvi Tuohinen
- Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Tanja Skytta
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | - Þórdís Jóna Hrafnkelsdóttir
- Department of Cardiology, Landspitali University Hospital, Reykjavík, Iceland and Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | - Elham Hedayati
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Breast Cancer, Sarcoma and Endocrine Tumors, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte V. Offersen
- Department of Experimental Clinical Oncology & Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Alexander R. Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and Imperial College London, London, UK
| | - Geeta Gulati
- Department of Cardiology, Oslo University Hospital, Postbox 4950, Ullevål, Nydalen, 0424 Oslo, Norway
- Department of Research, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
268
|
Nso N, Antwi-Amoabeng D, Beutler BD, Ulanja MB, Ghuman J, Hanfy A, Nimo-Boampong J, Atanga S, Doshi R, Enoru S, Gullapalli N. Cardiac adverse events of immune checkpoint inhibitors in oncology patients: A systematic review and meta-analysis. World J Cardiol 2020; 12:584-598. [PMID: 33312443 PMCID: PMC7701899 DOI: 10.4330/wjc.v12.i11.584] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/12/2020] [Accepted: 11/10/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are novel therapeutic agents used for various types of cancer. ICIs have revolutionized cancer treatment and improved clinical outcomes among cancer patients. However, immune-related adverse effects of ICI therapy are common. Cardiovascular immune-related adverse events (irAEs) are rare but potentially life-threatening complications.
AIM To estimate the incidence of cardiovascular irAEs among patients undergoing ICI therapy for various malignancies.
METHODS We conducted this systematic review and meta-analysis by searching PubMed, Cochrane CENTRAL, Web of Science, and SCOPUS databases for relevant interventional trials reporting cardiovascular irAEs. We performed a single-arm meta-analysis using OpenMeta [Analyst] software of the following outcomes: Myocarditis, pericardial effusion, heart failure, cardiomyopathy, atrial fibrillation, myocardial infarction, and cardiac arrest. We assessed the heterogeneity using the I2 test and managed to solve it with Cochrane’s leave-one-out method. The risk of bias was performed with the Cochrane’s risk of bias tool.
RESULTS A total of 26 studies were included. The incidence of irAEs follows: Myocarditis: 0.5% [95% confidence interval (CI): 0.1%-0.9%]; Pericardial effusion: 0.5% (95%CI: 0.1%-1.0%); Heart failure: 0.3% (95%CI: 0.0%-0.5%); Cardiomyopathy: 0.3% (95%CI: -0.1%-0.6%); atrial fibrillation: 4.6% (95%CI: 1.0%-14.1%); Myocardial infarction: 0.4% (95%CI: 0.0%-0.7%); and Cardiac arrest: 0.4% (95%CI: 0.1%-0.8%).
CONCLUSION The most common cardiovascular irAEs were atrial fibrillation, myocarditis, and pericardial effusion. Although rare, data from post market surveillance will provide estimates of the long-term prevalence and prognosis in patients with ICI-associated cardiovascular complications.
Collapse
Affiliation(s)
- Nso Nso
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Queens, NY 10029, United States
| | - Daniel Antwi-Amoabeng
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Bryce D Beutler
- Department of Radiology, University of Southern California, Keck School of Medicine, Los Angeles, CA 90033, United States
| | - Mark B Ulanja
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Jasmine Ghuman
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Ahmed Hanfy
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Joyce Nimo-Boampong
- Department of Internal Medicine, Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Sirri Atanga
- Department of Medicine, United Health Services Wilson Medical Center, Johnson City, NY 13790, United States
| | - Rajkumar Doshi
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| | - Sostanie Enoru
- Department of Cardiovascular Disease, SUNY Downstate Health Science University, Brooklyn, NY 11203, United States
| | - Nageshwara Gullapalli
- Department of Internal Medicine, University of Nevada, Reno School of Medicine, Reno, NV 89502, United States
| |
Collapse
|
269
|
Quagliariello V, Bonelli A, Caronna A, Conforti G, Iovine M, Carbone A, Berretta M, Botti G, Maurea N. SARS-CoV-2 Infection and Cardioncology: From Cardiometabolic Risk Factors to Outcomes in Cancer Patients. Cancers (Basel) 2020; 12:E3316. [PMID: 33182653 PMCID: PMC7697868 DOI: 10.3390/cancers12113316] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 11/05/2020] [Accepted: 11/08/2020] [Indexed: 02/07/2023] Open
Abstract
The coronavirus disease-2019 (COVID-19) is a highly transmissible viral illness caused by SARS-CoV-2, which has been defined by the World Health Organization as a pandemic, considering its remarkable transmission speed worldwide. SARS-CoV-2 interacts with angiotensin-converting enzyme 2 and TMPRSS2, which is a serine protease both expressed in lungs, the gastro-intestinal tract, and cardiac myocytes. Patients with COVID-19 experienced adverse cardiac events (hypertension, venous thromboembolism, arrhythmia, myocardial injury, fulminant myocarditis), and patients with previous cardiovascular disease have a higher risk of death. Cancer patients are extremely vulnerable with a high risk of viral infection and more negative prognosis than healthy people, and the magnitude of effects depends on the type of cancer, recent chemotherapy, radiotherapy, or surgery and other concomitant comorbidities (diabetes, cardiovascular diseases, metabolic syndrome). Patients with active cancer or those treated with cardiotoxic therapies may have heart damages exacerbated by SARS-CoV-2 infection than non-cancer patients. We highlight the cardiovascular side effects of COVID-19 focusing on the main outcomes in cancer patients in updated perspective and retrospective studies. We focus on the main cardio-metabolic risk factors in non-cancer and cancer patients and provide recommendations aimed to reduce cardiovascular events, morbidity, and mortality.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (G.C.); (M.I.); (A.C.)
| | - Annamaria Bonelli
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (G.C.); (M.I.); (A.C.)
| | - Antonietta Caronna
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (G.C.); (M.I.); (A.C.)
| | - Gabriele Conforti
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (G.C.); (M.I.); (A.C.)
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (G.C.); (M.I.); (A.C.)
| | - Andreina Carbone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (G.C.); (M.I.); (A.C.)
| | - Massimiliano Berretta
- Department of Medical Oncology-Centro di Riferimento Oncologico di Aviano (CRO), IRCCS, 33081 Aviano, Italy;
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (A.B.); (A.C.); (G.C.); (M.I.); (A.C.)
| |
Collapse
|
270
|
Xia W, Chen H, Chen D, Ye Y, Xie C, Hou M. PD-1 inhibitor inducing exosomal miR-34a-5p expression mediates the cross talk between cardiomyocyte and macrophage in immune checkpoint inhibitor-related cardiac dysfunction. J Immunother Cancer 2020; 8:jitc-2020-001293. [PMID: 33115945 PMCID: PMC7594538 DOI: 10.1136/jitc-2020-001293] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2020] [Indexed: 12/30/2022] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have been an important therapeutic advancement in the field of cancer medicine. Recent reports provided greater insights into the cardiovascular adverse events, which prohibited the use of ICIs. Cardiovascular adverse events occur in different forms, such as myocarditis and cardiomyopathy, myocardial fibrosis, heart failure and pericardial disease. Cardiac aging overlapped with the occurrence of some cardiac diseases. Exosomes mediate cell–cell cross talk in cardiac diseases by transferring a variety of biomolecules, including microRNAs (miRs). miR-34a-5p is a well-known miR associated with the cardiac senescence. This study aimed to investigate whether cardiovascular adverse effects of the programmed cell death 1 (PD-1) inhibitor, a widely used ICI, were related to exosomal-transferred miR-34a-5p in cardiac senescence in a mouse model. Methods and results The upregulation of miR-34a-5p in cardiomyocytes induced by exosomes derived from PD-1 inhibitor–treated macrophages, accompanied by cardiac senescence, caused cardiac injury in mouse hearts. miR-34a-5p was identified as an exosomal transfer RNA to induce cardiac senescence–related injury. Inhibiting miR-34a-5p in macrophages attenuated the exosomePD-1 inhibitor-induced pro-senescent effect in cardiomyocytes. TargetScan and luciferase assay showed that miR-34a-5p targeted the serine/threonine-protein phosphatase 1 regulatory subunit 10 (PNUTS) 3′-untranslated region. Conclusions Exosomes derived from PD-1 inhibitor–treated macrophages exerted a pro-senescent effect by modulating the miR-34a-5p/PNUTS signaling pathway. The findings might supply new targets to ameliorate cardiac injury in patients with cancer receiving PD-1 inhibitor treatment.
Collapse
Affiliation(s)
- Wenzheng Xia
- Neurosurgery, Wenzhou Medical University First Affiliated Hospital, Wenzhou, Zhejiang, China.,Neurosurgery, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hanbin Chen
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Didi Chen
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yijia Ye
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Congying Xie
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Meng Hou
- Radiation Oncology, First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
271
|
Castrillon JA, Eng C, Cheng F. Pharmacogenomics for immunotherapy and immune-related cardiotoxicity. Hum Mol Genet 2020; 29:R186-R196. [PMID: 32620943 PMCID: PMC7574958 DOI: 10.1093/hmg/ddaa137] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 06/25/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022] Open
Abstract
Immune checkpoint blockade (ICB) has become a standard of care in a subset of solid tumors. Although cancer survivorship has extended, rates of durable response of ICB remain poor; furthermore, cardiac adverse effects are emerging, which impact several mechanical aspects of the heart. Cardio-oncology programs implement a clinical assessment to curtail cardiovascular disease progression but are limited to the current clinical parameters used in cardiology. Pharmacogenomics provides the potential to unveil heritable and somatic genetic variations for guiding precision immunotherapy treatment to reduce the risk of immune-related cardiotoxicity. A better understanding of pharmacogenomics will optimize the current treatment selection and dosing of immunotherapy. Here, we summarize the recent pharmacogenomics studies in immunotherapy responsiveness and its related cardiotoxicity and highlight how patient genetics and epigenetics can facilitate researchers and clinicians in designing new approaches for precision immunotherapy. We highlight and discuss how single-cell technologies, human-induced pluripotent stem cells and systems pharmacogenomics accelerate future studies of precision cardio-oncology.
Collapse
Affiliation(s)
- Jessica A Castrillon
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
| | - Charis Eng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
- Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Genetics and Genome Sciences, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH 44195, USA
- Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| |
Collapse
|
272
|
Quagliariello V, Passariello M, Rea D, Barbieri A, Iovine M, Bonelli A, Caronna A, Botti G, De Lorenzo C, Maurea N. Evidences of CTLA-4 and PD-1 Blocking Agents-Induced Cardiotoxicity in Cellular and Preclinical Models. J Pers Med 2020; 10:jpm10040179. [PMID: 33086484 PMCID: PMC7711520 DOI: 10.3390/jpm10040179] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/29/2020] [Accepted: 10/09/2020] [Indexed: 12/16/2022] Open
Abstract
Background: Several strategies based on immune checkpoint inhibitors (ICIs) have been developed for cancer therapy, opening to advantages in cancer outcomes. However, several ICI-induced side effects have emerged in these patients, especially a rare but clinically significant cardiotoxicity with high rate of mortality. We studied the cytotoxic and pro-inflammatory properties of Ipilimumab and Nivolumab, the underlying pathways and cytokine storm involved. Methods: Co-cultures of human cardiomyocytes and lymphocytes were exposed to Ipilimumab or Nivolumab; cell viability and expression of leukotrienes, NLRP3, MyD88, and p65/NF-kB were performed. C57 mice were treated with Ipilimumab (15 mg/kg); analysis of fractional shortening, ejection fraction, radial and longitudinal strain were made before and after treatments through 2D-echocardiography. Expression of NLRP3, MyD88, p65/NF-kB, and 12 cytokines were analyzed in murine myocardium. Results: Nivolumab and Ipilimumab exert effective anticancer, but also significant cardiotoxic effects in co-cultures of lymphocytes and tumor or cardiac cells. Both ICIs increased NLRP3, MyD88, and p65/NF-kB expression compared to untreated cells, however, the most pro-inflammatory and cardiotoxic effects were seen after exposure to Ipilimumab. Mice treated with Ipilimumab showed a significant decrease in fractional shortening and radial strain with respect to untreated mice, coupled with a significant increase in myocardial expression of NLRP3, MyD88, and several interleukins. Conclusions: Nivolumab and Ipilimumab exert cytotoxic effects mediated by the NLRP3/IL-1β and MyD88 pathways, leading to pro-inflammatory cytokine storm in heart tissue.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | | | - Domenica Rea
- Animal Facility, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (D.R.); (A.B.)
| | - Antonio Barbieri
- Animal Facility, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (D.R.); (A.B.)
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Annamaria Bonelli
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Antonietta Caronna
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
| | - Gerardo Botti
- Scientific Direction, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy;
| | - Claudia De Lorenzo
- CEINGE—Biotecnologie Avanzate s.c.a.r.l., 80131 Naples, Italy;
- Department of Molecular Medicine and Medical Biotechnology, University of Naples “Federico II”, 80131 Napoli, Italy
- Correspondence: (C.D.L.); (N.M.)
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy; (V.Q.); (M.I.); (A.B.); (A.C.)
- Correspondence: (C.D.L.); (N.M.)
| |
Collapse
|
273
|
Metastatic Merkel Cell Carcinoma Masquerading as Multiple Immune-Related Adverse Events. Case Rep Dermatol Med 2020; 2020:8890845. [PMID: 33062347 PMCID: PMC7545440 DOI: 10.1155/2020/8890845] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 09/18/2020] [Indexed: 12/23/2022] Open
Abstract
Merkel cell carcinoma is a rare cutaneous neuroendocrine carcinoma with a high rate of regional and distant metastasis and mortality. Here, we report a novel case of Merkel cell carcinoma which presented as a primary lesion to the left cheek with regional lymph node involvement and was treated with pembrolizumab and radiation. Widely metastatic disease eventually revealed on autopsy clinically mimicked immune-related organ insult leading to management with immunosuppressants. The patient also had a biopsy-confirmed immune-related cutaneous adverse event during admission. The case highlights a rare circumstance in which disease progression masqueraded as multiple immune-related end-organ adverse events. Contribution of on-target anti-PD-1 toxicity remains a possibility.
Collapse
|
274
|
Abstract
PURPOSE OF REVIEW The coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), continues to spread with rising new cases and deaths. Patients with cancer represent a uniquely vulnerable population not only with higher susceptibility to COVID-19 but also at increased risk for its complications. This review focuses on the implications of COVID-19 in the cardiovascular health of patients with cancer. RECENT FINDINGS Patients more susceptible to COVID-19 with increased severity of disease include those with cancer and cardiovascular comorbidities. In addition, the cardiovascular complications of COVID-19 including acute myocardial injury, thromboembolism, cardiomyopathy, myocarditis, and pericardial disease overlap with many of those encountered during cancer treatment. Despite the absence of large studies of patients with both cancer and cardiovascular disease, the incidence of cardiovascular complications in cancer patients with COVID-19 is expected to be high. This has implications for cardiac monitoring, chemotherapy administration, and the diagnosis and treatment of cardiovascular disease during COVID-19.
Collapse
Affiliation(s)
- Nicolas L. Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77025 USA
| | | | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, 1400 Pressler Street, Unit 1451, Houston, TX 77025 USA
| |
Collapse
|
275
|
Safi M, Ahmed H, Al-Azab M, Xia YL, Shan X, Al-radhi M, Al-danakh A, Shopit A, Liu J. PD-1/PDL-1 Inhibitors and Cardiotoxicity; Molecular, Etiological and Management Outlines. J Adv Res 2020; 29:45-54. [PMID: 33842004 PMCID: PMC8020146 DOI: 10.1016/j.jare.2020.09.006] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2020] [Revised: 09/10/2020] [Accepted: 09/10/2020] [Indexed: 12/16/2022] Open
Abstract
Background The US Food and Drug Administration (FDA) has approved several immunotherapeutic drugs for cancer since 2010, and many more are still being evaluated in other clinical studies. These inhibitors significantly increase response rates and result in the treatment of patients with advanced cancer. However, cancer immunotherapy leads to essential cardiac toxicity properties that have become distinct from other cancer patients' care and are mostly related to their etiology. Aim of review As potential implications, the occurrence of cardiovascular adverse events is particularly challenging and needs a comprehensive understanding of overall cancer-related etiology, clinical outcomes with different variable severity, and management. Key scientific concepts of review In terms of improving the overall survival of patients with cancer, clinicians should be careful in selecting either programmed cell death-1 (PD-1) or its programmed cell death ligand (PDL-1) inhibitors by evaluating their risk and clinical benefit for early intervention and decrease the level of morbidity and mortality of their patients. This review focuses on the effectiveness of PD-1/PL-1 antibodies and associated cardiotoxicity adverse events, including etiological mechanisms, diagnosis, and treatment.
Collapse
Affiliation(s)
- Mohammed Safi
- Department of Oncology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Hyat Ahmed
- Department of Stomatology, Oral Pathology, Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Mahmoud Al-Azab
- Department of Immunology, Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, China
| | - Yun-long Xia
- Head of Department of Cardiology, Vice president of the First Affiliated Hospital of Dalian Medical University, 222 Zhongshan Road, Dalian 116021, Liaoning, China
| | - Xiu Shan
- First Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Mohammed Al-radhi
- Department of Urology, Second Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Abdullah Al-danakh
- Department of Urology, First Affiliated Hospital of Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Abdullah Shopit
- Department of Pharmacology, Dalian Medical University, Zhongshan Road No. 222, Dalian 116021, China
| | - Jiwei Liu
- Head of Department of Oncology First Affiliated Hospital of Dalian Medical University, Zhongshan Road Dalian, Dalian Liaoning Province 116044, China
- Corresponding author.
| |
Collapse
|
276
|
Neves KB, Montezano AC, Lang NN, Touyz RM. Vascular toxicity associated with anti-angiogenic drugs. Clin Sci (Lond) 2020; 134:2503-2520. [PMID: 32990313 DOI: 10.1042/cs20200308] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 09/15/2020] [Accepted: 09/21/2020] [Indexed: 02/07/2023]
Abstract
Over the past two decades, the treatment of cancer has been revolutionised by the highly successful introduction of novel molecular targeted therapies and immunotherapies, including small-molecule kinase inhibitors and monoclonal antibodies that target angiogenesis by inhibiting vascular endothelial growth factor (VEGF) signaling pathways. Despite their anti-angiogenic and anti-cancer benefits, the use of VEGF inhibitors (VEGFi) and other tyrosine kinase inhibitors (TKIs) has been hampered by potent vascular toxicities especially hypertension and thromboembolism. Molecular processes underlying VEGFi-induced vascular toxicities still remain unclear but inhibition of endothelial NO synthase (eNOS), reduced nitric oxide (NO) production, oxidative stress, activation of the endothelin system, and rarefaction have been implicated. However, the pathophysiological mechanisms still remain elusive and there is an urgent need to better understand exactly how anti-angiogenic drugs cause hypertension and other cardiovascular diseases (CVDs). This is especially important because VEGFi are increasingly being used in combination with other anti-cancer dugs, such as immunotherapies (immune checkpoint inhibitors (ICIs)), other TKIs, drugs that inhibit epigenetic processes (histone deacetylase (HDAC) inhibitor) and poly (adenosine diphosphate-ribose) polymerase (PARP) inhibitors, which may themselves induce cardiovascular injury. Here, we discuss vascular toxicities associated with TKIs, especially VEGFi, and provide an up-to-date overview on molecular mechanisms underlying VEGFi-induced vascular toxicity and cardiovascular sequelae. We also review the vascular effects of VEGFi when used in combination with other modern anti-cancer drugs.
Collapse
Affiliation(s)
- Karla B Neves
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Augusto C Montezano
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Ninian N Lang
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| | - Rhian M Touyz
- Institute of Cardiovascular and Medical Sciences, University of Glasgow, U.K
| |
Collapse
|
277
|
Mallouppas M, Walker JM, Guha A, Dobson R, Ghosh AK. Cardio-oncology for the general physician: 'old' and 'new' cardiovascular toxicities and how to manage them. Br J Hosp Med (Lond) 2020; 81:1-11. [PMID: 32990088 DOI: 10.12968/hmed.2020.0269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cardio-oncology is the care of cancer patients with cardiovascular disease. The need for a dedicated subspecialty emerged to address heart failure caused by drugs such as anthracyclines and anti-human epidermal growth factor receptor 2 (HER2) therapies, but over time has expanded into an exciting subspecialty with widening horizons. While still dealing with a lot of commonly recognised toxicities, such as heart failure, hypertension and coronary disease, new and revolutionary cancer therapies have been associated with challenging cardiovascular complications, requiring specialist input to manage effectively. Echocardiography is a key investigation, with advanced techniques such as three-dimensional and strain assessment allowing more accurate diagnosis and earlier detection of subtle changes. Cardiac magnetic resonance and biomarkers are useful adjuncts to aid diagnosis and management. With increasing cancer incidence and improved cancer survival rates, it is important that general cardiologists and physicians are aware of cardiac complications associated with cancer and how to manage them.
Collapse
Affiliation(s)
- Michael Mallouppas
- Cardio-Oncology Service, University College London Hospital NHS Foundation Trust, London, UK.,Hatter Cardiovascular Institute, University College London, London, UK
| | - J Malcolm Walker
- Cardio-Oncology Service, University College London Hospital NHS Foundation Trust, London, UK.,Hatter Cardiovascular Institute, University College London, London, UK
| | - Avirup Guha
- Harrington Heart and Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | | | - Arjun K Ghosh
- Cardio-Oncology Service, University College London Hospital NHS Foundation Trust, London, UK.,Hatter Cardiovascular Institute, University College London, London, UK.,Cardio-Oncology Service, Bart's Heart Centre, St Bartholomew's Hospital, London, UK
| |
Collapse
|
278
|
Chung WB, Youn JC, Youn HJ. Cardiovascular Complications of Novel Anti-Cancer Immunotherapy: Old Problems from New Agents? Korean Circ J 2020; 50:743-753. [PMID: 32725983 PMCID: PMC7440999 DOI: 10.4070/kcj.2020.0158] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2020] [Accepted: 04/22/2020] [Indexed: 12/19/2022] Open
Abstract
Many novel anti-cancer therapies have dramatically improved outcomes of various cancer patients. However, it also poses a risk for cardiovascular complications as well. For the novel anti-cancer agent with which physicians does not have enough clinical experiences to determine the characteristics of cardiovascular complications, it is important to assess risk factors for cardiotoxicity before starting anti-cancer therapy. High-risk patient should be consulted to cardiologist before initiating anti-cancer therapy and pre-emptive cardiac function monitoring plan might be prepared in advance. The biomarkers, electrocardiography and echocardiography are useful tools for the detection of subclinical cardiotoxicity during anti-cancer therapy. This review article tried to suggest the cardiac function monitoring strategies for newly encountered potential cardiotoxic anti-cancer agents and to summarize the cardiovascular complications of novel anti-cancer immunotherapies including immune checkpoint inhibitor (ICI) and chimeric antigen receptor (CAR) T-cell therapy. ICIs can cause fatal myocarditis, which usually occurs early after initiation, and prompt treatment with high-dose corticosteroid is necessary. CAR T-cell therapy can cause cytokine release syndrome, which may result in circulatory collapse. Supportive treatment as well as tocilizumab, an anti-interleukin-6 receptor antibody are cornerstones of treatment.
Collapse
Affiliation(s)
- Woo Baek Chung
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Chan Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Joong Youn
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea.
| |
Collapse
|
279
|
Like a Rolling Stone: Sting-Cgas Pathway and Cell-Free DNA as Biomarkers for Combinatorial Immunotherapy. Pharmaceutics 2020; 12:pharmaceutics12080758. [PMID: 32796670 PMCID: PMC7464249 DOI: 10.3390/pharmaceutics12080758] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 08/08/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022] Open
Abstract
Combining immune checkpoint inhibitors with other treatments likely to harness tumor immunity is a rising strategy in oncology. The exact modalities of such a combinatorial regimen are yet to be defined, and most attempts have relied so far on concomitant dosing, rather than sequential or phased administration. Because immunomodulating features are likely to be time-, dose-, and-schedule dependent, the need for biomarkers providing real-time information is critical to better define the optimal time-window to combine immune checkpoint inhibitors with other drugs. In this review, we present the various putative markers that have been investigated as predictive tools with immune checkpoint inhibitors and could be used to help further combining treatments. Whereas none of the current biomarkers, such as the PDL1 expression of a tumor mutational burden, is suitable to identify the best way to combine treatments, monitoring circulating tumor DNA is a promising strategy, in particular to check whether the STING-cGAS pathway has been activated by cytotoxics. As such, circulating tumor DNA could help defining the best time-window to administrate immune checkpoint inhibitors after that cytotoxics have been given.
Collapse
|
280
|
Gürdoğan M, Yalta K. Myocarditis associated with immune checkpoint inhibitors: Practical considerations in diagnosis and management. Anatol J Cardiol 2020; 24:68-75. [PMID: 32749254 PMCID: PMC7460679 DOI: 10.14744/anatoljcardiol.2020.79584] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/12/2020] [Indexed: 12/14/2022] Open
Abstract
Immune checkpoint inhibitors (ICI) have caused radical changes in the treatment scheme of many types of cancer in the past 10 years. ICIs are specific monoclonal antibodies that increase T-cell mediated immune response against cancer cells. Despite important advances in cancer treatment, uncontrolled activation of cytotoxic T cells has brought along many autoimmune clinical side effects, especially acute myocarditis. Although the incidence of ICI-related myocarditis is about 1%, it is remarkable in terms of mortality rate reaching 46% and demonstrating the necessity of rapid diagnosis and multidisciplinary approach. The present review aimed to summarize the heterogeneous symptomatology of ICI-associated myocarditis, clinical presentation ranging from elevated asymptomatic cardiac enzyme levels to cardiogenic shock, prominent diagnostic value of cardiac magnetic resonance imaging, and current information on the effectiveness of immunosuppressants in therapy.
Collapse
Affiliation(s)
- Muhammet Gürdoğan
- Department of Cardiology, Faculty of Medicine, Trakya University; Edirne-Turkey
| | - Kenan Yalta
- Department of Cardiology, Faculty of Medicine, Trakya University; Edirne-Turkey
| |
Collapse
|
281
|
Spallarossa P, Sarocchi M, Tini G, Arboscello E, Toma M, Ameri P, Porto I. How to Monitor Cardiac Complications of Immune Checkpoint Inhibitor Therapy. Front Pharmacol 2020; 11:972. [PMID: 32676031 PMCID: PMC7333217 DOI: 10.3389/fphar.2020.00972] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 06/15/2020] [Indexed: 12/12/2022] Open
Abstract
Immune-checkpoint inhibitors (ICIs) represent a successful paradigm in the treatment of cancer. ICIs elicit an immune response directed against cancer cells, by targeting the so-called immune checkpoints, key regulators of the immune system that when stimulated can dampen the immune response to an immunologic stimulus. Such response, however, is not entirely tumor-specific and may result in immune-related adverse events (irAEs), involving a number of organs and systems. Cardiovascular (CV) irAEs are rare, although potentially severe. In particular, several cases of ICI-related myocarditis with life-threatening course have been reported: the possibility of fulminant cases, thus, requires a high level of awareness among both oncologists and cardiologists. Aggressive work-up and management of symptomatic patients taking ICIs is fundamental for early recognition and initiation of specific immunosuppressive therapies. Notably, myocarditis occurs within few weeks from ICIs initiation, offering opportunity for a targeted screening. Troponin testing is the cornerstone of this screening, yet uncertainties remain regarding timing and candidates. Moreover, troponins positivity should be carefully interpreted. We herein review the main aspects of ICI-related myocarditis and suggest a practical approach. In particular, we focus on the opportunities that a baseline CV evaluation offers for subsequent management by collecting clinical and instrumental data, essential for the interpretation of troponin results, for differential diagnosis and for the formulation of a diagnostic and therapeutic workup.
Collapse
Affiliation(s)
- Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS San Martino Policlinic Hospital–IRCCS Italian Cardiovascular Network, Genova, Italy
| | - Matteo Sarocchi
- Cardiovascular Disease Unit, IRCCS San Martino Policlinic Hospital–IRCCS Italian Cardiovascular Network, Genova, Italy
| | - Giacomo Tini
- Cardiovascular Disease Unit, IRCCS San Martino Policlinic Hospital–IRCCS Italian Cardiovascular Network, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Eleonora Arboscello
- Department of Emergency, IRCCS San Martino Policlinic Hospital, University of Genova, Genova, Italy
| | - Matteo Toma
- Cardiovascular Disease Unit, IRCCS San Martino Policlinic Hospital–IRCCS Italian Cardiovascular Network, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS San Martino Policlinic Hospital–IRCCS Italian Cardiovascular Network, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Italo Porto
- Cardiovascular Disease Unit, IRCCS San Martino Policlinic Hospital–IRCCS Italian Cardiovascular Network, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| |
Collapse
|
282
|
Michel L, Totzeck M, Lehmann L, Finke D. Emerging role of immune checkpoint inhibitors and their relevance for the cardiovascular system. Herz 2020; 45:645-651. [DOI: 10.1007/s00059-020-04954-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
283
|
Palaskas N, Lopez‐Mattei J, Durand JB, Iliescu C, Deswal A. Immune Checkpoint Inhibitor Myocarditis: Pathophysiological Characteristics, Diagnosis, and Treatment. J Am Heart Assoc 2020; 9:e013757. [PMID: 31960755 PMCID: PMC7033840 DOI: 10.1161/jaha.119.013757] [Citation(s) in RCA: 279] [Impact Index Per Article: 55.8] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 12/03/2019] [Indexed: 12/21/2022]
Affiliation(s)
- Nicolas Palaskas
- Department of CardiologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Juan Lopez‐Mattei
- Department of CardiologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Jean Bernard Durand
- Department of CardiologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Cezar Iliescu
- Department of CardiologyThe University of Texas MD Anderson Cancer CenterHoustonTX
| | - Anita Deswal
- Department of CardiologyThe University of Texas MD Anderson Cancer CenterHoustonTX
- Section of CardiologyMichael E. DeBakey VA Medical CenterBaylor College of MedicineHoustonTX
| |
Collapse
|
284
|
Affiliation(s)
- Isabella M Grumbach
- Division of Cardiovascular Medicine Department of Internal Medicine Abboud Cardiovascular Research Center Carver College of Medicine Iowa City IA.,Free Radical and Radiation Biology Program Department of Radiation Oncology Holden Comprehensive Cancer Center University of Iowa Iowa City IA.,Department of Veterans Affairs Iowa City Health Care System Iowa City IA
| |
Collapse
|