1
|
Bai T, Wan Q, Yue C, Wang J, Deng S, Shen X, Wang H, Huang L, Wang D. Combined spatial metabolomics and 4D-DIA quantitative proteomics approaches to explore the relationship between lung cancer and the heart. Sci Rep 2025; 15:14878. [PMID: 40295682 PMCID: PMC12037715 DOI: 10.1038/s41598-025-97821-7] [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: 11/03/2024] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Lung cancer and cardiovascular disease pose persistent threats to human health, despite advancements in targeted therapy, percutaneous coronary intervention, and drug treatments. Challenges such as side effects, drug resistance, hospitalization rates, and mortality remain high. These diseases are closely linked, sharing common risk factors and intricately influencing each other. This study aims to investigate the interplay between lung disease and cardiovascular disease by examining changes in cardiac metabolites and protein expression using spatial metabolomics and 4D-DIA quantitative proteomics approaches in the setting of lung cancer. Nude mice were selected and A549 cells were injected axillary and metabolomics was used to observe the alterations in cardiac metabolism in the setting of lung cancer in nude mice.The findings reveal well-defined tumor structures. Further, spatial mass spectrometry imaging analysis demonstrates distinct metabolite distributions across cardiac regions, indicating significant differences between control and model groups. Through spatial metabolomics and proteomics analyses, key differential metabolites such as Gln-His-Val-Glu, LysoPC 22:6, and LPC (20:2/0:0), primarily amino acids, and glycerophospholipids, as well as differential proteins including Mknk1, Trafd1, Dab2ip, Tab1, Ripk3, G3PDH, and Mapk15, are identified. These results underscore the crucial role of these factors in cardiovascular injury. This study elucidates the intricate link between lung cancer and cardiovascular disease and identifies altered metabolites and proteins in the heart within a lung cancer environment. These insights are pivotal for informing future treatments and interventions for both diseases.
Collapse
Affiliation(s)
- Tingting Bai
- Inner Mongolia Medical University, Hohhot, Inner Mongolia, People's Republic of China
| | - Quan Wan
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Changcheng Yue
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Jingjing Wang
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Shichao Deng
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Xueying Shen
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Hongqing Wang
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Liyan Huang
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China
| | - Dong Wang
- Affiliated Hospital of Inner Mongolia Minzu University, No.1742 East Hollinghe Street, Tongliao, 028000, Inner Mongolia, People's Republic of China.
| |
Collapse
|
2
|
Liu R, Jia L, Yu L, Lai D, Li Q, Zhang B, Guo E, Xu K, Luo Q. Interaction between post-tumor inflammation and vascular smooth muscle cell dysfunction in sepsis-induced cardiomyopathy. Front Immunol 2025; 16:1560717. [PMID: 40276499 PMCID: PMC12018406 DOI: 10.3389/fimmu.2025.1560717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 02/28/2025] [Indexed: 04/26/2025] Open
Abstract
Background Sepsis-induced cardiomyopathy (SIC) presents a critical complication in cancer patients, contributing notably to heart failure and elevated mortality rates. While its clinical relevance is well-documented, the intricate molecular mechanisms that link sepsis, tumor-driven inflammation, and cardiac dysfunction remain inadequately explored. This study aims to elucidate the interaction between post-tumor inflammation, intratumor heterogeneity, and the dysfunction of VSMC in SIC, as well as to evaluate the therapeutic potential of exercise training and specific pharmacological interventions. Methods Transcriptomic data from NCBI and GEO databases were analyzed to identify differentially expressed genes (DEGs) associated with SIC. Weighted gene co-expression network analysis (WGCNA), gene ontology (GO), and KEGG pathway enrichment analyses were utilized to elucidate the biological significance of these genes. Molecular docking and dynamics simulations were used to investigate drug-target interactions, and immune infiltration and gene mutation analyses were carried out by means of platforms like TIMER 2.0 and DepMap to comprehend the influence of DVL1 on immune responsiveness. Results Through the utilization of the datasets, we discovered the core gene DVL1 that exhibited remarkable up-regulated expression both in SIC and in diverse kinds of cancers, which were associated with poor prognosis and inflammatory responses. Molecular docking revealed that Digoxin could bind to DVL1 and reduce oxidative stress in SIC. The DVL1 gene module related to SIC was identified by means of WGCNA, and the immune infiltration analysis demonstrated the distinctive immune cell patterns associated with DVL1 expression and the impact of DVL1 on immunotherapeutic resistance. Conclusions DVL1 is a core regulator of SIC and other cancers and, therefore, can serve as a therapeutic target. The present study suggests that targeted pharmacological therapies to enhance response to exercise regimens may be a novel therapeutic tool to reduce the inflammatory response during sepsis, particularly in cancer patients. The identified drugs, Digoxin, require further in vivo and clinical studies to confirm their effects on SIC and their potential efforts to improve outcomes in immunotherapy-resistant cancer patients.
Collapse
Affiliation(s)
- Rui Liu
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Lina Jia
- Hebei Medical University, Shijiazhuang, China
| | - Lin Yu
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Detian Lai
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Qingzhu Li
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Bingyu Zhang
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Enwei Guo
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Kailiang Xu
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| | - Qiancheng Luo
- Department of Critical Care Medicine, Shanghai Pudong New Area Gongli Hospital, Shanghai, China
| |
Collapse
|
3
|
Pearce L, Galán-Arriola C, Bell RM, Carr RD, Cunningham J, Davidson SM, Ghosh AK, Giesz S, Golforoush P, Gourine AV, Hermann DM, Heusch G, Ibanez B, Kalkhoran SB, Lecour S, Lukhna K, Ntsekhe M, Sack MN, Unwin RJ, Vilahur G, Walker JM, Yellon DM. Inter-organ communication: pathways and targets to cardioprotection and neuro-protection. A report from the 12th Hatter Cardiovascular Institute workshop. Basic Res Cardiol 2025; 120:287-299. [PMID: 39681732 PMCID: PMC11976342 DOI: 10.1007/s00395-024-01094-6] [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: 12/04/2024] [Revised: 12/04/2024] [Accepted: 12/04/2024] [Indexed: 12/18/2024]
Abstract
A long-standing aim in the setting of various pathologies including acute myocardial infarction, chronic kidney disease (CKD), and ischaemic stroke, has been to identify successful approaches to augment cellular and organ protection. Although the continual evolution and refinement of ideas over the past few decades has allowed the field to progress, we are yet to realise successful clinical translation of this concept. The 12th Hatter Cardiovascular Workshop identified a number of important points and key questions for future research relating to cardio- and neuro-protection and interorgan communication. Specific topics that were discussed include the 'cardio-metabolic-renal' axis of organ protection, the parasympathetic signalling hypothesis, the role of the coronary microvasculature in myocardial infarction, the RISK pathway of cardioprotection, extracellular vesicles and the way forward, the future for clinical studies of remote ischaemic conditioning, and new experimental models for cardio-oncology investigations.
Collapse
Affiliation(s)
- L Pearce
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - C Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - R M Bell
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - R D Carr
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
- School of Biomedical Sciences, Ulster University, Coleraine, UK
| | - J Cunningham
- Centre for Nephrology, University College London, London, UK
| | - S M Davidson
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - A K Ghosh
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - S Giesz
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - P Golforoush
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - A V Gourine
- Centre for Cardiovascular and Metabolic Neuroscience, Neuroscience, Physiology and Pharmacology, University College London, London, UK
| | - D M Hermann
- Chair of Vascular Neurology, Dementia and Ageing Research, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - G Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany
| | - B Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - S Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - S Lecour
- University of Cape Town, Cape Town, South Africa
| | - K Lukhna
- University of Cape Town, Cape Town, South Africa
| | - M Ntsekhe
- University of Cape Town, Cape Town, South Africa
| | - M N Sack
- Laboratory of Mitochondrial Biology and Metabolism, NHLBI, National Institutes of Health, Bethesda, MD, USA
| | - R J Unwin
- Centre for Nephrology, University College London, London, UK
| | - G Vilahur
- Institut de Recerca Sant Pau, IIB-Sant Pau, Hospital de la Santa Creu i Sant Pau, CIBERCV, Barcelona, Spain
| | - J M Walker
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK
| | - D M Yellon
- The Hatter Cardiovascular Institute, University College London, 67 Chenies Mews, London, WC1E 6HX, UK.
- University of Cape Town, Cape Town, South Africa.
| |
Collapse
|
4
|
Quagliariello V, Berretta M, Bisceglia I, Giacobbe I, Iovine M, Barbato M, Maurea C, Canale ML, Paccone A, Inno A, Scherillo M, Oliva S, Cadeddu Dessalvi C, Mauriello A, Fonderico C, Maratea AC, Gabrielli D, Maurea N. In the Era of Cardiovascular-Kidney-Metabolic Syndrome in Cardio-Oncology: From Pathogenesis to Prevention and Therapy. Cancers (Basel) 2025; 17:1169. [PMID: 40227756 PMCID: PMC11988012 DOI: 10.3390/cancers17071169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome represents a complex interplay between cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic disorders, significantly impacting cancer patients. The presence of CKM syndrome in cancer patients not only worsens their prognosis but also increases the risk of major adverse cardiovascular events (MACE), reduces quality of life (QoL), and affects overall survival (OS). Furthermore, several anticancer therapies, including anthracyclines, tyrosine kinase inhibitors, immune checkpoint inhibitors, and hormonal treatments, can exacerbate CKM syndrome by inducing cardiotoxicity, nephrotoxicity, and metabolic dysregulation. This review explores the pathophysiology of CKM syndrome in cancer patients and highlights emerging therapeutic strategies to mitigate its impact. We discuss the role of novel pharmacological interventions, including sodium-glucose cotransporter-2 inhibitors (SGLT2i), proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), and soluble guanylate cyclase (sGC) activators, as well as dietary and lifestyle interventions. Optimizing the management of CKM syndrome in cancer patients is crucial to improving OS, enhancing QoL, and reducing MACE. By integrating cardiometabolic therapies into oncologic care, we can create a more comprehensive treatment approach that reduces the burden of cardiovascular and renal complications in this vulnerable population. Further research is needed to establish personalized strategies for CKM syndrome prevention and treatment in cancer patients.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Carlo Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, 23807 Naples, Italy
| | | | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy
| | - Marino Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, 82100 Benevento, Italy
| | - Stefano Oliva
- Cardio-Oncology Unit, IRCCS Istituto Tumori, “Giovanni Paolo II”, 70124 Bari, Italy
| | | | - Alfredo Mauriello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Celeste Fonderico
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Anna Chiara Maratea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| |
Collapse
|
5
|
Nechita LC, Tutunaru D, Nechita A, Voipan AE, Voipan D, Tupu AE, Musat CL. AI and Smart Devices in Cardio-Oncology: Advancements in Cardiotoxicity Prediction and Cardiovascular Monitoring. Diagnostics (Basel) 2025; 15:787. [PMID: 40150129 PMCID: PMC11940913 DOI: 10.3390/diagnostics15060787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 03/29/2025] Open
Abstract
The increasing prevalence of cardiovascular complications in cancer patients due to cardiotoxic treatments has necessitated advanced monitoring and predictive solutions. Cardio-oncology is an evolving interdisciplinary field that addresses these challenges by integrating artificial intelligence (AI) and smart cardiac devices. This comprehensive review explores the integration of artificial intelligence (AI) and smart cardiac devices in cardio-oncology, highlighting their role in improving cardiovascular risk assessment and the early detection and real-time monitoring of cardiotoxicity. AI-driven techniques, including machine learning (ML) and deep learning (DL), enhance risk stratification, optimize treatment decisions, and support personalized care for oncology patients at cardiovascular risk. Wearable ECG patches, biosensors, and AI-integrated implantable devices enable continuous cardiac surveillance and predictive analytics. While these advancements offer significant potential, challenges such as data standardization, regulatory approvals, and equitable access must be addressed. Further research, clinical validation, and multidisciplinary collaboration are essential to fully integrate AI-driven solutions into cardio-oncology practices and improve patient outcomes.
Collapse
Affiliation(s)
- Luiza Camelia Nechita
- Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania
| | - Dana Tutunaru
- Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania
| | - Aurel Nechita
- Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania
| | - Andreea Elena Voipan
- Faculty of Automation, Computers, Electrical Engineering and Electronics, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania
| | - Daniel Voipan
- Faculty of Automation, Computers, Electrical Engineering and Electronics, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania
| | - Ancuta Elena Tupu
- Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania
| | - Carmina Liana Musat
- Faculty of Medicine and Pharmacy, ‘Dunarea de Jos’ University of Galati, 800008 Galati, Romania
| |
Collapse
|
6
|
An N, Zhang X, Lin H, Xu Q, Dai Q, Kong Y, Han S, Li X, Yang X, Xing Y, Shang H. The role and mechanism of TXNDC5 in cardio-oncology: Killing two birds with one stone? Curr Probl Cardiol 2025; 50:102951. [PMID: 39643150 DOI: 10.1016/j.cpcardiol.2024.102951] [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: 11/26/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Cardio-oncology has emerged as a new translational and clinical field owing to the growing repertory of cancer therapy. To date, there is a lack of effective pharmacological therapy to target cardiotoxicity. Cardio-oncology, which began by investigating the negative effects of cancer medicines on cardiovascular system, has since grown to include research into the similarities between cardiovascular disease (CVD) and cancer. Thioredoxin domain-containing protein 5 (TXNDC5) belongs to the protein disulfide isomerase (PDI) family. Many diseases, including CVD and cancer, improperly express TXNDC5. This review provides a comprehensive analysis of the expression patterns of TXNDC5 in diseases. It outlines the processes via which TXNDC5 contributes to the advancement of malignant diseases such as CVD and cancer. Additionally, it summarizes prospective therapeutic approaches that can be used to target TXNDC5 for the treatment of these diseases. This will offer novel perspectives for enhancing anticancer therapy and advancing cardio-oncology research and drug development.
Collapse
Affiliation(s)
- Na An
- DongZhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyuan Lin
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qianqian Xu
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qianqian Dai
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - YiFan Kong
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Li
- DongZhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China; Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China; College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
7
|
Sabaté-Tormos M, Bardají A, Peiró OM, Carrasquer A, Cediel G, Ferreiro JL. Cancer and myocardial injury in patients with suspected acute coronary syndrome. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2025; 11:21. [PMID: 39987454 PMCID: PMC11846249 DOI: 10.1186/s40959-025-00320-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/11/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Cancer and cardiovascular diseases are the leading causes of mortality worldwide, as they share common risk factors and exacerbate cardiovascular outcomes when they coexist. This study aimed to assess the clinical characteristics and cardiovascular outcomes of patients with a history of cancer and myocardial injury (MI) presenting with suspected acute coronary syndrome (ACS) in an emergency setting. METHODS This retrospective cohort study included 3,626 patients admitted to the emergency department with suspected ACS between 2012 and 2013. Patients were categorized on the basis of their cancer history and the presence of MI. Clinical variables and the associations between cancer history and MI with all-cause mortality were analyzed over a four-year follow-up period via univariate and multivariate Cox regression models. RESULTS Of the cohort, 10.6% (n = 384) had a history of cancer. Compared with other groups, cancer patients with MI were older, had more comorbidities, and presented a higher incidence of type 2 myocardial infarction (T2MI). At the four-year follow-up, all-cause mortality was significantly greater among cancer patients with MI (68.8%) than among cancer patients without MI (32.4%) and noncancer patients with or without MI (42.5% vs. 11.3%, respectively). Multivariate analysis identified cancer patients, particularly those with MI, as independent predictors of mortality. CONCLUSIONS Patients who present to emergency departments with suspected ACS, a history of cancer, or the presence of MI face greater cardiovascular risk and mortality than other patients do. The higher prevalence of T2MI in this population underscores the need for tailored management strategies.
Collapse
Affiliation(s)
- Marta Sabaté-Tormos
- Department of Cardiology, Joan XXIII University Hospital, Tarragona, Spain
- Pere Virgili Health Research Institute (IISPV), Tarragona, Spain
- Rovira I Virgili University, Tarragona, Spain
| | - Alfredo Bardají
- Department of Cardiology, Joan XXIII University Hospital, Tarragona, Spain.
- Pere Virgili Health Research Institute (IISPV), Tarragona, Spain.
- Rovira I Virgili University, Tarragona, Spain.
- Cardiology Service, Tarragona Joan XXIII University Hospital, Rovira Virgili University, IISPV, Calle Dr Mallafré Guasch 4, Tarragona, 43005, Spain.
| | - Oscar M Peiró
- Department of Cardiology, Joan XXIII University Hospital, Tarragona, Spain
- Pere Virgili Health Research Institute (IISPV), Tarragona, Spain
- Rovira I Virgili University, Tarragona, Spain
| | - Anna Carrasquer
- Department of Cardiology, Joan XXIII University Hospital, Tarragona, Spain
- Pere Virgili Health Research Institute (IISPV), Tarragona, Spain
- Rovira I Virgili University, Tarragona, Spain
| | - German Cediel
- Department of Cardiology, Joan XXIII University Hospital, Tarragona, Spain
- Pere Virgili Health Research Institute (IISPV), Tarragona, Spain
- Rovira I Virgili University, Tarragona, Spain
| | - Jose Luis Ferreiro
- Department of Cardiology, Joan XXIII University Hospital, Tarragona, Spain
- Pere Virgili Health Research Institute (IISPV), Tarragona, Spain
- Rovira I Virgili University, Tarragona, Spain
| |
Collapse
|
8
|
Murg SI, Matiș L, Moldovan AF, Popovici DI, Negru AG, Ghitea TC, Popescu MI. Association Between Advanced TNM Stages and Increased Risk of Cardiac Dysfunction in Patients with LVEF < 50. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:301. [PMID: 40005419 PMCID: PMC11857368 DOI: 10.3390/medicina61020301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 12/30/2024] [Accepted: 02/07/2025] [Indexed: 02/27/2025]
Abstract
Background and Objectives: Cardio-oncology addresses the growing concern of cardiovascular complications arising from cancer therapies. Although cancer treatments have greatly enhanced survival outcomes, they frequently carry substantial risks to cardiovascular health. This research examines the cardiovascular toxicity associated with HER2-targeted therapies, focusing on the interconnection between tumor characteristics, including histopathological profiles and TNM classification, and the development of cardiovascular complications. The objective is to identify key correlations that inform better prevention and management strategies for cardiotoxicity in oncology patients. Materials and Methods: This retrospective study analyzed cancer patients undergoing cytostatic treatments, particularly anthracyclines, radiotherapy, and HER2-targeted therapies. Cardiac function was monitored using echocardiographic assessments, including global longitudinal strain and left ventricular ejection fraction (LVEF). Patients were stratified based on TNM cancer staging and histopathological findings to evaluate correlations between treatment regimens and cardiovascular outcomes. Results: The analysis revealed a significant association between advanced TNM stages and reduced LVEF, with patients in stage T4 showing the highest prevalence of cardiac dysfunction. Cytostatic treatments, such as anthracyclines and HER2-targeted therapies, were identified as key contributors to cardiotoxicity, particularly in advanced-stage cancer patients. These findings emphasize the importance of regular cardiac monitoring to detect early signs of cardiotoxicity, as patients with pre-existing cardiovascular risk factors demonstrated a higher prevalence of complications. Conclusions: This study highlights the need for personalized treatment approaches and tailored cardioprotective strategies to improve outcomes and enhance the quality of life for oncology patients. Future studies should prioritize developing improved strategies to reduce the cardiovascular complications linked to contemporary cancer treatments.
Collapse
Affiliation(s)
- Sergiu Ioan Murg
- Doctoral School, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania;
| | - Loredana Matiș
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| | - Andrada Florina Moldovan
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| | - Dorel Ionel Popovici
- Department of Oncology, Faculty of Medicine, Victor Babeş University of Medicine and Pharmacy Timisoara, Eftimie Murgu Square 2, 300041 Timisoara, Romania;
| | - Alina Gabriela Negru
- Department of Cardiovascular Diseases of Timisoara, Victor Babeş University of Medicine and Pharmacy Timisoara, G. Adam Str. No. 13A, 300310 Timisoara, Romania;
| | - Timea Claudia Ghitea
- Pharmacy Department, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania
| | - Mircea Ioachim Popescu
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| |
Collapse
|
9
|
Liu X, Li Z. The role and mechanism of epigenetics in anticancer drug-induced cardiotoxicity. Basic Res Cardiol 2025; 120:11-24. [PMID: 38724618 DOI: 10.1007/s00395-024-01054-0] [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: 01/29/2024] [Revised: 04/20/2024] [Accepted: 05/03/2024] [Indexed: 05/23/2024]
Abstract
Cardiovascular disease is the main factor contributing to the global burden of diseases, and the cardiotoxicity caused by anticancer drugs is an essential component that cannot be ignored. With the development of anticancer drugs, the survival period of cancer patients is prolonged; however, the cardiotoxicity caused by anticancer drugs is becoming increasingly prominent. Currently, cardiovascular disease has emerged as the second leading cause of mortality among long-term cancer survivors. Anticancer drug-induced cardiotoxicity has become a frontier and hot topic. The discovery of epigenetics has given the possibility of environmental changes in gene expression, protein synthesis, and traits. It has been found that epigenetics plays a pivotal role in promoting cardiovascular diseases, such as heart failure, coronary heart disease, and hypertension. In recent years, increasing studies have underscored the crucial roles played by epigenetics in anticancer drug-induced cardiotoxicity. Here, we provide a comprehensive overview of the role and mechanisms of epigenetics in anticancer drug-induced cardiotoxicity.
Collapse
Affiliation(s)
- Xuening Liu
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zijian Li
- Department of Pharmacy, Peking University Third Hospital, Beijing, 100191, China.
- Department of Pharmacy Administration and Clinical Pharmacy, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital; State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing, 100191, China.
| |
Collapse
|
10
|
Sayour NV, Kucsera D, Alhaddad AR, Tóth VÉ, Gergely TG, Kovács T, Hegedűs ZI, Jakab ME, Ferdinandy P, Varga ZV. Effects of sex and obesity on immune checkpoint inhibition-related cardiac systolic dysfunction in aged mice. Basic Res Cardiol 2025; 120:207-223. [PMID: 39516409 PMCID: PMC11790738 DOI: 10.1007/s00395-024-01088-4] [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: 05/29/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
Despite accumulating data on underlying mechanisms, the influence of sex and prevalent cardio-metabolic co-morbidities on the manifestation and severity of immune checkpoint inhibitor (ICI)-induced cardiotoxicity has not been well defined. To elucidate whether sex and prevalent cardio-metabolic co-morbidities affect ICI-induced cardiotoxicity, we randomized 17-month-old male and female mice to receive control diet (CON) or high-fat diet (HFD) + L-NAME-a well-established mouse model of cardio-metabolic co-morbidities-for 17 weeks (n = 5-7), and evaluated markers of T-cell function in the spleen. As expected, HFD + L-NAME significantly increased body- and heart weight, and serum cholesterol levels, and caused no systolic dysfunction, however, led to diastolic dysfunction, cardiomyocyte hypertrophy, and increased fibrosis only in males compared to corresponding CON. Western blot analyses of splenic immune checkpoint protein levels showed differential expression depending on sex and prevalent cardio-metabolic co-morbidities, suggesting T-cell exhaustion in both sexes on HFD + L-NAME, but more pronounced in males. In a sub-study with a similar setup, we tested cardiotoxic manifestations of ICI by treating mice with anti-PD-1 monoclonal antibody (ICI) for the last 2 weeks of diet administration (n = 5-7). After 2 weeks of ICI treatment, cardiac systolic functions significantly decreased in CON, but not in HFD + L-NAME groups of both sexes compared to baseline (before ICI administration). In conclusion, in this exploratory study using aged mice, we describe for the first time that ICI-related systolic dysfunction is diminished in both sexes when obesity and hypercholesterolemia are present, possibly due to obesity-related T-cell exhaustion.
Collapse
Affiliation(s)
- Nabil V Sayour
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Dániel Kucsera
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Ayham R Alhaddad
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Viktória É Tóth
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Tamás G Gergely
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Tamás Kovács
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Zsombor I Hegedűs
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Márk E Jakab
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- Center for Pharmacology and Drug Research & Development, Semmelweis University, Budapest, Hungary.
- HCEMM-SE Cardiometabolic Immunology Research Group, Semmelweis University, Budapest, Hungary.
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
11
|
Buehning F, Lerchner T, Vogel J, Hendgen-Cotta UB, Totzeck M, Rassaf T, Michel L. Preclinical models of cardiotoxicity from immune checkpoint inhibitor therapy. Basic Res Cardiol 2025; 120:171-185. [PMID: 39039301 PMCID: PMC11790694 DOI: 10.1007/s00395-024-01070-0] [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: 03/05/2024] [Revised: 06/30/2024] [Accepted: 07/16/2024] [Indexed: 07/24/2024]
Abstract
Immune checkpoint inhibitor (ICI) therapy represents a ground-breaking paradigm in cancer treatment, harnessing the immune system to combat malignancies by targeting checkpoints such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein 1 (PD-1). The use of ICI therapy generates distinctive immune-related adverse events (irAEs) including cardiovascular toxicity, necessitating targeted research efforts. This comprehensive review explores preclinical models dedicated to ICI-mediated cardiovascular complications including myocarditis. Tailored preclinical models of ICI-mediated myocardial toxicities highlight the key role of CD8+ T cells, emphasizing the profound impact of immune checkpoints on maintaining cardiac integrity. Cytokines and macrophages were identified as possible driving factors in disease progression, and at the same time, initial data on possible cardiac antigens responsible are emerging. The implications of contributing factors including thoracic radiation, autoimmune disorder, and the presence of cancer itself are increasingly understood. Besides myocarditis, mouse models unveiled an accelerated progression of atherosclerosis, adding another layer for a thorough understanding of the diverse processes involving cardiovascular immune checkpoint signalling. This review aims to discuss current preclinical models of ICI cardiotoxicity and their potential for improving enhanced risk assessment and diagnostics, offering potential targets for innovative cardioprotective strategies. Lessons from ICI therapy can drive novel approaches in cardiovascular research, extending insights to areas such as myocardial infarction and heart failure.
Collapse
Affiliation(s)
- Florian Buehning
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tobias Lerchner
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Julia Vogel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Ulrike B Hendgen-Cotta
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany
| | - Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, Hufelandstraße 55, 45147, Essen, Germany.
| |
Collapse
|
12
|
Brauer J, Tumani M, Frey N, Lehmann LH. The cardio-oncologic burden of breast cancer: molecular mechanisms and importance of preclinical models. Basic Res Cardiol 2025; 120:91-112. [PMID: 39621070 PMCID: PMC11790711 DOI: 10.1007/s00395-024-01090-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 02/04/2025]
Abstract
Breast cancer, the most prevalent cancer affecting women worldwide, poses a significant cardio-oncological burden. Despite advancements in novel therapeutic strategies, anthracyclines, HER2 antagonists, and radiation remain the cornerstones of oncological treatment. However, each carries a risk of cardiotoxicity, though the molecular mechanisms underlying these adverse effects differ. Common mechanisms include DNA damage response, increased reactive oxygen species, and mitochondrial dysfunction, which are key areas of ongoing research for potential cardioprotective strategies. Since these mechanisms are also essential for effective tumor cytotoxicity, we explore tumor-specific effects, particularly in hereditary breast cancer linked to BRCA1 and BRCA2 mutations. These genetic variants impair DNA repair mechanisms, increase the risk of tumorigenesis and possibly for cardiotoxicity from treatments such as anthracyclines and HER2 antagonists. Novel therapies, including immune checkpoint inhibitors, are used in the clinic for triple-negative breast cancer and improve the oncological outcomes of breast cancer patients. This review discusses the molecular mechanisms underlying BRCA dysfunction and the associated pathological pathways. It gives an overview of preclinical models of breast cancer, such as genetically engineered mouse models, syngeneic murine models, humanized mouse models, and various in vitro and ex vivo systems and models to study cardiovascular side effects of breast cancer therapies. Understanding the underlying mechanism of cardiotoxicity and developing cardioprotective strategies in preclinical models are essential for improving treatment outcomes and reducing long-term cardiovascular risks in breast cancer patients.
Collapse
Affiliation(s)
- J Brauer
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - M Tumani
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - N Frey
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - L H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
13
|
Gergely TG, Drobni ZD, Sayour NV, Ferdinandy P, Varga ZV. Molecular fingerprints of cardiovascular toxicities of immune checkpoint inhibitors. Basic Res Cardiol 2025; 120:187-205. [PMID: 39023770 PMCID: PMC11790702 DOI: 10.1007/s00395-024-01068-8] [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: 05/30/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 07/20/2024]
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer therapy by unleashing the power of the immune system against malignant cells. However, their use is associated with a spectrum of adverse effects, including cardiovascular complications, which can pose significant clinical challenges. Several mechanisms contribute to cardiovascular toxicity associated with ICIs. First, the dysregulation of immune checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death protein-1 (PD-1) and its ligand (PD-L1), and molecular mimicry with cardiac autoantigens, leads to immune-related adverse events, including myocarditis and vasculitis. These events result from the aberrant activation of T cells against self-antigens within the myocardium or vascular endothelium. Second, the disruption of immune homeostasis by ICIs can lead to autoimmune-mediated inflammation of cardiac tissues, manifesting as cardiac dysfunction and heart failure, arrhythmias, or pericarditis. Furthermore, the upregulation of inflammatory cytokines, particularly tumor necrosis factor-alpha, interferon-γ, interleukin-1β, interleukin-6, and interleukin-17 contributes to cardiac and endothelial dysfunction, plaque destabilization, and thrombosis, exacerbating cardiovascular risk on the long term. Understanding the intricate mechanisms of cardiovascular side effects induced by ICIs is crucial for optimizing patient care and to ensure the safe and effective integration of immunotherapy into a broader range of cancer treatment protocols. The clinical implications of these mechanisms underscore the importance of vigilant monitoring and early detection of cardiovascular toxicity in patients receiving ICIs. Future use of these key pathological mediators as biomarkers may aid in prompt diagnosis of cardiotoxicity and will allow timely interventions.
Collapse
Affiliation(s)
- Tamás G Gergely
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Zsófia D Drobni
- Heart and Vascular Center, Semmelweis University, Budapest, Hungary
| | - Nabil V Sayour
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Péter Ferdinandy
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Zoltán V Varga
- Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary.
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary.
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary.
| |
Collapse
|
14
|
Efentakis P, Choustoulaki A, Kwiatkowski G, Varela A, Kostopoulos IV, Tsekenis G, Ntanasis-Stathopoulos I, Georgoulis A, Vorgias CE, Gakiopoulou H, Briasoulis A, Davos CH, Kostomitsopoulos N, Tsitsilonis O, Dimopoulos MA, Terpos E, Chłopicki S, Gavriatopoulou M, Andreadou I. Early microvascular coronary endothelial dysfunction precedes pembrolizumab-induced cardiotoxicity. Preventive role of high dose of atorvastatin. Basic Res Cardiol 2025; 120:263-286. [PMID: 38520533 PMCID: PMC11790778 DOI: 10.1007/s00395-024-01046-0] [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: 01/10/2024] [Revised: 03/07/2024] [Accepted: 03/08/2024] [Indexed: 03/25/2024]
Abstract
Immune checkpoint inhibitors (ICIs) exhibit remarkable antitumor activity and immune-related cardiotoxicity of unknown pathomechanism. The aim of the study was to investigate the ICI class-dependent cardiotoxicity in vitro and pembrolizumab's (Pem's) cardiotoxicity in vivo, seeking for translational prevention means. Cytotoxicity was investigated in primary cardiomyocytes and splenocytes, incubated with ipilimumab, Pem and avelumab. Pem's cross-reactivity was assessed by circular dichroism (CD) on biotechnologically produced human and murine PD-1 and in silico. C57BL6/J male mice received IgG4 or Pem for 2 and 5 weeks. Echocardiography, histology, and molecular analyses were performed. Coronary blood flow velocity mapping and cardiac magnetic resonance imaging were conducted at 2 weeks. Human EA.hy926 endothelial cells were incubated with Pem-conditioned media from human mononuclear cells, in presence and absence of statins and viability and molecular signaling were assessed. Atorvastatin (20 mg/kg, daily) was administered in vivo, as prophylaxis. Only Pem exerted immune-related cytotoxicity in vitro. Pem's cross-reactivity with the murine PD-1 was confirmed by CD and docking. In vivo, Pem initiated coronary endothelial and diastolic dysfunction at 2 weeks and systolic dysfunction at 5 weeks. At 2 weeks, Pem induced ICAM-1 and iNOS expression and intracardiac leukocyte infiltration. At 5 weeks, Pem exacerbated endothelial activation and triggered cardiac inflammation. Pem led to immune-related cytotoxicity in EA.hy926 cells, which was prevented by atorvastatin. Atorvastatin mitigated functional deficits, by inhibiting endothelial dysfunction in vivo. We established for the first time an in vivo model of Pem-induced cardiotoxicity. Coronary endothelial dysfunction precedes Pem-induced cardiotoxicity, whereas atorvastatin emerges as a novel prophylactic therapy.
Collapse
MESH Headings
- Animals
- Atorvastatin/administration & dosage
- Atorvastatin/pharmacology
- Mice, Inbred C57BL
- Humans
- Male
- Cardiotoxicity/prevention & control
- Antibodies, Monoclonal, Humanized/toxicity
- Mice
- Immune Checkpoint Inhibitors/toxicity
- Endothelium, Vascular/drug effects
- Endothelium, Vascular/physiopathology
- Endothelium, Vascular/metabolism
- Myocytes, Cardiac/drug effects
- Myocytes, Cardiac/metabolism
- Myocytes, Cardiac/pathology
- Coronary Vessels/drug effects
- Coronary Vessels/physiopathology
- Coronary Vessels/metabolism
- Disease Models, Animal
- Hydroxymethylglutaryl-CoA Reductase Inhibitors/administration & dosage
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Antineoplastic Agents, Immunological/toxicity
Collapse
Affiliation(s)
- Panagiotis Efentakis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Angeliki Choustoulaki
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Grzegorz Kwiatkowski
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
| | - Aimilia Varela
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioannis V Kostopoulos
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - George Tsekenis
- Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Ioannis Ntanasis-Stathopoulos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Anastasios Georgoulis
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece
| | - Constantinos E Vorgias
- Department of Biochemistry & Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | - Harikleia Gakiopoulou
- Department of Pathology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexandros Briasoulis
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Ourania Tsitsilonis
- Flow Cytometry Unit, Section of Animal and Human Physiology, Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Evangelos Terpos
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Stefan Chłopicki
- Jagiellonian Centre for Experimental Therapeutics (JCET), Jagiellonian University, Kraków, Poland
- Medical College, Jagiellonian University, Krakow, Poland
| | - Maria Gavriatopoulou
- Department of Clinical Therapeutics, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Panepistimiopolis, Zografou, 15771, Athens, Greece.
| |
Collapse
|
15
|
Lohr D, Thiele A, Stahnke M, Braun VM, Klopfleisch R, Klein O, Dresen S, Landmesser U, Foryst-Ludwig A, Kintscher U, Schreiber LM, Beyhoff N. Characterization of anthracycline-induced cardiotoxicity by diffusion tensor magnetic resonance imaging. Basic Res Cardiol 2025; 120:57-69. [PMID: 38483601 PMCID: PMC11790774 DOI: 10.1007/s00395-024-01039-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/05/2024] [Accepted: 02/05/2024] [Indexed: 02/04/2025]
Abstract
Anthracyclines are highly potent anti-cancer drugs, but their clinical use is limited by severe cardiotoxic side effects. The impact of anthracycline-induced cardiotoxicity (AIC) on left ventricular (LV) microarchitecture and diffusion properties remains unknown. This study sought to characterize AIC by cardiovascular magnetic resonance diffusion tensor imaging (DTI). Mice were treated with Doxorubicin (DOX; n = 16) for induction of AIC or saline as corresponding control (n = 15). Cardiac function was assessed via echocardiography at the end of the study period. Whole hearts (n = 8 per group) were scanned ex vivo by high-resolution DTI at 7 T. Results were correlated with histopathology and mass spectrometry imaging. Mice with AIC demonstrated systolic dysfunction (LVEF 52 ± 3% vs. 43 ± 6%, P < 0.001), impaired global longitudinal strain (-19.6 ± 2.0% vs. -16.6 ± 3.0%, P < 0.01), and cardiac atrophy (LV mass index [mg/mm], 4.3 ± 0.1 vs. 3.6 ± 0.2, P < 0.01). Regional sheetlet angles were significantly lower in AIC, whereas helix angle and relative helicity remained unchanged. In AIC, fractional anisotropy was increased (0.12 ± 0.01 vs. 0.14 ± 0.02, P < 0.05). DOX-treated mice displayed higher planar and less spherical anisotropy (CPlanar 0.07 ± 0.01 vs. 0.09 ± 0.01, P < 0.01; CSpherical 0.89 ± 0.01 vs. 0.87 ± 0.02, P < 0.05). CPlanar and CSpherical yielded good discriminatory power to distinguish between mice with and without AIC (c-index 0.91 and 0.84, respectively, P for both < 0.05). AIC is associated with regional changes in sheetlet angle but no major abnormalities of global LV microarchitecture. The geometric shape of the diffusion tensor is altered in AIC. DTI may provide a new tool for myocardial characterization in patients with AIC, which warrants future clinical studies to evaluate its diagnostic utility.
Collapse
Affiliation(s)
- David Lohr
- Chair of Molecular and Cellular Imaging, Comprehensive Heart Failure Center (CHFC), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Arne Thiele
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a joint cooperation of Max-Delbrück Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Nephrology and Intensive Care Medicine, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Max Stahnke
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Vera M Braun
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Pathology, College of Veterinary Medicine, Freie Universität Berlin, Berlin, Germany
| | - Oliver Klein
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin-Brandenburg Center for Regenerative Therapy (BCRT), Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Sandra Dresen
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Ulf Landmesser
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany
| | - Anna Foryst-Ludwig
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Ulrich Kintscher
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Laura M Schreiber
- Chair of Molecular and Cellular Imaging, Comprehensive Heart Failure Center (CHFC), University Hospital Wuerzburg, Wuerzburg, Germany
| | - Niklas Beyhoff
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Institute of Pharmacology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany.
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité - Medical Heart Center of Charité and German Heart Institute Berlin, Berlin, Germany.
| |
Collapse
|
16
|
Wernhart S, Rassaf T. Exercise, cancer, and the cardiovascular system: clinical effects and mechanistic insights. Basic Res Cardiol 2025; 120:35-55. [PMID: 38353711 PMCID: PMC11790717 DOI: 10.1007/s00395-024-01034-4] [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: 12/15/2023] [Revised: 01/21/2024] [Accepted: 01/21/2024] [Indexed: 03/05/2024]
Abstract
Cardiovascular diseases and cancer are the leading causes of death in the Western world and share common risk factors. Reduced cardiorespiratory fitness (CRF) is a major determinant of cardiovascular morbidity and cancer survival. In this review we discuss cancer- induced disturbances of parenchymal, cellular, and mitochondrial function, which limit CRF and may be antagonized and attenuated through exercise training. We show the impact of CRF on cancer survival and its attenuating effects on cardiotoxicity of cancer-related treatment. Tailored exercise programs are not yet available for each tumor entity as several trials were performed in heterogeneous populations without adequate cardiopulmonary exercise testing (CPET) prior to exercise prescription and with a wide variation of exercise modalities. There is emerging evidence that exercise may be a crucial pillar in cancer treatment and a tool to mitigate cardiotoxic treatment effects. We discuss modalities of aerobic exercise and resistance training and their potential to improve CRF in cancer patients and provide an example of a periodization model for exercise training in cancer.
Collapse
Affiliation(s)
- Simon Wernhart
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- West German Heart- and Vascular Center, Department of Cardiology and Vascular Medicine, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
17
|
Panuccio G, Correale P, d'Apolito M, Mutti L, Giannicola R, Pirtoli L, Giordano A, Labate D, Macheda S, Carabetta N, Abdelwahed YS, Landmesser U, Tassone P, Tagliaferri P, De Rosa S, Torella D. Immuno-related cardio-vascular adverse events associated with immuno-oncological treatments: an under-estimated threat for cancer patients. Basic Res Cardiol 2025; 120:153-169. [PMID: 39225869 PMCID: PMC11790807 DOI: 10.1007/s00395-024-01077-7] [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: 05/22/2024] [Revised: 08/16/2024] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Immunotherapy represents an emergent and heterogeneous group of anticancer treatments harnessing the human immune-surveillance system, including immune-checkpoint inhibitor monoclonal antibodies (mAbs), Chimeric Antigen Receptor T Cells (CAR-T) therapy, cancer vaccines and lymphocyte activation gene-3 (LAG-3) therapy. While remarkably effective against several malignancies, these therapies, often in combination with other cancer treatments, have showed unforeseen toxicity, including cardiovascular complications. The occurrence of immuno-mediated adverse (irAEs) events has been progressively reported in the last 10 years. These irAEs present an extended range of severity, from self-limiting to life-threatening conditions. Although recent guidelines in CardioOncology have provided important evidence in managing cancer treatments, they often encompass general approaches. However, a specific focus is required due to the particular etiology, unique risk factors, and associated side effects of immunotherapy. This review aims to deepen the understanding of the prevalence and nature of cardiovascular issues in patients undergoing immunotherapy, offering insights into strategies for risk stratification and management.
Collapse
Affiliation(s)
- Giuseppe Panuccio
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany.
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Pierpaolo Correale
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Maria d'Apolito
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luciano Mutti
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Applied Sciences and Biotechnology, Università dell'Aquila, L'Aquila, Italy
| | - Rocco Giannicola
- Medical Oncology Unit, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Luigi Pirtoli
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Antonio Giordano
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
- Department of Medical Biotechnology, University of Siena, 53100, Siena, Italy
| | - Demetrio Labate
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Sebastiano Macheda
- Unit of Intensive Care Medicine and Anesthesia, Grande Ospedale Metropolitano Bianchi Melacrino Morelli, 89124, Reggio Calabria, Italy
| | - Nicole Carabetta
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Youssef S Abdelwahed
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
| | - Ulf Landmesser
- Department of Cardiology, Angiology and Intensive Care Medicine, Deutsches Herzzentrum der Charité Berlin, 12200, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), 10785, Berlin, Germany
- Berlin Institute of Health (BIH), 10178, Berlin, Germany
| | - Pierfrancesco Tassone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine and Center of Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA, 19122, USA
| | - Pierosandro Tagliaferri
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| |
Collapse
|
18
|
Camilli M, Viscovo M, Maggio L, Bonanni A, Torre I, Pellegrino C, Lamendola P, Tinti L, Teofili L, Hohaus S, Lanza GA, Ferdinandy P, Varga Z, Crea F, Lombardo A, Minotti G. Sodium-glucose cotransporter 2 inhibitors and the cancer patient: from diabetes to cardioprotection and beyond. Basic Res Cardiol 2025; 120:241-262. [PMID: 38935171 PMCID: PMC11790819 DOI: 10.1007/s00395-024-01059-9] [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: 01/19/2024] [Revised: 05/18/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024]
Abstract
Sodium-glucose cotransporter 2 inhibitors (SGLT2i), a new drug class initially designed and approved for treatment of diabetes mellitus, have been shown to exert pleiotropic metabolic and direct cardioprotective and nephroprotective effects that extend beyond their glucose-lowering action. These properties prompted their use in two frequently intertwined conditions, heart failure and chronic kidney disease. Their unique mechanism of action makes SGLT2i an attractive option also to lower the rate of cardiac events and improve overall survival of oncological patients with preexisting cardiovascular risk and/or candidate to receive cardiotoxic therapies. This review will cover biological foundations and clinical evidence for SGLT2i modulating myocardial function and metabolism, with a focus on their possible use as cardioprotective agents in the cardio-oncology settings. Furthermore, we will explore recently emerged SGLT2i effects on hematopoiesis and immune system, carrying the potential of attenuating tumor growth and chemotherapy-induced cytopenias.
Collapse
Affiliation(s)
- Massimiliano Camilli
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy.
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy.
| | - Marcello Viscovo
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Luca Maggio
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Alice Bonanni
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Ilaria Torre
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Claudio Pellegrino
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Priscilla Lamendola
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
| | - Lorenzo Tinti
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Luciana Teofili
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Stefan Hohaus
- Sezione di Ematologia, Dipartimento di Scienze Radiologiche ed Ematologiche, Università Cattolica del Sacro Cuore, Rome, Italy
- Dipartimento di Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
| | - Gaetano Antonio Lanza
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
- MTA-SE System Pharmacology Research Group, Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
| | - Zoltan Varga
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary
- HCEMM-SU Cardiometabolic Immunology Research Group, Budapest, Hungary
- MTA-SE Momentum Cardio-Oncology and Cardioimmunology Research Group, Budapest, Hungary
| | - Filippo Crea
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Center of Excellence of Cardiovascular Sciences, Ospedale Isola Tiberina - Gemelli Isola, Rome, Italy
| | - Antonella Lombardo
- Department of Cardiovascular and Pulmonary Sciences, Catholic University of the Sacred Heart, Rome, Italy
- Department of Cardiovascular Medicine, Fondazione Policlinico Universitario A. Gemelli IRCCS, L.go A. Gemelli, 1, 00168, Rome, Italy
| | | |
Collapse
|
19
|
Peng S, Cai M, Kuang H, Lin A, Ma Q, Dai X, Luo P, Liu Y, Zhang G, Bai Y. From bench to bedside: elucidating VEGF(R) inhibitor-related heart failure in cancer treatment. J Transl Med 2025; 23:109. [PMID: 39849527 PMCID: PMC11756202 DOI: 10.1186/s12967-025-06133-x] [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: 10/29/2024] [Accepted: 01/14/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) and VEGF receptor (VEGFR) inhibitors play a pivotal role in treating various tumors; however, the clinical characteristics and molecular mechanisms of their associated heart failure (HF) remain incompletely understood. METHODS We investigated the epidemiological characteristics of VEGF or VEGFR inhibitors [VEGF(R)i]-related heart failure (VirHF) using the global pharmacovigilance database Vigibase. The phenotypic features and molecular mechanisms of VirHF were characterized using VEGF(R)i-treated mouse models through a combination of echocardiography, histopathological analysis, and transcriptome sequencing. Furthermore, we performed a retrospective analysis of cardiac function parameters in patients undergoing VEGF(R)i treatment at local hospitals. RESULTS In the analysis of 1871 VirHF cases, elderly patients (≥ 65 years) and female subjects demonstrated an elevated risk of occurrence. Experimental studies in mice revealed that both acute and chronic VEGF(R)i administration resulted in reduced left ventricular EF, cardiomyocyte hypertrophy, and myocardial fibrosis. Transcriptomic analysis identified significant dysregulation of multiple key signaling pathways, including DNA repair (R = 0.46), mitochondrial ATP synthesis (R = 0.39), glycogen metabolism regulation (R = 0.45), and proteasome-mediated protein degradation (R = 0.45). Moreover, significant upregulation was observed in inflammatory pathways, specifically those involving IL-1, IL-6, TNF-α, and IRF3/IRF7-mediated immune responses. Clinical cohort analyses demonstrated significant elevations in both cardiac injury biomarkers (NT-proBNP, CK-MB, cTnT) and inflammatory mediators (CRP) following VEGF(R)i administration. CONCLUSIONS Our findings present the first comprehensive characterization of VirHF clinical features and elucidate its underlying molecular mechanisms, thereby providing a theoretical framework for optimizing the clinical safety of VEGF(R)i therapy.
Collapse
Affiliation(s)
- Shengkun Peng
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, Sichuan, China
| | - MinHong Cai
- Healthcare-Associated Infection Management Office, Sichuan Academy of Medical Sciences and Sichuan People's Hospital, Chengdu, Sichuan, China
| | - Hongyu Kuang
- Department of Cardiology, University-town Hospital of Chongqing Medical University, Chongqing, China
- Department of Cardiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Qinghua Ma
- Department of Pediatrics, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xiaoqin Dai
- Department of Traditional Chinese Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Yijun Liu
- Department of Outpatient, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| | - Guo Zhang
- Department of Dean's office, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
20
|
Murg SI, Matiș L, Moldovan AF, Schwarz-Madar AF, Trifan DF, Ghitea TC, Popescu MI. Cardiotoxicity in Breast Cancer: Impact of Clinical Classifications and Treatment on Heart Health. Cancers (Basel) 2024; 16:4281. [PMID: 39766180 PMCID: PMC11674351 DOI: 10.3390/cancers16244281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/10/2024] [Accepted: 12/22/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Cardio-oncology has become essential in addressing cardiovascular complications from cancer therapies. While advancements in treatments have improved survival rates, they also increase cardiovascular risks. This study evaluates the cardiotoxic effects of cytostatic treatments, examining the relationship between tumor characteristics, such as histopathology and TNM classification, and cardiovascular complications, aiming to improve cardiotoxicity prevention and management in oncology patients. METHODS We conducted a retrospective analysis of cancer patients treated with anthracyclines, HER2-targeted therapies, and radiotherapy. Cardiac function was monitored via echocardiography, focusing on global longitudinal strain and left ventricular ejection fraction (LVEF). Cardiac troponins and natriuretic peptides were measured to detect subclinical cardiotoxicity, with patients stratified by TNM cancer stage and histopathology. RESULTS Our analysis identified a significant association between certain cytostatic treatments, such as anthracyclines and HER2-targeted therapies, and a reduction in LVEF, particularly in patients with advanced-stage cancer. Biomarker assessments indicated early signs of cardiotoxicity in patients before clinical symptoms emerged. The findings also demonstrated a higher prevalence of cardiovascular complications in patients with pre-existing risk factors. CONCLUSIONS This study highlights the importance of personalized treatment protocols in minimizing cardiotoxicity and improving the quality of life for oncology patients. Regular cardiac monitoring, combined with the use of biomarkers, can help identify high-risk patients early, allowing for timely interventions. Future research should focus on optimizing cardioprotective strategies to mitigate the cardiovascular risks associated with modern cancer therapies. CLINICAL TRIAL REGISTRATION N/A (retrospective study).
Collapse
Affiliation(s)
- Sergiu Ioan Murg
- Doctoral School, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (S.I.M.); (A.F.S.-M.)
| | - Loredana Matiș
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| | - Andrada Florina Moldovan
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| | - Andrada Florina Schwarz-Madar
- Doctoral School, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (S.I.M.); (A.F.S.-M.)
| | - Daniela Florina Trifan
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| | - Timea Claudia Ghitea
- Pharmacy Department, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania
| | - Mircea Ioachim Popescu
- Department of Clinical Discipline, Faculty of Medicine and Pharmacy, University of Oradea, 410068 Oradea, Romania; (L.M.); (A.F.M.); (M.I.P.)
| |
Collapse
|
21
|
Kirichenko YY, Ilgisonis IS, Chernichkina AD, Palienko AV, Buduscheva OI, Pakhtusov NN, Khabarova NV, Belenkov YN. [4‑Year Experience of the Cardio-Oncology Center of Sechenov University: Single-Center Epidemiological Study]. KARDIOLOGIIA 2024; 64:32-39. [PMID: 39526516 DOI: 10.18087/cardio.2024.10.n2768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
AIM To present the four-year experience and the accomplishments of the Scientific and Practical Cardio-Oncology Center of the Sechenov University. MATERIAL AND METHODS The records of patients referred for cardio-oncology consultation from January 2020 through March 2024 were retrospectively analyzed. The patients' cardiovascular (CV) status was assessed at baseline and after optimizing the cardiac therapy during the antitumor treatment. The endpoints were the completion of all antitumor therapy courses and the level of overall and CV mortality. RESULTS Among 233 enrolled patients (66% women), a considerable part belonged to the group of high/very high cardio-oncological risk (n=134, 57%). Various cardiovascular toxicities were observed in 22% of patients. At baseline, these patients significantly more frequently had heart failure and ischemic heart disease as well as previous radiation and chemotherapy. After the optimization of cardiac therapy, 88% of patients successfully completed all scheduled treatments. The overall mortality, including the CV mortality, was 14% (n=7). CONCLUSION Creation of cardio-oncological services allows considerably reducing the probability of adverse CV events during the antitumor therapy and successfully completing all scheduled treatments in most patients..
Collapse
|
22
|
Raisi-Estabragh Z, Murphy AC, Ramalingam S, Scherrer-Crosbie M, Lopez-Fernandez T, Reynolds KL, Aznar M, Lin AE, Libby P, Cordoba R, Bredsen-Masley C, Wechalekar A, Apperley J, Cheng RK, Manisty CH. Cardiovascular Considerations Before Cancer Therapy: Gaps in Evidence and JACC: CardioOncology Expert Panel Recommendations. JACC CardioOncol 2024; 6:631-654. [PMID: 39479317 PMCID: PMC11520216 DOI: 10.1016/j.jaccao.2024.07.017] [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: 09/11/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 11/02/2024] Open
Abstract
Baseline cardiovascular assessment before the initiation of potentially cardiotoxic cancer therapies is a key component of cardio-oncology, aiming to reduce cardiovascular complications and morbidity in patients and survivors. Recent clinical practice guidelines provide both general and cancer therapy-specific recommendations for baseline cardiovascular toxicity risk assessment and management, including the use of dedicated risk scores, cardiovascular imaging, and biomarker testing. However, the value of such interventions in altering disease trajectories has not been established, with many recommendations based on expert opinion or Level of Evidence: C, studies with a potential for high risk of bias. Advances in understanding underlying mechanisms of cardiotoxicity and the increased availability of genetic and immunologic profiling present new opportunities for personalized risk assessment. This paper evaluates the existing evidence on cardiovascular care of cancer patients before cardiotoxic cancer therapy and highlights gaps in evidence and priorities for future research.
Collapse
Affiliation(s)
- Zahra Raisi-Estabragh
- William Harvey Research Institute, National Institute for Health and Care Research Barts Biomedical Research Centre, Queen Mary University of London, London, United Kingdom
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
| | | | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy’s and St. Thomas’ NHS Foundation Trust, London, United Kingdom
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, The Hospital of the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Teresa Lopez-Fernandez
- Hospital Universitario La Paz, Instituto de Investigación La Paz-IdiPaz, Madrid, Spain
- Hospital Universitario Quiron Pozuelo, Madrid, Spain
| | - Kerry L. Reynolds
- Division of Oncology and Hematology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Marianne Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
| | - Amy E. Lin
- Department of Medicine, Division of Cardiology, Section of Cardio-Oncology and Immunology, University of California-San Francisco, San Francisco, California, USA
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Raul Cordoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute Instituto de Investigación Sanitaria de la Fundación Jiménez Díaz, Madrid, Spain
| | - Christine Bredsen-Masley
- Division of Hematology/Oncology, Department of Medicine, St. Michael’s Hospital, University of Toronto, Toronto, Ontario, Canada
| | - Ashu Wechalekar
- Department of Haematology, University College London Hospitals NHS Trust, London, United Kingdom
| | - Jane Apperley
- Department of Clinical Haematology, Hammersmith Hospital, Imperial College Healthcare NHS Trust, London, United Kingdom
- Centre for Haematology, Imperial College London, London, United Kingdom
| | - Richard K. Cheng
- Division of Cardiology, University of Washington Medical Center, Seattle, Washington, USA
| | - Charlotte H. Manisty
- Barts Heart Centre, St Bartholomew’s Hospital, Barts Health NHS Trust, West Smithfield, London, United Kindgom
- Institute of Cardiovascular Science, University College London, London, United Kingdom
| |
Collapse
|
23
|
Hung SK, Lee MS, Chiou WY, Liu DW, Yu CC, Chen LC, Lin RI, Chew CH, Hsu FC, Yang HJ, Chan MWY, Lin HY. Epigenetic modification in radiotherapy and immunotherapy for cancers. Tzu Chi Med J 2024; 36:396-406. [PMID: 39421493 PMCID: PMC11483092 DOI: 10.4103/tcmj.tcmj_3_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 04/20/2024] [Accepted: 06/18/2024] [Indexed: 10/19/2024] Open
Abstract
Radiotherapy (RT) is one of the primary treatment modalities in managing cancer patients. Recently, combined RT and immunotherapy (IT) (i.e., radio-IT [RIT]) have been aggressively investigated in managing cancer patients. However, several issues in conducting RIT are challenging, such as incorporating advanced irradiation techniques, predictive/prognostic biomarkers, and other treatment modalities. Several clinical efforts and novel biomarkers have been introduced and developed to solve these challenges. For example, stereotactic radiosurgery/stereotactic radiotherapy, stereotactic body radiotherapy/stereotactic ablative body radiotherapy, and FLASH-RT have been applied for delivering precise irradiation to lung and liver tumors in conjunction with IT. Besides, several novel IT agents and incorporations of other therapies, such as targeted and thermal therapies, have been further investigated. The present study reviewed the emerging challenges of RIT in modern oncology. We also evaluated clinical practice, bench research, and multimodality treatments. In addition to several clinically applicable biomarkers, we emphasize the roles of advanced irradiation techniques and epigenetic modification as predictive/prognostic biomarkers and potential therapeutic targets. For example, 6(m) A-based epigenetic agents demonstrate the potential to enhance the treatment effects of RIT. However, further prospective randomized trials should be conducted to confirm their roles.
Collapse
Affiliation(s)
- Shih-Kai Hung
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Moon-Sing Lee
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Wen-Yen Chiou
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Dai-Wei Liu
- Department of Radiation Oncology, Hualien Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Hualien, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chih-Chia Yu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Liang-Cheng Chen
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Ru-Inn Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Chia-Hui Chew
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
| | - Feng-Chun Hsu
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Hsuan-Ju Yang
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
| | - Michael W. Y. Chan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| | - Hon-Yi Lin
- Department of Radiation Oncology, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- Cancer Centre, Dalin Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Chiayi, Taiwan
- School of Medicine, Tzu Chi University, Hualien, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
24
|
Kleinbongard P, Andreadou I. Is There a Mitochondrial Protection via Remote Ischemic Conditioning in Settings of Anticancer Therapy Cardiotoxicity? Curr Heart Fail Rep 2024; 21:292-304. [PMID: 38512567 PMCID: PMC11333552 DOI: 10.1007/s11897-024-00658-w] [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] [Accepted: 03/13/2024] [Indexed: 03/23/2024]
Abstract
PURPOSE OF REVIEW To provide an overview of (a) protective effects on mitochondria induced by remote ischemic conditioning (RIC) and (b) mitochondrial damage caused by anticancer therapy. We then discuss the available results of studies on mitochondrial protection via RIC in anticancer therapy-induced cardiotoxicity. RECENT FINDINGS In three experimental studies in healthy mice and pigs, there was a RIC-mediated protection against anthracycline-induced cardiotoxicity and there was some evidence of improved mitochondrial function with RIC. The RIC-mediated protection was not confirmed in the two available studies in cancer patients. In adult cancer patients, RIC was associated with an adverse outcome. There are no data on mitochondrial function in cancer patients. Studies in tumor-bearing animals are needed to determine whether RIC does not interfere with the anticancer properties of the drugs and whether RIC actually improves mitochondrial function, ultimately resulting in improved cardiac function.
Collapse
Affiliation(s)
- Petra Kleinbongard
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
25
|
Jesus EB, Cavalcanti ACD, Oliveira JA. Nursing interventions for adult patients undergoing cancer cardiotoxic therapy: Scoping review. Int J Nurs Knowl 2024; 35:239-255. [PMID: 37459404 DOI: 10.1111/2047-3095.12435] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Accepted: 06/09/2023] [Indexed: 07/16/2024]
Abstract
PURPOSE To identify nursing interventions for the management of adult patients undergoing cardiotoxic oncologic therapy. METHODS This scoping review was performed in accordance with the JBI guidelines. The literature search took place between July and August 2022. Studies examining interventions for the management of adult cancer patients undergoing cardiotoxic therapy were included. The characteristics and results of the studies were synthesized and analyzed in a narrative way. FINDINGS In the nine included studies, it was verified that the interventions were implemented to guide the actions of the health team in general rather than specifically nursing staff. Nine nursing interventions related to the Classification of Nursing Interventions were included. CONCLUSIONS The nursing interventions identified focused on rigorous cardiovascular surveillance, risk assessment, and actions to identify and mitigate cardiotoxicity. IMPLICATIONS FOR NURSING PRACTICE It is believed that the implementation of the identified nursing interventions will lead to evidence-based nursing practice and will contribute to the development of care products and processes that assess the cardiological risks and cardiotoxicity.
Collapse
Affiliation(s)
- Evelyn Barcelos Jesus
- Professional Master's Degree in Nursing, Aurora de Afonso Costa Nursing School, Fluminense Federal University, Rio de Janeiro, Brazil
| | | | | |
Collapse
|
26
|
Voronkov NS, Maslov LN, Vyshlov EV, Mukhomedzyanov AV, Ryabov VV, Derkachev IA, Kan A, Gusakova SV, Gombozhapova AE, Panteleev OO. Do platelets protect the heart against ischemia/reperfusion injury or exacerbate cardiac ischemia/reperfusion injury? The role of PDGF, VEGF, and PAF. Life Sci 2024; 347:122617. [PMID: 38608835 DOI: 10.1016/j.lfs.2024.122617] [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: 01/25/2024] [Revised: 03/15/2024] [Accepted: 04/05/2024] [Indexed: 04/14/2024]
Abstract
BACKGROUND Acute myocardial infarction (AMI) is one of the main causes of death. It is quite obvious that there is an urgent need to develop new approaches for treatment of AMI. OBJECTIVE This review analyzes data on the role of platelets in the regulation of cardiac tolerance to ischemia/reperfusion (I/R). METHODS It was performed a search of topical articles using PubMed databases. FINDINGS Platelets activated by a cholesterol-enriched diet, thrombin, and myocardial ischemia exacerbate I/R injury of the heart. The P2Y12 receptor antagonists, remote ischemic postconditioning and conditioning alter the properties of platelets. Platelets acquire the ability to increase cardiac tolerance to I/R. Platelet-derived growth factors (PDGFs) increase tolerance of cardiomyocytes and endothelial cells to I/R. PDGF receptors (PDGFRs) were found in cardiomyocytes and endothelial cells. PDGFs decrease infarct size and partially abrogate adverse postinfarction remodeling. Protein kinase C, phosphoinositide 3-kinase, and Akt involved in the cytoprotective effect of PDGFs. Vascular endothelial growth factor increased cardiac tolerance to I/R and alleviated adverse postinfarction remodeling. The platelet-activating factor (PAF) receptor inhibitors increase cardiac tolerance to I/R in vivo. PAF enhances cardiac tolerance to I/R in vitro. It is possible that PAF receptor inhibitors could protect the heart by blocking PAF receptor localized outside the heart. PAF protects the heart through activation of PAF receptor localized in cardiomyocytes or endothelial cells. Reactive oxygen species and kinases are involved in the cardioprotective effect of PAF. CONCLUSION Platelets play an important role in the regulation of cardiac tolerance to I/R.
Collapse
Affiliation(s)
- Nikita S Voronkov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Leonid N Maslov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia.
| | - Evgeniy V Vyshlov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Alexander V Mukhomedzyanov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Vyacheslav V Ryabov
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Ivan A Derkachev
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Artur Kan
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Svetlana V Gusakova
- Department of Biophysics and Functional Diagnostics, Siberian State Medical University, 634050 Tomsk, Russia
| | - Alexandra E Gombozhapova
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| | - Oleg O Panteleev
- Department of Emergency Cardiology and Laboratory of Experimental Cardiology, Cardiology Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, 634012 Tomsk, Russia
| |
Collapse
|
27
|
Wernhart S, Rassaf T. Relevance of Cardiovascular Exercise in Cancer and Cancer Therapy-Related Cardiac Dysfunction. Curr Heart Fail Rep 2024; 21:238-251. [PMID: 38696059 PMCID: PMC11090948 DOI: 10.1007/s11897-024-00662-0] [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] [Accepted: 04/03/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE OF THE REVIEW Cancer therapy-related cardiac dysfunction (CTRCD) has been identified as a threat to overall and cancer-related survival. Although aerobic exercise training (AET) has been shown to improve cardiorespiratory fitness (CRF), the relationship between specific exercise regimens and cancer survival, heart failure development, and reduction of CTRCD is unclear. In this review, we discuss the impact of AET on molecular pathways and the current literature of sports in the field of cardio-oncology. RECENT FINDINGS Cardio-oncological exercise trials have focused on variations of AET intensity by using moderate continuous and high intensity interval training, which are applicable, safe, and effective approaches to improve CRF. AET increases CRF, reduces cardiovascular morbidity and heart failure hospitalization and should thus be implemented as an adjunct to standard cancer therapy, although its long-term effect on CTRCD remains unknown. Despite modulating diverse molecular pathways, it remains unknown which exercise regimen, including variations of AET duration and frequency, is most suited to facilitate peripheral and central adaptations to exercise and improve survival in cancer patients.
Collapse
Affiliation(s)
- Simon Wernhart
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
28
|
Wu Y, Li B, Yu X, Liu Y, Chui R, Sun K, Geng D, Ma L. Histone deacetylase 6 as a novel promising target to treat cardiovascular disease. CANCER INNOVATION 2024; 3:e114. [PMID: 38947757 PMCID: PMC11212282 DOI: 10.1002/cai2.114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 12/31/2023] [Accepted: 01/08/2024] [Indexed: 07/02/2024]
Abstract
Histone deacetylase 6 (HDAC6) belongs to a class of epigenetic targets that have been found to be a key protein in the association between tumors and cardiovascular disease. Recent studies have focused on the crucial role of HDAC6 in regulating cardiovascular diseases such as atherosclerosis, myocardial infarction, myocardial hypertrophy, myocardial fibrosis, hypertension, pulmonary hypertension, and arrhythmia. Here, we review the association between HDAC6 and cardiovascular disease, the research progress of HDAC6 inhibitors in the treatment of cardiovascular disease, and discuss the feasibility of combining HDAC6 inhibitors with other therapeutic agents to treat cardiovascular disease.
Collapse
Affiliation(s)
- Ya‐Xi Wu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical ScienceZhengzhou UniversityZhengzhouHenanChina
| | - Bing‐Qian Li
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical ScienceZhengzhou UniversityZhengzhouHenanChina
| | - Xiao‐Qian Yu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical ScienceZhengzhou UniversityZhengzhouHenanChina
| | - Yu‐Lin Liu
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical ScienceZhengzhou UniversityZhengzhouHenanChina
| | - Rui‐Hao Chui
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical ScienceZhengzhou UniversityZhengzhouHenanChina
| | - Kai Sun
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical ScienceZhengzhou UniversityZhengzhouHenanChina
| | - Dian‐Guang Geng
- Key Laboratory of Cardio‐Cerebrovascular Drugs'China Meheco Topfond Pharmaceutical Co.ZhumadianHenanChina
| | - Li‐Ying Ma
- State Key Laboratory of Esophageal Cancer Prevention and Treatment, Key Laboratory of Advanced Pharmaceutical Technology, Ministry of Education of China, School of Pharmaceutical Science and Institute of Pharmaceutical ScienceZhengzhou UniversityZhengzhouHenanChina
- Key Laboratory of Cardio‐Cerebrovascular Drugs'China Meheco Topfond Pharmaceutical Co.ZhumadianHenanChina
| |
Collapse
|
29
|
Kirichenko YY, Kulagina TY, Zhigulina OA, Ilgisonis IS, Belenkov YN. [Cardiac Dysfunction and Arterial Hypertension as Manifestations of Cardiovasculotoxicity of iVEGF-Containing Chemotherapy. Clinical Case]. KARDIOLOGIIA 2024; 64:33-38. [PMID: 38841787 DOI: 10.18087/cardio.2024.5.n2661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/11/2024] [Indexed: 06/07/2024]
Abstract
Significant advances in timely diagnosis and modern antitumor therapy have led to a considerable increase in the survival rate of cancer patients. On the other hand, the incidence of cardiovascular (CV) diseases and their complications is increasingly growing, including due to side effects of anticancer drugs. CV complications are the most common cause of non-oncological death of cancer patients. The development of polychemotherapy-induced arterial hypertension (AH) is closely associated with the use of certain groups of drugs, for example, inhibitors of vascular endothelial growth factor (iVEGF). Such AH is generally dose-dependent and reversible after interruption or termination of treatment. However, systemic AH, regardless of its genesis, is one of the key risk factors for many CV events (myocardial infarction, stroke, heart failure, arrhythmias) and kidney disease. Therefore, thorough blood pressure monitoring and its timely and adequate correction if needed are indicated when using certain groups of chemotherapy drugs. This article describes a clinical follow-up of a patient with induced AH associated with the iVEGF antitumor therapy for advanced uterine cancer with a rapid development of left ventricular myocardial dysfunction.
Collapse
Affiliation(s)
| | - T Yu Kulagina
- Academician Petrovsky Russian Research Center of Surgery
| | - O A Zhigulina
- Academician Petrovsky Russian Research Center of Surgery
| | | | | |
Collapse
|
30
|
Juthani R, Punatar S, Mittra I. New light on chemotherapy toxicity and its prevention. BJC REPORTS 2024; 2:41. [PMID: 39516565 PMCID: PMC11524128 DOI: 10.1038/s44276-024-00064-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 11/16/2024]
Abstract
Most patients with cancer receive chemotherapy. Unfortunately, chemotherapy is associated with a number of potentially life-threatening side effects. There is a need to ameliorate chemotoxicity to improve therapeutic outcomes and quality of life. Chemotoxicity arises from systemic DNA damage and inflammation in healthy cells due to chemotherapy drugs. Traditionally, these processes are believed to be caused by the direct death of normal cells by chemotherapeutic drugs. However, new research has challenged this dogma by suggesting that chemotoxicity is a secondary effect associated with the release of cell-free chromatin particles (cfChPs) from cells subjected to drug-induced death. Released cfChPs can freely enter into bystander healthy cells to inflict double-strand (dsDNA) breaks and activate inflammatory and apoptotic pathways. The drug-induced cell death and cfChPs release have cascading effects that exaggerate and prolong chemotoxicity. Furthermore, evidence has emerged from laboratory and preclinical studies, and two phase II clinical trials, indicating that chemotoxicity can be minimised by deactivating cfChPs. Three cfChPs-deactivating agents have been identified, of which the nutraceutical combination resveratrol and copper (R-Cu)-easily administered orally and with little toxicity-is the agent of choice for human therapeutic use. This article aims to provide practising medical oncologists with a perspective on this emerging research on chemotoxicity and its prevention and its potential implications for the future. Well-designed randomised clinical trials will be necessary to establish the true clinical value of these findings in day-to-day practice.
Collapse
Affiliation(s)
- Ronit Juthani
- Translational Research Laboratory, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
| | - Sachin Punatar
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India
- Department of Medical Oncology, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India
| | - Indraneel Mittra
- Translational Research Laboratory, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Kharghar, Navi Mumbai, 410210, India.
- Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400094, India.
| |
Collapse
|
31
|
Agnihotri A, Ramasubbu SK, Bandyopadhyay A, Bidarolli M, Nath UK, Das B. Prevalence, Attributes, and Risk Factors of QT-Interval-Prolonging Drugs and Potential Drug-Drug Interactions in Cancer Patients: A Prospective Study in a Tertiary Care Hospital. Cureus 2024; 16:e60492. [PMID: 38882995 PMCID: PMC11180424 DOI: 10.7759/cureus.60492] [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: 05/17/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction Cancer chemotherapy regimens include multiple classes of adjuvant drugs as supportive therapy. Because of the concurrent intake of other drugs (like antiemetics, antidepressants, analgesics, and antimicrobials), there is a heightened risk for possible QT interval prolongation. There is a dearth of evidence in the literature regarding the usage of QT-prolonging anticancer drugs and associated risk factors that have the propensity to prolong QT interval. The purpose was to explore the extent of the use of QT-interval-prolonging drugs and potential QT-prolonging drug-drug interactions (QT-DDIs) in cancer patients attending OPD in a tertiary-care hospital. Methods This was a hospital-based, cross-sectional, observational study. Risk stratification of QT-prolonging drugs for torsades de pointes (TdP) was done by the Arizona Center for Education and Research on Therapeutics (AzCERT)/CredibleMeds-lists, and potential QT-DDIs were determined with four online DDI-checker-software. Results In 1331 cancer patients, the overall prevalence of potential QT-prolonging drug utilization was 97.3%. Ondansetron, pantoprazole, domperidone, and olanzapine were the most frequent QT-prolonging drugs in cancer patients. The top six antineoplastics with potential QT-prolonging and torsadogenic actions were capecitabine, oxaliplatin, imatinib, bortezomib, 5-fluorouracil, and bendamustine. Evidence-based pragmatic QTc interval prolongation risk assessment tools are imperative for cancer patients. Conclusion This study revealed a high prevalence of QT-prolonging drugs and QT-DDIs among cancer patients who are treated with anticancer and non-anticancer drugs. As a result, it's critical to take precautions, stay vigilant, and avoid QT-prolonging in clinical situations. Evidence-based pragmatic QTc interval prolongation risk assessment tools are needed for cancer patients.
Collapse
Affiliation(s)
- Akash Agnihotri
- Department of Pharmacology, Amrita School of Medicine, Faridabad, IND
| | - Saravana Kumar Ramasubbu
- Department of Pharmacology, Andaman and Nicobar Islands Institute of Medical Sciences, Port Blair, IND
| | - Arkapal Bandyopadhyay
- Department of Pharmacology, All India Institute of Medical Sciences, Kalyani, Kalyani, IND
| | - Manjunath Bidarolli
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Uttam Kumar Nath
- Department of Medical Oncology and Hematology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| | - Biswadeep Das
- Department of Pharmacology, All India Institute of Medical Sciences, Rishikesh, Rishikesh, IND
| |
Collapse
|
32
|
Lopez-Garcia A, Macia E, Gomez-Talavera S, Castillo E, Morillo D, Tuñon J, Ibañez B, Cordoba R. Predictive Factors of Therapy-Related Cardiovascular Events in Patients with Lymphoma Receiving Anthracyclines. Med Sci (Basel) 2024; 12:23. [PMID: 38804379 PMCID: PMC11130872 DOI: 10.3390/medsci12020023] [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: 03/03/2024] [Revised: 04/11/2024] [Accepted: 04/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Cancer-therapy-related cardiac dysfunction (CTRCD) is a growing concern for public health, with a growing incidence due to improved survival rates of patients with hematological malignancies due to diagnostic and therapeutic advances. The identification of patients at risk for CTRCD is vital to developing preventive strategies. METHODS A single-center retrospective cohort study was conducted between 1 January 2017 and 15 February 2023. Medical records of patients with lymphoma treated with first-line anthracyclines were reviewed. Demographic data, cardiovascular risk factors, biomarkers of myocardial damage, and echocardiographic information were collected. RESULTS A total of 200 patients were included. The incidence of CTRCD was 17.4% (35/200). Patients with CTRCD were older than those without CTRCD, with a mean age of 65.17 years vs. 56.77 (p = 0.008). Dyslipidemia (DL) (31.4% vs. 13.4% p = 0.017) and previous cardiovascular disease (40% vs. 13.3%; p < 0.001) were more frequent in the group who developed an event. Mean baseline NT-proBNP levels in the subgroup with cardiovascular events were 388.73 kg/L ± 101.02, and they were 251.518 kg/L ± 26.22 in those who did not (p = 0.004). Differences in Troponin I levels were identified during and after treatment without exceeding the laboratory's upper reference limit. Patients were followed for a median of 51.83 months (0.76-73.49). The presence of a CTCRD event had a negative impact on overall mortality from any cause (HR = 2.23 (95% CI: 1.08-2.93); p = 0.031). CONCLUSIONS Early identification of risk factors is crucial to manage patients at risk for CTRCD.
Collapse
Affiliation(s)
- Alberto Lopez-Garcia
- Lymphoma Unit, Department of Hematology, Fundación Jiménez Díaz University Hospital, IIS-FJD Health Research Institute, Avenida Reyes Catolicos, 228040 Madrid, Spain
| | - Ester Macia
- Cardio-Oncology Unit, Department of Cardiology, Fundacion Jimenez Diaz University Hospital, Avenida Reyes Catolicos, 228040 Madrid, Spain
| | - Sandra Gomez-Talavera
- Cardio-Oncology Unit, Department of Cardiology, Fundacion Jimenez Diaz University Hospital, Avenida Reyes Catolicos, 228040 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Eva Castillo
- Department of Pharmacy, Fundación Jimenez Diaz University Hospital, Avenida Reyes Católicos, 228040 Madrid, Spain
| | - Daniel Morillo
- Lymphoma Unit, Department of Hematology, Fundación Jiménez Díaz University Hospital, IIS-FJD Health Research Institute, Avenida Reyes Catolicos, 228040 Madrid, Spain
| | - Jose Tuñon
- Cardio-Oncology Unit, Department of Cardiology, Fundacion Jimenez Diaz University Hospital, Avenida Reyes Catolicos, 228040 Madrid, Spain
| | - Borja Ibañez
- Cardio-Oncology Unit, Department of Cardiology, Fundacion Jimenez Diaz University Hospital, Avenida Reyes Catolicos, 228040 Madrid, Spain
- Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), 28029 Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), 28029 Madrid, Spain
| | - Raul Cordoba
- Lymphoma Unit, Department of Hematology, Fundación Jiménez Díaz University Hospital, IIS-FJD Health Research Institute, Avenida Reyes Catolicos, 228040 Madrid, Spain
| |
Collapse
|
33
|
Michel L, Rassaf T. [Heart and cancer: cardio-oncology]. Herz 2024; 49:111-117. [PMID: 38289420 DOI: 10.1007/s00059-024-05232-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 03/07/2024]
Abstract
Cardiovascular diseases and cancer have a complex relationship and show a reciprocal linkage and influence. Mutual risk factors, demographic changes and increasing multimorbidity result in an increase in the incidence of both diseases. Advances in oncological and cardiological treatment lead to a further increase in patients with cured or chronic diseases as a relevant comorbidity. The induction of cardiovascular side effects by cancer therapies leads to increased cardiovascular morbidity and mortality in cancer patients. Recent data also show that cardiovascular disease, through various factors, can also promote the development and progression of cancer. An understanding of the interrelationship between cardiovascular diseases and cancer can be seen as a major medical challenge for the future. To this end, scientific, structural, clinical and educational interfaces between cardiology and oncology are essential. This article outlines the complex relationships between cardiovascular diseases and cancer and defines current and future challenges for the best possible care of affected patients.
Collapse
Affiliation(s)
- Lars Michel
- Klinik für Kardiologie und Angiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Tienush Rassaf
- Klinik für Kardiologie und Angiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Hufelandstraße 55, 45147, Essen, Deutschland.
| |
Collapse
|
34
|
Michel L, Rassaf T. [Update cardio-oncology : Immune checkpoint inhibitor therapy]. Herz 2024; 49:81-90. [PMID: 38175285 DOI: 10.1007/s00059-023-05228-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/22/2023] [Indexed: 01/05/2024]
Abstract
Cardiovascular diseases and cancer are the most common causes of death in Germany. Cancer treatment can lead to significant cardiovascular side effects and thus form a link between the two disease groups. The focus of cardio-oncology is on the best possible prevention, diagnostics and treatment of cardiovascular complications caused by cancer treatment. It is crucial for cardio-oncology to adapt to the continuous development of new forms of oncological treatment with previously unknown cardiovascular side effects. One such new form of treatment is immune checkpoint inhibitor (ICI) therapy, which is regarded as the most important oncological milestone of the last decade due to its excellent oncological efficacy; however, the growing use has revealed a high risk of diverse cardiovascular side effects with high morbidity and mortality, so that cardio-oncological care of affected patients is of particular importance. This review summarizes the current scientific and clinical state of the pathophysiology, incidence, diagnosis and treatment of cardiovascular side effects of ICI therapy.
Collapse
Affiliation(s)
- Lars Michel
- Klinik für Kardiologie und Angiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland
| | - Tienush Rassaf
- Klinik für Kardiologie und Angiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Hufelandstr. 55, 45147, Essen, Deutschland.
| |
Collapse
|
35
|
Gong B, Li Y, Guo Y, Wang J, Liu W, Zhou G, Song J, Pan F, Yang L, Liang B. The impact of pulmonary artery to ascending aorta diameter ratio progression on the prognosis of NSCLC patients treated with immune checkpoint inhibitors. Front Immunol 2024; 15:1302233. [PMID: 38348049 PMCID: PMC10859503 DOI: 10.3389/fimmu.2024.1302233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 01/15/2024] [Indexed: 02/15/2024] Open
Abstract
Background Immunotherapy, represented by immune checkpoint inhibitors (ICIs), is a major breakthrough in cancer treatment. Studies have reported that the use of ICIs is associated with an increase in the pulmonary artery to ascending aorta diameter (PAD/AoD) ratio. However, the impact of PAD/AoD ratio progression on the prognosis of patients is unclear. Methods This retrospective cohort study included patients with stage III or IV non-small cell lung cancer (NSCLC) treated with ICIs at the Wuhan Union Hospital between March 1, 2020, and September 1, 2022. The baseline and post-treatment PAD/AoD ratios of patients were evaluated through chest CT scans. The primary outcome of this study was overall survival (OS), while the secondary outcomes included progression-free survival (PFS), objective response rate (ORR) and disease control rate (DCR). Results The PAD/AoD ratio increased after the initiation of ICIs (from 0.75 to 0.78; P < 0.001). A total of 441 patients were divided into severe group (n=221) and non-severe group (n=220) according to the median increase of PAD/AoD ratio (1.06). Compared with the non-severe group, the severe group had a lower DCR (87.8% vs. 96.0%, P = 0.005) and ORR (87.5% vs. 96.0%, P = 0.063). Over the entire duration of follow-up (median 22.0 months), 85 (38.5%) patients in the severe group and 30 (7.3%) patients in the non-severe group died. An increased PAD/AoD ratio was associated with shorter PFS (Hazard ratio (HR): 1.48 [95% CI, 1.14 to 1.93]; P = 0.003) and OS (HR: 3.50 [95% CI, 2.30 to 5.30]; P < 0.001). Similar results were obtained across subgroups. Conclusions ICI treatment exacerbates an increase in the PAD/AoD ratio in patients with cancer, and greater increase in the PAD/AoD ratio was associated with a worse prognosis. PAD/AoD ratio could be a biomarker to stratify prognosis of NSCLC patients treated with ICIs.
Collapse
Affiliation(s)
- Bingxin Gong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yi Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Jing Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Weiwei Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Guofeng Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Jiyu Song
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Feng Pan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| | - Bo Liang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, China
| |
Collapse
|
36
|
Moisuc DC, Constantinescu D, Marinca MV, Gafton B, Pavel-Tanasa M, Cianga P. Cyclophilin A: An Independent Prognostic Factor for Survival in Patients with Metastatic Colorectal Cancer Treated with Bevacizumab and Chemotherapy. Cancers (Basel) 2024; 16:385. [PMID: 38254874 PMCID: PMC10814009 DOI: 10.3390/cancers16020385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 01/11/2024] [Accepted: 01/14/2024] [Indexed: 01/24/2024] Open
Abstract
Colorectal cancer (CRC) ranks as second most common cause of cancer-related deaths. The CRC management considerably improved in recent years, especially due to biological therapies such as bevacizumab. The lack of predictive or prognostic biomarkers remains one of the major disadvantages of using bevacizumab in the CRC management. We performed a prospective study to analyze the prognostic and predictive roles of three potential serum biomarkers (Cyclophilin A (CypA), copeptin and Tie2) investigated by ELISA in 56 patients with metastatic CRC undergoing bevacizumab and chemotherapy between May 2019 and September 2021 at baseline and after one and six months of therapy. We showed that low levels of CypA at baseline and after one month of treatment were associated with better overall survival (OS) (42 versus 24 months, p = 0.029 at baseline; 42 versus 25 months, p = 0.039 after one month). For copeptin and Tie2, Kaplan-Meier curves showed no correlation between these biomarkers and OS or progression-free survival. When adjusting for baseline and post-treatment factors, a multivariate Cox analysis showed that low values of CypA at baseline and after one month of treatment were independent prognostic factors for OS and correlated with a better prognosis in metastatic CRC patients.
Collapse
Affiliation(s)
- Diana Cornelia Moisuc
- Immunology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.C.M.); (D.C.)
| | - Daniela Constantinescu
- Immunology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.C.M.); (D.C.)
- Immunology Department, “St. Spiridon” Hospital, 700111 Iasi, Romania
| | - Mihai Vasile Marinca
- Oncology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.V.M.); (B.G.)
- Oncology Department, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Bogdan Gafton
- Oncology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (M.V.M.); (B.G.)
- Oncology Department, Regional Institute of Oncology, 700483 Iasi, Romania
| | - Mariana Pavel-Tanasa
- Immunology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.C.M.); (D.C.)
- Immunology Department, “St. Spiridon” Hospital, 700111 Iasi, Romania
| | - Petru Cianga
- Immunology Department, “Grigore T. Popa” University of Medicine and Pharmacy, 700115 Iasi, Romania; (D.C.M.); (D.C.)
- Immunology Department, “St. Spiridon” Hospital, 700111 Iasi, Romania
| |
Collapse
|
37
|
Katolkar UN, Surana SJ. Exploring the Potential Role of Phytopharmaceuticals in Alleviating Toxicities of Chemotherapeutic Agents. Curr Protein Pept Sci 2024; 25:753-779. [PMID: 38919003 DOI: 10.2174/0113892037307940240606075208] [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: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 06/27/2024]
Abstract
BACKGROUND Chemotherapy is the mainstay of cancer treatment, bringing patients optimism about recurrence and survival. However, the clinical effectiveness of chemotherapeutic drugs is frequently jeopardized by their intrinsic toxicity, resulting in side effects affecting the quality of life of cancer patients. This analysis explores the ethnopharmacological impact of phytopharmaceuticals, highlighting their traditional use in many cultures. The present study, which takes its cues from indigenous knowledge, aims to close the knowledge gap between traditional medicine and modern medicine in reducing the toxicities of chemotherapy treatments. AIM The present in-depth study aims to highlight the current research and upcoming developments in phytopharmaceuticals for reducing the toxicity of chemotherapeutic drugs. Further, we address the mechanisms through which phytopharmaceuticals may reduce chemotherapy-induced side effects that include nausea, vomiting, myelosuppression, nephropathy, neuropathy, and cardiotoxicity using data from a variety of preclinical and clinical investigations. MATERIALS AND METHODS The literature search was carried out by employing search engines such as PubMed and Google Scholar with keywords such as cancer, chemotherapy, CNS toxicity, hematopoietic toxicity, renal toxicity, GI toxicity, CNS toxicity, and phytopharmaceuticals. RESULTS Bioactive chemicals found in plants, such as fruits, vegetables, herbs, and spices, are being studied for their capacity to improve the safety and acceptability of chemotherapy regimens. The current review also dives into the investigation of phytopharmaceuticals as adjuvant medicines in cancer treatment, which is a viable path for addressing the pressing need to lessen chemotherapy-induced toxicities. CONCLUSION The present review revealed that the potential of phytopharmaceuticals in alleviating chemotherapeutic drug toxicities would pave the way for better cancer treatment and patient outcomes, harmonizing with the larger trend towards personalized and holistic approaches to chemotherapy.
Collapse
Affiliation(s)
- Ujwal N Katolkar
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| | - Sanjay J Surana
- Department of Pharmacology, R.C. Patel Institute of Pharmaceutical Education and Research, Karwand Naka, Shirpur Dist. Dhule Maharashtra 425405, India
| |
Collapse
|
38
|
Jali AM, Alam MF, Hanbashi A, Mawkili W, Abdlasaed BM, Alshahrani S, Qahl AM, Alrashah ASS, Shahi HA. Sesamin's Therapeutic Actions on Cyclophosphamide-Induced Hepatotoxicity, Molecular Mechanisms, and Histopathological Characteristics. Biomedicines 2023; 11:3238. [PMID: 38137459 PMCID: PMC10741447 DOI: 10.3390/biomedicines11123238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 11/22/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Cyclophosphamide, an alkylating agent integral to specific cancer chemotherapy protocols, is often curtailed in application owing to its significant hepatotoxic side effects. Therefore, this study was conducted to assess the hepatoprotective potential of sesamin, a plant-originated antioxidant, using rat models. The rats were divided into five groups: a control group received only the vehicle for six days; a cyclophosphamide group received an intraperitoneal (i.p.) single injection of cyclophosphamide (150 mg/kg) on day four; a sesamin group received a daily high oral dose (20 mg/kg) of sesamin for six days; and two groups were pretreated with oral sesamin (10 and 20 mg/kg daily from day one to day six) followed by an i.p. injection of cyclophosphamide on day four. The final and last sesamin dose was administered 24 h before euthanasia. At the end of the experiment, blood and liver tissue were collected for biochemical and histopathological assessments. The results indicated significantly increased liver markers (AST, ALT, ALP, and BIL), cytokines (TNFα and IL-1β), caspase-3, and malondialdehyde (MDA) in the cyclophosphamide group as compared to the normal control. Additionally, there was a significant decline in antioxidants (GSH) and antioxidant enzymes (CAT and SOD), but the sesamin treatment reduced liver marker enzymes, cytokines, and caspase-3 and improved antioxidants and antioxidant enzymes. Thus, sesamin effectively countered these alterations and helped to normalize the histopathological alterations. In conclusion, sesamin demonstrated the potential for attenuating cyclophosphamide-induced hepatotoxicity by modulating cytokine networks, apoptotic pathways, and oxidative stress, suggesting its potential role as an adjunct in chemotherapy to reduce hepatotoxicity.
Collapse
Affiliation(s)
- Abdulmajeed M. Jali
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Mohammad Firoz Alam
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Ali Hanbashi
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX1 3QT, UK
| | - Wedad Mawkili
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Basher M. Abdlasaed
- Department of Biology, Faculty of Education, Alasmaray Islamic University, Zliten 218521, Libya;
| | - Saeed Alshahrani
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| | - Abdullah M. Qahl
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
- Pharmacy Department, Jazan University Hospital, Jazan University, Jazan 45142, Saudi Arabia
| | - Ahmad S. S. Alrashah
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
- Pharmacy Administration, Ministry of Health, Health Affairs General Directorate, Najran 66251, Saudi Arabia
| | - Hamad Al Shahi
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia; (A.M.J.); (A.H.); (W.M.); (S.A.); (A.M.Q.); (A.S.S.A.); (H.A.S.)
| |
Collapse
|
39
|
Kanno SI, Hara A. Everolimus prevents doxorubicin-induced apoptosis in H9c2 cardiomyocytes but not in MCF-7 cancer cells: Cardioprotective roles of autophagy, mitophagy, and AKT. Toxicol In Vitro 2023; 93:105698. [PMID: 37739323 DOI: 10.1016/j.tiv.2023.105698] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/11/2023] [Accepted: 09/14/2023] [Indexed: 09/24/2023]
Abstract
Cardiotoxicity is a severe side effect of the chemotherapeutic agent doxorubicin (DOX). We recently showed that DOX-induced cardiomyocyte apoptosis and death were attenuated through autophagy pre-induction. Herein, we assessed how the autophagy/mitophagy-inducing antitumor drug everolimus (EVL) affected DOX-induced cytotoxicity in the rat cardiomyocyte cell line H9c2 and human breast cancer cell line MCF-7. Apoptosis was assessed using annexin V assay. Autophagy and mitophagy were assessed using fluorescence assays. Cellular protein levels were determined using western blotting. Pretreatment with EVL (1 nM) before DOX exposure inhibited mammalian target of rapamycin (mTOR) activity, induced autophagy and mitophagy, and activated protein kinase B (AKT) in H9c2 cells. In mitochondria, DOX (1 μM) induced structural damage (decreased membrane potential and release of cytochrome c), increased superoxide levels, decreased apoptosis inhibitor Bcl-2, and increased apoptosis inducer Bax, leading to apoptosis and reduced viability in H9c2 cells. EVL pretreatment suppressed DOX-induced changes. EVL anti-apoptotic effects were inhibited by treatment with MK-2206, a selective AKT inhibitor. Furthermore, EVL suppressed DOX-induced cardiotoxicity through autophagy/mitophagy and AKT activation but did not attenuate DOX-induced apoptosis or reduction in viability in MCF-7 cells. Altogether, EVL can protect cardiomyocytes from DOX-induced apoptosis and toxicity without reducing DOX antitumor effects, allowing safer chemotherapy.
Collapse
Affiliation(s)
- Syu-Ichi Kanno
- Department of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan.
| | - Akiyoshi Hara
- Department of Clinical Pharmacotherapeutics, Tohoku Medical and Pharmaceutical University, 4-4-1 Komatsushima, Aoba-ku, Sendai 981-8558, Japan
| |
Collapse
|
40
|
Heusch G, Andreadou I, Bell R, Bertero E, Botker HE, Davidson SM, Downey J, Eaton P, Ferdinandy P, Gersh BJ, Giacca M, Hausenloy DJ, Ibanez B, Krieg T, Maack C, Schulz R, Sellke F, Shah AM, Thiele H, Yellon DM, Di Lisa F. Health position paper and redox perspectives on reactive oxygen species as signals and targets of cardioprotection. Redox Biol 2023; 67:102894. [PMID: 37839355 PMCID: PMC10590874 DOI: 10.1016/j.redox.2023.102894] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 09/04/2023] [Accepted: 09/15/2023] [Indexed: 10/17/2023] Open
Abstract
The present review summarizes the beneficial and detrimental roles of reactive oxygen species in myocardial ischemia/reperfusion injury and cardioprotection. In the first part, the continued need for cardioprotection beyond that by rapid reperfusion of acute myocardial infarction is emphasized. Then, pathomechanisms of myocardial ischemia/reperfusion to the myocardium and the coronary circulation and the different modes of cell death in myocardial infarction are characterized. Different mechanical and pharmacological interventions to protect the ischemic/reperfused myocardium in elective percutaneous coronary interventions and coronary artery bypass grafting, in acute myocardial infarction and in cardiotoxicity from cancer therapy are detailed. The second part keeps the focus on ROS providing a comprehensive overview of molecular and cellular mechanisms involved in ischemia/reperfusion injury. Starting from mitochondria as the main sources and targets of ROS in ischemic/reperfused myocardium, a complex network of cellular and extracellular processes is discussed, including relationships with Ca2+ homeostasis, thiol group redox balance, hydrogen sulfide modulation, cross-talk with NAPDH oxidases, exosomes, cytokines and growth factors. While mechanistic insights are needed to improve our current therapeutic approaches, advancements in knowledge of ROS-mediated processes indicate that detrimental facets of oxidative stress are opposed by ROS requirement for physiological and protective reactions. This inevitable contrast is likely to underlie unsuccessful clinical trials and limits the development of novel cardioprotective interventions simply based upon ROS removal.
Collapse
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Duisburg-Essen, Essen, Germany.
| | - Ioanna Andreadou
- Laboratory of Pharmacology, Faculty of Pharmacy, National and Kapodistrian University of Athens, Athens, Greece
| | - Robert Bell
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Edoardo Bertero
- Chair of Cardiovascular Disease, Department of Internal Medicine and Specialties, University of Genova, Genova, Italy
| | - Hans-Erik Botker
- Department of Cardiology, Institute for Clinical Medicine, Aarhus University, Aarhus N, Denmark
| | - Sean M Davidson
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - James Downey
- Department of Physiology, University of South Alabama, Mobile, AL, USA
| | - Philip Eaton
- William Harvey Research Institute, Queen Mary University of London, Heart Centre, Charterhouse Square, London, United Kingdom
| | - Peter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Semmelweis University, Budapest, Hungary; Pharmahungary Group, Szeged, Hungary
| | - Bernard J Gersh
- Department of Cardiovascular Medicine, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Mauro Giacca
- School of Cardiovascular and Metabolic Medicine & Sciences, King's College, London, United Kingdom
| | - Derek J Hausenloy
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom; Cardiovascular & Metabolic Disorders Program, Duke-National University of Singapore Medical School, National Heart Research Institute Singapore, National Heart Centre, Yong Loo Lin School of Medicine, National University Singapore, Singapore
| | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), IIS-Fundación Jiménez Díaz University Hospital, and CIBERCV, Madrid, Spain
| | - Thomas Krieg
- Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Christoph Maack
- Department of Translational Research, Comprehensive Heart Failure Center, University Clinic Würzburg, Würzburg, Germany
| | - Rainer Schulz
- Institute for Physiology, Justus-Liebig -Universität, Giessen, Germany
| | - Frank Sellke
- Division of Cardiothoracic Surgery, Alpert Medical School of Brown University and Rhode Island Hospital, Providence, RI, USA
| | - Ajay M Shah
- King's College London British Heart Foundation Centre of Excellence, London, United Kingdom
| | - Holger Thiele
- Heart Center Leipzig at University of Leipzig and Leipzig Heart Science, Leipzig, Germany
| | - Derek M Yellon
- The Hatter Cardiovascular Institute, University College London, London, United Kingdom
| | - Fabio Di Lisa
- Dipartimento di Scienze Biomediche, Università degli studi di Padova, Padova, Italy.
| |
Collapse
|
41
|
Zhou J, Jiang Z, Li Y, Shao X, Liao H. Cause of death during nasopharyngeal carcinoma survivorship: a population-based analysis. Front Oncol 2023; 13:1269118. [PMID: 37920157 PMCID: PMC10619912 DOI: 10.3389/fonc.2023.1269118] [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: 07/29/2023] [Accepted: 10/03/2023] [Indexed: 11/04/2023] Open
Abstract
Background Recently, the survival rate of nasopharyngeal carcinoma (NPC) patients has improved greatly due to developments in NPC treatments. But cause-specific mortality in NPC patients remains unclear. This study aims to investigate the common causes of death in NPC patients. Methods Eligible patients with NPC were included from the Surveillance, Epidemiology, and End Results (SEER) database. Standardized mortality ratios(SMRs) were calculated to compare death rates in NPC patients with those in the general population. Results A total of 3475 patients with NPC were included, of whom 1696 patients died during the follow-up period. 52.83% of deaths were caused by NPC, followed by other cancers (28.13%) and non-cancer causes (18.46%). The proportion of patients who died of NPC decreased over survival time. Moreover, non-cancer causes of death increase from 12.94% to 51.22% over time after 10 years of diagnosis. Heart diseases was the most common non-cancer cause of death in NPC patients. Conclusions Although NPC remains the leading cause of death after NPC diagnosis, other non-NPC causes of death represent an increased number of death in NPC patients. These findings support the involvement of multidisciplinary care for follow-up strategy in NPC patients.
Collapse
Affiliation(s)
- Jie Zhou
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Zhenyu Jiang
- Department of Nephrology, The First People's Hospital of Huzhou, First Affiliated Hospital of Huzhou University, Huzhou, China
| | - Yunhao Li
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| | - Xuwen Shao
- Department of Physical Examination Center, Zhejiang Xinda Hospital, Huzhou, China
| | - Haihong Liao
- Department of Oncology, Huzhou Central Hospital, Affiliated Central Hospital Huzhou University, Huzhou, China
| |
Collapse
|
42
|
Heusch G. Cardioprotection in cardio-oncology: a case for concern? Cardiovasc Res 2023; 119:e144-e145. [PMID: 37587745 DOI: 10.1093/cvr/cvad111] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 08/18/2023] Open
Affiliation(s)
- Gerd Heusch
- Institute for Pathophysiology, West German Heart and Vascular Center, University of Essen Medical School, University of Duisburg-Essen, Hufelandstr. 55, 45147 Essen, Germany
| |
Collapse
|
43
|
Cleary S, Rosen SD, Gilbert DC, Langley RE. Cardiovascular health: an important component of cancer survivorship. BMJ ONCOLOGY 2023; 2:e000090. [PMID: 39886493 PMCID: PMC11235026 DOI: 10.1136/bmjonc-2023-000090] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 08/30/2023] [Indexed: 01/04/2025]
Abstract
Advances in the detection and treatment of cancer have translated into improved cancer survival rates and a growing population of cancer survivors. These include those living with cancer and individuals free of the disease following treatment. Epidemiological studies demonstrate that cancer survivors are at an increased risk of cardiovascular disease (CVD), with cardiovascular (CV) mortality overtaking cancer mortality in some tumour types. Cancer and CVD share common aetiological risk factors, for example, age, tobacco use and obesity, as well as a shared inflammatory pathogenesis. The CV risks of mediastinal radiotherapy and chemotherapy, first observed in the 1970s with anthracyclines, have long been appreciated. More recently, targeted anticancer therapeutics (human epidermal growth factor receptor-2 targeted therpies, vascular endothelial growth factor inhibitors, second/third-generation BCR-ABL inhibitors, multiple myeloma therapies and combination RAF and MEK inhibitors in particular) as well as immunotherapies have added to the burden of treatment-related CV toxicity. Additionally, cancer therapy may indirectly impact on CV health by decreasing physical activity, increasing weight gain and accelerating the ageing process. Improving overall health outcomes by considering cardiological prevention and management in cancer survivorship is an area of increasing interest. CV risk factor assessment and management are recommended post-cancer treatment in accordance with primary prevention guidelines. The European Society of Cardiology 2022 guidelines also recommend enhanced surveillance after cancer treatments with a moderate to high risk of CV consequences. The aim of this article is to provide an overview of the interconnections between cancer and CVD, review current survivorship recommendations, and highlight key areas of ongoing and future research.
Collapse
Affiliation(s)
- Siobhan Cleary
- MRC Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, UK
- Department of Cancer and Surgery, Imperial College Healthcare NHS Trust, London, UK
| | - Stuart D Rosen
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Duncan C Gilbert
- MRC Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, UK
| | - Ruth E Langley
- MRC Clinical Trials Unit at University College London, Institute of Clinical Trials and Methodology, London, UK
| |
Collapse
|
44
|
Schütte W, Gütz S, Nehls W, Blum TG, Brückl W, Buttmann-Schweiger N, Büttner R, Christopoulos P, Delis S, Deppermann KM, Dickgreber N, Eberhardt W, Eggeling S, Fleckenstein J, Flentje M, Frost N, Griesinger F, Grohé C, Gröschel A, Guckenberger M, Hecker E, Hoffmann H, Huber RM, Junker K, Kauczor HU, Kollmeier J, Kraywinkel K, Krüger M, Kugler C, Möller M, Nestle U, Passlick B, Pfannschmidt J, Reck M, Reinmuth N, Rübe C, Scheubel R, Schumann C, Sebastian M, Serke M, Stoelben E, Stuschke M, Thomas M, Tufman A, Vordermark D, Waller C, Wolf J, Wolf M, Wormanns D. [Prevention, Diagnosis, Therapy, and Follow-up of Lung Cancer - Interdisciplinary Guideline of the German Respiratory Society and the German Cancer Society - Abridged Version]. Pneumologie 2023; 77:671-813. [PMID: 37884003 DOI: 10.1055/a-2029-0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
The current S3 Lung Cancer Guidelines are edited with fundamental changes to the previous edition based on the dynamic influx of information to this field:The recommendations include de novo a mandatory case presentation for all patients with lung cancer in a multidisciplinary tumor board before initiation of treatment, furthermore CT-Screening for asymptomatic patients at risk (after federal approval), recommendations for incidental lung nodule management , molecular testing of all NSCLC independent of subtypes, EGFR-mutations in resectable early stage lung cancer in relapsed or recurrent disease, adjuvant TKI-therapy in the presence of common EGFR-mutations, adjuvant consolidation treatment with checkpoint inhibitors in resected lung cancer with PD-L1 ≥ 50%, obligatory evaluation of PD-L1-status, consolidation treatment with checkpoint inhibition after radiochemotherapy in patients with PD-L1-pos. tumor, adjuvant consolidation treatment with checkpoint inhibition in patients withPD-L1 ≥ 50% stage IIIA and treatment options in PD-L1 ≥ 50% tumors independent of PD-L1status and targeted therapy and treatment option immune chemotherapy in first line SCLC patients.Based on the current dynamic status of information in this field and the turnaround time required to implement new options, a transformation to a "living guideline" was proposed.
Collapse
Affiliation(s)
- Wolfgang Schütte
- Klinik für Innere Medizin II, Krankenhaus Martha Maria Halle-Dölau, Halle (Saale)
| | - Sylvia Gütz
- St. Elisabeth-Krankenhaus Leipzig, Abteilung für Innere Medizin I, Leipzig
| | - Wiebke Nehls
- Klinik für Palliativmedizin und Geriatrie, Helios Klinikum Emil von Behring
| | - Torsten Gerriet Blum
- Helios Klinikum Emil von Behring, Klinik für Pneumologie, Lungenklinik Heckeshorn, Berlin
| | - Wolfgang Brückl
- Klinik für Innere Medizin 3, Schwerpunkt Pneumologie, Klinikum Nürnberg Nord
| | | | - Reinhard Büttner
- Institut für Allgemeine Pathologie und Pathologische Anatomie, Uniklinik Köln, Berlin
| | | | - Sandra Delis
- Helios Klinikum Emil von Behring, Klinik für Pneumologie, Lungenklinik Heckeshorn, Berlin
| | | | - Nikolas Dickgreber
- Klinik für Pneumologie, Thoraxonkologie und Beatmungsmedizin, Klinikum Rheine
| | | | - Stephan Eggeling
- Vivantes Netzwerk für Gesundheit, Klinikum Neukölln, Klinik für Thoraxchirurgie, Berlin
| | - Jochen Fleckenstein
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum des Saarlandes und Medizinische Fakultät der Universität des Saarlandes, Homburg
| | - Michael Flentje
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Würzburg, Würzburg
| | - Nikolaj Frost
- Medizinische Klinik mit Schwerpunkt Infektiologie/Pneumologie, Charite Universitätsmedizin Berlin, Berlin
| | - Frank Griesinger
- Klinik für Hämatologie und Onkologie, Pius-Hospital Oldenburg, Oldenburg
| | | | - Andreas Gröschel
- Klinik für Pneumologie und Beatmungsmedizin, Clemenshospital, Münster
| | | | | | - Hans Hoffmann
- Klinikum Rechts der Isar, TU München, Sektion für Thoraxchirurgie, München
| | - Rudolf M Huber
- Medizinische Klinik und Poliklinik V, Thorakale Onkologie, LMU Klinikum Munchen
| | - Klaus Junker
- Klinikum Oststadt Bremen, Institut für Pathologie, Bremen
| | - Hans-Ulrich Kauczor
- Klinikum der Universität Heidelberg, Abteilung Diagnostische Radiologie, Heidelberg
| | - Jens Kollmeier
- Helios Klinikum Emil von Behring, Klinik für Pneumologie, Lungenklinik Heckeshorn, Berlin
| | | | - Marcus Krüger
- Klinik für Thoraxchirurgie, Krankenhaus Martha-Maria Halle-Dölau, Halle-Dölau
| | | | - Miriam Möller
- Krankenhaus Martha-Maria Halle-Dölau, Klinik für Innere Medizin II, Halle-Dölau
| | - Ursula Nestle
- Kliniken Maria Hilf, Klinik für Strahlentherapie, Mönchengladbach
| | | | - Joachim Pfannschmidt
- Klinik für Thoraxchirurgie, Lungenklinik Heckeshorn, Helios Klinikum Emil von Behring, Berlin
| | - Martin Reck
- Lungeclinic Grosshansdorf, Pneumologisch-onkologische Abteilung, Grosshansdorf
| | - Niels Reinmuth
- Klinik für Pneumologie, Thorakale Onkologie, Asklepios Lungenklinik Gauting, Gauting
| | - Christian Rübe
- Klinik für Strahlentherapie und Radioonkologie, Universitätsklinikum des Saarlandes, Homburg/Saar, Homburg
| | | | | | - Martin Sebastian
- Medizinische Klinik II, Universitätsklinikum Frankfurt, Frankfurt
| | - Monika Serke
- Zentrum für Pneumologie und Thoraxchirurgie, Lungenklinik Hemer, Hemer
| | | | - Martin Stuschke
- Klinik und Poliklinik für Strahlentherapie, Universitätsklinikum Essen, Essen
| | - Michael Thomas
- Thoraxklinik am Univ.-Klinikum Heidelberg, Thorakale Onkologie, Heidelberg
| | - Amanda Tufman
- Medizinische Klinik und Poliklinik V, Thorakale Onkologie, LMU Klinikum München
| | - Dirk Vordermark
- Universitätsklinik und Poliklinik für Strahlentherapie, Universitätsklinikum Halle, Halle
| | - Cornelius Waller
- Klinik für Innere Medizin I, Universitätsklinikum Freiburg, Freiburg
| | | | - Martin Wolf
- Klinikum Kassel, Klinik für Onkologie und Hämatologie, Kassel
| | - Dag Wormanns
- Evangelische Lungenklinik, Radiologisches Institut, Berlin
| |
Collapse
|
45
|
Sanadgol G, Samimi S, Shirini D, Nakhaei P, Mohseni M, Alizadehasl A. Right ventricle toxicity in cancer treatment: a focused review on cardiac imaging. Future Cardiol 2023; 19:537-545. [PMID: 37830360 DOI: 10.2217/fca-2022-0024] [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: 10/14/2023] Open
Abstract
Background: The right ventricle (RV) remains the 'forgotten chamber' in the clinical assessment of cancer therapy-related cardiac dysfunction (CTRCD). Aim: We aimed to review the role that various cardiac imaging modalities play in RV assessment as part of the integrative management of patients undergoing cancer therapy. Discussion: RV assessment remains challenging by traditional 2D echocardiography. In this review we discuss other parameters such as right atrial strain, and other echocardiographic modalities such as 3D and stress echocardiography. We also elaborate on the specific role that cardiac magnetic resonance imaging and equilibrium radionuclide angiocardiography can play in assessing the RV. Conclusion: Biventricular function should be monitored following chemotherapy for early detection of subclinical CTRCD and possible solitary RV changes.
Collapse
Affiliation(s)
- Ghazal Sanadgol
- Shahid-Beheshti University of Medical Sciences, Tehran, 1983969411, Iran
| | - Sahar Samimi
- Tehran University of Medical Sciences, Tehran, 1416634793, Iran
| | - Dorsa Shirini
- Cardiovascular Research Center, Shahid Beheshti University of Medical, Tehran, 1983969411, Iran Sciences
| | - Pooria Nakhaei
- Heart Valve Disease Research Center, Rajaie Cardiovascular Medical & Research Center, Iran University of Medical Sciences, Tehran, 1995614331, Iran
| | - Mina Mohseni
- Department of Cardio-oncology Research, Rajaie Cardiovascular Medical & Research Center, Iran University of Medical Sciences, Tehran, 1995614331, Iran
| | - Azin Alizadehasl
- Professor of Cardiology, Echocardiologist, Cardio-oncologist, Cardio-oncology Research Center, Shaheed Rajaie Cardiovascular Medical & Research Center, Iran University of Medical Science, Tehran, 1995614331, Iran
| |
Collapse
|
46
|
Alessandra-Perini J, Machado DE, Palmero CY, Claudino MC, de Moura RS, Palumbo Junior A, Perini JA, Nasciutti LE. Euterpe oleracea extract (açaí) exhibits cardioprotective effects after chemotherapy treatment in a breast cancer model. BMC Complement Med Ther 2023; 23:301. [PMID: 37626388 PMCID: PMC10463785 DOI: 10.1186/s12906-023-04104-7] [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: 01/06/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
BACKGROUND Açaí, a Brazilian native fruit, has already been demonstrated to play a role in the progress of breast cancer and cardiotoxicity promoted by chemotherapy agents. Thus, the present study aimed to evaluate the combined use of açaí and the FAC-D chemotherapy protocol in a breast cancer model in vivo. METHODS Mammary carcinogenesis was induced in thirty female Wistar rats by subcutaneous injection of 25 mg/kg 7,12-dimethylbenzanthracene (DMBA) in the mammary gland. After sixty days, the rats were randomized into two groups: treated with 200 mg/kg of either açaí extract or vehicle, via gastric tube for 45 consecutive days. The FAC-D protocol was initiated after 90 days of induction by intraperitoneal injection for 3 cycles with a 7-day break each. After treatment, blood was collected for haematological and biochemical analyses, and tumours were collected for macroscopic and histological analyses. In the same way, heart, liver, and kidney samples were also collected for macroscopic and histological analyses. RESULTS Breast cancer was found as a cystic mass with a fibrotic pattern in the mammary gland. The histological analysis showed an invasive carcinoma area in both groups; however, in the saline group, there was a higher presence of inflammatory clusters. No difference was observed regarding body weight, glycaemia, aspartate aminotransferase (AST), alanine aminotransferase (ALT), creatinine, and urea in either group. However, açaí treatment decreased creatine kinase (CK), creatine kinase MB (CKMB), troponin I and C-reactive protein levels and increased the number of neutrophils and monocytes. Heart histopathology showed normal myocardium in the açaí treatment, while the saline group presented higher toxicity effects with loss of architecture of cardiac tissue. Furthermore, the açaí treatment presented greater collagen distribution, increased hydroxyproline concentration and lower H2AX immunostaining in the heart samples. CONCLUSION Açaí decreased the number of inflammatory cells in the tumor environment and exhibited protection against chemotherapy drug cardiotoxicity with an increased immune response in animals. Thus, açaí can be considered a promising low-cost therapeutic treatment that can be used in association with chemotherapy agents to avoid heart damage.
Collapse
Affiliation(s)
- Jéssica Alessandra-Perini
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil.
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil.
| | - Daniel Escorsim Machado
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil
| | - Celia Yelimar Palmero
- Laboratório Integrado de Morfologia, Instituto de Biodiversidade e Sustentabilidade, Universidade Federal do Rio de Janeiro 9UFRJ), Rio de Janeiro, RJ, Brazil
| | - Marllow Caetano Claudino
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil
| | - Roberto Soares de Moura
- Departamento de Farmacologia e Psicobiologia, Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Antônio Palumbo Junior
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| | - Jamila Alessandra Perini
- Laboratório de Pesquisa em Ciências Farmacêuticas (LAPESF), Universidade do Estado do Rio de Janeiro (UERJ), Manuel Caldeira de Alvarenga Avenue, 1.203, Rio de Janeiro, RJ, 23070-200, Brazil
| | - Luiz Eurico Nasciutti
- Instituto de Ciências Biomédicas (ICB), Universidade Federal do Rio de Janeiro (UFRJ), Rio de Janeiro, RJ, Brazil
| |
Collapse
|
47
|
Xia Y, Lu S, Huo C, Fan L, Lin M, Huang J. Non cancer causes of death after gallbladder cancer diagnosis: a population-based analysis. Sci Rep 2023; 13:13746. [PMID: 37612302 PMCID: PMC10447554 DOI: 10.1038/s41598-023-40134-4] [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] [Received: 03/04/2023] [Accepted: 08/05/2023] [Indexed: 08/25/2023] Open
Abstract
Mortality from non cancer causes in patients with gallbladder cancer (GBC) still unclear. This study evaluated the causes and risk factors of non cancer death during different follow-up periods after GBC diagnosis. Non cancer causes of death for GBC patients diagnosed between 2000 and 2017 in Surveillance, Epidemiology and End Results database were analyzed and standardized mortality rates (SMR) for each non cancer death were calculated. Predictors for non cancer death were identified through multivariate competing risk analysis. A total 11,927 GBC patients were identified for further analysis, 9393 died during follow up. The largest proportion of non cancer deaths occurred > 3 years after diagnosis (39.4%). Most common non cancer cause were cardiovascular disease (43.3%), followed by other cause of death (34.4%) and infectious diseases (8.6%). Compared with US general population, GBC patients has higher risk of death from disease of heart (SMR, 1.58; 95%CI, 1.41-1.75), septicemia (SMR,3.21; 95%CI, 2.27-4.40), diabetes mellitus (SMR,1.97; 95%CI, 1.43-2.63), alone with other causes. Non cancer causes accounted for a significant proportion of deaths during the follow-up period after GBC diagnosis. The risk of non cancer death is higher in GBC patients than in the general population. Our study provides comprehensive assessment of death from non cancer cause in GBC patients, which has important implications for health management in GBC patients.
Collapse
Affiliation(s)
- Yang Xia
- Department of Gastroenterology, The Affliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Shuangshuang Lu
- Department of Gastroenterology, The Affliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Chunyan Huo
- Department of Gastroenterology, The Affliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Li Fan
- Department of Gastroenterology, The Affliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
| | - Min Lin
- Department of Gastroenterology, The Affliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China.
| | - Jin Huang
- Department of Gastroenterology, The Affliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China.
| |
Collapse
|
48
|
Engan B, Diab S, Brun H, Raastad T, Torsvik IK, Omdal TR, Ghavidel FZ, Greve G, Ruud E, Edvardsen E, Leirgul E. Systolic myocardial function measured by echocardiographic speckle-tracking and peak oxygen consumption in pediatric childhood cancer survivors-a PACCS study. Front Cardiovasc Med 2023; 10:1221787. [PMID: 37476575 PMCID: PMC10354364 DOI: 10.3389/fcvm.2023.1221787] [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: 05/15/2023] [Accepted: 06/22/2023] [Indexed: 07/22/2023] Open
Abstract
Background Cancer therapy-related cardiotoxicity is a major cause of cardiovascular morbidity in childhood cancer survivors. The aims of this study were to investigate systolic myocardial function and its association to cardiorespiratory fitness in pediatric childhood cancer survivors. Methods In this sub-study of the international study "Physical Activity and fitness in Childhood Cancer Survivors" (PACCS), echocardiographic measures of left ventricular global longitudinal strain (LV-GLS) and right ventricular longitudinal strain (RV-LS) were measured in 128 childhood cancer survivors aged 9-18 years and in 23 age- and sex-matched controls. Cardiorespiratory fitness was measured as peak oxygen consumption achieved on treadmill and correlated to myocardial function. Results Mean LV-GLS was reduced in the childhood cancer survivors compared to the controls, -19.7% [95% confidence interval (CI) -20.1% to -19.3%] vs. -21.3% (95% CI: -22.2% to -20.3%) (p = 0.004), however, mainly within normal range. Only 13% of the childhood cancer survivors had reduced LV longitudinal strain z-score. Mean RV-LS was similar in the childhood cancer survivors and the controls, -23.2% (95% CI: -23.7% to -22.6%) vs. -23.3% (95% CI: -24.6% to -22.0%) (p = 0.8). In the childhood cancer survivors, lower myocardial function was associated with lower peak oxygen consumption [correlation coefficient (r) = -0.3 for LV-GLS]. Higher doses of anthracyclines (r = 0.5 for LV-GLS and 0.2 for RV-LS) and increasing time after treatment (r = 0.3 for LV-GLS and 0.2 for RV-LS) were associated with lower myocardial function. Conclusions Left ventricular function, but not right ventricular function, was reduced in pediatric childhood cancer survivors compared to controls, and a lower left ventricular myocardial function was associated with lower peak oxygen consumption. Furthermore, higher anthracycline doses and increasing time after treatment were associated with lower myocardial function, implying that long-term follow-up is important in this population at risk.
Collapse
Affiliation(s)
- Britt Engan
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Simone Diab
- Department of Pediatric Cardiology, Oslo University Hospital, Oslo, Norway
| | - Henrik Brun
- Department of Pediatric Cardiology, Oslo University Hospital, Oslo, Norway
- The Intervention Centre, Technology and Innovation Clinic, Oslo University Hospital, Oslo, Norway
| | - Truls Raastad
- Department of Sports Medicine, The Norwegian School of Sport Sciences, Oslo, Norway
| | - Ingrid Kristin Torsvik
- Department of Pediatric and Adolescent Medicine, Haukeland University Hospital, Bergen, Norway
| | - Tom Roar Omdal
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | | | - Gottfried Greve
- Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| | - Ellen Ruud
- Department of Pediatric Medicine, Oslo University Hospital, Oslo, Norway
| | - Elisabeth Edvardsen
- Department of Sports Medicine, The Norwegian School of Sport Sciences, Oslo, Norway
| | - Elisabeth Leirgul
- Department of Heart Disease, Haukeland University Hospital, Bergen, Norway
| |
Collapse
|
49
|
Mallouppas M, Chung R, Ghosh AK, Macklin A, Yellon DM, Walker JM. Anthracyclines and Biomarkers of Myocardial Injury: The Effect of Remote Ischemic Conditioning. JACC CardioOncol 2023; 5:343-355. [PMID: 37397080 PMCID: PMC10308041 DOI: 10.1016/j.jaccao.2023.03.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 03/02/2023] [Accepted: 03/06/2023] [Indexed: 07/04/2023] Open
Abstract
Background Remote ischemic conditioning (RIC) has been beneficial in laboratory studies of anthracycline cardiotoxicity, but its effects in patients is not established. Objectives The authors studied the effect of RIC on cardiac biomarkers and function during and after anthracycline chemotherapy. Methods The ERIC-Onc study (Effect of Remote Ischaemic Conditioning in Oncology Patients; NCT02471885) was a randomized, single-blind, sham-controlled study of RIC at each chemotherapy cycle. The primary endpoint was troponin T (TnT) during chemotherapy and up to 1 year. Secondary outcomes included cardiac function, major adverse cardiovascular events (MACE), and MACE or cancer death. Cardiac myosin-binding-protein C (cMyC) was investigated in parallel with TnT. Results The study was prematurely halted after the evaluation of 55 patients (RIC n = 28, sham n = 27). Biomarkers increased from baseline to cycle 6 of chemotherapy for all patients (median TnT 6 [IQR: 4-9] ng/L to 33 [IQR: 16-36)] ng/L; P ≤ 0.001; cMyC 3 (IQR: 2-5) ng/L to 47 (IQR: 18-49) ng/L; P ≤ 0.001). Mixed-effects regression analysis for repeated measures showed no difference in TnT between the 2 groups (RIC vs sham, mean difference 3.15 ng/L; 95% CI: -0.04 to 6.33; P = 0.053), or cMyC (RIC vs sham, mean difference 4.17 ng/L; 95% CI: -0.12 to 8.45; P = 0.056). There were more MACE and cancer deaths in the RIC group (11 vs 3; HR: 0.25; 95% CI: 0.07-0.90; P = 0.034), with more cancer deaths (8 vs 1; HR: 0.21; 95% CI: 0.04-0.95; P = 0.043) at 1 year. Conclusions TnT and cMyC significantly increased during anthracycline chemotherapy with 81% having a TnT ≥14 ng/L at cycle 6. RIC did not affect the rise in biomarkers, but there was a small increase in early cancer deaths, possibly related to the greater proportion of patients with metastatic disease randomized to the RIC group (54%vs 37%). (Effect of Remote Ischaemic Conditioning in Oncology Patients [ERIC-ONC]; NCT02471885).
Collapse
Affiliation(s)
| | | | | | | | - Derek M. Yellon
- Address for correspondence: Prof Derek Yellon, OR Prof Malcolm Walker, The Hatter Cardiovascular Institute, University College London Institute of Cardiovascular Science, 67 Chenies Mews, London WC1E 6HX, United Kingdom.
| | - J. Malcolm Walker
- Address for correspondence: Prof Derek Yellon, OR Prof Malcolm Walker, The Hatter Cardiovascular Institute, University College London Institute of Cardiovascular Science, 67 Chenies Mews, London WC1E 6HX, United Kingdom.
| |
Collapse
|
50
|
Yang Z, Leng K, Shi G. Causes of death among patients with hepatocellular carcinoma in United States from 2000 to 2018. Cancer Med 2023. [PMID: 37083308 DOI: 10.1002/cam4.5986] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 03/21/2023] [Accepted: 04/09/2023] [Indexed: 04/22/2023] Open
Abstract
BACKGROUND The gains in survival outcomes of US patients with hepatocellular carcinoma (HCC) have come at the expense of developing non-cancer-related morbidities, such as cardiovascular diseases (CVDs) and infections. However, population-based data on causes of death (CODs) in patients with HCC are scarce. METHODS A cancer registry database in the United States was used to analyze the CODs among patients with HCC. Death cause distribution and standardized mortality ratios were calculated to quantify the disease-specific death burden. RESULTS A total of 40,094 patients with a histological diagnosis of HCC were identified from the SEER-18 database between 2000 and 2018, of which 30,796 (76.8%) died during the follow-up period. The majority of these deaths (25,153, 81.7%) occurred within 2 years after diagnosis, 13.2% (4075) occurred within 2-5 years, and 5.1% (1568) occurred after 5 years. All age groups had a lower burden of female deaths than of male deaths during the study period. With respect to CODs, 23,824 (77.4%), 2289 (7.4%), and 4683 (15.2%) were due to HCC, other cancers, and non-cancer causes, respectively. Non-cancer-related deaths were more common among older patients and those with longer latency periods since diagnosis. The major causes of non-cancer-related deaths are other infectious and parasitic diseases, including HIV and CVDs. CONCLUSIONS CODs during HCC survivorship varied, and a growing number of survivors tended to die from causes other than HCC, with an increasing latency period since diagnosis. Comprehensive analyses of mortality patterns and temporal trends could underpin strategies to reduce these risks.
Collapse
Affiliation(s)
- Zhen Yang
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, People's Republic of China
- Department of Hepatopancreatobiliary Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, People's Republic of China
| | - Kaiming Leng
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, People's Republic of China
- Department of Hepatopancreatobiliary Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, People's Republic of China
| | - Guangjun Shi
- Department of Hepatopancreatobiliary Surgery, Qingdao Municipal Hospital, Qingdao University, Qingdao, People's Republic of China
- Department of Hepatopancreatobiliary Surgery, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, People's Republic of China
| |
Collapse
|