1
|
Maayah M, Grubman S, Allen S, Ye Z, Park DY, Vemmou E, Gokhan I, Sun WW, Possick S, Kwan JM, Gandhi PU, Hu JR. Clinical Interpretation of Serum Troponin in the Era of High-Sensitivity Testing. Diagnostics (Basel) 2024; 14:503. [PMID: 38472975 DOI: 10.3390/diagnostics14050503] [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: 01/01/2024] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 03/14/2024] Open
Abstract
Cardiac troponin (Tn) plays a central role in the evaluation of patients with angina presenting with acute coronary syndrome. The advent of high-sensitivity assays has improved the analytic sensitivity and precision of serum Tn measurement, but this advancement has come at the cost of poorer specificity. The role of clinical judgment is of heightened importance because, more so than ever, the interpretation of serum Tn elevation hinges on the careful integration of findings from electrocardiographic, echocardiographic, physical exam, interview, and other imaging and laboratory data to formulate a weighted differential diagnosis. A thorough understanding of the epidemiology, mechanisms, and prognostic implications of Tn elevations in each cardiac and non-cardiac etiology allows the clinician to better distinguish between presentations of myocardial ischemia and myocardial injury-an important discernment to make, as the treatment of acute coronary syndrome is vastly different from the workup and management of myocardial injury and should be directed at the underlying cause.
Collapse
Affiliation(s)
- Marah Maayah
- Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Scott Grubman
- Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Stephanie Allen
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Zachary Ye
- Department of Internal Medicine, Temple University Medical Center, Philadelphia, PA 19140, USA
| | - Dae Yong Park
- Department of Internal Medicine, Cook County Hospital, Chicago, IL 60612, USA
| | - Evangelia Vemmou
- Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Ilhan Gokhan
- Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Wendy W Sun
- Department of Emergency Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Stephen Possick
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Jennifer M Kwan
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| | - Parul U Gandhi
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
- Department of Cardiology, Veterans Affairs Connecticut Health Care System, West Haven, CT 06516, USA
| | - Jiun-Ruey Hu
- Section of Cardiovascular Medicine, Yale School of Medicine, Yale University, New Haven, CT 06520, USA
| |
Collapse
|
2
|
Jo W, Won T, Daoud A, Čiháková D. Immune checkpoint inhibitors associated cardiovascular immune-related adverse events. Front Immunol 2024; 15:1340373. [PMID: 38375475 PMCID: PMC10875074 DOI: 10.3389/fimmu.2024.1340373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/19/2024] [Indexed: 02/21/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are specialized monoclonal antibodies (mAbs) that target immune checkpoints and their ligands, counteracting cancer cell-induced T-cell suppression. Approved ICIs like cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed death-1 (PD-1), its ligand PD-L1, and lymphocyte activation gene-3 (LAG-3) have improved cancer patient outcomes by enhancing anti-tumor responses. However, some patients are unresponsive, and others experience immune-related adverse events (irAEs), affecting organs like the lung, liver, intestine, skin and now the cardiovascular system. These cardiac irAEs include conditions like myocarditis, atherosclerosis, pericarditis, arrhythmias, and cardiomyopathy. Ongoing clinical trials investigate promising alternative co-inhibitory receptor targets, including T cell immunoglobulin and mucin domain-containing protein 3 (Tim-3) and T cell immunoreceptor with immunoglobulin and ITIM domain (TIGIT). This review delves into the mechanisms of approved ICIs (CTLA-4, PD-1, PD-L1, and LAG-3) and upcoming options like Tim-3 and TIGIT. It explores the use of ICIs in cancer treatment, supported by both preclinical and clinical data. Additionally, it examines the mechanisms behind cardiac toxic irAEs, focusing on ICI-associated myocarditis and atherosclerosis. These insights are vital as ICIs continue to revolutionize cancer therapy, offering hope to patients, while also necessitating careful monitoring and management of potential side effects, including emerging cardiac complications.
Collapse
Affiliation(s)
- Wonyoung Jo
- Department of Biomedical Engineering, Johns Hopkins University, Whiting School of Engineering, Baltimore, MD, United States
| | - Taejoon Won
- Department of Pathobiology, University of Illinois Urbana-Champaign, College of Veterinary Medicine, Urbana, IL, United States
| | - Abdel Daoud
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, United States
| | - Daniela Čiháková
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins University, Bloomberg School of Public Health, Baltimore, MD, United States
- Department of Pathology, Johns Hopkins University, School of Medicine, Baltimore, MD, United States
| |
Collapse
|
3
|
Frascaro F, Bianchi N, Sanguettoli F, Marchini F, Meossi S, Zanarelli L, Tonet E, Serenelli M, Guardigli G, Campo G, Calabrò L, Pavasini R. Immune Checkpoint Inhibitors-Associated Myocarditis: Diagnosis, Treatment and Current Status on Rechallenge. J Clin Med 2023; 12:7737. [PMID: 38137806 PMCID: PMC10744238 DOI: 10.3390/jcm12247737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 12/02/2023] [Accepted: 12/15/2023] [Indexed: 12/24/2023] Open
Abstract
Immune checkpoint molecules like cytotoxic T-lymphocyte antigen 4 (CTLA-4), programmed cell death 1 (PD-1) or its ligand, programmed cell death ligand 1 (PD-L1), play a critical role in regulating the immune response, and immune checkpoint inhibitors (ICIs) targeting these checkpoints have shown clinical efficacy in cancer treatment; however, their use is associated with immune-related adverse events (irAEs), including cardiac complications. The prevalence of cardiac irAEs, particularly myocarditis, is relatively low, but they can become a severe and potentially life-threatening condition, usually occurring shortly after initiating ICI treatment; moreover, diagnosing ICI-related myocarditis can be challenging. Diagnostic tools include serum cardiac biomarkers, electrocardiography (ECG), echocardiography, cardiac magnetic resonance (CMR) and endomyocardial biopsy (EMB). The treatment of ICI-induced myocarditis involves high-dose corticosteroids, which have been shown to reduce the risk of major adverse cardiac events (MACE). In refractory cases, second-line immunosuppressive drugs may be considered, although their effectiveness is based on limited data. The mortality rates of ICI-induced myocarditis, particularly in severe cases, are high (38-46%). Therapy rechallenge after myocarditis is associated with a risk of recurrence and severe complications. The decision to rechallenge should be made on a case-by-case basis, involving a multidisciplinary team of cardiologists and oncologists. Further research and guidance are needed to optimize the management of cancer patients who have experienced such complications, evaluating the risks and benefits of therapy rechallenge. The purpose of this review is to summarize the available evidence on cardiovascular complications from ICI therapy, with a particular focus on myocarditis and, specifically, the rechallenge of immunotherapy after a cardiac adverse event.
Collapse
Affiliation(s)
- Federica Frascaro
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Nicola Bianchi
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Federico Sanguettoli
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Federico Marchini
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Sofia Meossi
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Luca Zanarelli
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Elisabetta Tonet
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Matteo Serenelli
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Gabriele Guardigli
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Gianluca Campo
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| | - Luana Calabrò
- Dipartimento di Medicina Translazionale e per la Romagna, Univerity of Ferrara, 44121 Ferrara, Italy;
- UO Medical Oncology, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy
| | - Rita Pavasini
- UO Cardiologia, Azienda Ospedaliero-Universitaria di Ferrara, 44124 Ferrara, Italy; (F.F.); (N.B.); (F.S.); (F.M.); (S.M.); (L.Z.); (E.T.); (M.S.); (G.G.); (G.C.)
| |
Collapse
|
4
|
Patel M, Hudson O, Han J, Kondapalli L, Arora G, Hawi R, Andrikopoulou E, Estes C, Johnson AM, Lenneman C. Update on Immunotherapy Cardiotoxicity: Checkpoint Inhibitors, CAR T, and Beyond. Curr Treat Options Oncol 2023; 24:1489-1503. [PMID: 37624557 DOI: 10.1007/s11864-023-01130-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2023] [Indexed: 08/26/2023]
Abstract
OPINION STATEMENT Immunotherapy is an innovative approach to cancer treatment that involves using the body's immune system to fight cancer. The landscape of immunotherapy is constantly evolving, as new therapies are developed and refined. Some of the most promising approaches in immunotherapy include immune checkpoint inhibitors (ICIs): these drugs target proteins on the surface of T-cells that inhibit their ability to attack cancer cells. By blocking these proteins, checkpoint inhibitors allow T-cells to recognize and destroy cancer cells more effectively. CAR T-cell therapy: this therapy involves genetically modifying a patient's own T-cells to recognize and attack cancer cells. CAR T-cell therapy exhibits favorable response in many patients with refractory hematological cancers with growing clinical trials in solid tumors. Immune system modulators: these drugs enhance the immune system's ability to fight cancer by stimulating the production of immune cells or inhibiting the activity of immune-suppressing cells. While immunotherapy has shown great promise in the treatment of cancer, it can also pose significant cardiac side effects. Some immunotherapy drugs like ICIs can cause myocarditis, which can lead to chest pain, shortness of breath, and heart failure. Other cardiac side effects of ICIs include arrhythmias, pericarditis, vasculitis, and accelerated atherosclerosis. It is important for patients receiving immunotherapy to be monitored closely for these side effects, as prompt treatment can help prevent serious complications. Patients should also report any symptoms to their healthcare providers right away, so that appropriate action can be taken. CAR T-cell therapy can also illicit an exaggerated immune response creating cytokine release syndrome (CRS) that may precipitate cardiovascular events: arrhythmias, myocardial infarction, and heart failure. Overall, while immune modulating therapy is a promising and expanding approach to cancer treatment, it is important to weigh the potential benefits against the risks and side effects, especially in patients with high risk for cardiovascular complications.
Collapse
Affiliation(s)
- Murti Patel
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Olivia Hudson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Jingnan Han
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Lavanya Kondapalli
- University of Colorado School of Medicine, Anschutz Medical Campus, Aurora, CO, USA
| | - Garima Arora
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Riem Hawi
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | | | - Courtney Estes
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Abigail M Johnson
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA
| | - Carrie Lenneman
- University of Alabama at Birmingham (UAB) Heersink School of Medicine, Birmingham, AL, USA.
| |
Collapse
|
5
|
Chitsazan M, Amin A, Ladel L, Baig A, Chitsazan M. Cardiovascular Toxicity Associated With Immune Checkpoint Inhibitor Therapy: A Comprehensive Review. Crit Pathw Cardiol 2023; 22:69-82. [PMID: 37363862 DOI: 10.1097/hpc.0000000000000327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2023]
Abstract
Immune checkpoint inhibitors (ICIs), a significant breakthrough treatment of cancer, exert their function through enhancing the immune system's ability to recognize and attack cancer cells. However, these revolutionary cancer treatments have been associated with a range of immune-related adverse effects, including cardiovascular toxicity. The most commonly reported cardiovascular toxicities associated with ICIs are myocarditis, pericarditis, arrhythmias, and vasculitis. These cardiovascular manifestations are often severe and can lead to life-threatening complications. Therefore, prompt identification and management of these toxicities is critical, and a multidisciplinary teamwork by cardiologists and oncologists are required to ensure optimal patient outcomes. In this review, we summarize the current knowledge on the mechanisms underlying ICI-associated cardiovascular toxicity, clinical presentations of these toxicities, potential risk factors, diagnosis, management, and surveillance strategies during ICI therapy. While ICIs have already transformed cancer treatment, further research is needed to better understand and manage their immune-related cardiovascular effects, and possibly, to identify biomarkers which can predict the occurrence of these cardiovascular complications.
Collapse
Affiliation(s)
| | - Ahmad Amin
- Medstar Union Memorial Hospital, Baltimore, MD
| | - Luisa Ladel
- From the Department of Medicine, Norwalk Hospital, Norwalk, CT
| | - Alyza Baig
- From the Department of Medicine, Norwalk Hospital, Norwalk, CT
| | - Mitra Chitsazan
- Rajaie Cardiovascular, Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Kazemzadeh K, Behrouzieh S, Rezaei N. Shedding light on the side effects of immunotherapies used for leukemia: an updated review of the literature. Expert Rev Anticancer Ther 2023; 23:1193-1204. [PMID: 37812581 DOI: 10.1080/14737140.2023.2267760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Accepted: 10/03/2023] [Indexed: 10/11/2023]
Abstract
INTRODUCTION Leukemia is an abnormal clonal development of leukemic cells originating from the bone marrow, which is widely known for its significant prevalence and mortality rate. Chemotherapy, surgery, radiation, and combination therapies have been its routine therapeutic methods; however, the advent of cancer immunotherapy is known as revolutionary for its higher efficacy and lesser toxicity. AREAS COVERED Immunotherapy boosts the body's immune system by using components from other living organisms. Although immunotherapy seems to be safer than chemotherapy, many studies have noticed different immune-related side effects in various body systems (e.g. cardiovascular, neurologic) which we have reviewed in this investigation as the main goal. We tried to describe immunotherapy-related side effects in human body systems in detail. EXPERT OPINION Being aware of these side effects leads to better clinical decision-making for each individual, and a one-step-ahead management in case of occurrence. We also briefly discussed the role of immunotherapy in treating leukemia as one of the most prevalent cancers in children and tried to emphasize that it is crucial to monitor adverse events as they may remain obscure until adolescence.
Collapse
Affiliation(s)
- Kimia Kazemzadeh
- Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
- Network of Neurosurgery and Artificial Intelligence (NONAI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Sadra Behrouzieh
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Network of Neurosurgery and Artificial Intelligence (NONAI), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Meng L, Wei Y, Xiao Y. Chemo-immunoablation of solid tumors: A new concept in tumor ablation. Front Immunol 2023; 13:1057535. [PMID: 36713427 PMCID: PMC9878389 DOI: 10.3389/fimmu.2022.1057535] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/09/2022] [Indexed: 01/13/2023] Open
Abstract
Chemical ablation was designed to inject chemical agents directly into solid tumors to kill cells and is currently only used clinically for the palliative treatment of tumors. The application and combination of different drugs, from anhydrous ethanol, and glacial acetic acid to epi-amycin, have been clinically tested for a long time. The effectiveness is unsatisfactory due to chemical agents' poor diffusion and concentration. Immunotherapy is considered a prospective oncologic therapeutic. Still, the clinical applications were limited by the low response rate of patients to immune drugs and the immune-related adverse effects caused by high doses. The advent of intratumoral immunotherapy has well addressed these issues. However, the efficacy of intratumoral immunotherapy alone is uncertain, as suggested by the results of preclinical and clinical studies. In this study, we will focus on the research of immunosuppressive tumor microenvironment with chemoablation and intratumoral immunotherapy, the synergistic effect between chemotherapeutic drugs and immunotherapy. We propose a new concept of intratumoral chemo-immunoablation. The concept opens a new perspective for tumor treatment from direct killing of tumor cells while, enhancing systemic anti-tumor immune response, and significantly reducing adverse effects of drugs.
Collapse
Affiliation(s)
- Liangliang Meng
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,Department of Radiology, Chinese PAP Hospital of Beijing, Beijing, China
| | - Yingtian Wei
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yueyong Xiao
- Department of Radiology, the First Medical Center, Chinese PLA General Hospital, Beijing, China,*Correspondence: Yueyong Xiao,
| |
Collapse
|
8
|
Son C, Moey MYY, Walker PR, Naqash AR, Peach MS, Ju AW. Cardiac toxicity in patients with lung cancer receiving thoracic radiotherapy and immunotherapy. Front Oncol 2023; 12:1025455. [PMID: 36698405 PMCID: PMC9868592 DOI: 10.3389/fonc.2022.1025455] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 12/13/2022] [Indexed: 01/11/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) are used to treat locally-advanced and metastatic lung cancer, which can lead to severe immunogenic-related cardiotoxicities. We assessed the risk of cardiotoxicity in ICI-treated lung cancer patients with or without cardiac radiation from thoracic radiotherapy. Methods Retrospective data was collected on Stage III-IV lung cancer patients who received ICIs between 2015 and 2018. All cardiotoxicities associated with ICI were assessed in correlation with the timing of radiotherapy (RT) in relation to ICI, and the mean RT heart dose. The rate of cardiac events in relation to RT timing and heart dose was compared using multiple logistic regression including the Framingham risk score and steroid use prior to ICI therapy. Results Of 194 ICI-treated patients evaluated, 55.2% (n=107/194) patients had received thoracic RT at a median dose of 60.4 Gy (range, 15-75). Cardiotoxicities such as non-ST elevated myocardial infarction and new onset supraventricular tachycardias were observed in 13 (12.2%) of those who had thoracic RT versus 9 (10.3%) who did not (p=0.87). 38 patients who received RT concurrently with ICI did not develop any cardiotoxicity whereas 14.1% (n=22/156) of those who did not receive concurrent RT developed cardiotoxicities (univariate, p=0.030; multivariate, p=0.055). There were no significant differences in the mean heart RT dose, Framingham risk score, and steroid treatment between patients that received concurrent RT with ICI versus non-concurrent RT/ICI. Conclusion ICI-related cardiotoxicities were not significantly associated with patients who received concurrent thoracic radiotherapy in this retrospective review. Further validation of prospective studies is needed to confirm these results.
Collapse
Affiliation(s)
- Christine Son
- Brody School of Medicine at East Carolina University, Greenville, NC, United States
| | - Melissa Y. Y. Moey
- Department of Cardiovascular Sciences at Vidant Medical Center/East Carolina University, Greenville, NC, United States
| | - Paul R. Walker
- Department of Hematology and Oncology at East Carolina University, Greenville, NC, United States
| | - Abdul R. Naqash
- Department of Hematology and Oncology at East Carolina University, Greenville, NC, United States,Medical Oncology/TSET Phase 1 Program OU Health Stephenson Cancer Center at the University of Oklahoma, Oklahoma City, OK, United States
| | - Matthew Sean Peach
- Department of Radiation and Oncology at East Carolina University, Greenville, NC, United States
| | - Andrew W. Ju
- Department of Radiation and Oncology at East Carolina University, Greenville, NC, United States,*Correspondence: Andrew W. Ju,
| |
Collapse
|
9
|
Liu H, Luo SX, Jie J, Peng L, Wang S, Song L. Immune checkpoint inhibitors related respiratory disorders in patients with lung cancer: A meta-analysis of randomized controlled trials. Front Immunol 2023; 14:1115305. [PMID: 36926326 PMCID: PMC10011157 DOI: 10.3389/fimmu.2023.1115305] [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: 12/03/2022] [Accepted: 02/14/2023] [Indexed: 03/08/2023] Open
Abstract
Background In recent years, immune checkpoint inhibitors (ICIs) had extremely rapid growth in anti-cancer and improved outcomes of many malignancies, specifically lung cancer. However, the incidence of ICIs-related adverse events also raised. Using this meta-analysis, ICIs-related respiratory disorders were investigated in lung cancer patients. Methods Using Cochrane Library, Embase, and PubMed databases, we performed an integrated search for randomized controlled trials (RCTs) to compare respiratory disorders among different regimens. The data was prepared with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) reporting guideline, and the quality of included studies was evaluated based on the Cochrane manual. Results In total, 22 RCTs were involved in this meta-analysis. Compared with ICIs, chemotherapy reduced the risk of interstitial lung disease (p = 0.03; SMD: 2.81; 95% CI: 1.08, 7.27), pleural effusion (p = 0.002; SMD: 2.12; 95% CI: 1.32, 3.42), and pneumonitis (p < 0.00001; SMD: 9.23; 95% CI: 4.57, 18.64). ICIs plus chemotherapy could provide a higher probability for patients to suffer pneumonitis than chemotherapy (p = 0.01; SMD: 1.96; 95% CI: 1.17, 3.28). In addition, single ICI brought a lower likelihood for patients suffering pneumonitis than double ICIs (p = 0.004; SMD: 2.17; 95% CI: 1.27, 3.69). Conclusion ICIs-based treatment, such as ICIs alone, ICIs plus chemotherapy and double ICIs, can raise the incidences of some respiratory disorders in patients with lung cancer. It suggests that ICIs should be conducted based on a comprehensive consideration to prevent ICIs-related respiratory disorders. To a certain degree, this study might be provided to the clinician as a reference for ICIs practice. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42022378901, identifier (CRD42022378901).
Collapse
Affiliation(s)
- Han Liu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Sean X Luo
- Department of Vascular Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jing Jie
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Liping Peng
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Shuai Wang
- Department of Vascular Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Song
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
10
|
Ganatra S, Dani SS, Yang EH, Zaha VG, Nohria A. Cardiotoxicity of T-Cell Antineoplastic Therapies: JACC: CardioOncology Primer. JACC CardioOncol 2022; 4:616-623. [PMID: 36636447 PMCID: PMC9830211 DOI: 10.1016/j.jaccao.2022.07.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 07/27/2022] [Indexed: 01/11/2023] Open
Abstract
T-cell therapies, such as chimeric antigen receptor (CAR) T-cell, bispecific T-cell engager (BiTE) and tumor-infiltrating lymphocyte (TIL) therapies, fight cancer cells harboring specific tumor antigens. However, activation of the immune response by these therapies can lead to a systemic inflammatory response, termed cytokine release syndrome (CRS), that can result in adverse events, including cardiotoxicity. Retrospective studies have shown that cardiovascular complications occur in 10% to 20% of patients who develop high-grade CRS after CAR T-cell therapy and can include cardiomyopathy, heart failure, arrhythmias, and myocardial infarction. While cardiotoxicities have been less commonly reported with BiTE and TIL therapies, systematic surveillance for cardiotoxicity has not been performed. Patients undergoing T-cell therapies should be screened for cardiovascular conditions that may not be able to withstand the hemodynamic perturbations imposed by CRS. Generalized management of CRS, including the use of the interleukin-6 antagonist, tocilizumab, for high-grade CRS, is used to mitigate the risk of cardiotoxicity.
Collapse
Key Words
- BCMA, B-cell maturation antigen
- BiTE therapy
- BiTE, bispecific T-cell engager
- CAR T-cell therapy
- CAR, chimeric antigen receptor
- CRS, cytokine release syndrome
- HF, heart failure
- ICSR, individual case safety report
- IL, interleukin
- LVEF, left ventricular ejection fraction
- MACE, major adverse cardiovascular event(s)
- TIL, tumor-infiltrating lymphocyte
- arrhythmia
- cardiomyopathy
- cardiotoxicity
- heart failure
- tumor-infiltrating lymphocytes
Collapse
Affiliation(s)
- Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital & Medical Center, Burlington, Massachusetts, USA
- Address for correspondence: Dr Sarju Ganatra, Department of Cardiovascular Medicine, Lahey Hospital & Medical Center, 41 Mall Road, Burlington, Massachusetts 01805, USA. @SarjuGanatraMD
| | - Sourbha S. Dani
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital & Medical Center, Burlington, Massachusetts, USA
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, UCLA-Cardio-Oncology Program, University of California-Los Angeles, Los Angeles, California, USA
| | - Vlad G. Zaha
- Cardio-Oncology Program, Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Division of Cardiology, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
- Parkland Health & Hospital System, Dallas, Texas, USA
| | - Anju Nohria
- Cardio-Oncology Program, Department of Cardiovascular Medicine, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
11
|
Zito C, Manganaro R, Ciappina G, Spagnolo CC, Racanelli V, Santarpia M, Silvestris N, Carerj S. Cardiotoxicity Induced by Immune Checkpoint Inhibitors: What a Cardio-Oncology Team Should Know and Do. Cancers (Basel) 2022; 14:cancers14215403. [PMID: 36358830 PMCID: PMC9653561 DOI: 10.3390/cancers14215403] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/27/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the therapeutic scenario for several malignancies. However, they can be responsible for immune-related adverse events (irAEs), involving several organs, with a pooled incidence ranging between 54% and 76%. The frequency of cardiovascular system involvement is <1%. Among the cardiovascular irAEs, myocarditis is the most common and the most dangerous but other, less common manifestations of ICI-related cardiotoxicity include pericardial disease, arrhythmias, Takotsubo-like syndrome, and acute myocardial infarction, all of which remain poorly explored. Both oncologists and cardiologists, as well as the patients, should be aware of the possible occurrence of one or more of these complications, which in some cases are fatal, in order to implement effective strategies of cardiac surveillance. In this review, we summarize the latest studies and recommendations on the pathogenesis, clinical manifestation, diagnosis, and management of ICI-related cardiotoxicity in order to realize a complete and updated overview on the main aspects of ICI-related cardiotoxicity, from surveillance to diagnosis to management, useful for both oncologists and cardiologists in their clinical practice. In particular, in the first part of the review, we realize a description of the pathogenetic mechanisms and risk factors of the main cardiovascular irAEs. Then, we focus on the management of ICI-related cardiotoxicity by analyzing five main points: (1) identifying and evaluating the type and severity of the cardiotoxicity; (2) deciding whether to withhold ICI therapy; (3) initiating steroid and immunosuppressive therapy; (4) starting conventional cardiac treatment; and (5) restarting ICI therapy. Finally, we discuss the existing evidence on surveillance for ICI-related cardiotoxicity and propose a surveillance strategy for both short- and long-term cardiotoxicity, according to the most recent guidelines.
Collapse
Affiliation(s)
- Concetta Zito
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Roberta Manganaro
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Giuliana Ciappina
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Calogera Claudia Spagnolo
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Vito Racanelli
- Department of Interdisciplinary Medicine, Medical School, University of Bari “Aldo Moro”, 70121 Bari, Italy
| | - Mariacarmela Santarpia
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
| | - Nicola Silvestris
- Medical Oncology Unit, Department of Human Pathology “G. Barresi”, University of Messina, 98125 Messina, Italy
- Correspondence:
| | - Scipione Carerj
- Cardiology Unit, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
12
|
Ling SP, Ming LC, Dhaliwal JS, Gupta M, Ardianto C, Goh KW, Hussain Z, Shafqat N. Role of Immunotherapy in the Treatment of Cancer: A Systematic Review. Cancers (Basel) 2022; 14:5205. [PMID: 36358624 PMCID: PMC9655090 DOI: 10.3390/cancers14215205] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 10/14/2022] [Accepted: 10/15/2022] [Indexed: 01/18/2024] Open
Abstract
Tremendous progress has been made in cancer research over the years, and, as a result, immunotherapy has emerged as an important therapy for the treatment of cancer, either as a stand-alone treatment or in conjunction with other cancer therapies. Immunotherapy has demonstrated encouraging outcomes and offers a viable strategy for not only enhancing the quality of life but also dramatically boosting the overall survival rate of cancer patients. The objective of this systematic review was to assess the efficacy of immunotherapy in the treatment of cancer. Databases such as PubMed and Science Direct were searched from their inception until September 2021, using the following keywords: cancer immunotherapy, cancer recurrence, cancer treatment options, and cancer therapies. The systematic review was conducted in accordance with the PRISMA protocol. There were a total of 599 articles; however, after applying the inclusion and exclusion criteria, the final review ended up with 34 publications. In conclusion, the studies have demonstrated that immunotherapy is a viable alternative treatment option for patients with recurrent or metastatic cancer, since the overall survival rate and progression-free survival rate were shown to be successful.
Collapse
Affiliation(s)
- Sia Pei Ling
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Long Chiau Ming
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Jagjit Singh Dhaliwal
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| | - Madhu Gupta
- School of Pharmaceutical Sciences, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India
| | - Chrismawan Ardianto
- Department of Pharmacy Practice, Faculty of Pharmacy, Universitas Airlangga, Surabaya 60115, Indonesia
| | - Khang Wen Goh
- Faculty of Data Science and Information Technology, INTI International University, Nilai 71800, Malaysia
| | - Zahid Hussain
- Faculty of Health, University of Canberra, Bruce, ACT 2617, Australia
| | - Naeem Shafqat
- PAPRSB Institute of Health Sciences, Universiti Brunei Darussalam, Gadong BE1410, Brunei
| |
Collapse
|
13
|
Li Y, Ming Y, Fu R, Li C, Wu Y, Jiang T, Li Z, Ni R, Li L, Su H, Liu Y. The pathogenesis, diagnosis, prevention, and treatment of CAR-T cell therapy-related adverse reactions. Front Pharmacol 2022; 13:950923. [PMID: 36313336 PMCID: PMC9616161 DOI: 10.3389/fphar.2022.950923] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Accepted: 09/29/2022] [Indexed: 11/13/2022] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is effective in the treatment of refractory/relapsed (r/r) hematological malignancies (r/r B-cell lymphoblastic leukemia, B-cell lymphoma, and multiple myeloma). In addition, it is being explored as a treatment option for solid tumors. As of 31 March 2022, seven CAR-T therapies for hematological malignancies have been approved worldwide. Although CAR-T therapy is an effective treatment for many malignancies, it also causes adverse effects. The incidence of cytokine release syndrome (CRS), the most common adverse reaction after infusion of CAR-T cells, is as high as 93%.CRS, is the leading risk factor of immune effector cell-associated neurotoxicity syndrome (ICANS), as well as cardiovascular, hematological, hepatorenal, skin, pulmonary, and gastrointestinal toxicity. Severe adverse reactions complicated by CRS severely impede the widespread application of CAR-T therapy. The CAR-T product was initially approved in 2017; however, only limited studies have investigated the adverse reactions owing to CAR-T therapy compared to that of clinically approved drugs. Thus, we aimed to elucidate the mechanisms, risk factors, diagnostic criteria, and treatment of toxicities concurrent with CRS, thereby providing a valuable reference for the safe, effective, and widespread application of CAR-T therapy.
Collapse
|
14
|
Perpinia AS, Kadoglou N, Vardaka M, Gkortzolidis G, Karavidas A, Marinakis T, Papachrysostomou C, Makaronis P, Vlachou C, Mantzourani M, Farmakis D, Konstantopoulos K. Pharmaceutical Prevention and Management of Cardiotoxicity in Hematological Malignancies. Pharmaceuticals (Basel) 2022; 15:ph15081007. [PMID: 36015155 PMCID: PMC9412591 DOI: 10.3390/ph15081007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/26/2022] [Accepted: 07/30/2022] [Indexed: 11/16/2022] Open
Abstract
Modern treatment modalities in hematology have improved clinical outcomes of patients with hematological malignancies. Nevertheless, many new or conventional anticancer drugs affect the cardiovascular system, resulting in various cardiac disorders, including left ventricular dysfunction, heart failure, arterial hypertension, myocardial ischemia, cardiac rhythm disturbances, and QTc prolongation on electrocardiograms. As these complications may jeopardize the significantly improved outcome of modern anticancer therapies, it is crucial to become familiar with all aspects of cardiotoxicity and provide appropriate care promptly to these patients. In addition, established and new drugs contribute to primary and secondary cardiovascular diseases prevention. This review focuses on the clinical manifestations, preventive strategies, and pharmaceutical management of cardiotoxicity in patients with hematologic malignancies undergoing anticancer drug therapy or hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
| | | | - Maria Vardaka
- Department of Hematology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | | | - Apostolos Karavidas
- Department of Cardiology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | - Theodoros Marinakis
- Department of Hematology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | | | - Panagiotis Makaronis
- Department of Cardiology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | - Charikleia Vlachou
- Department of Hematology, “G. Gennimatas” General Hospital, 11527 Athens, Greece
| | - Marina Mantzourani
- First Department of Internal Medicine, Medical School, National and Kapodistrian University of Athens, “Laiko” General Hospital, 11527 Athens, Greece
| | | | - Konstantinos Konstantopoulos
- Department of Hematology, Medical School, National and Kapodistrian University of Athens, “Laiko” General Hospital, 11527 Athens, Greece
| |
Collapse
|
15
|
Wu S, Bai H, Zhang L, He J, Luo X, Wang S, Fan G, Sun N. Cardiovascular adverse events induced by immune checkpoint inhibitors: A real world study from 2018 to 2022. Front Cardiovasc Med 2022; 9:969942. [PMID: 36035942 PMCID: PMC9399397 DOI: 10.3389/fcvm.2022.969942] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 07/13/2022] [Indexed: 12/19/2022] Open
Abstract
Background The reported rate of cardiovascular adverse events (CAE) caused by immune checkpoint inhibitors (ICI) is low but potentially fatal. Assess the risk of CAE in cancer patients and compare the incidence of CAE between Chinese developed ICIs and imported ICIs. Methods A retrospective analysis was performed on cancer patients treated with ICI for at least four cycles in the Second Affiliated Hospital of Dalian Medical University from January 2018 to March 2022. Baseline characteristics, physiological and biochemical values, electrocardiographic and echocardiographic findings were compared between patients with and without CAE. Results Among 495 patients treated with ICIs, CAEs occurred in 64 patients (12.93%). The median time to the event was 105 days (61–202). The patients with low neutrophil-to-lymphocyte ratio (L-NLR) were significantly associated with the risk of developing CAE (hazard ratio HR 3.64, 95% confidence ratio CI 1.86–7.15, P = 0.000). Patients with higher comorbidity burden significantly increased the risk of developing CAE (HR 1.30, 95% CI 1.05–1.61, P = 0.014). Those who received a combination of ICI and vascular endothelial growth factor receptor (VEGFR) inhibitors (HR 2.57, 95% CI 1.37–4.84, P = 0.003) or thoracic radiation therapy (HR 32.93, 95% CI 8.81–123.14, P = 0.000) were at a significantly increased risk of developing CAE. Compared to baseline values, creatine kinase is -oenzymes (CK-MB) (95% CI -9.73 to -2.20, P = 0.003) and cardiac troponin I (cTnI) (95% CI -1.06 to -0.06, P = 0.028) were elevated, and the QTc interval prolonged (95% CI -27.07 to -6.49, P = 0.002). Using nivolumab as a control, there was no difference in CAE risk among the eight ICIs investigated. However, the results of the propensity matching showed that programmed death-ligand 1 (PD-L1) inhibitors had lower CAE occurrence compared with programmed cell death protein 1 (PD-1) inhibitors (adjusted HR = 0.38, P = 0.045). Conclusion Patients who received concurrent VEGFR inhibitors and ICIs had a history of thoracic radiation therapy, L-NLR, and higher comorbidity burden had an increased risk of CAEs. Elevated cTnI, CK-MB, and QTc, can be used to monitor CAEs. There was no significant difference in CAE risks between Chinese domestic and imported ICIs. PD-L1 inhibitors had lower CAE occurrence than PD-1 inhibitors.
Collapse
|
16
|
Sorodoc V, Sirbu O, Lionte C, Haliga RE, Stoica A, Ceasovschih A, Petris OR, Constantin M, Costache II, Petris AO, Morariu PC, Sorodoc L. The Value of Troponin as a Biomarker of Chemotherapy-Induced Cardiotoxicity. LIFE (BASEL, SWITZERLAND) 2022; 12:life12081183. [PMID: 36013362 PMCID: PMC9410123 DOI: 10.3390/life12081183] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 11/16/2022]
Abstract
In cancer survivors, cardiac dysfunction is the main cause of mortality. Cardiotoxicity represents a decline in cardiac function associated with cancer therapy, and the risk factors include smoking, dyslipidemia, an age of over 60 years, obesity, and a history of coronary artery disease, diabetes, atrial fibrillation, or heart failure. Troponin is a biomarker that is widely used in the detection of acute coronary syndromes. It has a high specificity, although it is not exclusively associated with myocardial ischemia. The aim of this paper is to summarize published studies and to establish the role of troponin assays in the diagnosis of cardiotoxicity associated with various chemotherapeutic agents. Troponin has been shown to be a significant biomarker in the diagnosis of the cardiac dysfunction associated with several types of chemotherapeutic drugs: anthracyclines, anti-human epidermal growth factor receptor 2 treatment, and anti-vascular endothelial growth factor therapy. Based on the data available at this moment, troponin is useful for baseline risk assessment, the diagnosis of cardiotoxicity, and as a guide for the initiation of cardioprotective treatment. There are currently clear regulations regarding the timing of troponin surveillance depending on the patient’s risk of cardiotoxicity and the type of medication administered, but data on the cut-off values of this biomarker are still under investigation.
Collapse
Affiliation(s)
- Victorita Sorodoc
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Oana Sirbu
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Correspondence: (O.S.); (C.L.)
| | - Catalina Lionte
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Correspondence: (O.S.); (C.L.)
| | - Raluca Ecaterina Haliga
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Alexandra Stoica
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Alexandr Ceasovschih
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Ovidiu Rusalim Petris
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Mihai Constantin
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| | - Irina Iuliana Costache
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Department of Cardiology, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania
| | - Antoniu Octavian Petris
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
- Department of Cardiology, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania
| | - Paula Cristina Morariu
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
| | - Laurentiu Sorodoc
- Department of Internal Medicine, Clinical Emergency Hospital Sfântul Spiridon, 700111 Iasi, Romania; (V.S.); (R.E.H.); (A.S.); (A.C.); (O.R.P.); (M.C.); (P.C.M.); (L.S.)
- Faculty of Medicine, University of Medicine and Pharmacy Grigore T. Popa, 16 Universitatii Street, 700115 Iasi, Romania; (I.I.C.); (A.O.P.)
| |
Collapse
|
17
|
Cardiotoxicity of Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: Pathophysiology, Clinical Implications, and Echocardiographic Assessment. Int J Mol Sci 2022; 23:ijms23158242. [PMID: 35897819 PMCID: PMC9368621 DOI: 10.3390/ijms23158242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 07/18/2022] [Accepted: 07/23/2022] [Indexed: 02/04/2023] Open
Abstract
Contemporary anticancer immunotherapy with chimeric antigen receptor T-cell (CAR-T) therapy has dramatically changed the treatment of many hematologic malignancies previously associated with poor prognosis. The clinical improvement and the survival benefit unveiled the risk of cardiotoxicity, ranging from minimal effects to severe cardiac adverse events, including death. Immunotherapy should also be proposed even in patients with pre-existing cardiovascular risk factors, thereby increasing the potential harm of cardiotoxicity. CAR-T therapy frequently results in cytokine release syndrome (CRS), and inflammatory activation is sustained by circulating cytokines that foster a positive feedback mechanism. Prompt diagnosis and treatment of CAR-T cardiotoxicity might significantly improve outcomes and reduce the burden associated with cardiovascular complications. Clinical and echocardiographic examinations are crucial to perform a tailored evaluation and follow-up during CAR-T treatment. This review aims to summarize the pathophysiology, clinical implications, and echocardiographic assessment of CAR-T-related cardiotoxicity to enlighten new avenues for future research.
Collapse
|
18
|
Cancer du rein métastatique : gestion des toxicités des combinaisons. Bull Cancer 2022; 109:844-861. [DOI: 10.1016/j.bulcan.2022.04.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 11/23/2022]
|
19
|
Stein-Merlob AF, Ganatra S, Yang EH. T-cell Immunotherapy and Cardiovascular Disease: Chimeric Antigen Receptor T-cell and Bispecific T-cell Engager Therapies. Heart Fail Clin 2022; 18:443-454. [PMID: 35718418 DOI: 10.1016/j.hfc.2022.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell and bispecific T-cell engager (BiTE) therapies have revolutionized the treatment of refractory or relapsed leukemia and lymphoma. Increased use of these therapies has revealed signals of significant cardiotoxicity, including cardiomyopathy/heart failure, arrhythmia, myocardial injury, hemodynamic instability, and cardiovascular death mainly in the context of a profound inflammatory response to CAR T-cell antitumor effects known as cytokine release syndrome (CRS). Preexisting cardiovascular risk factors and disease may increase the risk of such cardiotoxicity. High index of suspicion and close monitoring is required for prompt recognition. Supportive hemodynamic care and targeted anti-IL-6 therapy, as well as possibly broader immunosuppression with corticosteroids, are the cornerstones of the management.
Collapse
Affiliation(s)
- Ashley F Stein-Merlob
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA; Division of Cardiology, Department of Medicine, UCLA-Cardio-Oncology Program, University of California at Los Angeles, Los Angeles, CA, USA. https://twitter.com/A_SteinMerlob
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA.
| | - Eric H Yang
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA, USA; Division of Cardiology, Department of Medicine, UCLA-Cardio-Oncology Program, University of California at Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
20
|
Patel A, Levenson J, Huang Z, Agha M, Dorritie K. CD-19 CART therapy and orthostatic hypotension: a single center retrospective cohort study. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2022; 8:6. [PMID: 35382903 PMCID: PMC8981866 DOI: 10.1186/s40959-022-00132-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 03/07/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND Chimeric antigen receptor T-cell (CART) therapy is a form of cellular immunotherapy used to treat hematologic malignancies. Major adverse cardiovascular events have been seen in CART patients who have high grade CRS, higher baseline creatinine, and troponin elevation. However, the incidence and factors associated with orthostatic hypotension after CART therapy have not previously been reported in the literature. METHODS We looked at patients who underwent CD-19 directed CART therapy at UPMC Shadyside hospital from April 1st 2018 to December 1st 2020. Patients were classified as having orthostatic hypotension if they had recorded orthostatic vital signs that were positive or provider notes indicated that vitals had been taken and were positive in the time period from discharge to 3 months post-CART. Data was analyzed with univariate and multivariate analysis using logistic regression. RESULTS 79% of patients had orthostatic hypotension after discharge from their CART hospitalization and 64% of those patients were symptomatic. Older age, lower BMI, lower ambulatory diastolic blood pressure and grade 2 CRS were associated with orthostatic hypotension in the univariate analysis. Older age and lower ambulatory systolic blood pressure were associated with orthostatic hypotension in the multivariate analysis. Symptomatic orthostatic hypotension was associated with a history of hypertension in both the univariate and multivariate analysis. Patients with symptoms also had a higher pre-CART ejection fraction but this association was not seen in the regression model. CONCLUSION There is a high incidence of orthostatic hypotension after CART therapy even after discharge. Therefore, orthostatic vitals signs and associated symptoms should be assessed in both the inpatient and outpatient setting. Older patients and patients with lower BMIs, lower ambulatory blood pressures, grade 2 CRS, or a history of hypertension may need closer monitoring.
Collapse
Affiliation(s)
- Ashish Patel
- Division of General Internal Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA.
| | - Joshua Levenson
- Division of Cardiology, UPMC Heart and Vascular Institute, University of Pittsburgh, Pittsburgh, PA, USA
| | - Ziyu Huang
- Biostatistics Facility, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Mounzer Agha
- Division of Hematology and Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kathleen Dorritie
- Division of Hematology and Oncology, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Herrmann J, Lenihan D, Armenian S, Barac A, Blaes A, Cardinale D, Carver J, Dent S, Ky B, Lyon AR, López-Fernández T, Fradley MG, Ganatra S, Curigliano G, Mitchell JD, Minotti G, Lang NN, Liu JE, Neilan TG, Nohria A, O'Quinn R, Pusic I, Porter C, Reynolds KL, Ruddy KJ, Thavendiranathan P, Valent P. Defining cardiovascular toxicities of cancer therapies: an International Cardio-Oncology Society (IC-OS) consensus statement. Eur Heart J 2022; 43:280-299. [PMID: 34904661 PMCID: PMC8803367 DOI: 10.1093/eurheartj/ehab674] [Citation(s) in RCA: 243] [Impact Index Per Article: 121.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/28/2021] [Accepted: 09/10/2021] [Indexed: 12/16/2022] Open
Abstract
The discipline of Cardio-Oncology has seen tremendous growth over the past decade. It is devoted to the cardiovascular (CV) care of the cancer patient, especially to the mitigation and management of CV complications or toxicities of cancer therapies, which can have profound implications on prognosis. To that effect, many studies have assessed CV toxicities in patients undergoing various types of cancer therapies; however, direct comparisons have proven difficult due to lack of uniformity in CV toxicity endpoints. Similarly, in clinical practice, there can be substantial differences in the understanding of what constitutes CV toxicity, which can lead to significant variation in patient management and outcomes. This document addresses these issues and provides consensus definitions for the most commonly reported CV toxicities, including cardiomyopathy/heart failure and myocarditis, vascular toxicity, and hypertension, as well as arrhythmias and QTc prolongation. The current document reflects a harmonizing review of the current landscape in CV toxicities and the definitions used to define these. This consensus effort aims to provide a structure for definitions of CV toxicity in the clinic and for future research. It will be important to link the definitions outlined herein to outcomes in clinical practice and CV endpoints in clinical trials. It should facilitate communication across various disciplines to improve clinical outcomes for cancer patients with CV diseases.
Collapse
Affiliation(s)
- Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, MN 55902, USA
| | - Daniel Lenihan
- International Cardio-Oncology Society, 465 Lucerne Ave., Tampa, FL 33606, USA
| | - Saro Armenian
- City of Hope Comprehensive Cancer Center, Department of Population Sciences, 500 E Duarte Rd, Duarte, CA 91010, USA
| | - Ana Barac
- MedStar Heart and Vascular Institute, Georgetown University, 10 Irving Street Northwest Suite NW, Washington, DC 20010, USA
| | - Anne Blaes
- University of Minnesota, Division of Hematology/Oncology, 420 Delaware Street SE, Minneapolis, MN 55455, USA
| | - Daniela Cardinale
- Cardioncology Unit, European Institute of Oncology, IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Joseph Carver
- Abraham Cancer Center, University of Pennsylvania, Philadelphia, 3400 Civic Center Boulevard, Pavilion 2nd Floor, Philadelphia, PA 19104, USA
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, 20 Duke Medicine Circle, Durham, NA 27704, USA
| | - Bonnie Ky
- Division of Cardiology, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Imperial College, Sydney St, London SW3 6NP, United Kingdom
| | - Teresa López-Fernández
- Division of Cardiology; Cardiac Imaging and Cardio-Oncology Unit; La Paz University Hospital, IdiPAZ Research Institute, CIBER CV, C. de Pedro Rico, 6, 28029 Madrid, Spain
| | - Michael G Fradley
- Division of Cardiology, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Department of Cardiovascular Medicine, Lahey Hospital and Medical Center, 41 Burlington Mall Road, Burlington, MA 01805, USA
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milano, Via Festa del Perdono 7. 20122 Milano, Italy
- European Institute of Oncology, IRCCS, Via Adamello 16, 20139 Milan, Italy
| | - Joshua D Mitchell
- Cardio-Oncology Center of Excellence, Washington University, 4921 Parkview Pl, St. Louis, MO 63110, USA
| | - Giorgio Minotti
- Department of Medicine, University Campus Bio-Medico, Via Álvaro del Portillo, 21, 00128 Roma, Italy
| | - Ninian N Lang
- British Heart Foundation Centre for Cardiovascular Sciences, University of Glasgow, 126 University Place, Glasgow, G12 8TA Scotland, United Kingdom
| | - Jennifer E Liu
- Memorial Sloan Kettering Cancer Center, Department of Medicine/Cardiology Service, 1275 York Ave, New York, NY 10065, USA
| | - Tomas G Neilan
- Cardio-oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
| | - Anju Nohria
- Cardio-Oncology Program, Brigham and Women’s Hospital and Dana Farber Cancer Institute, Harvard Medical School, 25 Shattuck Street, Boston, MA 02115, USA
| | - Rupal O'Quinn
- Division of Cardiology, University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA 19104, USA
| | - Iskra Pusic
- Washington University School of Medicine, Division of Oncology, 4921 Parkview Place, St. Louis, MO 63110, USA
| | - Charles Porter
- Cardiovascular Medicine, Cardio-Oncology Unit, University of Kansas Medical Center, 4000 Cambridge Street, Kansas City, KS 66160, USA
| | - Kerry L Reynolds
- Massachusetts General Hospital Cancer Center, Harvard Medical School, 55 Fruit St, Boston, MA 02114, USA
| | - Kathryn J Ruddy
- Department of Oncology, Mayo Clinic, 200 First Street SW, Rochester, MN, 55902, USA
| | - Paaladinesh Thavendiranathan
- Department of Medicine, Division of Cardiology, Ted Rogers Program in Cardiotoxicity Prevention, Peter Munk Cardiac Centre, University Health Network, University of Toronto, 585 University Ave, Toronto, ON M5G 2N2, Canada
| | - Peter Valent
- Department of Internal Medicine I, Division of Hematology and Hemostaseology and Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
22
|
Adhikari A, Asdaq SMB, Al Hawaj MA, Chakraborty M, Thapa G, Bhuyan NR, Imran M, Alshammari MK, Alshehri MM, Harshan AA, Alanazi A, Alhazmi BD, Sreeharsha N. Anticancer Drug-Induced Cardiotoxicity: Insights and Pharmacogenetics. Pharmaceuticals (Basel) 2021; 14:ph14100970. [PMID: 34681194 PMCID: PMC8539940 DOI: 10.3390/ph14100970] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/09/2021] [Accepted: 09/21/2021] [Indexed: 12/29/2022] Open
Abstract
The advancement in therapy has provided a dramatic improvement in the rate of recovery among cancer patients. However, this improved survival is also associated with enhanced risks for cardiovascular manifestations, including hypertension, arrhythmias, and heart failure. The cardiotoxicity induced by chemotherapy is a life-threatening consequence that restricts the use of several chemotherapy drugs in clinical practice. This article addresses the prevalence of cardiotoxicity mediated by commonly used chemotherapeutic and immunotherapeutic agents. The role of susceptible genes and radiation therapy in the occurrence of cardiotoxicity is also reviewed. This review also emphasizes the protective role of antioxidants and future perspectives in anticancer drug-induced cardiotoxicities.
Collapse
Affiliation(s)
- Archana Adhikari
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Syed Mohammed Basheeruddin Asdaq
- Department of Pharmacy Practice, College of Pharmacy, AlMaarefa University, Dariyah, Riyadh 13713, Saudi Arabia
- Correspondence: (S.M.B.A.); (M.C.)
| | - Maitham A. Al Hawaj
- Department of Pharmacy Practice, College of Clinical Pharmacy, King Faisal University, Hofuf 31982, Saudi Arabia;
| | - Manodeep Chakraborty
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
- Correspondence: (S.M.B.A.); (M.C.)
| | - Gayatri Thapa
- Pharmacology Department, Himalayan Pharmacy Institute Majhitar, Rangpo 737136, Sikkim, India; (A.A.); (G.T.)
| | - Nihar Ranjan Bhuyan
- Department of Pharmaceutical Analysis, Himalayan Pharmacy Institute, Majhitar, Rangpo 737136, Sikkim, India;
| | - Mohd. Imran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Northern Border University, Rafha 91911, Saudi Arabia;
| | | | - Mohammed M. Alshehri
- Pharmaceutical Care Department, Ministry of National Guard-Health Affairs, Riyadh 11426, Saudi Arabia;
| | - Aishah Ali Harshan
- Department of Pharmaceutical Care, Northern Area Armed Forces Hospital, King Khalid Military City Hospital, Hafr Al-Batin 39745, Saudi Arabia;
| | - Abeer Alanazi
- Department of Pharmaceutical Care, First Health Cluster in Eastern Province, King Fahad Specialist Hospital, Dammam 32253, Saudi Arabia;
| | | | - Nagaraja Sreeharsha
- Department of Pharmaceutical Sciences, College of Clinical Pharmacy, King Faisal University, Al-Ahsa-31982, Saudi Arabia;
- Department of Pharmaceutics, Vidya Siri College of Pharmacy, Off Sarjapura Road, Bengaluru 560035, Karnataka, India
| |
Collapse
|
23
|
Mechanisms and Insights for the Development of Heart Failure Associated with Cancer Therapy. CHILDREN-BASEL 2021; 8:children8090829. [PMID: 34572260 PMCID: PMC8468170 DOI: 10.3390/children8090829] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 09/11/2021] [Accepted: 09/18/2021] [Indexed: 01/01/2023]
Abstract
Cardiotoxicity is a well-recognized late effect among childhood cancer survivors. With various pediatric cancers becoming increasingly curable, it is imperative to understand the disease burdens that survivors may face in the future. In order to prevent or mitigate cardiovascular complications, we must first understand the mechanistic underpinnings. This review will examine the underlying mechanisms of cardiotoxicity that arise from traditional antineoplastic chemotherapies, radiation therapy, hematopoietic stem cell transplantation, as well as newer cellular therapies and targeted cancer therapies. We will then propose areas for prevention, primarily drawing from the anthracycline-induced cardiotoxicity literature. Finally, we will explore the role of human induced pluripotent stem cell cardiomyocytes and genetics in advancing the field of cardio-oncology.
Collapse
|
24
|
Cardiotoxicity associated with immune checkpoint inhibitors and CAR T-cell therapy. Am J Emerg Med 2021; 50:51-58. [PMID: 34274878 DOI: 10.1016/j.ajem.2021.07.014] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/01/2021] [Accepted: 07/02/2021] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION The expanding use of immunotherapy and the growing population of patients with cancer has led to an increase in the reporting of immune related adverse events (irAEs). The emergency clinician should be aware of these emerging toxicities, some of which can be fatal. In this review we discuss the cardiotoxic side effects of immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T-cell (CAR T-cell) therapy. DISCUSSION Recognizing the possible presentations of cardiotoxic irAEs is of utmost important as the diagnosis of cardiotoxicity associated with ICI and CAR T-cell can be difficult to make in the emergency department. The emergency clinician will have to presume the diagnosis and treat it without final confirmation in most cases. For this reason, if the diagnosis is suspected, early involvement of the cardiologist and oncologist is important to help guide management. Most irAEs will be treated with glucocorticoids, but in the case of CAR T-cell cardiotoxicity, Tocilizumab should be used as first line. CONCLUSION Although cardiotoxicity is rare, it is often life-threatening. Treatment should be initiated as soon as the diagnosis is suspected, and early involvement of the cardiologist and oncologist is imperative for optimal treatment.
Collapse
|
25
|
Borges GSM, Lages EB, Sicard P, Ferreira LAM, Richard S. Nanomedicine in Oncocardiology: Contribution and Perspectives of Preclinical Studies. Front Cardiovasc Med 2021; 8:690533. [PMID: 34277738 PMCID: PMC8277942 DOI: 10.3389/fcvm.2021.690533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 06/01/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer and cardiovascular diseases are the leading causes of death and morbidity worldwide. Strikingly, cardiovascular disorders are more common and more severe in cancer patients than in the general population, increasing incidence rates. In this context, it is vital to consider the anticancer efficacy of a treatment and the devastating heart complications it could potentially cause. Oncocardiology has emerged as a promising medical and scientific field addressing these aspects from different angles. Interestingly, nanomedicine appears to have great promise in reducing the cardiotoxicity of anticancer drugs, maintaining or even enhancing their efficacy. Several studies have shown the benefits of nanocarriers, although with some flaws when considering the concept of oncocardiology. Herein, we discuss how preclinical studies should be designed as closely as possible to clinical protocols, considering various parameters intrinsic to the animal models used and the experimental protocols. The sex and age of the animals, the size and location of the tumors, the doses of the nanoformulations administered, and the acute vs. the long-term effects of treatments are essential aspects. We also discuss the perspectives offered by non-invasive imaging techniques to simultaneously assess both the anticancer effects of treatment and its potential impact on the heart. The overall objective is to accelerate the development and validation of nanoformulations through high-quality preclinical studies reproducing the clinical conditions.
Collapse
Affiliation(s)
- Gabriel Silva Marques Borges
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Eduardo Burgarelli Lages
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.,PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France
| | - Pierre Sicard
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France.,IPAM, BioCampus, CNRS, INSERM, Université de Montpellier, Montpellier, France
| | - Lucas Antônio Miranda Ferreira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Sylvain Richard
- PhyMedExp, Université de Montpellier, INSERM, CNRS, Montpellier, France.,IPAM, BioCampus, CNRS, INSERM, Université de Montpellier, Montpellier, France
| |
Collapse
|
26
|
Ortega Pérez R, Pardo Sanz A, García Gutiérrez V, Zamorano Gómez JL. Cardiovascular Oncology: The Need for a Multidisciplinary Approach. Semin Thromb Hemost 2021; 47:907-913. [PMID: 34154001 DOI: 10.1055/s-0041-1726297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Improved cancer survivorship has led to an increase in cardiovascular (CV) complications in the oncologic population, mainly associated with therapeutic regimens. Hence, cardio-oncology has grown toward unifying the cancer care process in which the best prevention, early detection, treatment, and CV surveillance are offered to patients. This multidisciplinary approach allows us to optimize and agree upon clinical decisions to enhance clinical outcomes. Atrial fibrillation is one of the hot topics in the field because it is still challenging in cancer patients. The optimal antithrombotic therapy remains unclear. Nevertheless, evidence supports that specific recommendations are needed due to a hemorrhagic/thrombotic disbalance present within this subgroup of patients and a low rate of anticoagulation treatments compared with the general population. Further, cardiotoxicity management is currently transforming. Increasingly, early detection of subclinical alterations is raising awareness. When medical therapy is initiated early, fewer patients progress to ventricular dysfunction and the rate of patients completing cancer therapy gradually increases. New approaches are demonstrating better outcomes and these strategies will expectedly be established in clinical practice. Cardio-oncology enables us to find the best balance between cancer treatment and CV health protection. Nowadays, more and more physicians are being instructed in this discipline, which gradually exhibits a greater presence in conferences and scientific journals. However, given the need for physicians thoroughly trained in cardio-oncology, this subspecialty must be promoted further.
Collapse
Affiliation(s)
| | - Ana Pardo Sanz
- Department of Cardiology, University Hospital Ramón y Cajal, Madrid, Spain
| | | | | |
Collapse
|
27
|
Patel RP, Parikh R, Gunturu KS, Tariq RZ, Dani SS, Ganatra S, Nohria A. Cardiotoxicity of Immune Checkpoint Inhibitors. Curr Oncol Rep 2021; 23:79. [PMID: 33937956 PMCID: PMC8088903 DOI: 10.1007/s11912-021-01070-6] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 12/25/2022]
Abstract
Purpose of Review Immune checkpoint inhibitors (ICIs) have improved the survival of several cancers. However, they may cause a wide range of immune-related adverse events (irAEs). While most irAEs are manageable with temporary cessation of ICI and immunosuppression, cardiovascular toxicity can be associated with high rates of morbidity and mortality. As ICIs evolve to include high-risk patients with preexisting cardiovascular risk factors and disease, the risk and relevance of ICI-associated cardiotoxicity may be even higher. Recent Findings Several cardiovascular toxicities such as myocarditis, stress cardiomyopathy, and pericardial disease have been reported in association with ICIs. Recent findings also suggest an increased risk of atherosclerosis with ICI use. ICI-associated myocarditis usually occurs early after initiation and can be fulminant. A high index of suspicion is required for timely diagnosis. Prompt treatment with high-dose corticosteroids is shown to improve outcomes. Summary Although the overall incidence is rare, ICI cardiotoxicity, particularly myocarditis, is associated with significant morbidity and mortality, making it a major therapy-limiting adverse event. Early recognition and prompt treatment with the cessation of ICI therapy and initiation of high-dose corticosteroids are crucial to improve outcomes. Cardio-oncologists will need to play an important role not just in the management of acute cardiotoxicity but also to reduce the risk of long-term sequelae.
Collapse
Affiliation(s)
- Rushin P Patel
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Rohan Parikh
- Vascular Medicine Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Krishna S Gunturu
- Cancer Survivorship Program, Division of Hematology Oncology, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Rana Zouveenoor Tariq
- Division of Cardiovascular Medicine, Indiana University Health, Bloomington, IN, USA
| | - Sourbha S Dani
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA, USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA, 01805, USA.
| | - Anju Nohria
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|
28
|
Suero-Abreu GA, Ganatra S, Neilan TG. Cardiotoxicity Monitoring in Patients With Cancer: Focus on Safety and Clinical Relevance. JCO Oncol Pract 2021; 17:237-239. [PMID: 33793304 PMCID: PMC8258139 DOI: 10.1200/op.21.00075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 02/12/2021] [Indexed: 11/20/2022] Open
Affiliation(s)
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA
| | - Tomas G. Neilan
- Cardiovascular Imaging Research Center (CIRC) and Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
29
|
Chhabra N, Kennedy J. A Review of Cancer Immunotherapy Toxicity: Immune Checkpoint Inhibitors. J Med Toxicol 2021; 17:411-424. [PMID: 33826117 DOI: 10.1007/s13181-021-00833-8] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 02/08/2021] [Accepted: 02/16/2021] [Indexed: 02/07/2023] Open
Abstract
Cancer immunotherapy, which leverages features of the immune system to target neoplastic cells, has revolutionized the treatment of cancer. The use of these therapies has rapidly expanded in the past two decades. Immune checkpoint inhibitors represent one drug class within immunotherapy with its first agent FDA-approved in 2011. Immune checkpoint inhibitors act by disrupting inhibitory signals from neoplastic cells to immune effector cells, allowing activated T-cells to target these neoplastic cells. Unique adverse effects associated with immune checkpoint inhibitors are termed immune-related adverse effects (irAEs) and are usually immunostimulatory in nature. Almost all organ systems may be affected by irAEs including the dermatologic, gastrointestinal, pulmonary, endocrine, and cardiovascular systems. These effects range from mild to life-threatening, and their onset can be delayed several weeks or months. For mild irAEs, symptomatic care is usually sufficient. For higher grade irAEs, discontinuation of therapy and initiation of immunosuppressive therapy may be necessary. The management of patients with irAEs involves multidisciplinary care coordination with respect to the long-term goals the individual patient. Clinicians must be aware of the unique and sometimes fatal toxicologic profiles associated with immunotherapies to ensure prompt diagnosis and appropriate management.
Collapse
Affiliation(s)
- Neeraj Chhabra
- Cook County Health, Department of Emergency Medicine, Division of Medical Toxicology, 1950 W Polk Street, 7th Floor, Chicago, IL, 60612, USA. .,Toxikon Consortium, Chicago, IL, USA.
| | - Joseph Kennedy
- Cook County Health, Department of Emergency Medicine, Division of Medical Toxicology, 1950 W Polk Street, 7th Floor, Chicago, IL, 60612, USA.,Toxikon Consortium, Chicago, IL, USA
| |
Collapse
|
30
|
George AS, Fernandez CJ, Eapen D, Pappachan JM. Organ-specific Adverse Events of Immune Checkpoint Inhibitor Therapy, with Special Reference to Endocrinopathies. TOUCHREVIEWS IN ENDOCRINOLOGY 2021; 17:21-32. [PMID: 35118443 PMCID: PMC8320015 DOI: 10.17925/ee.2021.17.1.21] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 02/09/2021] [Indexed: 02/05/2023]
Abstract
Immune checkpoint inhibitors are potent and promising immunotherapeutic agents that are increasingly used for the management of various types of advanced cancers. The widespread approval of this group of drugs simultaneously revealed immune-related adverse events as unique side-effects. Endocrinopathies are one of the most common immune-related adverse events. The precise pathogenic mechanisms for these endocrinopathies are still unclear. Though few of the endocrinopathies are reversible, calling for only symptom control, most are irreversible, requiring multiple long-term hormone replacement therapies. However, in contrast to other organ-specific immune-related adverse events, patients with endocrinopathies can continue their immune checkpoint therapy, provided the hormone replacement therapy is adequate and the symptoms are controlled. Though patients who have developed immune-related adverse events demonstrate superior antitumor activity and overall survival, due to the high morbidity associated with the immune-related adverse events, researchers are trying to uncouple the antitumour activity associated with immune checkpoint inhibitor therapy from the immune-related adverse events, to preserve antitumour activity without adverse events.
Collapse
Affiliation(s)
- Annu Susan George
- Department of Medical Oncology, Lakeshore Hospital, Cochin, Kerala, India
| | - Cornelius J Fernandez
- Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK
| | - Dilip Eapen
- Department of Endocrinology, Pilgrim Hospital, United Lincolnshire Hospitals NHS Trust, Boston, UK
| | - Joseph M Pappachan
- Department of Endocrinology & Metabolism, Lancashire Teaching Hospitals NHS Trust, Preston, UK
- Manchester Metropolitan University, Manchester, UK
- The University of Manchester, Manchester, UK
| |
Collapse
|
31
|
Kichloo A, Albosta M, Dahiya D, Guidi JC, Aljadah M, Singh J, Shaka H, Wani F, Kumar A, Lekkala M. Systemic adverse effects and toxicities associated with immunotherapy: A review. World J Clin Oncol 2021; 12:150-163. [PMID: 33767971 PMCID: PMC7968107 DOI: 10.5306/wjco.v12.i3.150] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/21/2021] [Accepted: 02/20/2021] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy is rapidly evolving secondary to the advent of newer immunotherapeutic agents and increasing approval of the current agents by the United States Food and Drug Administration to treat a wide spectrum of cancers. Immunotherapeutic agents have gained immense popularity due to their tumor-specific action. Immunotherapy is slowly transforming into a separate therapeutic entity, and the fifth pillar of management for cancers alongside surgery, radiotherapy, chemotherapy, and targeted therapy. However, like any therapeutic entity it has its own adverse effects. With the increasing use of immuno-therapeutic agents, it is vital for physicians to acquaint themselves with these adverse effects. The aim of this review is to investigate the common systemic adverse effects and toxicities associated with the use of different classes of immunotherapeutic agents. We provide an overview of potential adverse effects and toxicities associated with different classes of immunotherapeutic agents organized by organ systems, as well as an extensive discussion of the current recommendations for treatment and clinical trial data. As we continue to see increasing usage of these agents in clinical practice, it is vital for physicians to familiarize themselves with these effects.
Collapse
Affiliation(s)
- Asim Kichloo
- Department of Internal Medicine, Central Michigan University, Saginaw, MI 48603, United States
- Department of Internal Medicine, Samaritan Medical Center, Watertown, NY 13601, United States
| | - Michael Albosta
- Department of Internal Medicine, Central Michigan University, Saginaw, MI 48603, United States
| | - Dushyant Dahiya
- Department of Internal Medicine, Central Michigan University, Saginaw, MI 48603, United States
| | - Jean Claude Guidi
- Department of Internal Medicine, Samaritan Medical Center, Watertown, NY 13601, United States
| | - Michael Aljadah
- Department of Internal Medicine, Medical College of Wisconsin, Milwaukee, WI 53201, United States
| | - Jagmeet Singh
- Department of Nephrology, Guthrie Robert Packer Hospital, Sayre, PA 18840, United States
| | - Hafeez Shaka
- Department of Internal Medicine, John H Stroger Jr. Hospital of Cook County, Chicago, IL 60612, United States
| | - Farah Wani
- Department of Family Medicine, Samaritan Medical Center, Watertown, NY 13601, United States
| | - Akshay Kumar
- Department of Surgery, University of Pittsburgh, Pittsburgh, PA 15213, United States
| | - Manidhar Lekkala
- Department of Hematology and Oncology, University of Rochester Medical Center, Rochester, NY 14642, United States
| |
Collapse
|
32
|
Burns EA, Gentille C, Trachtenberg B, Pingali SR, Anand K. Cardiotoxicity Associated with Anti-CD19 Chimeric Antigen Receptor T-Cell (CAR-T) Therapy: Recognition, Risk Factors, and Management. Diseases 2021; 9:20. [PMID: 33802788 PMCID: PMC8006027 DOI: 10.3390/diseases9010020] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 03/04/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Chimeric antigen receptor T-cells (CAR-T) are improving outcomes in pediatric and adult patients with relapsed or refractory B-cell acute lymphoblastic leukemias and subtypes of non-Hodgkin Lymphoma. As this treatment is being increasingly utilized, a better understanding of the unique toxicities associated with this therapy is warranted. While there is growing knowledge on the diagnosis and treatment of cytokine release syndrome (CRS), relatively little is known about the associated cardiac events that occur with CRS that may result in prolonged length of hospital stay, admission to the intensive care unit for pressor support, or cardiac death. This review focuses on the various manifestations of cardiotoxicity, potential risk factors, real world and clinical trial data on prevalence of reported cardiotoxicity events, and treatment recommendations.
Collapse
Affiliation(s)
- Ethan A. Burns
- Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA; (C.G.); (S.R.P.)
| | - Cesar Gentille
- Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA; (C.G.); (S.R.P.)
| | - Barry Trachtenberg
- Houston Methodist DeBakey Heart and Vascular Center, 6565 Fannin St, Houston, TX 77030, USA;
| | - Sai Ravi Pingali
- Houston Methodist Cancer Center, Houston Methodist Hospital, 6445 Main Street, Outpatient Center, 24th Floor, Houston, TX 77030, USA; (C.G.); (S.R.P.)
| | - Kartik Anand
- Callahan Cancer Center, Great Plains Health, 601 W Leota St, North Platte, NE 69101, USA
| |
Collapse
|
33
|
Compton F, He L, Sarode R, Wodajo A, Usmani A, Burner J, Berlacher M, De Simone N. Immune checkpoint inhibitor toxicity: A new indication for therapeutic plasma exchange? J Clin Apher 2021; 36:645-648. [PMID: 33648026 DOI: 10.1002/jca.21890] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/04/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022]
Abstract
Novel immune-modulating anticancer drugs are being used with increasing frequency. With increased use, there are more frequent cases of toxicities caused by these drugs, termed immune-related adverse events (irAEs). We present a case in which we successfully treated a case of severe, steroid-refractory, nivolumab-induced myocarditis with therapeutic plasma exchange (TPE). Nivolumab is an immune checkpoint inhibitor (ICI) which blocks programmed death receptor-1 (PD-1). This blockade allows for enhanced T-cell function and increased anti-tumor response. The patient presented with signs and symptoms of heart failure and was found to have a significantly depressed cardiac ejection fraction. Over the course of her five TPE procedures, she improved clinically and was discharged home with improved left ventricular ejection function. This case suggests an emerging role of TPE in the management of severe ICI-induced toxicity, such as myocarditis.
Collapse
Affiliation(s)
| | - Lin He
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ravi Sarode
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, USA.,Department of Internal Medicine (Hematology/Oncology), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Amelework Wodajo
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Amena Usmani
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - James Burner
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Mark Berlacher
- Department of Internal Medicine (Cardiology), UT Southwestern Medical Center, Dallas, Texas, USA
| | - Nicole De Simone
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
34
|
George AS, Fernandez CJ, Eapen D, Pappachan JM. Organ-specific Adverse Events of Immune Checkpoint Inhibitor Therapy, with Special Reference to Endocrinopathies. EUROPEAN ENDOCRINOLOGY 2021. [DOI: 10.17925/ee.2021.1.1.21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
35
|
Dal’bo N, Patel R, Parikh R, Shah SP, Guha A, Dani SS, Ganatra S. Cardiotoxicity of Contemporary Anticancer Immunotherapy. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2020; 22:62. [PMID: 33162729 PMCID: PMC7605901 DOI: 10.1007/s11936-020-00867-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/05/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE OF REVIEW Contemporary anticancer immunotherapy, particularly immune checkpoint inhibitors (ICI) and chimeric antigen receptor (CAR) T cell therapy, has changed the landscape of treatment for patients with a variety of malignancies who historically had a poor prognosis. However, both immune checkpoint inhibitors and CAR T cell therapy are associated with serious cardiovascular adverse effects. As immunotherapy evolves to include high-risk patients with preexisting cardiovascular risk factors and disease, the risk and relevance of its associated cardiotoxicity will be even higher. RECENT FINDINGS ICI can cause myocarditis, which usually occurs early after initiation, can be fulminant, and prompt treatment with high-dose corticosteroids is crucial. CAR T cell therapy frequently leads to cytokine release syndrome, which is associated with cardiomyopathy or arrhythmia development and may also result in circulatory collapse. Supportive treatment, as well as tocilizumab, an anti-interleukin-6 receptor antibody, is the cornerstone of treatment. Recent findings suggest that preexisting cardiovascular risk factors and disease may increase the risk of such cardiotoxicity, and prompt recognition, as well as treatment, may favorably alter the outcomes. SUMMARY ICI and CAR T cell therapy have improved cancer-related outcomes; however, they both are associated with potentially therapy-limiting cardiotoxicity. Cardio-oncologists are required to play an important role in patient selection, pretherapy cardiovascular optimization, and prompt recognition and treatment of cardiotoxicity.
Collapse
Affiliation(s)
- Natalie Dal’bo
- Division of Internal Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Rushin Patel
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Rohan Parikh
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Sachin P. Shah
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Avirup Guha
- Harrington Heart and Vascular Institute, Case Western Reserve University School of Medicine, Cleveland, USA
| | - Sourbha S. Dani
- Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, MA USA
| | - Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, 41 Mall Road, Burlington, MA 01805 USA
| |
Collapse
|
36
|
Chen X, Liu J, Liu J, Wang WJ, Lai WJ, Li SH, Deng YF, Zhou JZ, Yang SQ, Liu Y, Shou WN, Cao DY, Li XH. α-Galactosylceramide and its analog OCH differentially affect the pathogenesis of ISO-induced cardiac injury in mice. Acta Pharmacol Sin 2020; 41:1416-1426. [PMID: 32973325 DOI: 10.1038/s41401-020-00517-z] [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: 05/13/2020] [Accepted: 08/19/2020] [Indexed: 11/09/2022] Open
Abstract
Immunotherapies for cancers may cause severe and life-threatening cardiotoxicities. The underlying mechanisms are complex and largely elusive. Currently, there are several ongoing clinical trials based on the use of activated invariant natural killer T (iNKT) cells. The potential cardiotoxicity commonly associated with this particular immunotherapy has yet been carefully evaluated. The present study aims to determine the effect of activated iNKT cells on normal and β-adrenergic agonist (isoproterenol, ISO)-stimulated hearts. Mice were treated with iNKT stimulants, α-galactosylceramide (αGC) or its analog OCH, respectively, to determine their effect on ISO-induced cardiac injury. We showed that administration of αGC (activating both T helper type 1 (Th1)- and T helper type 2 (Th2)-liked iNKT cells) significantly accelerated the progressive cardiac injury, leading to enhanced cardiac hypertrophy and cardiac fibrosis with prominent increases in collagen deposition and TGF-β1, IL-6, and alpha smooth muscle actin expression. In contrast to αGC, OCH (mainly activating Th2-liked iNKT cells) significantly attenuated the progression of cardiac injury and cardiac inflammation induced by repeated infusion of ISO. Flow cytometry analysis revealed that αGC promoted inflammatory macrophage infiltration in the heart, while OCH was able to restrain the infiltration. In vitro coculture of αGC- or OCH-pretreated primary peritoneal macrophages with primary cardiac fibroblasts confirmed the profibrotic effect of αGC and the antifibrotic effect of OCH. Our results demonstrate that activating both Th1- and Th2-liked iNKT cells is cardiotoxic, while activating Th2-liked iNKT cells is likely cardiac protective, which has implied key differences among subpopulations of iNKT cells in their response to cardiac pathological stimulation.
Collapse
|
37
|
Pembrolizumab-Induced Myocarditis Leading to Persistent Atrial Arrhythmias and a Cascade of Complications: A Therapeutic Dilemma. Am J Ther 2020; 28:e606-e608. [PMID: 33136576 DOI: 10.1097/mjt.0000000000001287] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
38
|
Mena Sebastiá E, Tirapu Sola L, Villanueva Vázquez R, Losa Gaspà F, Freixa-Pamias R. [Cancer, onco-haematological treatment and cardiovascular toxicity]. HIPERTENSION Y RIESGO VASCULAR 2020; 38:72-82. [PMID: 32978077 DOI: 10.1016/j.hipert.2020.08.001] [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: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 11/16/2022]
Abstract
The increased availability of new cancer treatments in recent years has led to improved prognosis and increased life expectancy for cancer patients, but at the expense of increased cardiovascular risk. For this reason, multidisciplinary teams need to be formed for the joint evaluation of these patients to optimise the cardiovascular health and overall survival of these patients and minimise interruptions to onco-haematological treatments. A wide range of cardiovascular toxicities are associated with the various cancer treatments. The structured control of cardiovascular risk before, during and after oncological treatment will enable strategies for the prevention, early detection and early treatment of cardiotoxicities.
Collapse
Affiliation(s)
- E Mena Sebastiá
- Servicio de Cardiología, Hospital de Sant Joan Despí Moisès Broggi, Sant Joan Despí, España.
| | - L Tirapu Sola
- Servicio de Cardiología, Hospital de Sant Joan Despí Moisès Broggi, Sant Joan Despí, España
| | - R Villanueva Vázquez
- Servicio de Oncología Médica, Hospital de Sant Joan Despí Moisès Broggi, Institut Català d'Oncologia, Sant Joan Despí, España
| | - F Losa Gaspà
- Servicio de Oncología Médica, Hospital de Sant Joan Despí Moisès Broggi, Institut Català d'Oncologia, Sant Joan Despí, España
| | - R Freixa-Pamias
- Servicio de Cardiología, Hospital de Sant Joan Despí Moisès Broggi, Sant Joan Despí, España
| |
Collapse
|
39
|
Jafari A, Rezaei-Tavirani M, Karami S, Yazdani M, Zali H, Jafari Z. Cancer Care Management During the COVID-19 Pandemic. Risk Manag Healthc Policy 2020; 13:1711-1721. [PMID: 33061705 PMCID: PMC7520144 DOI: 10.2147/rmhp.s261357] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 09/02/2020] [Indexed: 01/08/2023] Open
Abstract
New cases of the novel coronavirus, also known as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), are increasing around the world. Currently, health care services are mainly focused on responding to and controlling the unique challenges of the coronavirus disease 2019 (COVID-19) pandemic. These changes, along with the higher susceptibility of patients with cancer to infections, have profound effects on other critical aspects of care and pose a serious challenge for the treatment of such patients. During the COVID-19 pandemic, it is important to provide strategies for managing the treatment of patients with cancer to limit COVID-19-associated risks at this difficult time. The present study set out to summarize the latest research on epidemiology, pathogenesis, and clinical features of COVID-19. We also address some of the current challenges associated with the management of patients with cancer during the COVID-19 pandemic and provide practical guidance to clinically deal with these challenges.
Collapse
Affiliation(s)
- Ameneh Jafari
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Student Research Committee, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Samira Karami
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohsen Yazdani
- Department of Bioinformatics, Institute of Biochemistry and Biophysics, University of Tehran, Tehran, Iran
| | - Hakimeh Zali
- Proteomics Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Jafari
- 9Dey Manzariye Hospital, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
40
|
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: 1.3] [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
|
41
|
Capone V, Cuomo V, Esposito R, Canonico ME, Ilardi F, Prastaro M, Esposito G, Santoro C. Epidemiology, prognosis, and clinical manifestation of cardiovascular disease in COVID-19. Expert Rev Cardiovasc Ther 2020; 18:531-539. [PMID: 32672482 DOI: 10.1080/14779072.2020.1797491] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION At the end of 2019, a novel coronavirus was identified as the cause of a pneumonia cluster in Wuhan, China. Since then, the contagion has rapidly spread all over the world resulting in a global pandemic. Since frequent cardiovascular (CV) system involvement has soon been detected in patients occurring coronavirus disease 2019 (COVID-19), we would provide a simple review available to cardiologists who are going to be involved in the management of COVID-19 patients from several levels: from diagnosis to prevention and management of CV complications. AREAS COVERED We investigate the role of CV diseases in COVID-19: from the incidence of CV comorbidities to their negative impact on prognosis. We also search Literature in order to identify the main CV manifestations in patients occurring virus infection and their management by cardiologists. EXPERT OPINION Specific treatments for CV involvement associated with COVID-19 are still debated. Results from ongoing trials are needed to further clarify issues about the therapeutic approach, which is constantly changing according to the continuous flow of published evidence. Finally, it seems necessary to sensitize all population to raise awareness on CV diseases in the COVID era, to hinder the underestimation of both new-onset acute CV diseases and the consequences of chronic mistreated CV diseases.
Collapse
Affiliation(s)
- Valentina Capone
- Department of Advanced Biomedical Sciences, Federico II University , Naples, Italy
| | - Vittoria Cuomo
- Department of Clinical Medicine and Surgery, Federico II University , Naples, Italy
| | - Roberta Esposito
- Department of Clinical Medicine and Surgery, Federico II University , Naples, Italy.,Mediterranea Cardiocentro , Naples, Italy
| | | | - Federica Ilardi
- Department of Advanced Biomedical Sciences, Federico II University , Naples, Italy.,Mediterranea Cardiocentro , Naples, Italy
| | - Maria Prastaro
- Department of Advanced Biomedical Sciences, Federico II University , Naples, Italy
| | - Giovanni Esposito
- Department of Advanced Biomedical Sciences, Federico II University , Naples, Italy
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University , Naples, Italy
| |
Collapse
|
42
|
Simbaqueba CC, Aponte MP, Kim P, Deswal A, Palaskas NL, Iliescu C, Jahangir E, Yang EH, Steiner RE, Lopez-Mattei J. Cardiovascular Complications of Chimeric Antigen Receptor T-Cell Therapy: The Cytokine Release Syndrome and Associated Arrhythmias. ACTA ACUST UNITED AC 2020; 3:113-120. [DOI: 10.36401/jipo-20-10] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/28/2020] [Indexed: 12/19/2022]
Abstract
ABSTRACT
In recent years, cancer treatment has evolved, and new therapies have been introduced with significant improvement in prognosis. The immunotherapies stand out owing to their efficacy and remission rate. Chimeric antigen receptor (CAR) T-cell therapy is a part of this new era of therapies. Chimeric antigen receptor T-cell therapy is a form of adoptive cellular therapy that uses a genetically encoded CAR in modified human T cells to target specific tumor antigens in a nonconventional, non-major histocompatibility complex (MHC) protein presentation. Chimeric antigen receptor T-cell therapy successfully identifies tumor antigens and through activation of T cells destroys tumoral cells. It has been found to efficiently induce remission in patients who have been previously treated for B-cell malignancies and have persistent disease. As the use of this novel therapy increases, its potential side effects also have become more evident, including major complications like cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Cytokine release syndrome is a major systemic inflammatory process as a result of massive cytokine production by the proliferating and activated CAR T cells in which multiple interleukins and immune cells contribute to the inflammatory response. Cytokine release syndrome has been associated with cardiovascular life-threatening complications including hypotension, shock, tachycardia, arrhythmias, left ventricular dysfunction, heart failure, and cardiovascular death. Arrhythmias, among its major complications, vary from asymptomatic prolonged corrected QT interval (QTc) to supraventricular tachycardia, atrial fibrillation, flutter, and ventricular arrhythmias like Torsade de pointes. This article focuses on the cardiovascular complications and arrhythmias associated with CRS and CAR T-cell therapy.
Collapse
Affiliation(s)
- Cesar Clavijo Simbaqueba
- Department of General Internal Medicine, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Maria Patarroyo Aponte
- Department of Cardiology, University of Texas Health System, Memorial Hermann Hospital, Houston, Texas
| | - Peter Kim
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Anita Deswal
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Nicolas L. Palaskas
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Cezar Iliescu
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Eiman Jahangir
- Department of Cardiology, Vanderbilt University Medical Center, Vanderbilt, Tennessee
| | - Eric H. Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California
| | - Raphael E. Steiner
- Department of Lymphoma-Myeloma, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Juan Lopez-Mattei
- Department of Cardiology, University of Texas MD Anderson Cancer Center, Houston, Texas
| |
Collapse
|
43
|
Clinical Practice and Research in Cardio-Oncology: Finding the "Rosetta Stone" for Establishing Program Excellence in Cardio-oncology. J Cardiovasc Transl Res 2020; 13:495-505. [PMID: 32444945 DOI: 10.1007/s12265-020-10010-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 04/17/2020] [Indexed: 02/06/2023]
Abstract
The burgeoning field of cardio-oncology (C-O) is now necessary for the delivery of excellent care for patients with cancer. Many factors have contributed to this increasing population of cancer survivors or those being treated with novel and targeted cancer therapies. There is a tremendous need to provide outstanding cardiovascular (CV) care for these patients; however, current medical literature actually provides a paucity of guidance. C-O therefore provides a novel opportunity for clinical, translational, and basic research to advance patient care. This review aims to be a primer for cardio-oncologists on how to develop a vibrant and comprehensive C-O program, use practical tools to assist in the construction of C-O services, and to proactively incorporate translational and clinical research into the training of future leaders as well as enhance clinical care.
Collapse
|
44
|
Raibagkar P. Immune checkpoint inhibitor associated neurotoxicity: Much more than meets the eye. Immunol Lett 2020; 221:72-74. [DOI: 10.1016/j.imlet.2020.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 02/20/2020] [Indexed: 11/25/2022]
|
45
|
Jin KT, Wang SB, Ying XJ, Lan HR, Lv JQ, Zhang LH, Motallebnezhad M, Mou XZ. Immune-mediated adverse effects of immune-checkpoint inhibitors and their management in cancer. Immunol Lett 2020; 221:61-71. [PMID: 32097671 DOI: 10.1016/j.imlet.2020.02.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 01/03/2020] [Accepted: 02/20/2020] [Indexed: 02/07/2023]
Abstract
Within the past decade, immune-checkpoint inhibitors (ICPIs), including anti-programmed cell death 1 (PD-1), anti-programmed cell death 1 ligand 1 (PD-L1), and anti-cytotoxic T lymphocyte antigen 4 (CTLA-4) antibodies, are undoubtfully the most remarkable advances in cancer therapy. The immune responses are modulated by these ICPIs via blocking the inhibitory PD-1/PD-L1 path and result in immune activation in the suppressive microenvironment of the tumor. While ICPIs result in benefits for numerous patients with malignancy and lead to disease control and survival, toxicity and safety problems have emerged as well. Although immune mediated adverse effects due to ICPIs could involve any organ system, skin, endocrine glands, and gastrointestinal tract, are one of the most commonly affected. Fortunately, in most of the cases, these immune‑mediated adverse effects (imAEs) are manageable, while in some cases these toxicities are fulminant and fatal and lead to the withdrawal of treatment. Numerous attempts have been started and are continuing to reduce the incidence rate of imAEs. Further studies are required for a better understanding of these imAEs, decrease the occurrence, and lighten the severity. In this work, we overview the imAEs and also, highlight the most important aspects of the imAEs management.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, PR China
| | - Shi-Bing Wang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China
| | - Xiao-Jiang Ying
- Department of Colorectal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, PR China
| | - Huan-Rong Lan
- Department of Breast and Thyroid Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, PR China
| | - Jie-Qing Lv
- Department of Gastrointestinal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, PR China
| | - Li-Hua Zhang
- Department of Colorectal Surgery, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, 312000, Zhejiang Province, PR China
| | - Morteza Motallebnezhad
- Student Research Committee, Iran University of Medical Sciences, Tehran, Iran; Department of Immunology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Xiao-Zhou Mou
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China; Clinical Research Institute, Zhejiang Provincial People's Hospital (People's Hospital of Hangzhou Medical College), Hangzhou 310014, Zhejiang Province, PR China.
| |
Collapse
|
46
|
Ganatra S, Carver JR, Hayek SS, Ky B, Leja MJ, Lenihan DJ, Lenneman C, Mousavi N, Park JH, Perales MA, Ryan TD, Scherrer-Crosbie M, Steingart RM, Yang EH, Zaha V, Barac A, Liu JE. Chimeric Antigen Receptor T-Cell Therapy for Cancer and Heart: JACC Council Perspectives. J Am Coll Cardiol 2019; 74:3153-3163. [PMID: 31856973 PMCID: PMC8211027 DOI: 10.1016/j.jacc.2019.10.049] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Revised: 10/09/2019] [Accepted: 10/31/2019] [Indexed: 12/23/2022]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has significantly advanced the treatment of patients with relapsed and refractory hematologic malignancies and is increasingly investigated as a therapeutic option of other malignancies. The main adverse effect of CAR T-cell therapy is potentially life-threatening cytokine release syndrome (CRS). Clinical cardiovascular (CV) manifestations of CRS include tachycardia, hypotension, troponin elevation, reduced left ventricular ejection fraction, pulmonary edema, and cardiogenic shock. Although insults related to CRS toxicity might be transient and reversible in most instances in patients with adequate CV reserve, they can be particularly challenging in higher-risk, often elderly patients with pre-existing CV disease. As the use of CAR T-cell therapy expands to include a wider patient population, careful patient selection, pre-treatment cardiac evaluation, and CV risk stratification should be considered within the CAR T-cell treatment protocol. Early diagnosis and management of CV complications in patients with CRS require awareness and multidisciplinary collaboration.
Collapse
Affiliation(s)
- Sarju Ganatra
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Lahey Hospital and Medical Center, Burlington, Massachusetts; Cardio-Oncology and Adult Cancer Survivorship Program, Dana Farber Cancer Institute, Boston, Massachusetts; Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts
| | - Joseph R Carver
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Salim S Hayek
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Bonnie Ky
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Abramson Cancer Center, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Monika J Leja
- Division of Cardiology, University of Michigan, Ann Arbor, Michigan
| | - Daniel J Lenihan
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Department of Medicine, Washington University, St. Louis, Missouri
| | - Carrie Lenneman
- Division of Cardiovascular Medicine, Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Negaresh Mousavi
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, McGill University Health Center, Montreal, Quebec, Canada
| | - Jae H Park
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York; Department of Medicine, Weill Cornell Medical College, New York, New York
| | - Miguel Angel Perales
- Department of Medicine, Weill Cornell Medical College, New York, New York; Adult Bone Marrow Transplant, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Thomas D Ryan
- Heart Institute, Cincinnati Children's Hospital Medical Center, Department of Pediatrics, Cincinnati College of Medicine, Cincinnati, Ohio
| | - Marielle Scherrer-Crosbie
- Cardio-Oncology Program and Cardiac Ultrasound Laboratory, Division of Cardiology, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Richard M Steingart
- Department of Medicine, Weill Cornell Medical College, New York, New York; Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Eric H Yang
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, Ronald Reagan University of California Los Angeles Medical Center, Los Angeles, California
| | - Vlad Zaha
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Department of Medicine, University of Texas Southwestern Medical Center, Divison of Cardiovascular Medicine, Department of Medicine, Dallas, Texas
| | - Ana Barac
- Cardio-Oncology Program, Department of Cardiology, MedStar Washington Hospital Center, MedStar Heart and Vascular Institute, Washington, DC.
| | - Jennifer E Liu
- Department of Medicine, Weill Cornell Medical College, New York, New York; Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| |
Collapse
|