1
|
Shaaban A, Scott SS, Greenlee AN, Binda N, Noor A, Webb A, Guo S, Purdy N, Pennza N, Habib A, Mohammad SJ, Smith SA. Atrial fibrillation in cancer, anticancer therapies, and underlying mechanisms. J Mol Cell Cardiol 2024; 194:118-132. [PMID: 38897563 PMCID: PMC11500699 DOI: 10.1016/j.yjmcc.2024.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 06/03/2024] [Accepted: 06/04/2024] [Indexed: 06/21/2024]
Abstract
Atrial fibrillation (AF) is a common arrhythmic complication in cancer patients and can be exacerbated by traditional cytotoxic and targeted anticancer therapies. Increased incidence of AF in cancer patients is independent of confounding factors, including preexisting myocardial arrhythmogenic substrates, type of cancer, or cancer stage. Mechanistically, AF is characterized by fast unsynchronized atrial contractions with rapid ventricular response, which impairs ventricular filling and results in various symptoms such as fatigue, chest pain, and shortness of breath. Due to increased blood stasis, a consequence of both cancer and AF, concern for stroke increases in this patient population. To compound matters, cardiotoxic anticancer therapies themselves promote AF; thereby exacerbating AF morbidity and mortality in cancer patients. In this review, we examine the relationship between AF, cancer, and cardiotoxic anticancer therapies with a focus on the shared molecular and electrophysiological mechanisms linking these disease processes. We also explore the potential role of sodium-glucose co-transporter 2 inhibitors (SGLT2i) in the management of anticancer-therapy-induced AF.
Collapse
Affiliation(s)
- Adnan Shaaban
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Shane S Scott
- Medical Scientist Training Program, Biomedical Sciences Graduate Program, The Ohio State University, Columbus, OH, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Ashley N Greenlee
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nkongho Binda
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA
| | - Ali Noor
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Averie Webb
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Shuliang Guo
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Najhee Purdy
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nicholas Pennza
- Ohio University Heritage College of Osteopathic Medicine, Athens, OH 45701, USA
| | - Alma Habib
- The Ohio State University College of Medicine, Department of Internal Medicine, Division of Hematology, Columbus, OH 43210, USA
| | - Somayya J Mohammad
- Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Sakima A Smith
- The Ohio State University College of Medicine, Department of Internal Medicine, Columbus, OH 43210, USA; Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University College of Medicine, Columbus, OH 43210, USA; Bob and Corrinne Frick Center for Heart Failure and Arrhythmia Research, The Dorothy M. Davis Heart and Lung Research Institute, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA.
| |
Collapse
|
2
|
Tamargo J, Villacastín J, Caballero R, Delpón E. Drug-induced atrial fibrillation. A narrative review of a forgotten adverse effect. Pharmacol Res 2024; 200:107077. [PMID: 38244650 DOI: 10.1016/j.phrs.2024.107077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia and is associated with an increased morbidity and mortality. There is clinical evidence that an increasing number of cardiovascular and non-cardiovascular drugs, mainly anticancer drugs, can induce AF either in patients with or without pre-existing cardiac disorders, but drug-induced AF (DIAF) has not received the attention that it might deserve. In many cases DIAF is asymptomatic and paroxysmal and patients recover sinus rhythm spontaneously, but sometimes, DIAF persists, and it is necessary to perform a cardioversion. Furthermore, DIAF is not mentioned in clinical guidelines on the treatment of AF. The risk of DIAF increases in elderly and in patients treated with polypharmacy and with risk factors and comorbidities that commonly coexist with AF. This is the case of cancer patients. Under these circumstances ascribing causality of DIAF to a given drug often represents a clinical challenge. We review the incidence, the pathophysiological mechanisms, risk factors, clinical relevance, and treatment of DIAF. Because of the limited information presently available, further research is needed to obtain a deeper insight into DIAF. Meanwhile, it is important that clinicians are aware of the problem that DIAF represents, recognize which drugs may cause DIAF, and consider the possibility that a drug may be responsible for a new-onset AF episode.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| | - Julián Villacastín
- Hospital Clínico San Carlos, CardioRed1, Universidad Complutense de Madrid, CIBERCV, 28040 Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain.
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| |
Collapse
|
3
|
Green CE, Chacon J, Godinich BM, Hock R, Kiesewetter M, Raynor M, Marwaha K, Maharaj S, Holland N. The Heart of the Matter: Immune Checkpoint Inhibitors and Immune-Related Adverse Events on the Cardiovascular System. Cancers (Basel) 2023; 15:5707. [PMID: 38136253 PMCID: PMC10742007 DOI: 10.3390/cancers15245707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 11/21/2023] [Accepted: 11/30/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer remains a prominent global cause of mortality, second only to cardiovascular disease. The past decades have witnessed substantial advancements in anti-cancer therapies, resulting in improved outcomes. Among these advancements, immunotherapy has emerged as a promising breakthrough, leveraging the immune system to target and eliminate cancer cells. Despite the remarkable potential of immunotherapy, concerns have arisen regarding associations with adverse cardiovascular events. This review examines the complex interplay between immunotherapy and cardiovascular toxicity and provides an overview of immunotherapy mechanisms, clinical perspectives, and potential biomarkers for adverse events, while delving into the intricate immune responses and evasion mechanisms displayed by cancer cells. The focus extends to the role of immune checkpoint inhibitors in cancer therapy, including CTLA-4, PD-1, and PD-L1 targeting antibodies. This review underscores the multifaceted challenges of managing immunotherapy-related cardiovascular toxicity. Risk factors for immune-related adverse events and major adverse cardiac events are explored, encompassing pharmacological, treatment-related, autoimmune, cardiovascular, tumor-related, social, genetic, and immune-related factors. The review also advocates for enhanced medical education and risk assessment tools to identify high-risk patients for preventive measures. Baseline cardiovascular evaluations, potential prophylactic strategies, and monitoring of emerging toxicity symptoms are discussed, along with the potential of adjunct anti-inflammatory therapies.
Collapse
Affiliation(s)
- Chase E. Green
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Jessica Chacon
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Brandon M. Godinich
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Rivers Hock
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Maria Kiesewetter
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Mark Raynor
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Komal Marwaha
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| | - Satish Maharaj
- Department of Internal Medicine, Division of Hematology/Oncology, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 4800 Alberta Ave., El Paso, TX 79905, USA
| | - Nathan Holland
- Department of Medical Education, Paul L. Foster School of Medicine, Texas Tech Health Sciences Center El Paso, 5001 El Paso Ave., El Paso, TX 79905, USA
| |
Collapse
|
4
|
Xie X, Li X, Liu G, Zhao H, Zhou Z, Xiong S. T cells expressing a HER2-specific chimeric antigen receptor as treatment for breast cancer. J Cancer Res Clin Oncol 2023; 149:11561-11570. [PMID: 37402965 DOI: 10.1007/s00432-023-04996-5] [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: 05/11/2023] [Accepted: 06/15/2023] [Indexed: 07/06/2023]
Abstract
PURPOSE Human endothelial growth factor receptor-2 (HER2) is a leucine kinase receptor that is closely related to cell growth and differentiation. It is very weakly expressed in a few epithelial cells in normal tissue. Abnormal expression of HER2 usually leads to sustained activation of downstream signaling pathways, enabling epithelial cell growth, proliferation, and differentiation; this disturbs normal physiological processes and causes tumor formation. Overexpression of HER2 is related to the occurrence and development of breast cancer. HER2 has become a well-established immunotherapy target for breast cancer. We chose to construct a second-generation CAR targeting HER 2 to test whether it kills breast cancer. METHODS We constructed a second-generation CAR molecule targeting HER2, and we generated cells expressing this second-generation CAR through lentivirus infection of T lymphocytes. LDH assay and flow cytometry were perform to detect the effect of cells and animal models. RESULTS The result indicated that the CARHER2 T cells could selectively kill cells with high Her2 expression. The PBMC-activated/CARHer2 cells had stronger in vivo tumor suppressive activity than PBMC-activated cells, and administration of PBMC-activated/CARHer2 cells significantly improved the survival of tumor-bearing mice, and induced the production of more Th1 cytokines in tumor-bearing NSG mice. CONCLUSIONS We prove that the generated T cells carrying the second-generation CARHer2 molecule could effectively guide immune effector cells to identify and kill HER2-positive tumor cells and inhibit tumors in model mice.
Collapse
Affiliation(s)
- Xinshan Xie
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Xiaobin Li
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Gang Liu
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Hui Zhao
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Zhenlong Zhou
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China
| | - Sheng Xiong
- College of Life Science and Technology, Jinan University, Guangzhou, 510630, China.
- Institute of Biomedicine and National Engineering Research Center of Genetic Medicine, Guangzhou, 510630, China.
| |
Collapse
|
5
|
Sadeq MA, Ashry MH, Ghorab RMF, Afify AY. Causes of death among patients with cutaneous melanoma: a US population-based study. Sci Rep 2023; 13:10257. [PMID: 37355743 PMCID: PMC10290704 DOI: 10.1038/s41598-023-37333-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023] Open
Abstract
Research on mortality outcomes and non-cancer-related causes of death in patients with cutaneous melanoma (CM) remains limited. This study aimed to identify the prevalence of non-cancer-related deaths following CM diagnosis. The data of 224,624 patients diagnosed with malignant CM in the United States between 2000 and 2019 were extracted from the Surveillance, Epidemiology, and End Results (SEER) database. We stratified our cohort based on their melanoma stage at diagnosis and further calculated standardized mortality ratios (SMRs) for each cause of death, comparing their relative risk to that of the general US population. The total number of fatalities among melanoma patients was 60,110, representing 26.8% of the total cases. The percentage of deaths is directly proportional to the disease stage, reaching 80% in distant melanoma. The highest fatalities among the localized melanoma group (25,332; 60.5%) occurred from non-cancer causes, followed by melanoma-attributable deaths (10,817; 25.8%). Conversely, melanoma is the leading cause of death in regional and distant melanoma cohorts. Cardiovascular and cerebrovascular diseases were the most prevalent non-cancer causes of death among the three disease-stage cohorts. Compared to the general population, we did not observe an increased risk of death due to non-cancer causes in the localized CM cohort, while patients diagnosed with regional and distant CMs had a statistically significant higher risk of death from all the reported major causes of death.
Collapse
Affiliation(s)
- Mohammed Ahmed Sadeq
- Faculty of Medicine, Misr University for Science and Technology, 6th of October, Giza, Egypt
| | | | | | | |
Collapse
|
6
|
Font J, Milliez P, Ouazar AB, Klok FA, Alexandre J. Atrial fibrillation, cancer and anticancer drugs. Arch Cardiovasc Dis 2023; 116:219-226. [PMID: 37002156 DOI: 10.1016/j.acvd.2023.02.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/27/2023] [Accepted: 02/27/2023] [Indexed: 04/18/2023]
Abstract
Active cancer is associated with an increased risk of atrial fibrillation (AF), which varies depending on the pre-existing substrate (particularly in older patients), the cancer type and stage, and the anticancer therapeutics being taken. To date, studies have not been able to identify the individual contribution of each factor. During anticancer drug therapy, AF may occur with a frequency of ≈ 15-20% according to several factors, including the patient's baseline cardiovascular toxicity risk and the AF-detection strategies used. Many anticancer drugs have been associated with AF or AF reporting, both in terms of incident and recurrent AF, but robust data are lacking. Only bruton tyrosine kinase inhibitor associated AF (mainly ibrutinib) has a high level of evidence, with a ≈ 3-4-fold higher risk of AF. AF in patients with active cancer is associated with a twofold higher risk of systemic thromboembolism or stroke, and the "TBIP" (Thromboembolic risk, Bleeding risk, drug-drug Interactions, Patient preferences) structured approach must be used to evaluate the need for anticoagulation therapy. AF in patients with active cancer is also associated with a sixfold higher risk of heart failure, and optimal symptom control must be targeted, usually with rate-control drugs (beta-blockers), but a rhythm-control strategy may be proposed in patients remaining symptomatic despite optimal rate-control. AF is generally manageable, with the continuation of anticancer drugs (including ibrutinib); interruption of cancer drugs must be avoided whenever possible and weighed against the risk of cancer progression.
Collapse
Affiliation(s)
- Jonaz Font
- Normandie University, UNICAEN, Inserm U1086 Anticipe, avenue du Général-Harris, 14000 Caen, France; CHU de Caen-Normandie, Department of Cardiology, avenue de la Côte de Nacre, 14000 Caen, France
| | - Paul Milliez
- CHU de Caen-Normandie, Department of Cardiology, avenue de la Côte de Nacre, 14000 Caen, France; Normandie University, UNICAEN, Inserm U1237 PhIND, GIP Cyceron, boulevard Henri-Becquerel, 14000 Caen, France
| | | | - Frederikus A Klok
- Department of Medicine - Thrombosis and Hemostasis, Leiden University Medical Center, Leiden, Netherlands
| | - Joachim Alexandre
- Normandie University, UNICAEN, Inserm U1086 Anticipe, avenue du Général-Harris, 14000 Caen, France; CHU de Caen-Normandie, PICARO Cardio-Oncology Program, Department of Pharmacology, avenue de la Côte de Nacre, 14000 Caen, France.
| |
Collapse
|
7
|
Burashnikov A. Atrial Fibrillation Induced by Anticancer Drugs and Underling Mechanisms. J Cardiovasc Pharmacol 2022; 80:540-546. [PMID: 34803149 PMCID: PMC10566596 DOI: 10.1097/fjc.0000000000001182] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 11/11/2021] [Indexed: 11/26/2022]
Abstract
ABSTRACT Cancer therapy has made major progress in the past several decades, but treatments are often accompanied by significant side effects. Arrhythmias are a widespread complication of some antineoplastic drugs, with atrial fibrillation (AF) being the most often encountered drug-associated arrhythmia. Preexisting AF risk factors are commonly present in cancer patients who develop drug-associated AF, and active cancer itself may cause or promote AF. Although anticancer drugs may induce AF in cancer patients without AF risk factors, it appears that most drug-associated AF develop when cancer drugs add or aggravate precancer-existing and/or cancer-related pro-AF factors/alterations, additively or synergistically producing AF. Abnormalities in intracellular calcium activity seem to be involved in the generation of anticancer drug-induced AF. In cancer survivors with cancer therapy-induced cardiomyopathy, AF often occurs, with most of the arrhythmias likely to develop secondary to the cardiomyopathy. AF may lead to modification or even cessation of cancer therapy. The management of AF in patients with cancer is currently conducted largely based on pragmatic assumptions. This review briefly discusses AF caused by anticancer drugs and the underlying mechanisms.
Collapse
Affiliation(s)
- Alexander Burashnikov
- Lankenau Institute for Medical Research, Wynnewood, PA; and
- Sidney Kimmel College of Medicine, Thomas Jefferson University, Philadelphia, PA
| |
Collapse
|
8
|
Lagan J, Naish JH, Fortune C, Campbell C, Chow S, Pillai M, Bradley J, Francis L, Clark D, Macnab A, Nucifora G, Dobson R, Schelbert EB, Schmitt M, Hawkins R, Miller CA. Acute and Chronic Cardiopulmonary Effects of High Dose Interleukin-2 Therapy: An Observational Magnetic Resonance Imaging Study. Diagnostics (Basel) 2022; 12:diagnostics12061352. [PMID: 35741162 PMCID: PMC9221588 DOI: 10.3390/diagnostics12061352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/23/2022] [Accepted: 05/24/2022] [Indexed: 11/20/2022] Open
Abstract
High dose interleukin-2 (IL-2) is known to be associated with cardiopulmonary toxicity. The goal of this study was to evaluate the effects of high dose IL-2 therapy on cardiopulmonary structure and function. Combined cardiopulmonary magnetic resonance imaging (MRI) was performed in 7 patients in the acute period following IL-2 therapy and repeated in 4 patients in the chronic period. Comparison was made to 10 healthy volunteers. IL-2 therapy was associated with myocardial and pulmonary capillary leak, tissue oedema and cardiomyocyte injury, which resulted in acute significant left ventricular (LV) dilatation, a reduction in LV ejection fraction (EF), an increase in LV mass and a prolongation of QT interval. The acute effects occurred irrespective of symptoms. In the chronic period many of the effects resolved, but LV hypertrophy ensued, driven by focal replacement and diffuse interstitial myocardial fibrosis and increased cardiomyocyte mass. In conclusion, IL-2 therapy is ubiquitously associated with acute cardiopulmonary inflammation, irrespective of symptoms, which leads to acute LV dilatation and dysfunction, increased LV mass and QT interval prolongation. Most of these effects are reversible but IL-2 therapy is associated with chronic LV hypertrophy, driven by interstitial myocardial fibrosis and increased cardiomyocyte mass. The findings have important implications for the monitoring and long term impact of newer immunotherapies. Future studies are needed to improve risk stratification and develop cardiopulmonary-protective strategies.
Collapse
Affiliation(s)
- Jakub Lagan
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Josephine H. Naish
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Christien Fortune
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Christopher Campbell
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (C.C.); (S.C.); (M.P.); (R.H.)
| | - Shien Chow
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (C.C.); (S.C.); (M.P.); (R.H.)
- The Clatterbridge Cancer Centre NHS Foundation Trust, Wirral, Bebingtonm CH63 4JY, UK
| | - Manon Pillai
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (C.C.); (S.C.); (M.P.); (R.H.)
| | - Joshua Bradley
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Lenin Francis
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
| | - David Clark
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
| | - Anita Macnab
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
| | - Gaetano Nucifora
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
| | - Rebecca Dobson
- Liverpool Heart and Chest Hospital NHS Foundation Trust, Thomas Drive, Liverpool L14 3PE, UK;
| | - Erik B. Schelbert
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA;
- UPMC Cardiovascular Magnetic Resonance Center, Heart and Vascular Institute, Pittsburgh, PA 15213, USA
- Clinical and Translational Science Institute, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Matthias Schmitt
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Robert Hawkins
- The Christie NHS Foundation Trust, Wilmslow Road, Manchester M20 4BX, UK; (C.C.); (S.C.); (M.P.); (R.H.)
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - Christopher A. Miller
- Wythenshawe Hospital, Manchester University NHS Foundation Trust, Wythenshawe Hospital, Southmoor Road, Wythenshawe, Manchester M23 9LT, UK; (J.L.); (C.F.); (J.B.); (L.F.); (D.C.); (A.M.); (G.N.); (M.S.)
- Division of Cardiovascular Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
- Wellcome Centre for Cell-Matrix Research, Division of Cell-Matrix Biology & Regenerative Medicine, School of Biology, Faculty of Biology, Medicine & Health, Manchester Academic Health Science Centre, University of Manchester, Oxford Road, Manchester M13 9PT, UK
- Correspondence: ; Tel.: +44-161-291-2034; Fax: +44-161-291-2389
| |
Collapse
|
9
|
Makunda N, Vallabhaneni S, Lefebvre B, Fradley MG. Cardiotoxicity of Systemic Melanoma Treatments. Curr Treat Options Oncol 2022; 23:240-253. [PMID: 35192138 DOI: 10.1007/s11864-021-00924-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/16/2021] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT Melanoma is the least common but most dangerous skin cancer, accounting for 75% of all deaths from a primary cutaneous malignancy, with incidence rates rising significantly over the last decade. Traditional treatments for melanoma including interferon and cytotoxic chemotherapy had marginal efficacy. With the advent of targeted and immunotherapies, the prognosis for patients with advanced melanoma has significantly improved including those with metastatic disease to the heart. BRAF and MEK inhibitors as well as immune checkpoint inhibitors have become front line therapy for eligible patients with metastatic melanoma and have led to long-term durable response and in some cases can be curative. Despite these oncologic advances, various treatment-limiting side effects can occur. In particular, cardiovascular toxicities can contribute to overall morbidity and mortality in these patients. Toxicities range from asymptomatic QT prolongation and mild LV dysfunction to fulminant myocarditis and potentially life-threatening arrhythmias. A multidisciplinary approach to the care of these patients which includes cardio-oncology evaluation is necessary to develop both risk mitigation and treatment strategies to ensure patients continue receiving necessary and effective melanoma treatments while minimizing long-term adverse cardiovascular effects.
Collapse
Affiliation(s)
- Neha Makunda
- Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | - Srilakshmi Vallabhaneni
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Benedicte Lefebvre
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA
| | - Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, 3400 Civic Center Blvd, Philadelphia, PA, 19104, USA.
| |
Collapse
|
10
|
Nettersheim FS, Picard FSR, Hoyer FF, Winkels H. Immunotherapeutic Strategies in Cancer and Atherosclerosis-Two Sides of the Same Coin. Front Cardiovasc Med 2022; 8:812702. [PMID: 35097027 PMCID: PMC8792753 DOI: 10.3389/fcvm.2021.812702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 12/20/2021] [Indexed: 11/16/2022] Open
Abstract
The development and clinical approval of immunotherapies has revolutionized cancer therapy. Although the role of adaptive immunity in atherogenesis is now well-established and several immunomodulatory strategies have proven beneficial in preclinical studies, anti-atherosclerotic immunotherapies available for clinical application are not available. Considering that adaptive immune responses are critically involved in both carcinogenesis and atherogenesis, immunotherapeutic approaches for the treatment of cancer and atherosclerosis may exert undesirable but also desirable side effects on the other condition, respectively. For example, the high antineoplastic efficacy of immune checkpoint inhibitors, which enhance effector immune responses against tumor cells by blocking co-inhibitory molecules, was recently shown to be constrained by substantial proatherogenic properties. In this review, we outline the specific role of immune responses in the development of cancer and atherosclerosis. Furthermore, we delineate how current cancer immunotherapies affect atherogenesis and discuss whether anti-atherosclerotic immunotherapies may similarly have an impact on carcinogenesis.
Collapse
Affiliation(s)
- Felix Sebastian Nettersheim
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Molecular Medicine Cologne (CMMC), University of Cologne, Cologne, Germany
| | - Felix Simon Ruben Picard
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Friedrich Felix Hoyer
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Holger Winkels
- Department of Cardiology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| |
Collapse
|
11
|
Ostroumova OD, Chernyaeva MS, Kochetkov AI, Vorobieva AE, Bakhteeva DI, Korchagina SP, Bondarets OV, Boyko ND, Sychev DA. Drug-Induced Atrial Fibrillation / Atrial Flutter. RATIONAL PHARMACOTHERAPY IN CARDIOLOGY 2022. [DOI: 10.20996/1819-6446-2021-12-11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Drug-induced atrial fibrillation / flutter (DIAF) is a serious and potentially life-threatening complication of pharmacotherapy. Purpose of the work: systematization and analysis of scientific literature data on drugs, the use of which can cause the development of DIAF, as well as on epidemiology, pathophysiological mechanisms, risk factors, clinical picture, diagnosis and differential diagnosis, treatment and prevention of DIAF. Analysis of the literature has shown that many groups of drugs can cause the development of DIAF, with a greater frequency while taking anticancer drugs, drugs for the treatment of the cardiovascular, bronchopulmonary and central nervous systems. The mechanisms and main risk factors for the development of DIAF have not been finally established and are known only for certain drugs, therefore, this section requires further study. The main symptoms of DIAF are due to the severity of tachycardia and their influence on the parameters of central hemodynamics. For diagnosis, it is necessary to conduct an electrocardiogram (ECG) and Holter monitoring of an ECG and echocardiography. Differential diagnosis should be made with AF, which may be caused by other causes, as well as other rhythm and conduction disturbances. Successful treatment of DIAF is based on the principle of rapid recognition and immediate discontinuation of drugs (if possible), the use of which potentially caused the development of adverse drug reactions (ADR). The choice of management strategy: heart rate control or rhythm control, as well as the method of achievement (medication or non-medication), depends on the specific clinical situation. For the prevention of DIAF, it is necessary to instruct patients about possible symptoms and recommend self-monitoring of the pulse. It is important for practitioners to be wary of the risk of DIAF due to the variety of drugs that can potentially cause this ADR.
Collapse
Affiliation(s)
- O. D Ostroumova
- Russian Medical Academy of Continuing Professional Education
| | - M. S. Chernyaeva
- Central State Medical Academy of the Administrative Department of the President; Hospital for War Veterans No. 2
| | - A. I. Kochetkov
- Russian Medical Academy of Continuing Professional Education
| | - A. E. Vorobieva
- Moscow State University of Medicine and Dentistry named after A.I. Evdokimova
| | | | | | - O. V. Bondarets
- Moscow State University of Medicine and Dentistry named after A.I. Evdokimova
| | | | - D. A. Sychev
- Russian Medical Academy of Continuing Professional Education
| |
Collapse
|
12
|
Essa H, Wright DJ, Dobson R, Lip GYH. Chemotherapy-Induced Arrhythmia - Underrecognized and Undertreated. Am J Med 2021; 134:1224-1231.e1. [PMID: 34216562 DOI: 10.1016/j.amjmed.2021.05.026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 05/05/2021] [Accepted: 05/07/2021] [Indexed: 10/21/2022]
Abstract
Cancer is one of the leading causes of death worldwide. Chemotherapy-induced arrhythmia is a potential complication of treatment that confers increased morbidity and mortality. The relationship between chemotherapeutic agents and arrhythmias is poorly established. Atrial fibrillation, ventricular ectopic beats, and prolonged QTc are the most common arrhythmias suffered by cancer patients undergoing chemotherapy. The treatment of atrial fibrillation in cancer is complicated by complex drug-drug interactions and a lack of evidence guiding practice. Furthermore, the normal risk assessment scores utilized in the decision-making for anticoagulation in the normal population are not validated in the cancer population. Multiple agents are implicated in prolonging the QTc, and this can often have adverse consequences for both the patient and the treatment of their cancer. This can manifest as torsades de pointes and sudden cardiac death. It is advised that, during treatment, oncologists should have close liaison with cardio-oncologists to ensure optimum patient management.
Collapse
Affiliation(s)
- Hani Essa
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool Heart & Chest Hospital, UK.
| | - David J Wright
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool Heart & Chest Hospital, UK
| | - Rebecca Dobson
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool Heart & Chest Hospital, UK
| | - Gregory Y H Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool Heart & Chest Hospital, UK; Aalborg Thrombosis Research Unit, Department of Clinical Medicine, Aalborg University, Denmark
| |
Collapse
|
13
|
Fradley MG, Damrongwatanasuk R, Chandrasekhar S, Alomar M, Kip KE, Sarnaik AA. Cardiovascular Toxicity and Mortality Associated With Adoptive Cell Therapy and Tumor-infiltrating Lymphocytes for Advanced Stage Melanoma. J Immunother 2021; 44:86-89. [PMID: 33044384 DOI: 10.1097/cji.0000000000000341] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 08/25/2020] [Indexed: 11/26/2022]
Abstract
Adoptive cellular therapy (ACT) with tumor-infiltrating lymphocytes (TILs) has emerged as an effective treatment option for unresectable stage III/IV metastatic melanoma. Acute toxicities, particularly cardiovascular (CV), can have a significant effect on the completion of therapy. We abstracted information on 43 patients who received ACT-TIL treatment for melanoma at the Moffitt Cancer Center between 2010 and 2016. The Student t tests and χ2 tests were used to compare patient characteristics by presence versus absence of specific CV complications. In this cohort, 32.6% developed hypotension requiring treatment with intravenous fluids and pressors, 14% atrial fibrillation, and 2.3% troponin elevations suggestive of myocardial damage. No patients developed clinical heart failure, and among the patients that underwent echocardiography, there was no significant difference in mean left ventricular ejection fraction before or after therapy (62.9% vs. 63.5%, respectively, P=0.79). There was also no statistically significant difference in survival between those with and without CV complications (overall survival=61.9%, mean: 26.0 mo and progression-free survival=45.2%, mean: 18.1 mo). CV toxicities are common in ACT-TIL protocols; however, survival does not appear to be significantly affected. Further research is needed to define mechanisms and potential prevention strategies to help clinicians manage these complications and mitigate risk.
Collapse
Affiliation(s)
- Michael G Fradley
- Cardio-Oncology Center of Excellence, Division of Cardiology, University of Pennsylvania, Philadelphia, PA
| | - Rongras Damrongwatanasuk
- Cardio-Oncology Program, Division of Cardiology, University of South Florida and Moffitt Cancer Center
| | - Sanjay Chandrasekhar
- Cardio-Oncology Program, Division of Cardiology, University of South Florida and Moffitt Cancer Center
| | - Mohammed Alomar
- Cardio-Oncology Program, Division of Cardiology, University of South Florida and Moffitt Cancer Center
| | - Kevin E Kip
- Department of Biostatistics, University of South Florida
| | - Amod A Sarnaik
- Cutaneous Oncology Program, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
14
|
Cardiotoxic mechanisms of cancer immunotherapy - A systematic review. Int J Cardiol 2020; 323:179-187. [PMID: 32800915 DOI: 10.1016/j.ijcard.2020.08.033] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/26/2020] [Accepted: 08/07/2020] [Indexed: 02/08/2023]
Abstract
Cancer immunotherapy is a success story of translational medicine that has led to improved survival in patients with different difficult-to-treat types of cancer, such as metastasized melanoma, non-small cell lung cancer or renal cell carcinoma. These novel therapeutic agents exert their antitumor effects by activating the patients' immune system against cancer cells. Immunotherapy can be divided into active agents, such as anti-tumour vaccines or adoptive T-cell transfer, and passive immunotherapies like monoclonal antibodies, checkpoint inhibitors, cytokine therapy, bispecific T-cell engagers. After initial experimental use, broad clinical application revealed a number of important cardiovascular side effects of immunotherapeutics, which limit treatment options and decrease patients' prognosis and quality of life. With the rising rate of new immunotherapeutics at a hand, the number of patients receiving cancer immunotherapy will constantly increase, resulting in improved long-term survival rates. This review aims to summarize available cancer immunotherapies, their mechanism of action, currently known cardiovascular toxicities and their treatment. Further optimization of patient care will depend on the combined efforts by oncologists, cardiologists and cardiac surgeons to identify patients at risk and the implementation of interdisciplinary screening and treatment strategies. It is therefore crucial to familiarize heart specialists with novel cancer therapeutics and their potential adverse effects.
Collapse
|
15
|
Alexandre J, Salem JE, Moslehi J, Sassier M, Ropert C, Cautela J, Thuny F, Ederhy S, Cohen A, Damaj G, Vilque JP, Plane AF, Legallois D, Champ-Rigot L, Milliez P, Funck-Brentano C, Dolladille C. Identification of anticancer drugs associated with atrial fibrillation: analysis of the WHO pharmacovigilance database. EUROPEAN HEART JOURNAL. CARDIOVASCULAR PHARMACOTHERAPY 2020; 7:312-320. [PMID: 32353110 DOI: 10.1093/ehjcvp/pvaa037] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2020] [Revised: 03/05/2020] [Accepted: 04/22/2020] [Indexed: 02/04/2023]
Abstract
AIMS The explosion of novel anticancer therapies has meant emergence of cardiotoxicity signals including atrial fibrillation (AF). Reliable data concerning the liability of anticancer drugs in inducing AF are scarce. Using the World Health Organization individual case safety report database, VigiBase®, we aimed to determine the association between anticancer drugs and AF. METHODS AND RESULTS A disproportionality analysis evaluating the multivariable-adjusted reporting odds ratios for AF with their 99.97% confidence intervals was performed for 176 U.S. Food and Drug Administration (FDA)- or European Medicines Agency (EMA)-labelled anticancer drugs in VigiBase®, followed by a descriptive analysis of AF cases for the anticancer drugs identified in VigiBase®. ClinicalTrial registration number: NCT03530215. A total of 11 757 AF cases associated with at least one anticancer drug were identified in VigiBase® of which 95.8% were deemed serious. Nineteen anticancer drugs were significantly associated with AF of which 14 (74%) are used in haematologic malignancies and 9 (45%) represented new AF associations not previously confirmed in literature including immunomodulating agents (lenalidomide, pomalidomide), several kinase inhibitors (nilotinib, ponatinib, midostaurin), antimetabolites (azacytidine, clofarabine), docetaxel (taxane), and obinutuzumab, an anti-CD20 monoclonal antibody. CONCLUSION Although cancer malignancy itself may generate AF, we identified 19 anticancer drugs significantly associated with a significant increase in AF over-reporting. This pharmacovigilance study provides evidence that anticancer drugs themselves could represent independent risk factors for AF development. Dedicated prospective clinical trials are now required to confirm these 19 associations. This list of suspected anticancer drugs should be known by physicians when confronted to AF in cancer patients, particularly in case of haematologic malignancies.
Collapse
Affiliation(s)
- Joachim Alexandre
- PICARO Cardio-oncology Program, Department of Pharmacology, Normandie University, UNICAEN, CHU de Caen Normandie, EA 4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, F-14000 Caen, France
| | - Joe-Elie Salem
- Department of Pharmacology, AP-HP, Pitié-Salpêtrière Hospital, CIC-1421, INSERM, UMR ICAN 1166, Sorbonne Université, APHP.6 Cardio-oncology Program, F-75013 Paris, France.,Department of Medicine, Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Javid Moslehi
- Department of Medicine, Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Marion Sassier
- Department of Pharmacology, CHU de Caen Normandie, PICARO Cardio-oncology Program, F-14000 Caen, France
| | - Camille Ropert
- Department of Cardiology, CHU de Caen Normandie, F-14000 Caen, France
| | - Jennifer Cautela
- Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Aix-Marseille University, Assistance Publique-Hôpitaux de Marseille, Mediterranean University Cardio-Oncology Center, Hôpital Nord, Marseille 13915, France.,Centre de Recherche Cardiovasculaire et Nutrition, Inserm 1263, Inra, Marseille 13915, France.,Groupe Méditerranéen de Cardio-Oncologie, Marseille 13915, France.,Oncosafety Network of the Early Phases Cancer Trials Center, Marseille 13915, France
| | - Franck Thuny
- Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, Aix-Marseille University, Assistance Publique-Hôpitaux de Marseille, Mediterranean University Cardio-Oncology Center, Hôpital Nord, Marseille 13915, France.,Centre de Recherche Cardiovasculaire et Nutrition, Inserm 1263, Inra, Marseille 13915, France.,Groupe Méditerranéen de Cardio-Oncologie, Marseille 13915, France.,Oncosafety Network of the Early Phases Cancer Trials Center, Marseille 13915, France
| | - Stéphane Ederhy
- Hôpitaux Universitaires Paris-Est, Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine, Service de cardiologie, Unico, Unité de cardio-oncologie APHP.6, GRC Groupe de recherche clinique en cardio oncologie, Inserm 856, Université Pierre et Marie Curie, Paris, France
| | - Ariel Cohen
- Hôpitaux Universitaires Paris-Est, Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine, Service de cardiologie, Unico, Unité de cardio-oncologie APHP.6, GRC Groupe de recherche clinique en cardio oncologie, Inserm 856, Université Pierre et Marie Curie, Paris, France
| | - Ghandi Damaj
- Department of Hematology, CHU de Caen Normandie, F-14000 Caen, France
| | | | - Anne-Flore Plane
- Department of Cardiology, CHU de Caen Normandie, F-14000 Caen, France
| | - Damien Legallois
- Department of Cardiology, Normandie University, UNICAEN, CHU de Caen Normandie, EA 4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, F-14000 Caen, France
| | - Laure Champ-Rigot
- Department of Cardiology, Normandie University, UNICAEN, CHU de Caen Normandie, EA 4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, F-14000 Caen, France
| | - Paul Milliez
- Department of Cardiology, Normandie University, UNICAEN, CHU de Caen Normandie, EA 4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, F-14000 Caen, France
| | - Christian Funck-Brentano
- Department of Medicine, Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, TN 37240, USA
| | - Charles Dolladille
- PICARO Cardio-oncology Program, Department of Pharmacology, Normandie University, UNICAEN, CHU de Caen Normandie, EA 4650, Signalisation, électrophysiologie et imagerie des lésions d'ischémie-reperfusion myocardique, F-14000 Caen, France
| |
Collapse
|
16
|
Alexandre J, Moslehi JJ, Bersell KR, Funck-Brentano C, Roden DM, Salem JE. Anticancer drug-induced cardiac rhythm disorders: Current knowledge and basic underlying mechanisms. Pharmacol Ther 2018; 189:89-103. [PMID: 29698683 DOI: 10.1016/j.pharmthera.2018.04.009] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Significant advances in cancer treatment have resulted in decreased cancer related mortality for many malignancies with some cancer types now considered chronic diseases. Despite these improvements, there is increasing recognition that many cancer patients or cancer survivors can develop cardiovascular diseases, either due to the cancer itself or as a result of anticancer therapy. Much attention has focused on heart failure; however, other cardiotoxicities, notably cardiac rhythm disorders, can occur without underlying cardiomyopathy. Supraventricular tachycardias occur in cancer patients treated with cytotoxic chemotherapy (anthracyclines, gemcitabine, cisplatin and alkylating-agents) or kinase-inhibitors (KIs) such as ibrutinib. Ventricular arrhythmias, with a subset of them being torsades-de-pointes (TdP) favored by QTc prolongation have been reported: this may be the result of direct hERG-channel inhibition or a more recently-described mechanism of phosphoinositide-3-kinase inhibition. The major anticancer drugs responsible for QTc prolongation in this context are KIs, arsenic trioxide, anthracyclines, histone deacetylase inhibitors, and selective estrogen receptor modulators. Anticancer drug-induced cardiac rhythm disorders remain an underappreciated complication even by experienced clinicians. Moreover, the causal relationship of a particular anticancer drug with cardiac arrhythmia occurrence remains challenging due in part to patient comorbidities and complex treatment regimens. For example, any cancer patient may also be diagnosed with common diseases such as hypertension, diabetes or heart failure which increase an individual's arrhythmia susceptibility. Further, anticancer drugs are generally usually used in combination, increasing the challenge around establishing causation. Thus, arrhythmias appear to be an underappreciated adverse effect of anticancer agents and the incidence, significance and underlying mechanisms are now being investigated.
Collapse
Affiliation(s)
- Joachim Alexandre
- CHU Caen, PICARO Cardio-oncology Program, Department of Pharmacology, F-14033 Caen, France; Normandie Univ, UNICAEN, CHU Caen, EA 4650, Signalisation, Électrophysiologie et Imagerie des Lésions d'Ischémie-Reperfusion Myocardique, 14000 Caen, France
| | - Javid J Moslehi
- Vanderbilt University Medical Center, Cardio-oncology Program, Department of Medicine, Nashville, Tennessee, USA
| | - Kevin R Bersell
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Christian Funck-Brentano
- Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Pitié-Salpêtrière Hospital, Department of Pharmacology, F-75013 Paris, France
| | - Dan M Roden
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Joe-Elie Salem
- Vanderbilt University Medical Center, Cardio-oncology Program, Department of Medicine, Nashville, Tennessee, USA; Department of Pharmacology, Vanderbilt University Medical Center, Nashville, TN, USA; Sorbonne Université, INSERM CIC Paris-Est, AP-HP, ICAN, Pitié-Salpêtrière Hospital, Department of Pharmacology, F-75013 Paris, France.
| |
Collapse
|
17
|
Lissoni P, Galli MA, Tancini G, Barni S. Prevention by L-Carnitine of Interleukin-2-Related Cardiac Toxicity during Cancer Immunotherapy. TUMORI JOURNAL 2018; 79:202-4. [PMID: 8236505 DOI: 10.1177/030089169307900309] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Aims and Background Cardiac toxicity has been observed during IL-2 cancer immunotherapy. Because of its trophic action on the myocardial tissue, the use of L-carnitine has been evaluated during IL-2 therapy in advanced cancer patients with clinically important cardiac diseases. Methods The study included 30 cancer patients, who were randomized to treatment with IL-2 alone or IL-2 plus L-carnitine (1000 mg/day orally). IL-2 was injected subcutaneously at a daily dose of 6 million IU for 5 days/week for 4-6 weeks. Results The percentage of cardiac complications was significantly lower in patients concomitantly treated with L-carnitine than those receiving IL-2 alone (0/15 vs 4/15; P < 0.05), whereas no difference was seen in mean creatine phosphokinase levels on study. Conclusions The results would suggest that L-carnitine may be used successfully to prevent cardiac complications during IL-2 immunotherapy in cancer patients with clinically relevant cardiac disorders. Since cardiac metabolism depends mainly on fatty acid oxidation, the stimulatory role of L-carnitine on fatty acid oxidation could explain at least in part its ability to prevent heart disturbances in response to IL-2 administration.
Collapse
Affiliation(s)
- P Lissoni
- Division of Radiation Oncology, San Gerardo Hospital, Monza, Milan, Italy
| | | | | | | |
Collapse
|
18
|
Lissoni P, Barni S, Cattaneo G, Archili C, Perego M, Tancini G. Evaluation of the Cardiovascular Toxicity Related to Cancer Immunotherapy with Interleukin-2 by Monitoring Atrial Natriuretic Peptide Secretion: A Case Report. TUMORI JOURNAL 2018; 76:603-5. [PMID: 2149472 DOI: 10.1177/030089169007600620] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Increased capillary permeability and severe hypotension represent the two major cardiovascular complications of IL-2 immunotherapy. The mechanisms responsible for IL-2 cardiovascular toxicity are still obscure. Since increased vascular permeability and vasodilatation may be also induced by the cardiac hormone atrial natriuretic peptide (ANP), we have evaluated ANP concentrations in relation to mean arterial pressure in one patient with metastatic renal carcinoma, treated with a 24-h intravenous infusion of IL-2 at a dose of 3 × 106 Cetus U/m2/day for 5 days. The results showed that episodes of important hypotension were associated with abnormally high plasma levels of ANP. Owing to its vasodilator activity, exagerated ANP secretion, perhaps due to an inappropriate cardiac endocrine function in response to hemodynamic changes induced by IL-2, may play a role in hypotension, which occurs during IL-2 immunotherapy for cancer.
Collapse
Affiliation(s)
- P Lissoni
- Divisione di Radioterapia Oncologica, Ospedale San Gerardo, Monza, Milano, Italy
| | | | | | | | | | | |
Collapse
|
19
|
Lissoni P, Barni S, Cattaneo G, Archili C, Crispino S, Tancini G, D'Angelo L, Magni S, Fiorelli G. Activation of the Complement System during Immunotherapy of Cancer with Interleukin-2: A Possible Explanation of the Capillary Leak Syndrome. Int J Biol Markers 2018; 5:195-7. [PMID: 2093734 DOI: 10.1177/172460089000500405] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The capillary leak syndrome, responsible for fluid loss into the interstitial space, represents one of the major cardiovascular toxicities of IL-2 during the immunotherapy of cancer. The mechanisms involved in the increased vascular permeability have still to be better understood. The present study was carried out to investigate the role of the complement system in mediating the IL-2 vascular toxicity. The study was performed in metastatic renal cancer patients, treated with IL-2 through a 24-hour i.v. infusion at a daily dose of 3 × 106 U/m2 for 5 consecutive days, corresponding to one IL-2 course. Six IL-2 courses were evaluated. C3 and C4 were measured daily during IL-2 infusion, and 2 and 5 days after its interruption. IL-2 administration induced a significant decrease in both C3 and C4 mean levels, which became within the normal range 5 days after the end of IL-2 infusion. These results show that IL-2 administration may directly activate the complement system through the classical pathway, which might play a role in determining the increased vascular permeability.
Collapse
Affiliation(s)
- P Lissoni
- Division of Oncological Radiotherapy, San Gerardo Hospital, Monza, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
|
21
|
Wang DY, Okoye GD, Neilan TG, Johnson DB, Moslehi JJ. Cardiovascular Toxicities Associated with Cancer Immunotherapies. Curr Cardiol Rep 2017; 19:21. [PMID: 28220466 PMCID: PMC10176498 DOI: 10.1007/s11886-017-0835-0] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
PURPOSE OF REVIEW We review the cardiovascular toxicities associated with cancer immune therapies and discuss the cardiac manifestations, potential mechanisms, and management strategies. RECENT FINDINGS The recent advances in cancer immune therapy with immune checkpoint inhibitors and adoptive cell transfer have improved clinical outcomes in numerous cancers. The rising use of cancer immune therapy will lead to a higher incidence in immune-related adverse events. Recent studies have highlighted several reports of severe cases of acute cardiotoxic events with immune therapy including fulminant myocarditis. We believe that immune-mediated myocarditis is a driving mechanism behind these cardiovascular toxicities and requires vigilant screening and prompt management with corticosteroids and immune-modulating drugs, especially with combination immune therapies. While the incidence of serious cardiovascular toxicities with immune therapy appears low, these can be life-threatening especially when manifesting as acute immune-mediated myocarditis. Further collaborative studies are needed to effectively identify, characterize, and manage these events.
Collapse
Affiliation(s)
- Daniel Y Wang
- Divisions of Oncology, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg., Nashville, TN, 37232, USA
| | - Gosife Donald Okoye
- Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Thomas G Neilan
- Division of Cardiovascular Medicine, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.,Department of Medicine, Cardio-Oncology Program, Massachusetts General Hospital, Boston, MA, USA
| | - Douglas B Johnson
- Divisions of Oncology, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg., Nashville, TN, 37232, USA.
| | - Javid J Moslehi
- Divisions of Oncology, Department of Medicine, Vanderbilt University Medical Center, 2220 Pierce Ave, 777 Preston Research Bldg., Nashville, TN, 37232, USA.,Department of Medicine, Cardiovascular Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.,Department of Medicine, Cardio-Oncology Program, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
22
|
Lotze MT. Transplantation and Adoptive Cellular Therapy of Cancer: The Role of T-Cell Growth Factors. Cell Transplant 2017. [DOI: 10.1177/096368979300200106] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The ability to transfer cultured lymphocytes required the availability and the understanding of the use of the T-cell growth factors IL-2, IL-4, IL-7, and IL-12. Application of these cytokines in vitro and in vivo has allowed the modern development of adoptive transfer of tumor reactive lymphocytes to the modern immunotherapy of patients with cancer. In a randomized prospective study of IL-2 administration compared with IL-2 and lymphokine-activated killer (LAK) cells, no increase in response rate was observed. In a total of 90 patients randomized to receive LAK and IL-2 and 91 patients randomized to receive IL-2 alone, there were a total of 24 responses in patients receiving cells and IL-2 and 16 responses in those receiving IL-2 alone (no significant difference). There was some suggestion that complete responses were observed more often in melanoma patients treated with LAK and IL-2. The most interesting aspect of this study is the prolonged duration of responses, lasting for many months or years. Unfortunately, given the large numbers of variables that were examined, it became very difficult to demonstrate a clear-cut association between clinical outcome (response) and any variable that was routinely measured. Significant antitumor responses have been observed greater than expected with IL-2 alone, with the administration of tumor-infiltrating lymphocytes to patients with melanoma. We currently use hollow fiber devices (Cellco, Germantown, MD) to expand cells up through the many doublings required to generate approximately 1-2 × 1011 cells over a period of 6 wk in culture. In a recent review of the results in patients with melanoma treated on such regimens in combination with high-dose IL-2, an approximately 20-50% response rate has been observed. The factors associated with response are still unclear. Although we initially felt that it was associated with specific lysis, subsequent studies from our group suggest that the relevant factor is specific cytokine (INF-γ, GM-CSF, TNF) production upon tumor stimulation. Additional studies will need to be done to clarify these issues.
Collapse
Affiliation(s)
- Michael T. Lotze
- Department of Surgery, University of Pittsburgh Medical Center and the Pittsburgh Cancer Institute, Pittsburgh, PA 15261, USA
| |
Collapse
|
23
|
Chandra S, Carver J. Myocardial Ischemia and Cancer Therapy. CARDIO-ONCOLOGY 2017:123-137. [DOI: 10.1016/b978-0-12-803547-4.00008-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
24
|
Caorsi C, Quintana E, Valdés S, Muñoz C. Continuous cardiac output and hemodynamic monitoring: high temporal correlation between plasma TNF-α and hemodynamic changes during a sepsis-like state in cancer immunotherapy. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519030090020301] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Through continuous cardiac output monitoring, we investigated the temporal relationship between hemodynamic changes and plasma cytokines in a cancer patient who developed collateral sepsis to immunotherapy. A 52-year-old male with metastatic renal cell carcinoma received interleukin-2 (IL-2) infusion completing 72 h of administration. The patient developed 3 sepsis-like states including systemic inflammatory response syndrome (SIRS), shock, and multiple organ dysfunction syndrome (MODS). Hemodynamic parameters including systemic vascular resistance index (SVRI), left ventricular stroke work index (LVSWI) and cardiac index (CI) were measured over 60 h. Peripheral blood was drawn when SVRI dropped 20% in the patient and plasma cytokines including TNF-α , IL-6 and IL-1β were measured using ELISA. After 60 h of immunotherapy, the patient showed a 63.4% decrease in SVRI, 54.5% decrease in LVSWI and 65.4% increase in CI. The evaluation of systemic cytokines revealed different kinetic patterns: (i) a sustained increase in TNFα levels through all 3 sepsis-like states; (ii) IL-6 increased preferentially during SIRS and shock, while up/down-responses were found during MODS; (iii) IL-1β was undetectable during the entire study period. A high temporal relationship between hemodynamic changes and plasma TNF-α, but not IL-6, was found. Although there are factors other than cytokines that can alter vascular resistance, this finding could represent an approach to evaluate the course of hemodynamia and probably the systemic cytokine expression after IL-2 administration in renal cancer.
Collapse
Affiliation(s)
- Carlos Caorsi
- Intensive Care Unit, Clínica Las Condes, Santiago, Chile
| | | | - Sergio Valdés
- Intensive Care Unit, Clínica Las Condes, Santiago, Chile
| | - Carlos Muñoz
- Immunology Unit, INTA, University of Chile, Santiago, Chile,
| |
Collapse
|
25
|
Viganego F, Singh R, Fradley MG. Arrhythmias and Other Electrophysiology Issues in Cancer Patients Receiving Chemotherapy or Radiation. Curr Cardiol Rep 2016; 18:52. [DOI: 10.1007/s11886-016-0730-0] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
26
|
Chazova IE, Oshchepkova EV, Kantorova AY. [Comorbidity of cardiovascular diseases and cancers: Problems in the diagnosis of cardiotoxic effects of chemo- and radiation therapy]. TERAPEVT ARKH 2015; 87:4-10. [PMID: 26591546 DOI: 10.17116/terarkh20158794-10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Cardiovascular diseases (CVD) and cancers are the leaders in their prevalence and the major causes of death in economically developed countries, determining their high sociomedical significance in society. Improvement of methods for the early diagnosis and treatment of cancers has contributed to increases in relapse-free survival and life expectancy in these patients. At the same time, the new problems have emerged particularly in the development of various cardiovascular events/diseases when treating cancer, which may predict worse prognosis in patients and be an independent cause of death. To search for new markers of cardiotoxicity at early stages and to develop effective methods for the prevention and personalized treatment of cancer and CVD are the problems that can be solved only by joint efforts of cardiologists and oncologists.
Collapse
Affiliation(s)
- I E Chazova
- Russian Cardiology Research-and-Production Complex, Ministry of Health of Russia, Moscow
| | - E V Oshchepkova
- Russian Cardiology Research-and-Production Complex, Ministry of Health of Russia, Moscow
| | - A Yu Kantorova
- Russian Cardiology Research-and-Production Complex, Ministry of Health of Russia, Moscow
| |
Collapse
|
27
|
Oprea AD, Russell RR, Russell KS, Abu-Khalaf M. Chemotherapy Agents With Known Cardiovascular Side Effects and Their Anesthetic Implications. J Cardiothorac Vasc Anesth 2015; 31:2206-2226. [PMID: 26952170 DOI: 10.1053/j.jvca.2015.06.020] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Indexed: 01/11/2023]
|
28
|
Plasma angiopoietin 2 concentrations are related to impaired lung function and organ failure in a clinical cohort receiving high-dose interleukin 2 therapy. Shock 2015; 42:115-20. [PMID: 24727870 DOI: 10.1097/shk.0000000000000188] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
INTRODUCTION The pathophysiology and therapeutic options in sepsis-induced lung injury remain elusive. High-dose interleukin 2 therapy (HDIL-2) is an important protocol for advanced malignancies but is limited by systemic inflammation and pulmonary edema that is indistinguishable from sepsis. In preclinical models, IL-2 stimulates angiopoietin 2 (AngP-2) secretion, which increases endothelial permeability and causes pulmonary edema. However, these relationships have not been fully elucidated in humans. Furthermore, the relevance of plasma AngP-2 to organ function is not clear. We hypothesized that plasma AngP-2 concentrations increase during HDIL-2 and are relevant to clinical pathophysiology. METHODS We enrolled 13 subjects with metastatic melanoma or renal cell carcinoma admitted to receive HDIL-2 and collected blood and spirometry data daily. The plasma concentrations of AngP-2 and IL-6 were measured with enzyme-linked immunosorbent assay. RESULTS At baseline, the mean AngP-2 concentration was 2.5 (SD, 1.0) ng/mL. Angiopoietin 2 concentrations increased during treatment: the mean concentration on the penultimate day was 16.0 (SD, 4.5) ng/mL and increased further to 18.6 (SD, 4.9) ng/mL (P < 0.05 vs. penultimate) during the last day of therapy. The forced expiratory volume in 1 s decreased during treatment. Interestingly, plasma AngP-2 concentrations correlated negatively with forced expiratory volume in 1 s (Spearman r = -0.78, P < 0.0001). Plasma AngP-2 concentrations also correlated with plasma IL-6 concentrations (r = 0.61, P < 0.0001) and Sequential Organ Failure Assessment scores (r = 0.68, P < 0.0001). CONCLUSIONS Plasma AngP-2 concentrations increase during HDIL-2 administration and correlate with pulmonary dysfunction. High-dose IL-2 may serve as a clinical model of sepsis and acute lung injury. Further investigation is warranted.
Collapse
|
29
|
Cabrera-Bueno F, Medina-Palomo C, Ruiz-Salas A, Flores A, Rodríguez-Losada N, Barrera A, Jiménez-Navarro M, Alzueta J. Serum levels of interleukin-2 predict the recurrence of atrial fibrillation after pulmonary vein ablation. Cytokine 2015; 73:74-8. [PMID: 25743240 DOI: 10.1016/j.cyto.2015.01.026] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2014] [Revised: 10/16/2014] [Accepted: 01/23/2015] [Indexed: 10/23/2022]
Abstract
AIMS Interleukin-2 has a significant antitumor activity in some types of cancer, and has been associated with the development of atrial fibrillation (AF). In addition, IL-2 serum levels in recent onset AF have been related with pharmaceutical cardioversion outcomes. We evaluated the hypothesis that a relationship exists between inflammation and the outcome of catheter ablation of AF. METHODS We studied 44 patients with paroxysmal AF who underwent catheter ablation. Patients with structural heart disease, coronary artery or valve disease, active inflammatory disease, known or suspected neoplasm, endocrinopathies, or exposure to anti-inflammatory drugs were excluded. All study participants underwent evaluation with a standardized protocol, including echocardiography, and cytokine levels of interleukin-2, interleukin-4, interleukin-6, interleukin-10, tumour necrosis factor-alpha, and gamma-interferon determination before procedure. Clinical and electrocardiographic follow-up were performed with Holter-ECG at 3, 6 and 12months in order to know if sinus rhythm was maintained. RESULTS After catheter ablation of the 44 patients included (53±10years, 27.3% female), all patients returned to sinus rhythm. During the first year of follow-up seven patients (15.9%) experienced recurrence of AF. The demographics, clinical and echocardiographic features, and pharmacological treatments of these patients were similar to those who maintained sinus rhythm. The only independent factor predictive of recurrence of AF was an elevated level of IL-2 (OR 1.18, 95% CI 1.12-1.38). CONCLUSIONS High serum levels of interleukin-2, a pro-inflammatory non-vascular cytokine, are associated with the recurrence of AF in patients undergoing catheter ablation.
Collapse
Affiliation(s)
- Fernando Cabrera-Bueno
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain.
| | - Carmen Medina-Palomo
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain
| | - Amalio Ruiz-Salas
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain
| | - Ana Flores
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain
| | - Noela Rodríguez-Losada
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain
| | - Alberto Barrera
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain
| | - Manuel Jiménez-Navarro
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain
| | - Javier Alzueta
- UGC del Corazón, Servicio de Cardiología, Hospital Clínico Universitario Virgen de la Victoria de Málaga, IMIBA (Instituto de Investigación Biomédica de Málaga), Universidad de Málaga, Spain
| |
Collapse
|
30
|
Tamargo J, Caballero R, Delpón E. Cancer Chemotherapy and Cardiac Arrhythmias: A Review. Drug Saf 2015; 38:129-52. [DOI: 10.1007/s40264-014-0258-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
31
|
Abstract
Cytokines, currently known to be more than 130 in number, are small MW (<30 kDa) key signaling proteins that modulate cellular activities in immunity, infection, inflammation and malignancy. Key to understanding their function is recognition of their pleiotropism and often overlapping and functional redundancies. Classified here into 9 main families, most of the 20 approved cytokine preparations (18 different cytokines; 3 pegylated), all in recombinant human (rh) form, are grouped in the hematopoietic growth factor, interferon, platelet-derived growth factor (PDGF) and transforming growth factor β (TGFβ) families. In the hematopoietin family, approved cytokines are aldesleukin (rhIL-2), oprelvekin (rhIL-11), filgrastim and tbo-filgrastim (rhG-CSF), sargramostim (rhGM-CSF), metreleptin (rh-leptin) and the rh-erythropoietins, epoetin and darbepoietin alfa. Anakinra, a recombinant receptor antagonist for IL-1, is in the IL-1 family; recombinant interferons alfa-1, alfa-2, beta-1 and gamma-1 make up the interferon family; palifermin (rhKGF) and becaplermin (rhPDGF) are in the PDGF family; and rhBMP-2 and rhBMP-7 represent the TGFβ family. The main physicochemical features, FDA-approved indications, modes of action and side effects of these approved cytokines are presented. Underlying each adverse events profile is their pleiotropism, potency and capacity to release other cytokines producing cytokine 'cocktails'. Side effects, some serious, occur despite cytokines being endogenous proteins, and this therefore demands caution in attempts to introduce individual members into the clinic. This caution is reflected in the relatively small number of cytokines currently approved by regulatory agencies and by the fact that 14 of the FDA-approved preparations carry warnings, with 10 being black box warnings.
Collapse
|
32
|
Dutcher JP, Schwartzentruber DJ, Kaufman HL, Agarwala SS, Tarhini AA, Lowder JN, Atkins MB. High dose interleukin-2 (Aldesleukin) - expert consensus on best management practices-2014. J Immunother Cancer 2014; 2:26. [PMID: 31546315 PMCID: PMC6889624 DOI: 10.1186/s40425-014-0026-0] [Citation(s) in RCA: 143] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2014] [Accepted: 07/16/2014] [Indexed: 12/28/2022] Open
Abstract
Interleukin-2 (IL-2) was historically one of the few treatments for adults with stage IV solid tumors that could produce complete responses (CRs) that were often durable for decades without further therapy. The majority of complete responders with metastatic renal cell carcinoma (mRCC) and metastatic melanoma (mM) could probably be classified as "cures". Recent publications have suggested improved efficacy, perhaps due to improved patient selection based on a better understanding of clinical features predicting outcomes. Guidelines for clinical management were established from experience at the National Cancer Institute (NCI) and an affiliation of institutions known as the Cytokine Working Group (CWG), who were among the first to utilize HD IL-2 treatment outside of the NCI. As new centers have opened, further management variations have emerged based upon center-specific experience, to optimize administration of IL-2 and provide high quality care for patients at each individual site. Twenty years of evolution in differing environments has led to a plethora of clinical experience and effective management approaches. The goal of this review is to summarize the spectrum of HD IL-2 treatment approaches, describing various effective strategies that incorporate newer adjunctive treatments for managing the side effects of IL-2 in patients with mRCC and mM. The goal for IL-2 therapy is typically to administer the maximum number of doses of IL-2 without putting the patient at unacceptable risk for severe, irreversible toxicity. This review is based upon a consensus meeting and includes guidelines on pre-treatment screening, criteria for administration and withholding doses, and defines consensus criteria for safe administration and toxicity management. The somewhat heterogeneous best practices of 2014 will be compared and contrasted with the guidelines provided in 2001 and the package inserts from 1992 and 1998.
Collapse
Affiliation(s)
- Janice P Dutcher
- Associate Director, Cancer Research Foundation, Chappaqua, NY, USA.
| | - Douglas J Schwartzentruber
- Associate Director of Clinical Operations, Professor of Surgery, IU Simon Cancer Center, 550 N University Blvd, Indianapolis, 46202, IN, USA
| | - Howard L Kaufman
- Chief Surgical Officer and Associate Director for Clinical Science, Professor of Surgery, Rutgers Cancer Center Institute of New Jersey, 195 Little Albany Street, Room 2007, New Brunswick, 08901, NJ, USA
| | - Sanjiv S Agarwala
- Chief of Medical Oncology and Professor of Medicine, St. Luke's Cancer Center, Bethlehem, 18015, PA, USA
| | - Ahmad A Tarhini
- Associate Professor of Medicine and Translational Science, University of Pittsburgh Cancer Institute, Suite 555, 5150 Centre Ave, Pittsburgh, 15232, PA, USA
| | - James N Lowder
- Senior Medical Director, Prometheus Laboratories Inc, 9410 Carroll Park Drive, San Diego, 92121, CA, USA
| | - Michael B Atkins
- Deputy Director, Professor of Medicine, Georgetown-Lombardi Comprehensive Cancer Center, 3970 Reservoir Rd NW, NRB-E501, Washington, 20057, DC, USA
| |
Collapse
|
33
|
Abstract
Atrial fibrillation (AF) is a common cardiac arrhythmia that is associated with severe consequences, including symptoms, haemodynamic instability, increased cardiovascular mortality and stroke. While other arrhythmias such as torsades de pointes and sinus bradycardia are more typically thought of as drug induced, AF may also be precipitated by drug therapy, although ascribing causality to drug-associated AF is more difficult than with other drug-induced arrhythmias. Drug-induced AF is more likely to occur in patients with risk factors and co-morbidities that commonly co-exist with AF, such as advanced age, alcohol consumption, family history of AF, hypertension, thyroid dysfunction, sleep apnoea and heart disease. New-onset AF has been associated with cardiovascular drugs such as adenosine, dobutamine and milrinone. In addition, medications such as corticosteroids, ondansetron and antineoplastic agents such as paclitaxel, mitoxantrone and doxorubicin have been reported to induce AF. Whether bisphosphonate drugs are associated with new-onset AF remains controversial and requires further study. The potential contribution of specific drug therapy should be considered when patients present with new-onset AF.
Collapse
Affiliation(s)
- Yaman Kaakeh
- Department of Pharmacy Practice, College of Pharmacy, Purdue University, West Lafayette Indianapolis, IN, USA
| | | | | | | |
Collapse
|
34
|
Abstract
INTRODUCTION Atrial fibrillation (AF) is the most common arrhythmia and an important cause of hospitalization, morbidity, and mortality. A myriad of drugs can induce AF. However, drug-induced AF (DIAF) receives little attention. Thus, this review is an attempt to attract the attention on this adverse effect. AREAS COVERED Published reports of drug-induced AF (DIAF) are reviewed in this paper, from January 1974 to December 2011, using the PubMed/Medline database and lateral references. EXPERT OPINION In most cases, DIAF is paroxysmal and terminates spontaneously, but sometimes AF persists and it is necessary to perform a cardioversion to restore sinus rhythm and avoid progression to persistent AF. Because of the short duration of DIAF, in addition to physicians/patients not being knowledgeable about this side effect, the real incidence and clinical consequences of DIAF are presently unknown. DIAF is an increasing problem, as some widely prescribed drugs can present this adverse effect. The risk is expected to increase in the elderly and in patients with comorbidities. It is important that physicians understand the significance of DIAF, to increase the collaboration between cardiac and non-cardiac professionals, and to educate patients to make them aware of this adverse side effect.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, 28040 Madrid, Spain.
| | | | | |
Collapse
|
35
|
Hong RA, Iimura T, Sumida KN, Eager RM. Cardio-oncology/onco-cardiology. Clin Cardiol 2010; 33:733-7. [PMID: 21184556 PMCID: PMC6653579 DOI: 10.1002/clc.20823] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2010] [Accepted: 06/24/2010] [Indexed: 02/06/2023] Open
Abstract
An understanding of onco-cardiology or cardio-oncology is critical for the effective care of cancer patients. Virtually all antineoplastic agents are associated with cardiotoxicity, which can be divided into 5 categories: direct cytotoxic effects of chemotherapy and associated cardiac systolic dysfunction, cardiac ischemia, arrhythmias, pericarditis, and chemotherapy-induced repolarization abnormalities. Radiation therapy can also lead to coronary artery disease and fibrotic changes to the valves, pericardium, and myocardium. All patients being considered for chemotherapy, especially those who have prior cardiac history, should undergo detailed cardiovascular evaluation to optimize the treatment. Serial assessment of left ventricular systolic function and cardiac biomarkers might also be considered in selected patient populations. Cardiotoxic effects of chemotherapy might be decreased by the concurrent use of angiotensin-converting enzyme inhibitors, angiotensin receptor blockers, or beta-blockers. Antiplatelet or anticoagulation therapy might be considered in patients with a potential hypercoagulable state associated with chemotherapy or cancer. Open dialogue between both cardiologists and oncologists will be required for optimal patient care.
Collapse
Affiliation(s)
- Robert A. Hong
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| | - Takeshi Iimura
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| | - Kenneth N. Sumida
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| | - Robert M. Eager
- Internal Medicine Residency Program, The Queen's Medical Center, Honolulu, Hawaii
| |
Collapse
|
36
|
Guglin M, Aljayeh M, Saiyad S, Ali R, Curtis AB. Introducing a new entity: chemotherapy-induced arrhythmia. Europace 2009; 11:1579-86. [DOI: 10.1093/europace/eup300] [Citation(s) in RCA: 138] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
37
|
|
38
|
Inducible Nitric Oxide Synthase (iNOS) is Not Required for IL-2–induced Hypotension and Vascular Leak Syndrome in Mice. J Immunother 2008; 31:325-33. [DOI: 10.1097/cji.0b013e31816112e8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
39
|
Curti BD, Longo DL. Intensive Care of the Cancer Patient. Crit Care Med 2008. [DOI: 10.1016/b978-032304841-5.50083-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
40
|
Gallagher DC, Bhatt RS, Parikh SM, Patel P, Seery V, McDermott DF, Atkins MB, Sukhatme VP. Angiopoietin 2 Is a Potential Mediator of High-Dose Interleukin 2–Induced Vascular Leak. Clin Cancer Res 2007; 13:2115-20. [PMID: 17404094 DOI: 10.1158/1078-0432.ccr-06-2509] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE High-dose interleukin 2 (HDIL2) produces durable tumor regressions in 10% of patients with metastatic renal cell carcinoma and melanoma. However, a major toxicity is vascular leak syndrome (VLS). We previously reported elevated serum angiopoietin 2 (Ang2) in septic patients with vascular leak and hypothesized that Ang2 might also contribute to HDIL2 VLS. EXPERIMENTAL DESIGN Blood was collected from 14 patients receiving HDIL2 and from 4 patients receiving HDIL2 and bevacizumab, an antibody against vascular endothelial growth factor (VEGF). The effect of Ang2 was studied in vitro by incubating high Ang2 patient serum with cultured endothelial cells. RESULTS Pretreatment Ang2 levels were in the reference range (median, 3.3 ng/mL) and rose with each day of IL-2 therapy (median peak, 29.7 ng/mL). No trend was seen in free VEGF levels during therapy. Patients treated with HDIL2 and bevacizumab all developed VLS and elevated Ang2. High Ang2 patient sera induced propermeability structural changes in endothelial cells, an effect reversed by blockade with the competitive ligand angiopoietin 1 (Ang1). CONCLUSIONS Ang2 may be a mediator of HDIL2 VLS as evidenced by (a) an increase in Ang2 in all patients on HDIL2; (b) the effect of high Ang2 patient serum on cultured endothelial cells; (c) rescue of those structural changes by Ang1. The lack of correlation between VLS and serum VEGF levels in patients treated with HDIL2 alone or in combination with bevacizumab suggests that VEGF is not a major contributor to VLS or Ang2 release. These data suggest that the inhibition of Ang2 may mitigate VLS in patients receiving HDIL2.
Collapse
Affiliation(s)
- Diana C Gallagher
- Division of Pulmonary and Critical Care, Department of Medicine, Beth Israel Deaconess Medical Center, Boston, Massachusetts 02215, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
|
42
|
Jones RL, Ewer MS. Cardiac and cardiovascular toxicity of nonanthracycline anticancer drugs. Expert Rev Anticancer Ther 2006; 6:1249-69. [PMID: 17020459 DOI: 10.1586/14737140.6.9.1249] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Anthracyclines are a well-known cause of cardiotoxicity, but a number of other drugs used to treat cancer can also result in cardiac and cardiovascular adverse effects. Cardiotoxicity can result in the alteration of cardiac rhythm, changes in blood pressure and ischemia, and can also alter the ability of the heart to contract and/or relax. The clinical spectrum of these toxicities can range from subclinical abnormalities to catastrophic life-threatening, and sometimes fatal, sequelae. These events may occur acutely or may only become apparent months or years following completion of oncological treatment. Ischemia and rhythm abnormalities are treated symptomatically in most cases. Knowledge of these toxicities can aid clinicians to choose the optimal and least toxic regimen suitable for an individual patient.
Collapse
Affiliation(s)
- Robin L Jones
- Royal Marsden Hospital, Department of Medicine, Fulham Road, London SW3 6JJ, UK.
| | | |
Collapse
|
43
|
Samlowski WE, McGregor JR, Jurek M, Baudys M, Zentner GM, Fowers KD. ReGel® Polymer-based Delivery of Interleukin-2 as a Cancer Treatment. J Immunother 2006; 29:524-35. [PMID: 16971808 DOI: 10.1097/01.cji.0000211306.05869.25] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
ReGel is an aqueous, filter sterilizable ABA tri-block polymer consisting of poly-(lactide-co-glycolide) and polyethylene glycol. We tested the suitability of this polymer to provide sustained interleukin-2 (IL-2) delivery for cancer immunotherapy. ReGel/IL-2 is liquid at or below room temperature, and is easily injectable through narrow gauge needles, but undergoes a reversible thermal transition into a bioerodible depot at body temperature. We demonstrated that ReGel/IL-2 releases IL-2 over 72 to 96 hours in vitro, without loss of bioactivity. Pharmacokinetic studies after peritumoral injection of 0.1 mL ReGel/IL-2 in mice demonstrated an early burst of IL-2 release, followed by more sustained release kinetics over 96 hours (T(1/2)beta 48 h). Less than 1.5% of the injected dose was detectable in blood or kidneys during the first 48 hours. A single peritumoral dose of ReGel/IL-2 [1 to 4 million international units (MIU) ReGel/IL-2, split into 4 quadrant injections] was administered to mice bearing subcutaneous RD-995 spindle cell carcinoma. Only the highest dose of ReGel/IL-2 tested (4.0 MIU) resulted in significant hypotension on day 3 after injection. Weekly treatment of Meth A fibrosarcoma and RENCA renal carcinoma with ReGel/IL-2 (2 MIU/dose) induced a significant reduction in tumor growth and improved survival. Reduction in tumor growth at implants remote from treated lesions was also observed, suggesting systemic activation of antitumor immunity. These findings establish that peritumoral injection of ReGel/IL-2 is an effective delivery system for cancer immunotherapy, while decreasing IL-2 toxicity. This polymer delivery system is likely to be broadly applicable for sustained delivery of other cytokines and peptides.
Collapse
Affiliation(s)
- Wolfram E Samlowski
- Multidisciplinary Melanoma Program, Huntsman Cancer Institute, 2000 Circle of Hope Drive, Salt Lake City, UT 84112, USA.
| | | | | | | | | | | |
Collapse
|
44
|
Alak AM, Moy S. Determination of NG-Monomethyl-L-arginine in Human and Dog Serum Using Pre-Column o-Phthaldialdehyde Derivatization and High Performance Liquid Chromatography. J LIQ CHROMATOGR R T 2006. [DOI: 10.1080/10826079708005546] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Affiliation(s)
- A. M. Alak
- a Fujisawa Research Institute of America, Inc. Northwestern University/Evanston Research Park , 1801 Maple Avenue, Evanston, Illinois, 60201
| | - S. Moy
- a Fujisawa Research Institute of America, Inc. Northwestern University/Evanston Research Park , 1801 Maple Avenue, Evanston, Illinois, 60201
| |
Collapse
|
45
|
Rossi L, Martin BM, Hortin GL, White RL, Foster M, Moharram R, Stroncek D, Wang E, Marincola FM, Panelli MC. Inflammatory protein profile during systemic high dose interleukin-2 administration. Proteomics 2006; 6:709-20. [PMID: 16342236 DOI: 10.1002/pmic.200500004] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Systemic interleukin-2 (IL-2) administration induces an assortment of downstream effects whose biological and therapeutic significance remains unexplored mostly because of the methodological inability to globally address their complexity. Protein array analysis of sera from patients with renal cell carcinoma obtained prior and during high-dose IL-2 therapy had previously revealed extensive alterations in expression of the soluble factors analyzed, whose discovery was limited by the number of capture antibodies selected for protein detection. Here, we expanded the analysis to SELDI-TOF-MS and quantitative protein analysis (nephelometry). All cytokines/chemokines detected by protein arrays were below the SELDI detection limit, while novel IL-2-specific changes in expression of acute-phase reactants and high-density lipoprotein metabolites could be identified. Serum amyloid protein A (SAA) and C-reactive protein expression were consistently up-regulated after four doses of IL-2, while other proteins were down-regulated. These findings were confirmed by SELDI immunoaffinity capture and nephelometry. Immunoaffinity capture revealed different, otherwise undetectable, isoforms of SAA. A linear correlation between peak area by SELDI and protein concentration by nephelometry was observed. Overall distinct yet complementary information was obtained using different platforms, which may better illustrate complex phenomena such as the systemic response to biological response modifiers.
Collapse
Affiliation(s)
- Leonardo Rossi
- Department of Human Morphology and Applied Biology, University of Pisa, Pisa, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Floyd JD, Nguyen DT, Lobins RL, Bashir Q, Doll DC, Perry MC. Cardiotoxicity of cancer therapy. J Clin Oncol 2005; 23:7685-96. [PMID: 16234530 DOI: 10.1200/jco.2005.08.789] [Citation(s) in RCA: 234] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Because cancer is a leading cause of mortality in the United States, the number of therapeutic modalities available for the treatment of neoplastic processes has increased. This has resulted in a large number of patients being exposed to a wide variety of cancer therapy. Historically, it has been well recognized that antineoplastic agents may have adverse effects on multiple organs and normal tissues. The most commonly associated toxicities occur in tissues composed of rapidly dividing cells and may spontaneously reverse with minimal long-term toxicity. However, the myocardium consists of cells that have limited regenerative capability, which may render the heart susceptible to permanent or transient adverse effects from chemotherapeutic agents. Such toxicity encompasses a heterogeneous group of disorders, ranging from relatively benign arrhythmias to potentially lethal conditions such as myocardial ischemia/infarction and cardiomyopathy. In some instances, the pathogenesis of these toxic effects has been elucidated, whereas in others the precise etiology remains unknown. We review herein the various syndromes of cardiac toxicity that are reported to be associated with antineoplastic agents and discuss their putative mechanisms and treatment.
Collapse
Affiliation(s)
- Justin D Floyd
- University of Missouri-Columbia, Ellis Fischel Cancer Center, 115 Business Loop 70 W, Columbia, MO 65203, USA
| | | | | | | | | | | |
Collapse
|
47
|
Suh RD, Goldin JG, Wallace AB, Sheehan RE, Heinze SB, Gitlitz BJ, Figlin RA. Metastatic renal cell carcinoma: CT-guided immunotherapy as a technically feasible and safe approach to delivery of gene therapy for treatment. Radiology 2004; 231:359-64. [PMID: 15128982 DOI: 10.1148/radiol.2312021754] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To assess the technical feasibility and safety of weekly outpatient percutaneous computed tomographic (CT)-guided intratumoral injections of interleukin-2 (IL-2) plasmid DNA in a wide variety of superficial and deep tumor sites. MATERIALS AND METHODS Twenty-nine patients with metastatic renal cell carcinoma and a total of 30 lesions measuring 1.0 cm(2) or greater in accessible thoracic (n = 15) or abdominal (n = 15) locations underwent up to three cycles of six weekly intratumoral IL-2 plasmid DNA injections. CT was used to guide needle placement and injection. After injection cycle 1, patients whose tumors demonstrated stable (< or =25% increase and < or =50% decrease in product of lesion diameters) or decreased size (>50% decrease in product of lesion diameters) advanced to injection cycle 2. Patients whose lesions decreased in size by more than 50% over the course of injection cycle 2 were eligible to begin injection cycle 3. An acceptable safety and technical feasibility profile for this technique was deemed to be (a) a safety and feasibility profile similar to that of single-needle biopsy and (b) an absence of serious adverse events (as defined in Title 21 of the Code of Federal Regulations) and/or unacceptable toxicities (as graded according to the National Cancer Institute Common Toxicity Criteria). RESULTS A total of 284 intratumoral injections were performed, with a mean of 9.8 injections (range, 6-18 injections) received by each patient. Technical success (needle placement and injection of gene therapy agent) was achieved in all cases. Complications were experienced after 42 (14.8%) of the 284 injections. The most common complication was pneumothorax (at 32 [28.6%] of 112 intrathoracic injections), for which only one patient required catheter drainage. Complications occurred randomly throughout injection cycles and did not appear to increase as patients received more injections (P =.532). No patient experienced serious adverse events or unacceptable toxicities. CONCLUSION Percutaneous CT-guided intratumoral immunotherapy injections are technically feasible and can be safely performed.
Collapse
Affiliation(s)
- Robert D Suh
- Department of Radiological Sciences, UCLA Medical Center, 10833 Le Conte Ave, B2-168 CHS, Los Angeles, CA 90095-1721, USA.
| | | | | | | | | | | | | |
Collapse
|
48
|
Samlowski WE, Petersen R, Cuzzocrea S, Macarthur H, Burton D, McGregor JR, Salvemini D. A nonpeptidyl mimic of superoxide dismutase, M40403, inhibits dose-limiting hypotension associated with interleukin-2 and increases its antitumor effects. Nat Med 2003; 9:750-5. [PMID: 12730689 DOI: 10.1038/nm874] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2003] [Accepted: 03/21/2003] [Indexed: 11/09/2022]
Abstract
Interleukin-2 (IL-2) is used to treat metastatic renal cell carcinoma and malignant melanoma, but its use is limited by the severe hypotension it produces. We have shown here that M40403, a superoxide dismutase (SOD) mimetic, blocked IL-2-induced hypotension and allowed the dose of IL-2 to be increased in mice. The reversal of IL-2-mediated hypotension was associated with an increase in plasma catecholamines. In addition, M40403 increased lymphokine-activated killer (LAK) cell cytotoxicity in vitro and in vivo, through inhibition of macrophage superoxide production. Treatment of methylcholanthrene-induced (Meth A) ascites tumors with IL-2 and > or =3 mg per kg body weight M40403 induced 50% complete remissions lasting for more than 200 d, which was longer than those of untreated mice (15-d median survival) or mice treated with IL-2 alone (22-d median). Growth of subcutaneous implants of RENCA renal carcinoma was also inhibited by the combination of IL-2 and M40403. These results established that M40403 prevented IL-2 from causing dose-limiting hypotension, while enhancing its anticancer activity.
Collapse
Affiliation(s)
- Wolfram E Samlowski
- Melanoma Program, Huntsman Cancer Institute, University of Utah, 2000 Circle of Hope Drive, Salt Lake City, Utah 84112, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Epstein AL, Mizokami MM, Li J, Hu P, Khawli LA. Identification of a protein fragment of interleukin 2 responsible for vasopermeability. J Natl Cancer Inst 2003; 95:741-9. [PMID: 12759392 DOI: 10.1093/jnci/95.10.741] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The cytokine interleukin 2 (IL-2) is involved in the activation of T cells and has been shown to play a central role in cancer immunotherapy. The full therapeutic potential of IL-2, however, has not been realized because of its dose-limiting systemic toxicity. We sought to identify a region of IL-2 that is responsible for the induction of vasopermeability (leaky tumor endothelium), a property associated with the toxicity of the molecule. METHODS Intact IL-2 or overlapping synthetic peptides of IL-2 that were chemically conjugated to tumor-targeting monoclonal antibodies (TNT-1 or Lym-1) were injected into groups of mice (n = 4) that had previously been xenotransplanted with human tumor cells (ME-180 cervical carcinoma and Raji lymphoma). Two hours later, mice received intravenous injections of radiolabeled tracer antibody, and 3 days later they were subjected to biodistribution analysis to measure the ability of each immunoconjugate to enhance tumor uptake of the tracer antibody (i.e., vasopermeability activity). The cytokine activity of the immunoconjugates was determined by assaying their ability to promote the proliferation of a mouse IL-2-dependent cell line. RESULTS Pretreatment of mice with an antibody/IL-2 immunoconjugate resulted in an approximately fourfold increase in radiolabeled tracer antibody uptake in the xenograft tumor as compared with uptake in mice injected with antibody alone. One synthetic fragment consisting of amino acids 22-58 contained 100% of the vasopermeability activity of IL-2 and was designated permeability-enhancing peptide (PEP). PEP had vasopermeability activity only when conjugated to a tumor-targeting antibody, had maximal activity as a dimer, and was devoid of cytokine activity. CONCLUSIONS The identification of PEP should aid in the discovery of ways to decrease the toxicity of IL-2. Moreover, PEP is a promising candidate for the generation of agents that can enhance the delivery of antibodies and drugs to tumors.
Collapse
Affiliation(s)
- Alan L Epstein
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | |
Collapse
|
50
|
Dutcher J, Atkins MB, Margolin K, Weiss G, Clark J, Sosman J, Logan T, Aronson F, Mier J. Kidney cancer: the Cytokine Working Group experience (1986-2001): part II. Management of IL-2 toxicity and studies with other cytokines. Med Oncol 2002; 18:209-19. [PMID: 11917945 DOI: 10.1385/mo:18:3:209] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The Cytokine Working Group (CWG) was initially established in 1986 as the Extramural IL-2/LAK Working Group. With funding from the National Cancer Institute (NCI), the CWG was mandated to confirming data regarding the efficacy of the high-dose interleukin-2 (IL2)/lymphokine-activated killer cell (LAK cell) regimen piloted at the NCI in the treatment of renal cell cancer. Since those initial studies, the CWG has conducted a series of clinical trials, often with correlative immunologic investigations, to evaluate combination immunotherapy in attempts to enhance the efficacy of IL-2 or to reduce toxicity. Subsequently, the CWG conducted trials to demonstrate the activity of lower-dose outpatient combination cytokine regimens to help determine their role in the armamentarium of treatment for metastatic renal cell cancer. This has culminated in a phase III randomized trial comparing the activity of high-dose IL-2 with the activity of outpatient IL-2 plus interferon-alpha. The CWG also has honed the management of both high-dose IL-2 and outpatient IL-2 regimens to make these safer in the hands of experienced clinicians. In addition, the CWG has produced a series of carefully conducted clinical trials of new cytokines, again attempting to define their clinical efficacy as anticancer agents. These include studies of IL-4, IL-6, and IL-12. Currently, the CWG is conducting studies with new approaches to IL-2 therapy, as well as planning trials with new agents for treatment of renal cell cancer. This review describes these efforts conducted over the past 15 yr.
Collapse
Affiliation(s)
- J Dutcher
- Our Lady of Mercy/New York Medical College, Bronx 10466, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|