1
|
Jing H, Cao X, Li K, Liu Y, Meng M, Liu S, Ye M, Zhang J, Wu Y. PLA2G2D promotes immune escape in non-small cell lung cancer by regulating T cell immune function through PD-L1-expressing extracellular vesicles. Scand J Immunol 2024; 100:e13393. [PMID: 38922971 DOI: 10.1111/sji.13393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 05/31/2024] [Accepted: 06/02/2024] [Indexed: 06/28/2024]
Abstract
It is urgent to explore factors affecting immunotherapy efficacy to benefit non-small cell lung cancer (NSCLC) patient survival. Bioinformatics predicted genes associated with programmed cell death ligand 1 (PD-L1) expression and analysed phospholipase A2 group IID (PLA2G2D) expression in NSCLC. BODIPY 493/503 dye staining and kits detected lipids, triglycerides, and phospholipids in H1299 cells, respectively. Extracellular vesicles (EVs) were extracted for morphology and size assessment using electron microscopy. Western blot assayed CD9, CD63, HSP90, EVs-PD-L1, PD-L1, and PLA2G2D expression. CCK-8, LDH, and ELISA tested proliferation and toxicity of CD8+ T cells, interleukin-2, and interferon-gamma secretion, respectively. PLA2G2D, PD-L1, and Ki67 expression was detected by immunohistochemistry. Immunofluorescence assayed PLA2G2D localisation and CD8+ T cell content. Flow cytometry assessed PD-L1 and CD8 expression. In NSCLC, upregulated EVs-PD-L1 and clinical characteristics showed a strong correlation. H1299 cells with overexpression PD-L1 significantly reduced proliferation, toxicity of CD8+ T cells, and interleukin-2 and interferon-gamma levels. Bioinformatics revealed positive correlations between PLA2G2D and overexpressed PD-L1. PLA2G2D was expressed in macrophages and dendritic cells in NSCLC tissue. Overexpression PLA2G2D (oe-PLA2G2D) increased lipids, triglycerides, and phospholipids contents in H1299 cells. oe-PLA2G2D significantly reduced proliferation, toxicity of CD8+ T cells, and interleukin-2 and interferon-gamma levels. si-PD-L1 restored inhibition of oe-PLA2G2D on CD8+ T cells. oe-PLA2G2D significantly increased mice tumour volume and weight, upregulated expression of blood EVs-PD-L1 and tissue PD-L1, PLA2G2D, Ki67, and decreased CD8+ T cell content. PLA2G2D facilitated immune escape in NSCLC by regulating CD8+ T cell immune function by upregulating EVs-PD-L1.
Collapse
Affiliation(s)
- Hui Jing
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
- Department of Respiratory and Critical Care Medicine, The Xuzhou School of Clinical Medicine of Nanjing Medical University, Xuzhou, China
| | - Xubo Cao
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Ke Li
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Yuanyuan Liu
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Meng Meng
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Shuan Liu
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Mengjie Ye
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Jinghao Zhang
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| | - Yanmin Wu
- Department of Respiratory and Critical Care Medicine, Xuzhou Central Hospital, Xuzhou Hospital Affiliated to Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
2
|
Sharma A, Alexander G, Chu JH, Markopoulos A, Maloul G, Ayub MT, Fidler MJ, Okwuosa TM. Immune Checkpoint Inhibitors and Cardiotoxicity: A Comparative Meta-Analysis of Observational Studies and Randomized Controlled Trials. J Am Heart Assoc 2024; 13:e032620. [PMID: 38761070 PMCID: PMC11179795 DOI: 10.1161/jaha.123.032620] [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: 09/12/2023] [Accepted: 02/20/2024] [Indexed: 05/20/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have uncommon associations with cardiotoxicity, yet these cardiotoxic effects are associated with high mortality. An accurate assessment of risk for cardiotoxicity is essential for clinical decision-making, but data from randomized controlled trials often differ from real-world observational studies. METHODS AND RESULTS A systematic search of PubMed, Embase, Cochrane Library, and Scopus was performed, including phase II and III randomized controlled trials (RCTs) and observational studies (OSs) reporting myocarditis or pericardial disease, myocardial infarction, or stroke with an immunotherapy. Odds ratios (ORs) were used to pool results between ICIs and other cancer therapy in RCTs and OSs. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses guideline was followed. In total, 54 RCTs (N=38 264) and 24 OSs (N=12 561 455) were included. In RCTs, ICI use resulted in higher risk of myocarditis (OR, 3.55 [95% CI, 2.10-5.98]), pericardial disease (OR, 2.73 [95% CI, 1.57-4.77]), and myocardial infarction (OR, 1.83 [95% CI, 1.03-3.25]), compared with non-ICI (placebo or chemotherapy). In OSs, ICI use was not associated with myocarditis, pericardial disease, or myocardial infarction compared with controls; however, combination ICIs demonstrated higher risk of myocarditis compared with single ICI use (OR, 3.07 [95% CI, 1.28-7.39]). Stroke risk was not increased with use of ICIs in RCTs. CONCLUSIONS We demonstrated increased risk of ICI myocarditis, pericardial disease, and myocardial infarction in RCTs but not OSs. Results of this study suggest there are differences between ICI cardiotoxicity risk, possibly suggesting differences in diagnoses and management, in clinical trials versus the OSs.
Collapse
Affiliation(s)
- Akash Sharma
- Department of Medicine University at Buffalo-Catholic Health System Buffalo NY
- Center for Global Health Research Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University Chennai India
| | - Grace Alexander
- Department of Internal Medicine University of Iowa Hospitals & Clinics Iowa City IA
| | - Jian H Chu
- Division of Cardiology, Department of Medicine University of Oklahoma Oklahoma City OK
| | | | | | - Muhammad Talha Ayub
- Heart and Vascular Institute, University of Pittsburgh Medical Center Pittsburgh PA
| | - Mary J Fidler
- Division of Hematology/Oncology/Stem cell transplant Rush University Medical Center Chicago IL
| | - Tochukwu M Okwuosa
- Division of Cardiology, Department of Internal Medicine Rush University Medical Center Chicago IL
| |
Collapse
|
3
|
Mudra SE, Rayes DL, Agrawal A, Kumar AK, Li JZ, Njus M, McGowan K, Kalam KA, Charalampous C, Schleicher M, Majid M, Syed A, Yesilyaprak A, Klein AL. Immune checkpoint inhibitors and pericardial disease: a systematic review. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:29. [PMID: 38760863 PMCID: PMC11100143 DOI: 10.1186/s40959-024-00234-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/03/2024] [Indexed: 05/19/2024]
Abstract
INTRODUCTION Despite the growing use of immune checkpoint inhibitors (ICI) in cancer treatment, data regarding ICI-associated pericardial disease are primarily derived from case reports and case series. ICI related pericardial disease can be difficult to diagnose and is associated with significant morbidity. We conducted a systematic review to further characterize the epidemiology, clinical presentation, and outcomes of this patient population. METHODS A search of four databases resulted in 31 studies meeting inclusion criteria. Patients > 18 years old who presented with ICI mediated pericardial disease were included. Intervention was medical + surgical therapy and outcomes were development of cardiac tamponade, morbidity, and mortality. RESULTS Thirty- eight patients across 31 cases were included. Patients were majority male (72%) with a median age of 63. Common symptoms included dyspnea (59%) and chest pain (32%), with 41% presenting with cardiac tamponade. Lung cancer (81%) was the most prevalent, and nivolumab (61%) and pembrolizumab (34%) were the most used ICIs. Pericardiocentesis was performed in 68% of patients, and 92% experienced symptom improvement upon ICI cessation. Overall mortality was 16%. DISCUSSION This study provides the most comprehensive analysis of ICI-mediated pericardial disease to date. Patients affected were most commonly male with lung cancer treated with either Nivolumab or Pembrolizumab. Diagnosis may be challenging in the setting of occult presentation with normal EKG and physical exam as well as delayed onset from therapy initiation. ICI-associated pericardial disease demonstrates high morbidity and mortality, as evidenced by a majority of patients requiring pericardiocentesis.
Collapse
Affiliation(s)
- Sarah E Mudra
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
| | - Danny L Rayes
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
| | - Ankit Agrawal
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Ave., Desk J1-5, Cleveland, OH, 44195, USA
| | - Ashwin K Kumar
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Ave., Desk J1-5, Cleveland, OH, 44195, USA
| | - Jason Z Li
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
| | - Meredith Njus
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
| | - Kevin McGowan
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
| | - Kazi A Kalam
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
| | - Charalompos Charalampous
- Department of Internal Medicine, MedStar Georgetown University Hospital, MedStar Health, Washington, DC, USA
| | - Mary Schleicher
- Floyd D. Loop Memorial Library, Cleveland Clinic, Cleveland, OH, USA
| | - Muhammad Majid
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Ave., Desk J1-5, Cleveland, OH, 44195, USA
| | - Alvena Syed
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Ave., Desk J1-5, Cleveland, OH, 44195, USA
| | - Abdullah Yesilyaprak
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Ave., Desk J1-5, Cleveland, OH, 44195, USA
| | - Allan L Klein
- Center for the Diagnosis and Treatment of Pericardial Diseases, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, 9500 Euclid Ave., Desk J1-5, Cleveland, OH, 44195, USA.
| |
Collapse
|
4
|
Gosangi B, Wang Y, Rubinowitz AN, Kwan J, Traube L, Gange C, Bader AS. Cardiothoracic complications of immune checkpoint inhibitors. Clin Imaging 2023; 102:98-108. [PMID: 37659356 DOI: 10.1016/j.clinimag.2023.08.001] [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: 02/19/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 09/04/2023]
Abstract
A paradigm shift in cancer treatment occurred with the advent of immune checkpoint inhibitors (ICI). ICI therapy has improved tumor response and increased overall survival in patients with solid tumors and hematologic malignancies. While ICI therapy has improved overall patient outcomes in oncology, it has also introduced novel adverse effects called immune-related adverse effects (irAEs). Studies have shown that the development of irAEs is associated with improved overall survival, but certain irAEs like pneumonitis and myocarditis are life threatening, and could result in death if not identified and treated early. Therefore, it is important for radiologists to be aware of complications arising from ICI administration, especially those related to the heart and lungs as they are associated with greater mortality. This paper will review the imaging features of cardiothoracic toxicities, recurrent and chronic irAEs, and atypical tumor responses associated with irAEs.
Collapse
Affiliation(s)
- Babina Gosangi
- Yale School of Medicine, New Haven, CT 06510, United States of America.
| | - Yifan Wang
- Yale School of Medicine, New Haven, CT 06510, United States of America
| | - Ami N Rubinowitz
- Yale School of Medicine, New Haven, CT 06510, United States of America
| | - Jennifer Kwan
- Yale School of Medicine, New Haven, CT 06510, United States of America
| | - Leah Traube
- Yale School of Medicine, New Haven, CT 06510, United States of America
| | - Christopher Gange
- Yale School of Medicine, New Haven, CT 06510, United States of America
| | - Anna S Bader
- Yale School of Medicine, New Haven, CT 06510, United States of America
| |
Collapse
|
5
|
Suarez ZK, Finke AC, Hospedales E, Perez E, Sharifzadeh A, Foster J, Ferris A. An unusual case of checkpoint-inhibitor-induced pleuropericarditis. J Oncol Pharm Pract 2023; 29:1525-1528. [PMID: 37254508 DOI: 10.1177/10781552231179369] [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] [Indexed: 06/01/2023]
Abstract
INTRODUCTION Pembrolizumab is an immune checkpoint inhibitor that promotes effector T-cell functions on malignant cells by binding to programmed cell death protein 1 (PD-1). Pembrolizumab is well tolerated in most cases with an adverse event profile consisting mainly of pruritus, fatigue, and anorexia. Cardiotoxicity comprises 1% of the total adverse events. CASE REPORT We present a case of a 64-year-old female with non-small cell lung cancer (NSCLC) who developed pleuropericarditis following pembrolizumab therapy. MANAGEMENT & OUTCOME The patient was successfully managed with colchicine, furosemide, and timely initiation of methylprednisolone with the improvement of her symptoms. The decision to discontinue pembrolizumab was made, and six months after this intervention, the patient has remained asymptomatic. DISCUSSION Clinicians should recognize these potential immune-mediated adverse effects to provide effective and timely management and optimize patient care.
Collapse
Affiliation(s)
- Zoilo K Suarez
- Internal Medicine Department, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, USA
| | - Ana C Finke
- Internal Medicine Department, Universidad Iberoamericana, Santo Domingo, Dominican Republic
| | - Emilio Hospedales
- Internal Medicine Department, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, USA
| | - Ernesto Perez
- Internal Medicine Department, Kendall Regional Medical Center, Kendall, FL, USA
| | - Arya Sharifzadeh
- Internal Medicine Department, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, USA
| | - Jennifer Foster
- Internal Medicine Department, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, USA
| | - Allison Ferris
- Internal Medicine Department, Florida Atlantic University Charles E. Schmidt College of Medicine, Boca Raton, FL, USA
| |
Collapse
|
6
|
Hussain A, Prevatt O, Piercy J, Ahmed N, Marlowe S, Damaa G. A Rare Case of Non-Small-Cell Lung Carcinoma Complicated by Massive Pericardial Effusion and Cardiac Tamponade: A Case Report. Cureus 2023; 15:e44047. [PMID: 37746410 PMCID: PMC10517708 DOI: 10.7759/cureus.44047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
This case report presents a late middle-aged man with a right infra-hilar lung mass and pericardial effusion (PE). The patient was diagnosed with metastatic small-cell lung carcinoma, with metastases to the liver, pancreas, and cerebellum. The pericardial fluid cytology confirmed the presence of malignant cells most compatible with non-small-cell carcinoma. The patient received carbo/etoposide chemotherapy, and his treatment plan included adding atezolizumab and radiation therapy. Despite the excellent efficacy of immunotherapy, immune-related adverse events (IRAEs), including cardiac toxicity, were noted in some patients. PE related to immune checkpoint inhibitor (ICI) use is rare but potentially severe. This case highlights the importance of vigilant monitoring for cardiovascular symptoms during immunotherapy and the significance of pericardial fluid analysis in diagnosing malignant pericardial disease. Prompt diagnosis and appropriate treatment can lead to improved patient outcomes in cases of lung cancer-associated cardiac complications.
Collapse
Affiliation(s)
- Akbar Hussain
- Internal Medicine, Appalachian Regional Healthcare, Harlan, USA
| | - Opal Prevatt
- Internal Medicine, Appalachian Regional Healthcare, Harlan, USA
| | - Jonathan Piercy
- Internal Medicine, Appalachian Regional Healthcare, Harlan, USA
| | - Nazneen Ahmed
- Internal Medicine, Appalachian Regional Healthcare, Harlan, USA
| | - Stanley Marlowe
- Internal Medicine, Appalachian Regional Healthcare, Harlan, USA
| | - Georges Damaa
- Internal Medicine, Appalachian Regional Healthcare, Harlan, USA
| |
Collapse
|
7
|
Inno A, Tarantini L, Parrini I, Spallarossa P, Maurea N, Bisceglia I, Silvestris N, Russo A, Gori S. Cardiovascular Effects of Immune Checkpoint Inhibitors: More Than Just Myocarditis. Curr Oncol Rep 2023; 25:743-751. [PMID: 37017825 DOI: 10.1007/s11912-023-01411-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2023] [Indexed: 04/06/2023]
Abstract
PURPOSE OF REVIEW Immune checkpoint inhibitors have reshaped the treatment of cancer, but they are characterized by peculiar toxicity consisting of immune-related adverse events that may potentially affect any organ or system. In this review, we summarize data on clinical presentation, diagnosis, pathogenesis, and management of the main immune-related cardiovascular toxicities of immune checkpoint inhibitors. RECENT FINDINGS The most relevant immune-related cardiovascular toxicity is myocarditis, but other non-negligible reported events include non-inflammatory heart failure, conduction abnormalities, pericardial disease, and vasculitis. More recently, growing evidence suggests a role for immune checkpoint inhibitors in accelerating atherosclerosis and promoting plaque inflammation, thus leading to myocardial infarction. Immune checkpoint inhibitors are associated with several forms of cardiovascular toxicity; thus, an accurate cardiovascular baseline evaluation and periodical monitoring are required. Furthermore, the optimization of cardiovascular risk factors before, during, and after treatment may contribute to mitigating both short-term and long-term cardiovascular toxicity of these drugs.
Collapse
Affiliation(s)
- Alessandro Inno
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, Via Don A Sempreboni 5, 37024, Negrar Di Valpolicella, VR, Italy.
| | - Luigi Tarantini
- Cardiologia Ospedaliera, AUSL - IRCCS in Tecnologie Avanzate E Modelli Assistenziali in Oncologia, Reggio Emilia, Italy
| | - Iris Parrini
- Dipartimento Di Cardiologia, Ospedale Mauriziano, Turin, Italy
| | - Paolo Spallarossa
- Clinica Di Malattie Dell'Apparato Cardiovascolare, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Nicola Maurea
- Struttura Complessa Cardiologia, Istituto Nazionale Tumori Di Napoli IRCCS Fondazione G. Pascale, Naples, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, Rome, Italy
| | - Nicola Silvestris
- Oncologia Medica, Dipartimento Di Patologia Umana "G. Barresi", Università Di Messina, Messina, Italy
| | - Antonio Russo
- Dipartimento Di Discipline Chirurgiche, Oncologiche E Stomatologiche, Università Di Palermo, Palermo, Italy
| | - Stefania Gori
- Oncologia Medica, IRCCS Ospedale Sacro Cuore Don Calabria, Via Don A Sempreboni 5, 37024, Negrar Di Valpolicella, VR, Italy
| |
Collapse
|
8
|
Uczkowski D, Ashraf H, Sekhri A, Samad A. Pembrolizumab induced pericardial tamponade: A case report. Clin Case Rep 2023; 11:e7298. [PMID: 37143451 PMCID: PMC10152068 DOI: 10.1002/ccr3.7298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/06/2023] [Accepted: 04/19/2023] [Indexed: 05/06/2023] Open
Abstract
Key Clinical Message The occurrence of a large pericardial effusion is not a commonly noted adverse event associated with pembrolizumab and our report demonstrates that a rapid development can be diagnosed with close monitoring and triage to acute medical settings. Abstract Pembrolizumab is an immune checkpoint inhibitor used in various types of cancers. Pericardial tamponade is a rare side effect reported in only very few case reports. Early recognition and therapeutic intervention is vital in all cases. We report a case of a 54-year-old male with Stage 3 lung adenocarcinoma who developed cardiac tamponade secondary to pembrolizumab and subsequently required pericardial window.
Collapse
Affiliation(s)
- Dariusz Uczkowski
- Overlook Medical Center Atlantic Health SystemSummitNew JerseyUSA
- Morristown Medical Center Atlantic Health SystemMorristownNew JerseyUSA
| | - Hamza Ashraf
- Overlook Medical Center Atlantic Health SystemSummitNew JerseyUSA
- Morristown Medical Center Atlantic Health SystemMorristownNew JerseyUSA
| | - Arunabh Sekhri
- Overlook Medical Center Atlantic Health SystemSummitNew JerseyUSA
- Morristown Medical Center Atlantic Health SystemMorristownNew JerseyUSA
| | - Arbaz Samad
- Overlook Medical Center Atlantic Health SystemSummitNew JerseyUSA
- Morristown Medical Center Atlantic Health SystemMorristownNew JerseyUSA
| |
Collapse
|
9
|
Ndjana Lessomo FY, Wang Z, Mukuka C. Comparative cardiotoxicity risk of pembrolizumab versus nivolumab in cancer patients undergoing immune checkpoint inhibitor therapy: A meta-analysis. Front Oncol 2023; 13:1080998. [PMID: 37064101 PMCID: PMC10090546 DOI: 10.3389/fonc.2023.1080998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
ObjectiveRecently, several researchers have reported the incidence of cardiac-related toxicities occurring with nivolumab (Opdivo) and pembrolizumab (Keytruda). There is still a need for balance between oncology treatment efficacy and reduction of cardiotoxicity burden in immune checkpoint inhibitor (ICI)-treated patients. Thus, the primary aim was to determine whether pembrolizumab or nivolumab would present with a greater risk for cardiotoxicity reports.Materials and methodsThis meta-analysis was performed with respect to the MOOSE reporting guidelines. Studies were retrieved by searching PubMed, Embase, and Google Scholar; the search terms were Keytruda or Pembrolizumab, PD1 inhibitors, anti-PD1 drugs, Nivolumab or Opdivo, and cardiotoxicities or cardiac toxicity. The study was restricted to original articles investigating ICI-induced cardiac immune-related adverse events (irAEs). The targeted population was cancer patients treated with either pembrolizumab or nivolumab monotherapy, of which those with records of any cardiac events following the therapy were labeled as events. The measures used to achieve the comparison were descriptive proportions, probabilities, and meta-analysis pooled odds ratios (ORs).ResultsFifteen studies were included in this meta-analysis. Nivolumab accounted for 55.7% cardiotoxicity and pembrolizumab, for 27.31% (P = 0.027). The meta-analysis was based on the Mantel–Haenszel method, and the random-effect model yielded a pooled OR = 0.73 (95% CI [0.43–1.23] P = 0.24), with considerable heterogeneity (I2 = 99% P = 0). Hence, the difference in cardiotoxicity odds risk between pembrolizumab and nivolumab was not statistically significant. On subgroup analysis based on cardiotoxicity type, the “myocarditis” subgroup in which there was no statistical heterogeneity was associated with a significant cardiotoxicity risk increase with pembrolizumab (OR = 1.30 [1.07;1.59], P< 0.05; I2 = 0%, Ph = 0.4).ConclusionTo our knowledge, this is the first meta-analysis to compare the cardiotoxicity potentials of nivolumab and pembrolizumab. In contrast to previous reports, the overall findings here demonstrated that nivolumab-induced cardiotoxicity was more commonly reported in the literature than pembrolizumab; however, myocarditis seemed more likely to occur with pembrolizumab therapy.
Collapse
Affiliation(s)
| | - Zhiquan Wang
- Cardiovascular Internal Medicine Department, Zhongnan Hospital of Wuhan University, Wuhan, China
- *Correspondence: Zhiquan Wang, ,
| | - Chishimba Mukuka
- Internal Medicine Department, MANSA General Hospital, Mansa, Luapula, Zambia
| |
Collapse
|
10
|
Kanbayashi Y, Shimizu T, Anzai M, Kawai R, Uchida M. Evaluation of Cardiac Adverse Events with Nivolumab Using a Japanese Real-World Database. Clin Drug Investig 2023; 43:177-184. [PMID: 36780109 DOI: 10.1007/s40261-023-01246-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 02/14/2023]
Abstract
BACKGROUND Nivolumab has been used for the treatment of various types of cancers and has achieved improvements in overall survival. However, nivolumab can cause a variety of adverse events (AEs). Among these, cardiac-specific AEs have received little attention in clinical trials, despite their life-threatening potential. OBJECTIVE The present study aimed to determine the risk of nivolumab-induced cardiac AEs, time to onset, incidence rates, and post hoc outcomes using the Japanese Adverse Drug Event Report database. METHODS We analyzed data for the period between April 2004 and March 2021. Data on cardiac AEs were extracted and relative risk of AEs was estimated using the reporting odds ratio (ROR). RESULTS We analyzed 1,772,494 reports and identified 18,721 reports of AEs caused by nivolumab. Of these, 409 reports involved cardiac AEs. Signals were detected for four cardiac AEs: myocarditis; pericardial effusion; pericarditis; and immune-mediated myocarditis. Among these, myocarditis was the most frequently reported (35.0%) and included fatal cases. A histogram of times to onset showed nivolumab-associated AEs occurring 41-127 days after starting administration, with outlier cases of myocarditis or pericardial effusion occurring after more than one year, both with catastrophic consequences. CONCLUSION This study focused on cardiac AEs caused by nivolumab as post-marketing AEs. Myocarditis and pericardial effusion have been associated with some fatal cases after administration of nivolumab. Patients should be monitored for signs of onset for these AEs, not only at the start of administration, but also over an extended period after nivolumab administration.
Collapse
Affiliation(s)
- Yuko Kanbayashi
- Department of Education and Research Center for Clinical Pharmacy, Faculty of Pharmacy, Osaka Medical and Pharmaceutical University, 4-20-1 Nasahara, Takatsuki, Osaka, 569-1094, Japan.
| | - Tadashi Shimizu
- School of Pharmacy, Hyogo Medical University, 1-3-6 Minatojima, Kobe, Hyogo, 650-8530, Japan
| | - Miku Anzai
- Department of Education and Research Center for Pharmacy Practice, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, 97-1 Kodominamihokotate, Kyotanabe, Kyoto, 610-0395, Japan
| | - Rika Kawai
- Department of Education and Research Center for Pharmacy Practice, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, 97-1 Kodominamihokotate, Kyotanabe, Kyoto, 610-0395, Japan
| | - Mayako Uchida
- Department of Education and Research Center for Pharmacy Practice, Faculty of Pharmaceutical Sciences, Doshisha Women's College of Liberal Arts, 97-1 Kodominamihokotate, Kyotanabe, Kyoto, 610-0395, Japan
| |
Collapse
|
11
|
Jin C, Qi J, Wang Q, Pu C, Tan M. Cardiotoxicity of lung cancer-related immunotherapy versus chemotherapy: a systematic review and network meta-analysis of randomized controlled trials. Front Oncol 2023; 13:1158690. [PMID: 37124488 PMCID: PMC10141653 DOI: 10.3389/fonc.2023.1158690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 03/31/2023] [Indexed: 05/02/2023] Open
Abstract
Background Previous clinical randomized controlled trials (RCTs) have demonstrated that immune checkpoint inhibitors (ICIs) cause various toxicities during cancer treatment, but the effects of different inhibitors in combination with chemotherapy for cardiotoxicity remain controversial. The aim of the present study was to assess cardiotoxicity caused by programmed cell death protein 1 (PD-1), programmed cell death-Ligand 1 (PD-L1), and cytotoxic T lymphocyte associate protein-4 (CTLA-4) in combination with chemotherapy to treat lung cancer. Methods The following ICIs were included in the present study: durvalumab, avelumab, ipilimumab, atezolizumab, pembrolizumab, cemiplimab, and nivolumab. The relevant information was extracted using a predefined data extraction table, and the risk of bias was assessed in randomized controlled trials using the Cochrane Bias Risk tool. The main outcomes were hypertension, heart failure, pericardial effusion, and other adverse cardiac events. The random effects model was used to conduct a paired meta-analysis, and a random effects network meta-analysis was then performed within a Bayesian framework. Results In total, 17 RCTs were included in the present study. There were 11,063 individuals in the experimental and control groups, with an average age greater than 60 years. Based on the evaluation of all drug classes in RCTs, CTLA-4+chemotherapy (RR, -0.69 [95% CI, 2.91-1.52] and PD-L1 (RR, -0.21 [95% CI, -1.03-0.60]) were less cardiotoxic than the control arm, which indicated they were safer options for adverse cardiac events. PD-L1 alone was less cardiotoxic than PD-1 alone (RR, -0.57 [95% CI, -1.96-0.82]). Further, the dual immunotarget inhibitor, PD-1+CTLA-4, had the lowest SUCRA value and had the highest cardiotoxicity (SUCRA=9). Conclusion When classified according to drug type, CTLA-4+chemotherapy is associated with fewer cardiac adverse events compared to other treatments. Dual immunotarget inhibitors are more likely to have adverse cardiac reactions. Therefore, clinicians should consider this evidence when developing an ICI immunotherapy regimen for lung cancer. Systematic review registration https://www.crd.york.ac.uk/prospero, identifier CRD42023360931.
Collapse
Affiliation(s)
- Chengwei Jin
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Jia Qi
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Qilei Wang
- Department of Cardiology, Zibo Central Hospital, Zibo, China
| | - Chenwei Pu
- Department of Respiratory and Critical Care Medicine, Zibo Central Hospital, Shandong, China
| | - Mingming Tan
- Department of Respiratory and Critical Care Medicine, Zibo Central Hospital, Shandong, China
- *Correspondence: Mingming Tan,
| |
Collapse
|
12
|
Li X, Peng W, Wu J, Yeung SCJ, Yang R. Advances in immune checkpoint inhibitors induced-cardiotoxicity. Front Immunol 2023; 14:1130438. [PMID: 36911712 PMCID: PMC9995967 DOI: 10.3389/fimmu.2023.1130438] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 02/13/2023] [Indexed: 02/25/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) are approved as the first-line drug for treating many cancers and has shown significant survival benefits; however, it also causes immune-related adverse events (irAEs) while activating the immune system, involving multiple organs. Among them, cardiovascular immune-related adverse events (CV-irAE) are rare, but common causes of death in ICIs treated cancer patients, which manifest as myocardial, pericardial, vascular and other cardiovascular toxicities. Therefore, it is important that irAEs, especially CV-irAE should be carefully recognized and monitored during the whole ICIs treatment because early detection and treatment of CV-irAE can significantly reduce the mortality of such patients. Consequently, it is urgent to fully understand the mechanism and management strategies of CV-irAE. The effects of ICIs are multifaceted and the exact mechanism of CV-irAE is still elusive. Generally, T cells identify tumor cell antigens as well as antigen in cardiomyocytes that are the same as or homologous to those on tumor cells, thus causing myocardial damage. In addition, ICIs promote formation of cardiac troponin I (cTnI) that induces cardiac dysfunction and myocardial dilatation; moreover, ICIs also increase the production of cytokines, which promote infiltration of inflammation-linked molecules into off-target tissues. Currently, the management and treatment of cardiovascular toxicity are largely dependent on glucocorticoids, more strategies for prevention and treatment of CV-irAE, such as predictive markers are being explored. This review discusses risk factors, potential pathophysiological mechanisms, clinical manifestations, and management and treatment of CV-irAE, guiding the development of more effective prevention, treatment and management strategies in the future.
Collapse
Affiliation(s)
- Xiang Li
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Wenying Peng
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Jiao Wu
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Sai-Ching Jim Yeung
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, TX, United States
| | - Runxiang Yang
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| |
Collapse
|
13
|
Malaty MM, Amarasekera AT, Li C, Scherrer-Crosbie M, Tan TC. Incidence of immune checkpoint inhibitor mediated cardiovascular toxicity: A systematic review and meta-analysis. Eur J Clin Invest 2022; 52:e13831. [PMID: 35788986 DOI: 10.1111/eci.13831] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 06/24/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICI) are a novel class of anti-cancer therapy becoming increasingly associated with fatal cardiovascular toxicities (CVTs). The aim is to determine the incidence of CVTs in cohorts treated with ICIs as sole anti-cancer therapy. METHODS A systematic literature search of scientific and medical databases was performed using PRISMA principles to identify relevant cohorts (PROSPERO registration CRD42021272470). Data for specific CVTs (pericardial disease, myocarditis, heart failure, arrhythmia, myocardial infarction/ischaemia and angina), CVT-related death and CV risk factors were extracted. Presence of CVTs in ICI-monotherapy versus combination-ICI therapy, and programmed death 1/programmed death ligand 1- (PD1/PDL1-) versus cytotoxic T-lymphocyte-associated protein 4- (CTLA4-) inhibitor groups were dichotomised and meta-analysed using random-effect models. RESULTS Forty-eight studies (11,207 patients) were identified, from which 146 CVTs were observed (incidence 1.30%). ICI-monotherapy led to more CVTs than combination therapy (119/9009; 1.32% vs. 18/2086; 0.86%). Across monotherapies, PD1/PDL1-inhibitors had lower incidence of CVTs compared to CTLA4-inhibitors (62/6950; 0.89% vs. 57/2059; 2.77%). Based on eight studies that were meta-analysed, no significant difference was observed comparing monotherapy versus combination-ICI therapy (RR-0.69, 95% CI -1.47 to 0.09) for all CVTs, or PD1/PDL1- to CTLA4-inhibitors (RR-0.27, 95% CI -2.06 to 1.53), for all CVTs including CVT-death. CV risk factors could not be attributed to an ICI group as data was population based rather than individual based. CONCLUSION ICI-mediated CVTs are rare and potentially fatal. The role of CV risk factors in their development remains unclear.
Collapse
Affiliation(s)
- Michael M Malaty
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia
| | - Anjalee Thanuja Amarasekera
- School of Medicine, Western Sydney University, Sydney, Australia.,Department of Cardiology, Westmead Hospital, Western Sydney Local Health District, Sydney, Australia.,Westmead Applied Research Centre (WARC), Faculty of Medicine and Health, University of Sydney, Sydney, Australia
| | - Cindy Li
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Timothy C Tan
- Department of Cardiology, Blacktown Hospital, Western Sydney Local Health District, Sydney, Australia.,School of Medicine, Western Sydney University, Sydney, Australia.,Department of Cardiology, Westmead Hospital, Western Sydney Local Health District, Sydney, Australia
| |
Collapse
|
14
|
Chen R, Zhou M, Zhu F. Immune Checkpoint Inhibitors Related to Cardiotoxicity. J Cardiovasc Dev Dis 2022; 9:jcdd9110378. [PMID: 36354777 PMCID: PMC9697232 DOI: 10.3390/jcdd9110378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/17/2022] [Accepted: 10/26/2022] [Indexed: 11/06/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have now emerged as a mainstay of treatment for various cancers. Along with development of ICIs, immune-related adverse effects (irAEs) have aroused wide attention. The cardiac irAE, one of the rare but potentially fatal effects, have been reported recently. However, the clinical comprehension of cardiac irAEs remains limited and guidelines are inadequate for cardio-oncologists to tackle the problem. In this review, we have summarized current classifications of, manifestations of, potential mechanisms of, and treatment for ICI-related myocardial injury in order to provide some clues for the understanding of cardiac irAEs in clinical work.
Collapse
Affiliation(s)
- Ru Chen
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Zhou
- Department of Pulmonary and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Key Laboratory of Respiratory Diseases, National Ministry of Health of the People’s Republic of China and National Clinical Research Center for Respiratory Disease, Wuhan 430022, China
- Correspondence: (M.Z.); (F.Z.)
| | - Feng Zhu
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Correspondence: (M.Z.); (F.Z.)
| |
Collapse
|
15
|
Jin YJ, Jin YF, Zhu XY, Zhang BL, Chen C. Intermediate risk pulmonary embolism concomitant with or without lung cancer: a wide spectrum of features. Clin Exp Hypertens 2022; 44:589-594. [PMID: 35766216 DOI: 10.1080/10641963.2022.2093892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
OBJECTIVES We aimed to investigate the differences in clinical features between pulmonary embolism (PE) patients concomitant with lung cancer and without lung cancer (LC) and gain further understanding of the impact of lung cancer on pulmonary embolism. METHODS This retrospective study sampled 114 patients diagnosed with pulmonary embolism from January 2017 to April 2021 in the First Affiliated Hospital of Soochow University. The patients were categorized into the LC group (n = 22) or non-LC group (n = 92). Myocardial injury, coagulation and blood cell parameters, along with imaging findings, were analyzed for the two groups. The primary outcome measure was the 90-day mortality. RESULTS Of the 114 patients with pulmonary embolism in the present study, the 90 intermediate-risk patients were enrolled for further investigations. Compared to the non-LC group, patients in the LC group had milder myocardial injury, more severe coagulation function disorder, a higher incidence of central PE and a smaller change in diameter of the main pulmonary artery. We found that the occurrence of pericardial effusion created the risk of lung cancer in patients with pulmonary embolism, but there was no increase in the 90-day mortality for non-LC group versus LC group. CONCLUSION Intermediate risk PE patients concomitant with lung cancer seem to be more likely to present specific clinical features, accordingly, clinicians must pay great attention to PE patients concomitant with lung cancer and implement effective treatments to simultaneously manage the two conditions.
Collapse
Affiliation(s)
- Yu-Jia Jin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, JS, China
| | - Yi-Fan Jin
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, JS, China
| | - Xin-Yun Zhu
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, JS, China
| | - Bei-Lei Zhang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, JS, China
| | - Cheng Chen
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Soochow University, Suzhou, JS, China
| |
Collapse
|
16
|
Li H, Han D, Zhang L, Feng X, Li H, Yang F, Song L, Li X. PD-1/L1 inhibitors may increase the risk of pericardial disease in non-small-cell lung cancer patients: a meta-analysis and systematic review. Immunotherapy 2022; 14:577-592. [PMID: 35373580 DOI: 10.2217/imt-2021-0223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: The advent of PD-1/L1 inhibitors has changed the landscape for patients with non-small-cell lung cancer (NSCLC). Meanwhile, the adverse events of PD-1/L1 inhibitors have been focused. Methods: The Cochrane Central Register of Controlled Trials, PubMed and Embase databases and ClinicalTrials.gov were searched from inception to February 2021. Results: 18 studies involving 11,394 patients with NSCLC were included. PD-1/L1 inhibitor monotherapy was associated (relative risk, 95% confidence interval) with an increased risk of pericardial effusion (2.72 [1.45-5.12]; p = 0.002) and cardiac tamponade (2.76 [1.15-6.62]; p = 0.023), whereas PD-1/L1 inhibitors combined with chemotherapy did not increase the risk of pericardial effusion and cardiac tamponade (3.08 [0.93-10.21]; p = 0.066 and 3.27 [0.37-28.94]; p = 0.288, respectively). Conclusion: For patients with NSCLC, treatment with PD-1/L1 inhibitor monotherapy increases the risk of pericardial effusion and cardiac tamponade, but PD-1/L1 inhibitors combined with chemotherapy do not.
Collapse
Affiliation(s)
- Honglin Li
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Deting Han
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Lei Zhang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiaoteng Feng
- Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Huijie Li
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Feiran Yang
- First Clinical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lucheng Song
- Department of Gerontology, The First Affiliated Hospital of Shandong First Medical University, Jinan, Shandong, China
| | - Xiurong Li
- Department of Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| |
Collapse
|
17
|
Ronen D, Bsoul A, Lotem M, Abedat S, Yarkoni M, Amir O, Asleh R. Exploring the Mechanisms Underlying the Cardiotoxic Effects of Immune Checkpoint Inhibitor Therapies. Vaccines (Basel) 2022; 10:vaccines10040540. [PMID: 35455289 PMCID: PMC9031363 DOI: 10.3390/vaccines10040540] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 03/15/2022] [Accepted: 03/24/2022] [Indexed: 02/01/2023] Open
Abstract
Adaptive immune response modulation has taken a central position in cancer therapy in recent decades. Treatment with immune checkpoint inhibitors (ICIs) is now indicated in many cancer types with exceptional results. The two major inhibitory pathways involved are cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and programmed cell death protein 1 (PD-1). Unfortunately, immune activation is not tumor-specific, and as a result, most patients will experience some form of adverse reaction. Most immune-related adverse events (IRAEs) involve the skin and gastrointestinal (GI) tract; however, any organ can be involved. Cardiotoxicity ranges from arrhythmias to life-threatening myocarditis with very high mortality rates. To date, most treatments of ICI cardiotoxicity include immune suppression, which is also not cardiac-specific and may result in hampering of tumor clearance. Understanding the mechanisms behind immune activation in the heart is crucial for the development of specific treatments. Histological data and other models have shown mainly CD4 and CD8 infiltration during ICI-induced cardiotoxicity. Inhibition of CTLA4 seems to result in the proliferation of more diverse T0cell populations, some of which with autoantigen recognition. Inhibition of PD-1 interaction with PD ligand 1/2 (PD-L1/PD-L2) results in release from inhibition of exhausted self-recognizing T cells. However, CTLA4, PD-1, and their ligands are expressed on a wide range of cells, indicating a much more intricate mechanism. This is further complicated by the identification of multiple co-stimulatory and co-inhibitory signals, as well as the association of myocarditis with antibody-driven myasthenia gravis and myositis IRAEs. In this review, we focus on the recent advances in unraveling the complexity of the mechanisms driving ICI cardiotoxicity and discuss novel therapeutic strategies for directly targeting specific underlying mechanisms to reduce IRAEs and improve outcomes.
Collapse
Affiliation(s)
- Daniel Ronen
- Department of Internal Medicine D, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Aseel Bsoul
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
| | - Michal Lotem
- Department of Oncology, Hadassah Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Suzan Abedat
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
| | - Merav Yarkoni
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Offer Amir
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
| | - Rabea Asleh
- Cardiovascular Research Center, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel; (A.B.); (S.A.); (O.A.)
- Department of Cardiology, Heart Institute, Hadassah University Medical Center, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem 9112001, Israel;
- Correspondence: ; Tel.: +972-2-6776564; Fax: +972-2-6411028
| |
Collapse
|
18
|
Braden J, Lee JH. Immune Checkpoint Inhibitor Induced Pericarditis and Encephalitis in a Patient Treated With Ipilimumab and Nivolumab for Metastatic Melanoma: A Case Report and Review of the Literature. Front Oncol 2021; 11:749834. [PMID: 34956874 PMCID: PMC8696256 DOI: 10.3389/fonc.2021.749834] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 11/16/2021] [Indexed: 12/19/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have dramatically improved outcomes in melanoma. Common ICI toxicities have become familiar to clinicians; however, rare delayed toxicities remain challenging given the paucity of data with such presentations. We present the unique case of a 61-year-old with metastatic melanoma with two rare, delayed ICI-induced toxicities. After resection of a large symptomatic parietal metastases, this patient received two doses of combination ipilimumab and nivolumab. Five weeks following his second dose, he developed ICI-induced pericarditis with associated pericardial effusion and early signs of tamponade. Corticosteroids were not administered due to a concurrent cerebral abscess. Administration of colchicine, ibuprofen, judicious monitoring, and cessation of immunotherapy led to the complete resolution of the effusion over several weeks. Seven months following his last dose of immunotherapy, the patient developed ICI-associated grade four autoimmune encephalitis, presenting as status epilepticus. High-dose steroid initiation led to rapid clinical improvement. The patient remains in near-complete response on imaging with no recurrence of pericardial effusion and partial resolution of neurological symptoms. ICI-induced pericardial disease and encephalitis carry substantial mortality rates and prompt diagnosis and management is critical. Clinicians must therefore remain vigilant for these rare toxicities regardless of duration of drug exposure or time since cessation of therapy.
Collapse
Affiliation(s)
- Jorja Braden
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, Australia
| | - Jenny H Lee
- Department of Medical Oncology, Chris O'Brien Lifehouse, Sydney, NSW, Australia.,Department of Biomedical Sciences, Macquarie University, Sydney, NSW, Australia
| |
Collapse
|
19
|
Sun JY, Qu Q, Lou YX, Hua Y, Sun GZ, Sun W, Kong XQ. Cardiotoxicity in cancer immune-checkpoint therapy: Mechanisms, clinical evidence, and management strategies. Int J Cardiol 2021; 344:170-178. [PMID: 34563597 DOI: 10.1016/j.ijcard.2021.09.041] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/30/2021] [Revised: 08/30/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022]
Abstract
Immune-checkpoint inhibitors (ICIs), a unique antibody-based therapeutic strategy, have revolutionized the treatment landscape of solid and hematological cancers. Despite the proven benefits of ICIs, the cardiotoxicity from unspecific immune activation (uncommon but potentially fatal) is a continuing concern. Accumulating preclinical research has demonstrated that ICIs initiate inflammation in the myocardium, while clinically significant cardiotoxicity were reported in few patients receiving ICI therapy, probably due to the low incidence and unspecific symptoms. The subtle signs and symptoms (e.g., chest pain, dizziness, and dyspnea) were likely attributed to cancer and/or non-cardiac events by previous studies, thus limiting the understanding of the incidence, outcomes, risk factors, and management of ICI-related cardiotoxicity. The heterogeneous clinical presentation and complex diagnostic procedure further make it challenging to accurately identify ICI-related cardiac events in clinical trials. Therefore, ICI-related cardiotoxicity, whose incidence is probably underestimated, has not been well recognized. In this article, we provide an overview of potential mechanisms underlying ICI-related cardiotoxicity and review accumulating clinical evidence of ICI-related cardiotoxicity, with a focus on myocarditis. Moreover, we discuss possible strategies to manage ICI-related cardiotoxicity and highlight the importance of developing cardio-oncology.
Collapse
Affiliation(s)
- Jin-Yu Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Qiang Qu
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Yu-Xuan Lou
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Yang Hua
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Guo-Zhen Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China
| | - Wei Sun
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China..
| | - Xiang-Qing Kong
- Department of Cardiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210000, China..
| |
Collapse
|
20
|
Benjamin L, Jean-Charles G, Laurence M, Adrien R, Terry L, Régis D. Malignant pericardial effusion complicated by cardiac tamponade under atezolizumab. SAGE Open Med Case Rep 2021; 9:2050313X211036005. [PMID: 34377486 PMCID: PMC8323422 DOI: 10.1177/2050313x211036005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Accepted: 07/12/2021] [Indexed: 11/16/2022] Open
Abstract
Immune-related adverse events including cardiac toxicity are increasingly described in patients receiving immune checkpoint inhibitors. We described a malignant pericardial effusion complicated by a cardiac tamponade in an advanced non-small cell lung cancer patient who had received five infusions of atezolizumab, a PDL-1 monoclonal antibody, in combination with cabozantinib. The definitive diagnosis was quickly made by cytology examination showing typical cell abnormalities and high fluorescence cell information provided by the hematology analyzer. The administration of atezolizumab and cabozantinib was temporarily discontinued due to cardiogenic hepatic failure following cardiac tamponade. After the re-initiation of the treatment, pericardial effusion relapsed. In this patient, the analysis of the pericardial fluid led to the final diagnosis of pericardial tumor progression. This was afterwards confirmed by the finding of proliferating intrapericardial tissue by computed tomography scan and ultrasound. This report emphasizes the value of cytology analysis performed in a hematology laboratory as an accurate and immediate tool for malignancy detection in pericardial effusions.
Collapse
Affiliation(s)
- Lardinois Benjamin
- Laboratory Department, CHU UCL Namur Site de
Sainte-Elisabeth, Université catholique de Louvain, Namur, Belgium
| | - Goeminne Jean-Charles
- Oncology Department, CHU UCL Namur Site de
Sainte-Elisabeth, Université catholique de Louvain, Namur, Belgium
| | - Miller Laurence
- Laboratory Department, CHU UCL Namur Site de
Sainte-Elisabeth, Université catholique de Louvain, Namur, Belgium
| | - Randazzo Adrien
- Laboratory Department, CHU UCL Namur Site de
Sainte-Elisabeth, Université catholique de Louvain, Namur, Belgium
| | - Laurent Terry
- Laboratory Department, CHU UCL Namur Site de
Sainte-Elisabeth, Université catholique de Louvain, Namur, Belgium
| | - Debois Régis
- Laboratory Department, CHU UCL Namur Site de
Sainte-Elisabeth, Université catholique de Louvain, Namur, Belgium
| |
Collapse
|
21
|
Jiao Y, Qian C, Fei S. Commentary: Overall Survival in Heart Disease-Related Death in Non-Small Cell Lung Cancer Patients: Nonimmunotherapy Versus Immunotherapy Era: Population-Based Study. Front Oncol 2021; 11:639042. [PMID: 33968737 PMCID: PMC8100200 DOI: 10.3389/fonc.2021.639042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 02/18/2021] [Indexed: 11/13/2022] Open
Affiliation(s)
- Yiping Jiao
- School of Automation, Southeast University, Nanjing, China
| | - Chengqi Qian
- Jiangsu Chunyu Education Group Co., Ltd., Nanjing, China
| | - Shumin Fei
- School of Automation, Southeast University, Nanjing, China
| |
Collapse
|
22
|
Inno A, Maurea N, Metro G, Carbone A, Russo A, Gori S. Immune checkpoint inhibitors-associated pericardial disease: a systematic review of case reports. Cancer Immunol Immunother 2021; 70:3041-3053. [DOI: 10.1007/s00262-021-02938-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 04/07/2021] [Indexed: 12/18/2022]
|
23
|
Poto R, Marone G, Pirozzi F, Galdiero MR, Cuomo A, Formisano L, Bianco R, Della Corte CM, Morgillo F, Napolitano S, Troiani T, Tocchetti CG, Mercurio V, Varricchi G. How can we manage the cardiac toxicity of immune checkpoint inhibitors? Expert Opin Drug Saf 2021; 20:685-694. [PMID: 33749484 DOI: 10.1080/14740338.2021.1906860] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Cancer immunotherapies with monoclonal antibodies (mAbs) against immune checkpoints (i.e. CTLA-4 and PD-1/PD-L1) have revolutionized antineoplastic treatments. Immune checkpoint inhibitors (ICIs) approved for cancer immunotherapy are mAbs anti-CTLA-4 (ipilimumab), anti-PD-1 (nivolumab, pembrolizumab, and cemiplimab), and anti-PD-L1 (atezolizumab, avelumab, and durvalumab). Treatment with ICIs can be associated with immune-related adverse events (irAEs), including an increased risk of developing myocarditis. These findings are compatible with the observation that, CTLA-4, PD-1, and PD-L1 pathways play a central role in the modulation of autoimmunity.Areas covered: In this paper, we start from examining the pathogenesis of cardiovascular adverse events from ICIs, and then we focus on risk factors and strategies to prevent and manage this cardiotoxicity.Expert opinion: There is a growing need for a multidisciplinary approach of ICI-associated cardiotoxicity, involving oncologists, cardiologists, and immunologists. Prevention and effective management of ICIs cardiotoxicity starts with an in-depth screening and surveillance strategies of high-risk patients, in order to improve early detection and appropriate management in a personalized approach.
Collapse
Affiliation(s)
- Remo Poto
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Giancarlo Marone
- Department of Public Health, Section of Hygiene, University of Naples Federico II, Naples, Italy.,Monaldi Hospital Pharmacy, Naples, Italy
| | - Flora Pirozzi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Maria Rosaria Galdiero
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Luigi Formisano
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | - Roberto Bianco
- Department of Clinical Medicine and Surgery, Federico II University, Naples, Italy.,Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy
| | | | - Floriana Morgillo
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Stefania Napolitano
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Teresa Troiani
- Department of Precision Medicine, Luigi Vanvitelli University of Campania, Naples, Italy
| | - Carlo G Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Interdepartmental Center of Clinical and Translational Research (CIRCET), Federico II University, Naples, Italy.,Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | - Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, Naples, Italy
| | - Gilda Varricchi
- Department of Translational Medical Sciences, Federico II University, Naples, Italy.,Center for Basic and Clinical Immunology Research (CISI), Federico II University, Naples, Italy.,WAO Center of Excellence, Naples, Italy.,Institute of Experimental Endocrinology and Oncology "G. Salvatore" (IEOS), National Research Council (CNR), Naples, Italy
| |
Collapse
|
24
|
Stein-Merlob AF, Rothberg MV, Ribas A, Yang EH. Cardiotoxicities of novel cancer immunotherapies. Heart 2021; 107:1694-1703. [PMID: 33722826 DOI: 10.1136/heartjnl-2020-318083] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/28/2021] [Accepted: 02/13/2021] [Indexed: 12/28/2022] Open
Abstract
Immunotherapy revolutionised oncology by harnessing the native immune system to effectively treat a wide variety of malignancies even at advanced stages. Off-target immune activation leads to immune-related adverse events affecting multiple organ systems, including the cardiovascular system. In this review, we discuss the current literature describing the epidemiology, mechanisms and proposed management of cardiotoxicities related to immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR) T-cell therapies and bispecific T-cell engagers. ICIs are monoclonal antibody antagonists that block a co-inhibitory pathway used by tumour cells to evade a T cell-mediated immune response. ICI-associated cardiotoxicities include myocarditis, pericarditis, atherosclerosis, arrhythmias and vasculitis. ICI-associated myocarditis is the most recognised and potentially fatal cardiotoxicity with mortality approaching 50%. Recently, ICI-associated dysregulation of the atherosclerotic plaque immune response with prolonged use has been linked to early progression of atherosclerosis and myocardial infarction. Treatment strategies include immunosuppression with corticosteroids and supportive care. In CAR T-cell therapy, autologous T cells are genetically engineered to express receptors targeted to cancer cells. While stimulating an effective tumour response, they also elicit a profound immune reaction called cytokine release syndrome (CRS). High-grade CRS causes significant systemic abnormalities, including cardiovascular effects such as arrhythmias, haemodynamic compromise and cardiomyopathy. Treatment with interleukin-6 inhibitors and corticosteroids is associated with improved outcomes. The evidence shows that, although uncommon, immunotherapy-related cardiovascular toxicities confer significant risk of morbidity and mortality and benefit from rapid immunosuppressive treatment. As new immunotherapies are developed and adopted, it will be imperative to closely monitor for cardiotoxicity.
Collapse
Affiliation(s)
- Ashley F Stein-Merlob
- Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Michael V Rothberg
- University of California Los Angeles David Geffen School of Medicine, Los Angeles, California, USA
| | - Antoni Ribas
- Division of Hematology-Oncology, Jonsson Comprehensive Cancer Center, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| | - Eric H Yang
- UCLA-Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, California, USA
| |
Collapse
|
25
|
Stein-Merlob AF, Rothberg MV, Holman P, Yang EH. Immunotherapy-Associated Cardiotoxicity of Immune Checkpoint Inhibitors and Chimeric Antigen Receptor T Cell Therapy: Diagnostic and Management Challenges and Strategies. Curr Cardiol Rep 2021; 23:11. [PMID: 33483873 PMCID: PMC7821837 DOI: 10.1007/s11886-021-01440-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/08/2021] [Indexed: 12/12/2022]
Abstract
PURPOSE OF REVIEW Immunotherapies have demonstrated robust clinical efficacy in treating malignancies with increasing use and FDA approvals. We review the epidemiology, risk factors, diagnosis, and treatment of immunotherapy-associated cardiovascular toxicities. RECENT FINDINGS Cardiotoxicity is reported in patients receiving immune checkpoint inhibitors (ICI) and chimeric antigen receptor (CAR) T cell therapies. The incidence of ICI-related cardiotoxicity is above 1% and includes myocarditis, pericardial disease, arrhythmia, acute coronary syndrome, and vasculitis. The incidence of CAR T cell-associated cardiotoxicities was shown to be as high as 26% and thought to be primarily mediated by cytokine release syndrome. The presentations of cardiotoxicities are variable but are associated with significant morbidity and mortality and benefit from prompt initiation of immunosuppressive therapy. There is increasing evidence for cardiotoxicities following cancer immunotherapy. Available evidence suggests that pretreatment evaluation, close monitoring, and early intervention may reduce cardiovascular morbidity and improve outcomes in the cancer immunotherapy population.
Collapse
Affiliation(s)
- Ashley F. Stein-Merlob
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Michael V. Rothberg
- David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Patrick Holman
- Department of Medicine, University of California, Los Angeles, Los Angeles, CA USA
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, Los Angeles, CA USA
- UCLA-Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California, Los Angeles, 100 Medical Plaza, Suite 630, Los Angeles, CA 90095 USA
| |
Collapse
|
26
|
Venturini E, Iannuzzo G, D’Andrea A, Pacileo M, Tarantini L, Canale M, Gentile M, Vitale G, Sarullo F, Vastarella R, Di Lorenzo A, Testa C, Parlato A, Vigorito C, Giallauria F. Oncology and Cardiac Rehabilitation: An Underrated Relationship. J Clin Med 2020; 9:E1810. [PMID: 32532011 PMCID: PMC7356735 DOI: 10.3390/jcm9061810] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 06/04/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
Cancer and cardiovascular diseases are globally the leading causes of mortality and morbidity. These conditions are closely related, beyond that of sharing many risk factors. The term bidirectional relationship indicates that cardiovascular diseases increase the likelihood of getting cancer and vice versa. The biological and biochemical pathways underlying this close relationship will be analyzed. In this new overlapping scenario, physical activity and exercise are proven protective behaviors against both cardiovascular diseases and cancer. Many observational studies link an increase in physical activity to a reduction in either the development or progression of cancer, as well as to a reduction in risk in cardiovascular diseases, a non-negligible cause of death for long-term cancer survivors. Exercise is an effective tool for improving cardio-respiratory fitness, quality of life, psychological wellbeing, reducing fatigue, anxiety and depression. Finally, it can counteract the toxic effects of cancer therapy. The protection obtained from physical activity and exercise will be discussed in the various stages of the cancer continuum, from diagnosis, to adjuvant therapy, and from the metastatic phase to long-term effects. Particular attention will be paid to the shelter against chemotherapy, radiotherapy, cardiovascular risk factors or new onset cardiovascular diseases. Cardio-Oncology Rehabilitation is an exercise-based multi-component intervention, starting from the model of Cardiac Rehabilitation, with few modifications, to improve care and the prognosis of a patient's cancer. The network of professionals dedicated to Cardiac Rehabilitation is a ready-to-use resource, for implementing Cardio-Oncology Rehabilitation.
Collapse
Affiliation(s)
- E. Venturini
- Cardiac Rehabilitation Unit, Azienda USL Toscana Nord-Ovest, Cecina Civil Hospital, 57023 LI Cecina, Italy
| | - G. Iannuzzo
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (G.I.); (M.G.)
| | - A. D’Andrea
- Unit of Cardiology and Intensive Care, “Umberto I” Hospital, Viale San Francesco, Nocera Inferiore, 84014 SA, Italy; (A.D.); (M.P.)
| | - M. Pacileo
- Unit of Cardiology and Intensive Care, “Umberto I” Hospital, Viale San Francesco, Nocera Inferiore, 84014 SA, Italy; (A.D.); (M.P.)
| | - L. Tarantini
- Division of Cardiology, Ospedale San Martino ULSS1 Dolomiti, 32100 Belluno, Italy;
| | - M.L. Canale
- Department of Cardiology, Azienda USL Toscana Nord-Ovest, Ospedale Versilia, Lido di Camaiore, 55041 LU, Italy;
| | - M. Gentile
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy; (G.I.); (M.G.)
| | - G. Vitale
- Cardiovascular Rehabilitation Unit, Buccheri La Ferla Fatebenefratelli Hospital, 90123 Palermo, Italy; (G.V.); (F.M.S.)
| | - F.M. Sarullo
- Cardiovascular Rehabilitation Unit, Buccheri La Ferla Fatebenefratelli Hospital, 90123 Palermo, Italy; (G.V.); (F.M.S.)
| | - R. Vastarella
- UOSD Scompenso Cardiaco e Cardiologia Riabilitativa, AORN Ospedale dei Colli-Monaldi, 80131 Naples, Italy;
| | - A. Di Lorenzo
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (A.D.L.); (C.T.); (A.P.); (C.V.); (F.G.)
| | - C. Testa
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (A.D.L.); (C.T.); (A.P.); (C.V.); (F.G.)
| | - A. Parlato
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (A.D.L.); (C.T.); (A.P.); (C.V.); (F.G.)
| | - C. Vigorito
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (A.D.L.); (C.T.); (A.P.); (C.V.); (F.G.)
| | - F. Giallauria
- Department of Translational Medical Sciences, Federico II University of Naples, 80131 Naples, Italy; (A.D.L.); (C.T.); (A.P.); (C.V.); (F.G.)
| |
Collapse
|