1
|
Shi H, Zou Y, Li Y, Li Y, Liu B. Neuregulin-1 reduces Doxorubicin-induced cardiotoxicity by upregulating YAP to inhibit senescence. Int Immunopharmacol 2024; 143:113278. [PMID: 39405937 DOI: 10.1016/j.intimp.2024.113278] [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: 07/29/2024] [Revised: 09/16/2024] [Accepted: 09/25/2024] [Indexed: 10/30/2024]
Abstract
The cardiotoxicity of Doxorubicin (Dox) limits its clinical application, creating an urgent need to investigate its underlying mechanism and develop effective therapies. Senescence plays an important role in Dox-induced cardiotoxicity (DIC). Recently, Neuregulin-1 (NRG1) was found to regulate Yes-associated protein (YAP), which was reported to inhibit senescence, suggesting that NRG1 might be used to treat DIC by inhibiting senescence through YAP regulation. We examined the changes and regulatory roles of YAP and senescence in Dox cardiotoxicity and whether NRG1 could reduce DIC in chronic DIC mice and Dox-treated H9c2 cells. Our study revealed that sustained small doses of Dox impaired cardiac function and H9c2 cell viability, induced myocardial senescence, and inhibited YAP expression. Conversely, high levels of YAP inhibited Dox-induced senescence in H9c2 cells, indicating that Dox promotes myocardial senescence by inhibiting YAP. In addition, we found that exogenous NRG1 inhibited the phosphorylation of LATS1 and MST1, thereby inhibiting YAP phosphorylation and promote the nuclear translocation of YAP, inhibiting senescence and attenuating Dox-induced cardiotoxicity. YAP knockdown or inhibition of YAP binding to TEA domain transcription factor protein (TEAD)blocks the protective effects of NRG1. In conclusion, our study suggests that Dox-induced myocardial senescence through YAP inhibition is one of the pathological mechanisms of its cardiotoxicity. Additionally, NRG1 reduces DIC by upregulating YAP to inhibit senescence.
Collapse
Affiliation(s)
- Henghe Shi
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yifei Zou
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yinghao Li
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Yangxue Li
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, No. 4026 Yatai Street, Changchun, Jilin Province 130041, China.
| |
Collapse
|
2
|
Yang F, Zhang G, An N, Dai Q, Cho W, Shang H, Xing Y. Interplay of ferroptosis, cuproptosis, and PANoptosis in cancer treatment-induced cardiotoxicity: Mechanisms and therapeutic implications. Semin Cancer Biol 2024; 106-107:106-122. [PMID: 39299410 DOI: 10.1016/j.semcancer.2024.09.003] [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: 04/29/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/22/2024]
Abstract
With the prolonged survival of individuals with cancer, the emergence of cardiovascular diseases (CVD) induced by cancer treatment has become a significant concern, ranking as the second leading cause of death among cancer survivors. This review explores three distinct types of programmed cell death (PCD): ferroptosis, cuproptosis, and PANoptosis, focusing on their roles in chemotherapy-induced cardiotoxicity. While ferroptosis and cuproptosis are triggered by excess iron and copper (Cu), PANoptosis is an inflammatory PCD with features of pyroptosis, apoptosis, and necroptosis. Recent studies reveal intricate connections among these PCD types, emphasizing the interplay between cuproptosis and ferroptosis. Notably, the role of intracellular Cu in promoting ferroptosis through GPX4 is highlighted. Additionally, ROS-induced PANoptosis is influenced by ferroptosis and cuproptosis, suggesting a complex interrelationship. This review provides insights into the molecular mechanisms of these PCD modalities and their distinct contributions to chemotherapy-induced cardiotoxicity. Furthermore, we discuss the potential application of cardioprotective drugs in managing these PCD types. This comprehensive analysis aims to advance the understanding, diagnosis, and therapeutic strategies for cardiotoxicity associated with cancer treatment.
Collapse
Affiliation(s)
- Fan Yang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Guoxia Zhang
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China; Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Na An
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - Qianqian Dai
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China
| | - William Cho
- Department of Clinical Oncology, Queen Elizabeth Hospital, Kowloon, Hong Kong, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of the Ministry of Education, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing 100700, China.
| | - Yanwei Xing
- Guang'anmen Hospital, Chinese Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
3
|
Austin D, Maier RH, Akhter N, Sayari M, Ogundimu E, Maddox JM, Vahabi S, Humphreys AC, Graham J, Oxenham H, Haney S, Cresti N, Verrill M, Osborne W, Wright KL, Goranova R, Bailey JR, Kalakonda N, Macheta M, Kilner MF, Young ME, Morley NJ, Neelakantan P, Gilbert G, Thomas BK, Graham RJ, Fujisawa T, Mills NL, Hildreth V, Prichard J, Kasim AS, Hancock HC, Plummer C. Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma: The PROACT Clinical Trial. JACC CardioOncol 2024; 6:684-696. [PMID: 39479329 PMCID: PMC11520224 DOI: 10.1016/j.jaccao.2024.07.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 11/02/2024] Open
Abstract
Background Cardiotoxicity is a concern for cancer survivors undergoing anthracycline chemotherapy. Enalapril has been explored for its potential to mitigate cardiotoxicity in cancer patients. The dose-dependent cardiotoxicity effects of anthracyclines can be detected early through the biomarker cardiac troponin. Objectives The PROACT (Preventing Cardiac Damage in Patients Treated for Breast Cancer and Lymphoma) clinical trial assessed the effectiveness of enalapril in preventing cardiotoxicity, manifesting as myocardial injury and cardiac function impairment, in patients undergoing high-dose anthracycline-based chemotherapy for breast cancer or non-Hodgkin lymphoma. Methods This prospective, multicenter, open-label, randomized controlled trial employed a superiority design with observer-blinded endpoints. A total of 111 participants, scheduled for 6 cycles of chemotherapy with a planned dose of ≥300 mg/m2 doxorubicin equivalents, were randomized to receive either enalapril (titrated up to 20 mg daily) or standard care without enalapril. Results Myocardial injury, indicated by cardiac troponin T (≥14 ng/L), during and 1 month after chemotherapy, was observed in 42 (77.8%) of 54 patients in the enalapril group vs 45 (83.3%) of 54 patients in the standard care group (OR: 0.65; 95% CI: 0.23-1.78). Injury detected by cardiac troponin I (>26.2 ng/L) occurred in 25 (47.2%) of 53 patients on enalapril compared with 24 (45.3%) of 53 in standard care (OR: 1.10; 95% CI: 0.50-2.38). A relative decline of more than 15% from baseline in left ventricular global longitudinal strain was observed in 10 (21.3%) of 47 patients on enalapril and 9 (21.9%) of 41 in standard care (OR: 0.95; 95% CI: 0.33-2.74). An absolute decline of >10% to <50% in left ventricular ejection fraction was seen in 2 (4.1%) of 49 patients on enalapril vs none in patients in standard care. Conclusions Adding enalapril to standard care during chemotherapy did not prevent cardiotoxicity in patients receiving high-dose anthracycline-based chemotherapy. (PROACT: Can we prevent Chemotherapy-related Heart Damage in Patients With Breast Cancer and Lymphoma?; NCT03265574).
Collapse
Affiliation(s)
- David Austin
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rebecca H. Maier
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Nasima Akhter
- School of Health and Life Sciences, Teesside University, Middlesbrough, United Kingdom
| | - Mohammad Sayari
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Emmanuel Ogundimu
- Department of Mathematical Science, Durham University, Durham, United Kingdom
| | - Jamie M. Maddox
- Department of Haematology, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Sharareh Vahabi
- Bristol Heart Institute, University Hospitals Bristol and Weston NHS Foundation Trust, Bristol, United Kingdom
| | - Alison C. Humphreys
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Janine Graham
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Helen Oxenham
- Department of Cardiology, North Tees and Hartlepool NHS Foundation Trust, Stockton-on-Tees, United Kingdom
| | - Sophie Haney
- Cancer Services, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
- Cancer Services, County Durham and Darlington NHS Foundation Trust, Darlington, United Kingdom
| | - Nicola Cresti
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Mark Verrill
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Wendy Osborne
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Kathryn L. Wright
- Freeman Hospital, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Cancer Services, South Tyneside and Sunderland NHS Foundation Trust, Sunderland, United Kingdom
| | - Rebecca Goranova
- Department of Oncology, Derriford Hospital, University Hospitals Plymouth NHS Trust, Plymouth, United Kingdom
| | - James R. Bailey
- Department of Haematology, Hull University Teaching Hospitals NHS Trust, Queen’s Centre for Oncology and Haematology, Castle Hill Hospital, Hull, United Kingdom
| | - Nagesh Kalakonda
- Department of Molecular and Clinical Cancer Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Mac Macheta
- Department of Haematology, Blackpool Teaching Hospitals, Blackpool, United Kingdom
| | - Mari F. Kilner
- Pathology Services, Northumbria Healthcare NHS Foundation Trust, North Shields, United Kingdom
| | - Moya E. Young
- Department of Haematology, East Kent Hospitals University NHS Foundation Trust, Canterbury, United Kingdom
| | - Nick J. Morley
- Department of Haematology, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom
| | - Pratap Neelakantan
- Department of Haematology, Royal Berkshire NHS Foundation Trust, Reading, United Kingdom
| | - Georgia Gilbert
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Byju K. Thomas
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Richard J. Graham
- Academic Cardiovascular Unit, James Cook University Hospital, South Tees Hospitals NHS Foundation Trust, Middlesbrough, United Kingdom
| | - Takeshi Fujisawa
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Nicholas L. Mills
- Centre for Cardiovascular Science, University of Edinburgh, Edinburgh, United Kingdom
| | - Victoria Hildreth
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Jonathan Prichard
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Adetayo S. Kasim
- Department of Anthropology, Durham University, Durham, United Kingdom
| | - Helen C. Hancock
- Newcastle Clinical Trials Unit, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Chris Plummer
- Department of Cardiology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
- Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
4
|
Jiang R, Lou L, Shi W, Chen Y, Fu Z, Liu S, Sok T, Li Z, Zhang X, Yang J. Statins in Mitigating Anticancer Treatment-Related Cardiovascular Disease. Int J Mol Sci 2024; 25:10177. [PMID: 39337662 PMCID: PMC11432657 DOI: 10.3390/ijms251810177] [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: 08/20/2024] [Revised: 09/14/2024] [Accepted: 09/20/2024] [Indexed: 09/30/2024] Open
Abstract
Certain anticancer therapies inevitably increase the risk of cardiovascular events, now the second leading cause of death among cancer patients. This underscores the critical need for developing effective drugs or regimens for cardiovascular protection. Statins possess properties such as antioxidative stress, anti-inflammatory effects, antifibrotic activity, endothelial protection, and immune modulation. These pathological processes are central to the cardiotoxicity associated with anticancer treatment. There is prospective clinical evidence confirming the protective role of statins in chemotherapy-induced cardiotoxicity. Numerous preclinical studies have demonstrated that statins can ameliorate heart and endothelial damage caused by radiotherapy, although clinical studies are scarce. In the animal models of trastuzumab-induced cardiomyopathy, statins provide protection through anti-inflammatory, antioxidant, and antifibrotic mechanisms. In animal and cell models, statins can mitigate inflammation, endothelial damage, and cardiac injury induced by immune checkpoint inhibitors. Chimeric antigen receptor (CAR)-T cell therapy-induced cardiotoxicity and immune effector cell-associated neurotoxicity syndrome are associated with uncontrolled inflammation and immune activation. Due to their anti-inflammatory and immunomodulatory effects, statins have been used to manage CAR-T cell therapy-induced immune effector cell-associated neurotoxicity syndrome in a clinical trial. However, direct evidence proving that statins can mitigate CAR-T cell therapy-induced cardiotoxicity is still lacking. This review summarizes the possible mechanisms of anticancer therapy-induced cardiotoxicity and the potential mechanisms by which statins may reduce related cardiac damage. We also discuss the current status of research on the protective effect of statins in anticancer treatment-related cardiovascular disease and provide directions for future research. Additionally, we propose further studies on using statins for the prevention of cardiovascular disease in anticancer treatment.
Collapse
Affiliation(s)
- Rong Jiang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Lian Lou
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Wen Shi
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Yuxiao Chen
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhaoming Fu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Shuo Liu
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Thida Sok
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Zhihang Li
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Xuan Zhang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| | - Jian Yang
- Department of Cardiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310003, China
| |
Collapse
|
5
|
Logotheti S, Pavlopoulou A, Rudsari HK, Galow AM, Kafalı Y, Kyrodimos E, Giotakis AI, Marquardt S, Velalopoulou A, Verginadis II, Koumenis C, Stiewe T, Zoidakis J, Balasingham I, David R, Georgakilas AG. Intercellular pathways of cancer treatment-related cardiotoxicity and their therapeutic implications: the paradigm of radiotherapy. Pharmacol Ther 2024; 260:108670. [PMID: 38823489 DOI: 10.1016/j.pharmthera.2024.108670] [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: 11/11/2023] [Revised: 05/16/2024] [Accepted: 05/25/2024] [Indexed: 06/03/2024]
Abstract
Advances in cancer therapeutics have improved patient survival rates. However, cancer survivors may suffer from adverse events either at the time of therapy or later in life. Cardiovascular diseases (CVD) represent a clinically important, but mechanistically understudied complication, which interfere with the continuation of best-possible care, induce life-threatening risks, and/or lead to long-term morbidity. These concerns are exacerbated by the fact that targeted therapies and immunotherapies are frequently combined with radiotherapy, which induces durable inflammatory and immunogenic responses, thereby providing a fertile ground for the development of CVDs. Stressed and dying irradiated cells produce 'danger' signals including, but not limited to, major histocompatibility complexes, cell-adhesion molecules, proinflammatory cytokines, and damage-associated molecular patterns. These factors activate intercellular signaling pathways which have potentially detrimental effects on the heart tissue homeostasis. Herein, we present the clinical crosstalk between cancer and heart diseases, describe how it is potentiated by cancer therapies, and highlight the multifactorial nature of the underlying mechanisms. We particularly focus on radiotherapy, as a case known to often induce cardiovascular complications even decades after treatment. We provide evidence that the secretome of irradiated tumors entails factors that exert systemic, remote effects on the cardiac tissue, potentially predisposing it to CVDs. We suggest how diverse disciplines can utilize pertinent state-of-the-art methods in feasible experimental workflows, to shed light on the molecular mechanisms of radiotherapy-related cardiotoxicity at the organismal level and untangle the desirable immunogenic properties of cancer therapies from their detrimental effects on heart tissue. Results of such highly collaborative efforts hold promise to be translated to next-generation regimens that maximize tumor control, minimize cardiovascular complications, and support quality of life in cancer survivors.
Collapse
Affiliation(s)
- Stella Logotheti
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece; Biomedical Physics in Radiation Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| | - Athanasia Pavlopoulou
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | | | - Anne-Marie Galow
- Institute for Genome Biology, Research Institute for Farm Animal Biology (FBN), 18196 Dummerstorf, Germany
| | - Yağmur Kafalı
- Izmir Biomedicine and Genome Center, Izmir, Turkey; Izmir International Biomedicine and Genome Institute, Dokuz Eylul University, Izmir, Turkey
| | - Efthymios Kyrodimos
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Aris I Giotakis
- First Department of Otorhinolaryngology, Head and Neck Surgery, Hippocrateion General Hospital Athens, National and Kapodistrian University of Athens, Athens, Greece
| | - Stephan Marquardt
- Institute of Translational Medicine for Health Care Systems, Medical School Berlin, Hochschule Für Gesundheit Und Medizin, 14197 Berlin, Germany
| | - Anastasia Velalopoulou
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ioannis I Verginadis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Constantinos Koumenis
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thorsten Stiewe
- Institute of Molecular Oncology, Philipps-University, 35043 Marburg, Germany; German Center for Lung Research (DZL), Universities of Giessen and Marburg Lung Center (UGMLC), 35043 Marburg, Germany; Genomics Core Facility, Philipps-University, 35043 Marburg, Germany; Institute for Lung Health (ILH), Justus Liebig University, 35392 Giessen, Germany
| | - Jerome Zoidakis
- Department of Biotechnology, Biomedical Research Foundation, Academy of Athens, Athens, Greece; Department of Biology, National and Kapodistrian University of Athens, Athens, Greece
| | | | - Robert David
- Department of Cardiac Surgery, Rostock University Medical Center, 18057 Rostock, Germany; Department of Life, Light & Matter, Interdisciplinary Faculty, Rostock University, 18059 Rostock, Germany
| | - Alexandros G Georgakilas
- DNA Damage Laboratory, Physics Department, School of Applied Mathematical and Physical Sciences, National Technical University of Athens (NTUA), Zografou, 15780, Athens, Greece.
| |
Collapse
|
6
|
Liu X, Yao X, Chen L. Expanding roles of circRNAs in cardiovascular diseases. Noncoding RNA Res 2024; 9:429-436. [PMID: 38511061 PMCID: PMC10950605 DOI: 10.1016/j.ncrna.2024.02.001] [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: 12/26/2023] [Revised: 02/01/2024] [Accepted: 02/04/2024] [Indexed: 03/22/2024] Open
Abstract
CircRNAs are a class of single-stranded RNAs characterized by covalently looped structures. Emerging advances have promoted our understanding of circRNA biogenesis, nuclear export, biological functions, and functional mechanisms. Roles of circRNAs in diverse diseases have been increasingly recognized in the past decade, with novel approaches in bioinformatics analysis and new strategies in modulating circRNA levels, which have made circRNAs the hot spot for therapeutic applications. Moreover, due to the intrinsic features of circRNAs such as high stability, conservation, and tissue-/stage-specific expression, circRNAs are believed to be promising prognostic and diagnostic markers for diseases. Aiming cardiovascular disease (CVD), one of the leading causes of mortality worldwide, we briefly summarize the current understanding of circRNAs, provide the recent progress in circRNA functions and functional mechanisms in CVD, and discuss the future perspectives both in circRNA research and therapeutics based on existing knowledge.
Collapse
Affiliation(s)
- Xu Liu
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Xuelin Yao
- Department of Endocrinology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
- Department of Endocrinology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Liang Chen
- Department of Cardiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| |
Collapse
|
7
|
Staropoli N, Scionti F, Farenza V, Falcone F, Luciano F, Renne M, Di Martino MT, Ciliberto D, Tedesco L, Crispino A, Labanca C, Cucè M, Esposito S, Agapito G, Cannataro M, Tassone P, Tagliaferri P, Arbitrio M. Identification of ADME genes polymorphic variants linked to trastuzumab-induced cardiotoxicity in breast cancer patients: Case series of mono-institutional experience. Biomed Pharmacother 2024; 174:116478. [PMID: 38547766 DOI: 10.1016/j.biopha.2024.116478] [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: 02/03/2024] [Revised: 03/18/2024] [Accepted: 03/19/2024] [Indexed: 05/01/2024] Open
Abstract
BACKGROUND Long-term survival induced by anticancer treatments discloses emerging frailty among breast cancer (BC) survivors. Trastuzumab-induced cardiotoxicity (TIC) is reported in at least 5% of HER2+BC patients. However, TIC mechanism remains unclear and predictive genetic biomarkers are still lacking. Interaction between systemic inflammation, cytokine release and ADME genes in cancer patients might contribute to explain mechanisms underlying individual susceptibility to TIC and drug response variability. We present a single institution case series to investigate the potential role of genetic variants in ADME genes in HER2+BC patients TIC experienced. METHODS We selected data related to 40 HER2+ BC patients undergone to DMET genotyping of ADME constitutive variant profiling, with the aim to prospectively explore their potential role in developing TIC. Only 3 patients ("case series"), who experienced TIC, were compared to 37 "control group" matched patients cardiotoxicity-sparing. All patients underwent to left ventricular ejection fraction (LVEF) evaluation at diagnosis and during anti-HER2 therapy. Each single probe was clustered to detect SNPs related to cardiotoxicity. RESULTS In this retrospective analysis, our 3 cases were homogeneous in terms of clinical-pathological characteristics, trastuzumab-based treatment and LVEF decline. We identified 9 polymorphic variants in 8 ADME genes (UGT1A1, UGT1A6, UGT1A7, UGT2B15, SLC22A1, CYP3A5, ABCC4, CYP2D6) potentially associated with TIC. CONCLUSION Real-world TIC incidence is higher compared to randomized clinical trials and biomarkers with potential predictive value aren't available. Our preliminary data, as proof of concept, could suggest a predictive role of pharmacogenomic approach in the identification of cardiotoxicity risk biomarkers for anti-HER2 treatment.
Collapse
Affiliation(s)
- Nicoletta Staropoli
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy; Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Francesca Scionti
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Valentina Farenza
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Federica Falcone
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Francesco Luciano
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Maria Renne
- Surgery Unit, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy
| | - Maria Teresa Di Martino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Domenico Ciliberto
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy
| | - Ludovica Tedesco
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Antonella Crispino
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Caterina Labanca
- Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Maria Cucè
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy
| | - Stefania Esposito
- Pharmacy Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Campus Salvatore Venuta, Catanzaro, Italy
| | - Giuseppe Agapito
- Department of Law, Economics and Sociology, Magna Graecia University of Catanzaro, Catanzaro 88100, Italy; Data Analytics Research Center, Magna Graecia University of Catanzaro, Catanzaro 88100, Italy
| | - Mario Cannataro
- Department of Medical and Surgical Science, Magna Graecia University of Catanzaro, Catanzaro 88100, Italy
| | - Pierfrancesco Tassone
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy; Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy
| | - Pierosandro Tagliaferri
- Medical Oncology Unit, R. Dulbecco (Mater Domini facility), Teaching Hospital, Magna Græcia University and Cancer Center, Campus Salvatore Venuta, Catanzaro, Italy; Department of Experimental and Clinical Medicine, Magna Graecia University, Catanzaro, Italy.
| | - Mariamena Arbitrio
- Institute for Biomedical Research and Innovation (IRIB), National Research Council of Italy (CNR), Catanzaro 88100, Italy.
| |
Collapse
|
8
|
Chan JSK, Chan RNC, Lee YHA, Satti DI, Dee EC, Ng K, Achim A, Ng CF, Liu T, Matthews GDK, Tse G, Vassiliou VS. Cardiovascular health of patients with cancer: Challenges abound. Trends Cardiovasc Med 2024:S1050-1738(24)00036-7. [PMID: 38657744 DOI: 10.1016/j.tcm.2024.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Patients with cancer have elevated cardiovascular risks compared to those without cancer. As cancer incidence increases and cancer-related mortality decreases, cardiovascular diseases in patients with a history of cancer will become increasingly important. This in turn is reflected by the exponentially increasing amount of cardio-oncology research in recent years. This narrative review aims to summarize the key existing literature in several main areas of cardio-oncology, including the epidemiology, natural history, prevention, management, and determinants of the cardiovascular health of patients with cancer, and identify relevant gaps in evidence for further research.
Collapse
Affiliation(s)
- Jeffrey Shi Kai Chan
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China
| | - Raymond Ngai Chiu Chan
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China
| | - Yan Hiu Athena Lee
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China; Division of Urology, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China; SH Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Danish Iltaf Satti
- Cardio-Oncology Research Unit, Cardiovascular Analytics Group, PowerHealth Research Institute, Hong Kong, PR China; Division of Cardiology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward Christopher Dee
- Department of Radiation Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Kenrick Ng
- Department of Medical Oncology, Barts Cancer Centre, London, UK
| | - Alexandru Achim
- Department of Internal Medicine, Invasive Cardiology Division, University of Szeged, Szeged, Hungary; Department of Cardiology, "Niculae Stancioiu" Heart Institute, University of Medicine and Pharmacy "Iuliu Hatieganu", Cluj-Napoca, Romania
| | - Chi Fai Ng
- Division of Urology, Department of Surgery, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, PR China; SH Ho Urology Centre, The Chinese University of Hong Kong, Hong Kong, PR China
| | - Tong Liu
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, PR China
| | - Gareth D K Matthews
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich, UK; Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich, UK
| | - Gary Tse
- Tianjin Key Laboratory of Ionic-Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of Cardiology, Second Hospital of Tianjin Medical University, Tianjin 300211, PR China; Kent and Medway Medical School, Canterbury, Kent CT2 7NT, UK; School of Nursing and Health Studies, Hong Kong Metropolitan University, Hong Kong, PR China.
| | - Vassilios S Vassiliou
- Norwich Medical School, University of East Anglia, Norwich Research Park, Rosalind Franklin Road, Norwich, UK; Department of Cardiology, Norfolk and Norwich University NHS Foundation Trust, Colney Lane, Norwich, UK
| |
Collapse
|
9
|
Vakilpour A, Lefebvre B, Lai C, Scherrer-Crosbie M. Heartbreaker: Detection and prevention of cardiotoxicity in hematological malignancies. Blood Rev 2024; 64:101166. [PMID: 38182490 DOI: 10.1016/j.blre.2023.101166] [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: 10/30/2023] [Revised: 12/12/2023] [Accepted: 12/30/2023] [Indexed: 01/07/2024]
Abstract
Cancer survivors are at significant risk of cardiovascular (CV) morbidity and mortality; patients with hematologic malignancies have a higher rate of death due to heart failure compared to all other cancer subtypes. The majority of conventional hematologic cancer treatments is associated with increased risk of acute and long-term CV toxicity. The incidence of cancer therapy induced CV toxicity depends on the combination of patient characteristics and on the type, dose, and duration of the therapy. Early diagnosis of CV toxicity, appropriate referral, more specific cardiac monitoring follow-up and timely interventions in target patients can decrease the risk of CV adverse events, the interruption of oncological therapy, and improve the patient's prognosis. Herein, we summarize the CV effects of conventional treatments used in hematologic malignancies with a focus on definitions and incidence of the most common CV toxicities, guideline recommended early detection approaches, and preventive strategies before and during cancer treatments.
Collapse
Affiliation(s)
- Azin Vakilpour
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA.
| | - Bénédicte Lefebvre
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; The Thalheimer Center for Cardio-oncology, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| | - Catherine Lai
- Division of Hematology-Oncology, Perelman Center for Advanced Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Marielle Scherrer-Crosbie
- Division of Cardiovascular Diseases, Department of Medicine, Hospital of the University of Pennsylvania, Philadelphia, PA, USA; The Thalheimer Center for Cardio-oncology, Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
10
|
Tamargo J, Villacastín J, Caballero R, Delpón E. Drug-induced atrial fibrillation. A narrative review of a forgotten adverse effect. Pharmacol Res 2024; 200:107077. [PMID: 38244650 DOI: 10.1016/j.phrs.2024.107077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 01/22/2024]
Abstract
Atrial fibrillation (AF) is the most common cardiac arrhythmia and is associated with an increased morbidity and mortality. There is clinical evidence that an increasing number of cardiovascular and non-cardiovascular drugs, mainly anticancer drugs, can induce AF either in patients with or without pre-existing cardiac disorders, but drug-induced AF (DIAF) has not received the attention that it might deserve. In many cases DIAF is asymptomatic and paroxysmal and patients recover sinus rhythm spontaneously, but sometimes, DIAF persists, and it is necessary to perform a cardioversion. Furthermore, DIAF is not mentioned in clinical guidelines on the treatment of AF. The risk of DIAF increases in elderly and in patients treated with polypharmacy and with risk factors and comorbidities that commonly coexist with AF. This is the case of cancer patients. Under these circumstances ascribing causality of DIAF to a given drug often represents a clinical challenge. We review the incidence, the pathophysiological mechanisms, risk factors, clinical relevance, and treatment of DIAF. Because of the limited information presently available, further research is needed to obtain a deeper insight into DIAF. Meanwhile, it is important that clinicians are aware of the problem that DIAF represents, recognize which drugs may cause DIAF, and consider the possibility that a drug may be responsible for a new-onset AF episode.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| | - Julián Villacastín
- Hospital Clínico San Carlos, CardioRed1, Universidad Complutense de Madrid, CIBERCV, 28040 Madrid, Spain
| | - Ricardo Caballero
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain.
| | - Eva Delpón
- Department of Pharmacology and Toxicology, School of Medicine, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Gregorio Marañón, CIBERCV, 28040 Madrid, Spain
| |
Collapse
|
11
|
Galimzhanov A, Istanbuly S, Tun HN, Ozbay B, Alasnag M, Ky B, Lyon AR, Kayikcioglu M, Tenekecioglu E, Panagioti M, Kontopantelis E, Abdel-Qadir H, Mamas MA. Cardiovascular outcomes in breast cancer survivors: a systematic review and meta-analysis. Eur J Prev Cardiol 2023; 30:2018-2031. [PMID: 37499186 DOI: 10.1093/eurjpc/zwad243] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 06/19/2023] [Accepted: 07/21/2023] [Indexed: 07/29/2023]
Abstract
AIMS It is unclear whether the future risk of cardiovascular events in breast cancer (Bc) survivors is greater than in the general population. This meta-analysis quantifies the risk of cardiovascular disease development in Bc patients, compared to the risk in a general matched cancer-free population, and reports the incidence of cardiovascular events in patients with Bc. METHODS AND RESULTS We searched PubMed, Scopus, and Web of Science databases (up to 23 March 2022) for observational studies and post hoc analyses of randomized controlled trials. Cardiovascular death, heart failure (HF), atrial fibrillation (AF), coronary artery disease (CAD), myocardial infarction (MI), and stroke were the individual endpoints for our meta-analysis. We pooled incidence rates (IRs) and risk in hazard ratios (HRs), using random-effects meta-analyses. Heterogeneity was reported through the I2 statistic, and publication bias was examined using funnel plots and Egger's test in the meta-analysis of risk. One hundred and forty-two studies were identified in total, 26 (836 301 patients) relevant to the relative risk and 116 (2 111 882 patients) relevant to IRs. Compared to matched cancer-free controls, Bc patients had higher risk for cardiovascular death within 5 years of cancer diagnosis [HR = 1.09; 95% confidence interval (CI): 1.07, 1.11], HF within 10 years (HR = 1.21; 95% CI: 1.1, 1.33), and AF within 3 years (HR = 1.13; 95% CI: 1.05, 1.21). The pooled IR for cardiovascular death was 1.73 (95% CI 1.18, 2.53), 4.44 (95% CI 3.33, 5.92) for HF, 4.29 (95% CI 3.09, 5.94) for CAD, 1.98 (95% CI 1.24, 3.16) for MI, 4.33 (95% CI 2.97, 6.30) for stroke of any type, and 2.64 (95% CI 2.97, 6.30) for ischaemic stroke. CONCLUSION Breast cancer exposure was associated with the increased risk for cardiovascular death, HF, and AF. The pooled incidence for cardiovascular endpoints varied depending on population characteristics and endpoint studied. REGISTRATION CRD42022298741.
Collapse
Affiliation(s)
- Akhmetzhan Galimzhanov
- Department of Propedeutics of Internal Disease, Semey Medical University, Semey, Kazakhstan
- Keele Cardiovascular Research Group, Center for Prognosis Research, Keele University, Stoke on Trent, Keele, UK
| | - Sedralmontaha Istanbuly
- Keele Cardiovascular Research Group, Center for Prognosis Research, Keele University, Stoke on Trent, Keele, UK
- Faculty of Medicine, University of Aleppo, Aleppo, Syrian Arab Republic
| | - Han Naung Tun
- Larner College of Medicine, University of Vermont, Burlington, VT, USA
| | - Benay Ozbay
- Basaksehir Cam and Sakura State Hospital Department of Cardiology, Istanbul, Turkey
- Division of Cardiology, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | | | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and National Heart and Lung Institute, Imperial College London, London, UK
| | - Meral Kayikcioglu
- Department of Cardiology, Faculty of Medicine, 60521 Ege University, Izmir, Turkey
| | - Erhan Tenekecioglu
- Department of Cardiology, Bursa Yuksek İhtisas Training and Research Hospital, Health Sciences University, Bursa, Turkey
- Department of Cardiology, Erasmus MC, Thorax Center, Erasmus University, Rotterdam, The Netherlands
| | - Maria Panagioti
- Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, National Institute for Health Research School for Primary Care Research, University of Manchester, Manchester, UK
| | - Evangelos Kontopantelis
- Division of Population Health, Health Services Research and Primary Care, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, National Institute for Health Research School for Primary Care Research, University of Manchester, Manchester, UK
| | - Husam Abdel-Qadir
- Department of Medicine and Institute of Health Policy, Management and Evaluation, University of Toronto, Canada
| | - Mamas A Mamas
- Keele Cardiovascular Research Group, Center for Prognosis Research, Keele University, Stoke on Trent, Keele, UK
| |
Collapse
|
12
|
Costa A, Ameri P. Sexual dimorphism in doxorubicin cardiotoxicity: two sides of a complex coin. Am J Physiol Heart Circ Physiol 2023; 325:H949-H951. [PMID: 37712921 DOI: 10.1152/ajpheart.00566.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Accepted: 09/12/2023] [Indexed: 09/16/2023]
Affiliation(s)
- Ambra Costa
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
| | - Pietro Ameri
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| |
Collapse
|
13
|
Hu Y, Ma P, Chen L, Liu J, Shang Y, Jian W. Multi-parameter cardiac magnetic resonance imaging detects anthracycline-induced cardiotoxicity in rabbits model. Heliyon 2023; 9:e21845. [PMID: 38058655 PMCID: PMC10695840 DOI: 10.1016/j.heliyon.2023.e21845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 10/29/2023] [Accepted: 10/30/2023] [Indexed: 12/08/2023] Open
Abstract
Background Cardiac magnetic resonance (CMR) quantitative T1 and T2 mapping offers a non-invasive means to evaluate early cardiotoxicity changes. This study aimed to pinpoint the earliest CMR indicators of myocardial injury in Anthracycline-induced cardiotoxicity (AIC) and to elucidate the connections between these CMR indicators and associated pathological indicators. Methods A total of 34 rabbits were administered doxorubicin at a dosage of 1 mg/kg/weekly. The study incorporated six 3T CMR scan time points: baseline, and at intervals of four, six, eight, twelve, and sixteen weeks. Cine, T1 and T2 mapping sequences assessed the left ventricular ejection fraction (LVEF), native T1, extracellular volume fraction (ECV), and T2 values. Following each time point, three rabbits were sacrificed for histological analysis involving Hematoxylin and eosin (H&E), Masson, TUNEL, and microvascular density (MVD) stains. Spearman correlations and linear mixed model analysis served in the statistical analysis. Results Diverse degrees of alternation were recorded in LVEF, native T1, T2, and ECV over time. LVEF declined to 49.0 ± 2.6 % at 12 weeks from the baseline of 53.4 ± 3.2 %, p < 0.001. Native T1 values increase from the baseline (1396.5 ± 79.2 ms) until 8 weeks (1498.8 ± 95.4 ms, p < 0.001). T2 values increased from the baseline (36.6 ± 3.3 ms) within 4 weeks of initiation (37.5 ± 3.4, p = 0.02) and remained elevated through 16 weeks (42.8 ± 0.3, p < 0.01). ECV was elevated at 8 weeks (33.9 ± 3.8 %, p = 0.005) compared to the baseline (30.2 ± 2.5 %). By week 12, myocardial edema and increased CVF were apparent (p = 0.04 and = 0.001, respectively). The area under ROC curve for positive CMR presence and the gold standards were 0.87 (T2-ROC, 4 weeks) and 0.92 (LVEF&BNP-ROC, 12 weeks). Conclusion T1 and T2 mapping are effective tools for cardiotoxicity detection and monitoring. The prolongation of T2 value emerged as the most consistent and early-onset indicator.
Collapse
Affiliation(s)
- Yurou Hu
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Peisong Ma
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lin Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Juan Liu
- Department of Ultrasound, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Yongning Shang
- Department of Ultrasound, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Wang Jian
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
- 7T Magnetic Resonance Imaging Translational Medical Center, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| |
Collapse
|
14
|
Nunes JHC, Cella PS, Guimarães TAS, Buçu IP, Deminice R. Chemotherapy periodization to maximize resistance training adaptations in oncology. Cancer Chemother Pharmacol 2023; 92:357-367. [PMID: 37582913 DOI: 10.1007/s00280-023-04576-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: 04/05/2023] [Accepted: 08/06/2023] [Indexed: 08/17/2023]
Abstract
INTRODUCTION Engaging in exercise programs during cancer treatment is challenging due to the several chemotherapy-induced side effects. Using a pre-clinical model that mimics chemotherapy treatment, we investigated if a periodized-within-chemotherapy training strategy can maximize resistance training (RT) adaptations such as increasing muscle mass and strength. METHODS Swiss mice were randomly allocated into one of the following five groups (n = 14): control (C), resistance training (RT), chemotherapy-treated non-exercised group (Ch), resistance training chemotherapy treated (RTCh), and resistance training periodized-within-chemotherapy (RTPCh). Doxorubicin (i.p.) was weekly injected for a total of 3 weeks (total dose of 12 mg/kg). Resistance training consisted of ladder climbing with progressive intensity, three times a week for 3 weeks, during chemotherapy treatment. RTPCh prescriptions considered "bad day" adjustments while RTCh did not. "Bad day" adjustments considered the presence or absence of clinical signs (e.g., severe weight loss, diarrhea, mice refusing to train) to replace RT sessions. At the end of the third week, animals were euthanized. RESULTS Weekly doxorubicin injection promoted progressive body weight loss, muscle atrophy, strength loss, local oxidative stress, and elevated inflammatory mediators, such as TNF-α and IL-6. Non-periodized-within-chemotherapy RT promoted mild protection against doxorubicin-induced skeletal muscle disturbances; moreover, when periodized-within-chemotherapy was applied, RT prevented elevated skeletal muscle inflammatory mediators and oxidative damage markers and promoted muscle mass and strength gains. CONCLUSION Considering chemotherapy-induced side effects is a crucial aspect when prescribing resistance exercise during cancer, it will maximize the effectiveness of exercise in enhancing muscle mass and strength.
Collapse
Affiliation(s)
- Jonathan H C Nunes
- Department of Physical Education, Faculty of Physical Education and Sport, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445 Km 380, Campus Universitário, Londrina, Paraná, Brazil
| | - Paola S Cella
- Department of Physical Education, Faculty of Physical Education and Sport, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445 Km 380, Campus Universitário, Londrina, Paraná, Brazil
| | - Tatiana A S Guimarães
- Department of Physical Education, Faculty of Physical Education and Sport, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445 Km 380, Campus Universitário, Londrina, Paraná, Brazil
| | - Icaro P Buçu
- Department of Physical Education, Faculty of Physical Education and Sport, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445 Km 380, Campus Universitário, Londrina, Paraná, Brazil
| | - Rafael Deminice
- Department of Physical Education, Faculty of Physical Education and Sport, State University of Londrina, Rodovia Celso Garcia Cid, Pr 445 Km 380, Campus Universitário, Londrina, Paraná, Brazil.
| |
Collapse
|
15
|
Chen L, Byer SH, Holder R, Wu L, Burkey K, Shah Z. Wnt10b protects cardiomyocytes against doxorubicin-induced cell death via MAPK modulation. PLoS One 2023; 18:e0277747. [PMID: 37856516 PMCID: PMC10586692 DOI: 10.1371/journal.pone.0277747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 11/02/2022] [Indexed: 10/21/2023] Open
Abstract
BACKGROUND Doxorubicin, an anthracycline chemotherapeutic known to incur heart damage, decreases heart function in up to 11% of patients. Recent investigations have implicated the Wnt signaling cascade as a key modulator of cardiac tissue repair after myocardial infarction. Wnt upregulation in murine models resulted in stimulation of angiogenesis and suppression of fibrosis after ischemic insult. However, the molecular mechanisms of Wnt in mitigating doxorubicin-induced cardiac insult require further investigation. Identifying cardioprotective mechanisms of Wnt is imperative to reducing debilitating cardiovascular adverse events in oncologic patients undergoing treatment. METHODS Exposing human cardiomyocyte AC16 cells to varying concentrations of Wnt10b and DOX, we observed key metrics of cell viability. To assess the viability and apoptotic rates, we utilized MTT and TUNEL assays. We quantified cell and mitochondrial membrane stability via LDH release and JC-1 staining. To investigate how Wnt10b mitigates doxorubicin-induced apoptosis, we introduced pharmacologic inhibitors of key enzymes involved in apoptosis: FR180204 and SB203580, ERK1/2 and p38 inhibitors. Further, we quantified apoptotic executor enzymes, caspase 3/7, via immunofluorescence. RESULTS AC16 cells exposed solely to doxorubicin were shrunken with distorted morphology. Cardioprotective effects of Wnt10b were demonstrated via a reduction in apoptosis, from 70.1% to 50.1%. LDH release was also reduced between doxorubicin and combination groups from 2.27-fold to 1.56-fold relative to the healthy AC16 control group. Mitochondrial membrane stability was increased from 0.67-fold in the doxorubicin group to 5.73 in co-treated groups relative to control. Apoptotic protein expression was stifled by Wnt10b, with caspase3/7 expression reduced from 2.4- to 1.3-fold, and both a 20% decrease in p38 and 40% increase in ERK1/2 activity. CONCLUSION Our data with the AC16 cell model demonstrates that Wnt10b provides defense mechanisms against doxorubicin-induced cardiotoxicity and apoptosis. Further, we explain a mechanism of this beneficial effect involving the mitochondria through simultaneous suppression of pro-apoptotic p38 and anti-apoptotic ERK1/2 activities.
Collapse
Affiliation(s)
- Lei Chen
- Department of Cardiovascular Medicine, University of Kansas School of Medicine, Kansas City, KS, United States of America
| | - Stefano H. Byer
- Department of Cardiovascular Medicine, University of Kansas School of Medicine, Kansas City, KS, United States of America
| | - Rachel Holder
- Department of Cardiovascular Medicine, University of Kansas School of Medicine, Kansas City, KS, United States of America
| | - Lingyuan Wu
- Department of Cardiovascular Medicine, University of Kansas School of Medicine, Kansas City, KS, United States of America
| | - Kyley Burkey
- Department of Cardiovascular Medicine, University of Kansas School of Medicine, Kansas City, KS, United States of America
| | - Zubair Shah
- Department of Cardiovascular Medicine, University of Kansas School of Medicine, Kansas City, KS, United States of America
| |
Collapse
|
16
|
Pavlovic D, Niciforovic D, Papic D, Milojevic K, Markovic M. CDK4/6 inhibitors: basics, pros, and major cons in breast cancer treatment with specific regard to cardiotoxicity - a narrative review. Ther Adv Med Oncol 2023; 15:17588359231205848. [PMID: 37841752 PMCID: PMC10571689 DOI: 10.1177/17588359231205848] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/18/2023] [Indexed: 10/17/2023] Open
Abstract
Breast cancer is characterized by the uncontrolled proliferation of breast cells, with a high incidence reported in 2020 to have affected over 2 million women. In recent years, the conventional methods of treating breast cancer have involved radiotherapy and chemotherapy. However, the emergence of CDK4/6 inhibitors has shown potential as a promising cancer therapy. Cyclin-dependent kinases (CDK) inhibitors are a class of molecules that impede the formation of an active kinase complex, thereby hindering its activity and consequently halting the progression of the cell cycle. It was discovered that they have a significant impact on impeding the progression of the cancer. This is evident with the Food and Drug Administration's approval of drugs such as palbociclib, ribociclib, and abemaciclib for hormone receptor-positive metastatic breast cancer in combination with specific endocrine therapies. In spite of enormous success in breast cancer treatment, certain obstacles have emerged, such as therapy resistance, side effects, and most of all, cardiotoxicity. Some of these drawbacks have been successfully overcome by dosage reduction, different combinations of the drugs, and the assessment of each patient's condition and suitability prior to treatment. Yet other drawbacks still require tenacious research, especially certain cases of cardiotoxicities. This article delves into the biological mechanisms of CDK4/6 in the cell cycle and cancer, as well as the clinical advantages and most common adverse events (AEs) associated with CDK4/6 inhibitors. The primary objective of this review is to provide a comprehensive analysis of cardiotoxic AEs and elucidate the underlying pathophysiological mechanisms responsible for the cardiotoxicity of CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Dragica Pavlovic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, 69 Svetozar Markovic Street, Kragujevac 34000, Serbia
| | - Danijela Niciforovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Dragana Papic
- Department of Genetics, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| | - Katarina Milojevic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
| | - Marina Markovic
- Center for Internal Oncology, University Clinical Center Kragujevac, Kragujevac, Serbia
- Department of Internal Medicine, Faculty of Medical Sciences, University of Kragujevac, Kragujevac, Serbia
| |
Collapse
|
17
|
Lyu Y, Bennamoun M, Sharif N, Lip GYH, Dwivedi G. Artificial Intelligence in the Image-Guided Care of Atrial Fibrillation. Life (Basel) 2023; 13:1870. [PMID: 37763273 PMCID: PMC10532509 DOI: 10.3390/life13091870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/19/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
Atrial fibrillation arises mainly due to abnormalities in the cardiac conduction system and is associated with anatomical remodeling of the atria and the pulmonary veins. Cardiovascular imaging techniques, such as echocardiography, computed tomography, and magnetic resonance imaging, are crucial in the management of atrial fibrillation, as they not only provide anatomical context to evaluate structural alterations but also help in determining treatment strategies. However, interpreting these images requires significant human expertise. The potential of artificial intelligence in analyzing these images has been repeatedly suggested due to its ability to automate the process with precision comparable to human experts. This review summarizes the benefits of artificial intelligence in enhancing the clinical care of patients with atrial fibrillation through cardiovascular image analysis. It provides a detailed overview of the two most critical steps in image-guided AF management, namely, segmentation and classification. For segmentation, the state-of-the-art artificial intelligence methodologies and the factors influencing the segmentation performance are discussed. For classification, the applications of artificial intelligence in the diagnosis and prognosis of atrial fibrillation are provided. Finally, this review also scrutinizes the current challenges hindering the clinical applicability of these methods, with the aim of guiding future research toward more effective integration into clinical practice.
Collapse
Affiliation(s)
- Yiheng Lyu
- Department of Computer Science and Software Engineering, School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA 6009, Australia; (Y.L.); (M.B.)
- Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Mohammed Bennamoun
- Department of Computer Science and Software Engineering, School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA 6009, Australia; (Y.L.); (M.B.)
| | - Naeha Sharif
- Department of Computer Science and Software Engineering, School of Physics, Mathematics and Computing, The University of Western Australia, Perth, WA 6009, Australia; (Y.L.); (M.B.)
| | - Gregory Y. H. Lip
- Liverpool Centre for Cardiovascular Science, University of Liverpool, Liverpool L69 3BX, UK
- Liverpool John Moores University, Liverpool L3 5UX, UK
- Liverpool Heart and Chest Hospital, Liverpool L14 3PE, UK
- Danish Center for Health Services Research, Department of Clinical Medicine, Aalborg University, 9220 Aalborg, Denmark
| | - Girish Dwivedi
- Harry Perkins Institute of Medical Research, The University of Western Australia, Perth, WA 6009, Australia
- Department of Cardiology, Fiona Stanley Hospital, Perth, WA 6150, Australia
- Medical School, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
18
|
Chan SHY, Khatib Y, Webley S, Layton D, Salek S. Identification of cardiotoxicity related to non-small cell lung cancer (NSCLC) treatments: A systematic review. Front Pharmacol 2023; 14:1137983. [PMID: 37383708 PMCID: PMC10294714 DOI: 10.3389/fphar.2023.1137983] [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: 01/05/2023] [Accepted: 03/27/2023] [Indexed: 06/30/2023] Open
Abstract
Introduction: In the last few decades, there has been a rapid development in cancer therapies and improved detection strategies, hence the death rates caused by cancer have decreased. However, it has been reported that cardiovascular disease has become the second leading cause of long-term morbidity and fatality among cancer survivors. Cardiotoxicity from anticancer drugs affects the heart's function and structure and can occur during any stage of the cancer treatments, which leads to the development of cardiovascular disease. Objectives: To investigate the association between anticancer drugs for non-small cell lung cancer (NSCLC) and cardiotoxicity as to whether: different classes of anticancer drugs demonstrate different cardiotoxicity potentials; different dosages of the same drug in initial treatment affect the degree of cardiotoxicity; and accumulated dosage and/or duration of treatments affect the degree of cardiotoxicity. Methods: This systematic review included studies involving patients over 18 years old with NSCLC and excluded studies in which patients' treatments involve radiotherapy only. Electronic databases and registers including Cochrane Library, National Cancer Institute (NCI) Database, PubMed, Scopus, Web of Science, ClinicalTrials.gov and the European Union Clinical Trials Register were systematically searched from the earliest available date up until November 2020. A full version protocol of this systematic review (CRD42020191760) had been published on PROSPERO. Results: A total of 1785 records were identified using specific search terms through the databases and registers; 74 eligible studies were included for data extraction. Based on data extracted from the included studies, anticancer drugs for NSCLC that are associated with cardiovascular events include bevacizumab, carboplatin, cisplatin, crizotinib, docetaxel, erlotinib, gemcitabine and paclitaxel. Hypertension was the most reported cardiotoxicity as 30 studies documented this cardiovascular adverse event. Other reported treatment-related cardiotoxicities include arrhythmias, atrial fibrillation, bradycardia, cardiac arrest, cardiac failure, coronary artery disease, heart failure, ischemia, left ventricular dysfunction, myocardial infarction, palpitations, and tachycardia. Conclusion: The findings of this systematic review have provided a better understanding of the possible association between cardiotoxicities and anticancer drugs for NSCLC. Whilst variation is observed across different drug classes, the lack of information available on cardiac monitoring can result in underestimation of this association. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42020191760, identifier PROSPERO CRD42020191760.
Collapse
Affiliation(s)
- Stefanie Ho Yi Chan
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Yasmin Khatib
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Sherael Webley
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| | - Deborah Layton
- IQVIA UK, London, United Kingdom
- PEPI Consultancy Limited, Southampton, United Kingdom
- University of Keele, Keele, United Kingdom
| | - Sam Salek
- School of Life and Medical Sciences, University of Hertfordshire, Hatfield, United Kingdom
| |
Collapse
|
19
|
Nam J, Schirmer AU, Loh C, Drewry DH, Macias E. Targeting the Divergent Roles of STK3 Inhibits Breast Cancer Cell Growth and Opposes Doxorubicin-Induced Cardiotoxicity In Vitro. Cancers (Basel) 2023; 15:2817. [PMID: 37345153 DOI: 10.3390/cancers15102817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Revised: 04/26/2023] [Accepted: 05/06/2023] [Indexed: 06/23/2023] Open
Abstract
Breast cancer (BCa) is the most prevalent type of cancer in women. Several therapies used in the treatment of breast cancer are associated with clinically important rates of cardiovascular toxicity during or after treatment exposure, including anthracyclines. There is a need for new BCa therapeutics and treatments that mitigate chemotherapy-induced cardiotoxicity in BCa. In this study, we examine the effects of Serine/Threonine Kinase 3 (STK3) inhibition in the context of BCa therapy and cardioprotection from doxorubicin. STK3 (also known as MST2) is a key member of the Hippo Tumor-Suppressor Pathway, which regulates cell growth and proliferation by inhibiting YAP/TAZ co-transcription factors. Canonically, STK3 should restrict BCa growth; however, we observed that STK3 is amplified in BCa and associated with worse patient outcomes, suggesting a noncanonical pro-tumorigenic role. We found BCa cell lines have varying dependence on STK3. SUM52PE cells had the highest expression and dependence on STK3 in genetic and pharmacological assays. MCF-7 and MDA-MB-231 were less sensitive to STK3 targeting in standard proliferation assays, but were STK3 dependent in colony formation and matrigel invasion assays. In contrast, STK3 inhibition mitigated the toxic effects of doxorubicin in H9C2 rat cardiomyocytes by increasing YAP expression. Importantly, STK3 inhibition in BCa cells did not interfere with the therapeutic effects of doxorubicin. Our studies highlight STK3 is a potential molecular target for BCa with dual therapeutic effects: suppression of BCa growth and progression, and chemoprotection in cardiomyocytes.
Collapse
Affiliation(s)
- Jiung Nam
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Amelia U Schirmer
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Chelsea Loh
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| | - David H Drewry
- Structural Genomics Consortium and Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Lineberger Comprehensive Cancer Center, Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Everardo Macias
- Department of Pathology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
20
|
Costa Godinho LRL, Cella PS, Guimarães TAS, Palma GHD, Nunes JHC, Deminice R. Creatine Supplementation Potentiates Exercise Protective Effects against Doxorubicin-Induced Hepatotoxicity in Mice. Antioxidants (Basel) 2023; 12:antiox12040823. [PMID: 37107198 PMCID: PMC10135080 DOI: 10.3390/antiox12040823] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2023] [Revised: 03/20/2023] [Accepted: 03/23/2023] [Indexed: 03/30/2023] Open
Abstract
We tested the hypothesis that creatine supplementation may potentiate exercise’s protective effects against doxorubicin-induced hepatotoxicity. Thirty-eight Swiss mice were randomly allocated into five groups: control (C, n = 7), exercised (Ex, n = 7), treated with doxorubicin (Dox, n = 8), treated with doxorubicin and exercised (DoxEx, n = 8), and treated with doxorubicin, exercised, and supplemented with creatine (DoxExCr, n = 8). Doxorubicin was administered weekly (i.p.) for a total dose of 12 mg/kg. Creatine supplementation (2% added to the diet) and strength training (climbing stairs, 3 times a week) were performed for a total of 5 weeks. The results demonstrated that doxorubicin caused hepatotoxicity, which was evidenced by increased (p < 0.05) hepatic markers of inflammation (i.e., TNF-α and IL-6) and oxidative damage, while the redox status (GSH/GSSG) was reduced. The plasma concentrations of liver transaminases were also significantly (p < 0.05) elevated. Furthermore, doxorubicin-treated animals presented hepatic fibrosis and histopathological alterations such as cellular degeneration and the infiltration of interstitial inflammatory cells. Exercise alone partly prevented doxorubicin-induced hepatotoxicity; thus, when combined with creatine supplementation, exercise was able to attenuate inflammation and oxidative stress, morphological alterations, and fibrosis. In conclusion, creatine supplementation potentiates the protective effects of exercise against doxorubicin-induced hepatotoxicity in mice.
Collapse
|
21
|
Oikawa M, Ishida T, Takeishi Y. Cancer therapeutics-related cardiovascular dysfunction: Basic mechanisms and clinical manifestation. J Cardiol 2023; 81:253-259. [PMID: 35589463 DOI: 10.1016/j.jjcc.2022.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 04/14/2022] [Indexed: 02/01/2023]
Abstract
Although recent advances in cancer treatment improve cancer prognosis, cancer therapeutics-related cardiovascular dysfunction (CTRCD) significantly contributes to the global burden of cardiovascular disease. CTRCD causes two crucial issues: first, premature treatment interruption or discontinuation of chemotherapy; second, the development of congestive heart failure during and after cancer treatment. Thus, early detection and prompt treatment of CTRCD may improve the prognosis in cancer patients. This review covers representative anticancer drugs, including anthracyclines, human epidermal growth factor 2 inhibitors, tyrosine kinase inhibitors, proteasome inhibitors, and immune checkpoint inhibitors. We focus on the molecular mechanisms of CTRCD and various approaches to diagnosis, prevention, monitoring, and treatment.
Collapse
Affiliation(s)
- Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University, 1-Hikarigaoka, Fukushima, Fukushima prefecture 960-1295, Japan.
| | - Takafumi Ishida
- Department of Cardiovascular Medicine, Fukushima Medical University, 1-Hikarigaoka, Fukushima, Fukushima prefecture 960-1295, Japan
| | - Yasuchika Takeishi
- Department of Cardiovascular Medicine, Fukushima Medical University, 1-Hikarigaoka, Fukushima, Fukushima prefecture 960-1295, Japan
| |
Collapse
|
22
|
García-Cruz JC, García-Cruz E, Baranda-Tovar FM. Cardiotoxicity and cancer: prudential judgment in medical practice. Rev Salud Publica (Bogota) 2023; 22:346-350. [PMID: 36753162 DOI: 10.15446/rsap.v22n3.84770] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 05/30/2020] [Indexed: 11/09/2022] Open
Abstract
The objective of this paper is to propose a conceptual tool for consideration by medical professionals and cardiologists, based on the concept of prudential judgment or Aristotelian phronesis to confront the problems of cardiotoxicity resulting from cancer treatments. We start by analyzing the case of a young female patient who received two types of therapies: the first with anthracyclines (adriamycin), which produces type I damage, as stated in the consensus of 2014; and the second treatment, one month later, with trastuzumab, an agent that produces type II damage not dependent on dose. In this case, the patient manifested acute cardiac insufficiency, with a decrease of LVEF to 28% on the echocardiogram and to 27% on magnetic resonance imaging. Reports have indicated that treatment with beta blockers and the suspension or decrease of the dose limits damage, but during preclinical stages. Awareness and early attention to subclinical damage have thus become extremely relevant to substantiate treatments based not only on clinical evidence but also on the ability of medical professionals to rely on prudential judgment--which moves away from the medical practices that are developed on a daily basis in order to influence and reduce the cases of irreversible heart failure known as cardiotoxicity.
Collapse
Affiliation(s)
- Juan C García-Cruz
- JG: Bachelor in Communication and Journalism. M. Sc. Philosophy of Science (UNAM/Mexico). M.Sc. Philosophy, Science and Values (UPV/Spain), Ph. D. Philosophy of Science (UNAM/Mexico). CONACYT-Universidad Autónoma Metropolitana Xochimilco. Mexico City, Mexico.
| | - Edgar García-Cruz
- EG: Medical Surgeon. Specialty in Internal Medicine, Subspecialty in Clinical Cardiology, High specialty in Intensive Cardiology Therapy, Echocardiography, Adult Congenital Heart Diseases (Spain). Instituto Nacional de Cardiología "Ignacio Chávez". Mexico City, Mexico.
| | - Francisco M Baranda-Tovar
- FB: General Medicine. Specialty in Internal Medicine, Subspecialty in Clinical Cardiology, Intensive Cardiovascular Therapy, Postgraduate in Heart Transplantation (Argentina), Heart Failure, and Transplants (Spain). Instituto Nacional de Cardiología "Ignacio Chávez". Mexico City, Mexico.
| |
Collapse
|
23
|
Li L, Chen R, Zhou D, Sun J, Wang L, Zhu L, Shen H, Xie W, Ye X. The efficacy and cardiac toxicity of different-dose pegylated liposomal doxorubicin in elderly patients with diffuse large B lymphoma. Cancer Med 2023; 12:4184-4194. [PMID: 36200320 PMCID: PMC9972167 DOI: 10.1002/cam4.5280] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/25/2022] [Accepted: 08/12/2022] [Indexed: 11/08/2022] Open
Abstract
OBJECTIVES In order to explore the impact of pegylated liposomal doxorubicin (PLD) dose intensity on survival outcomes of newly diagnosed elderly patients with diffuse large B-cell lymphoma (DLBCL), we performed a retrospective study to compare the efficacy and adverse effects of RCEOP (70 mg/m2 ), RCdOP (20-30 mg/m2 ) and RCDOP (30-45 mg/m2 ). The optimal PLD dose of patients with different clinical characteristics of subgroups was explored to provide a clue for the selection of clinical PLD dose. METHODS A total of 335 DLBCL patients (60-85 years old) who were newly diagnosed and completed at least four cycles of RCE(D)OP were selected. The patients were mainly divided into RCEOP (126 cases) (epirubicin 70 mg/m2 ), RCdOP (151 cases) (PLD 20-30 mg/m2 ) and RCdOP (58 cases) (PLD 30-45 mg/m2 ). The effects of different doses of PLD on clinical efficacy, cardiotoxicity and prognosis of patients were retrospectively analyzed. Subgroup analysis was performed to compare the clinical characteristics of different subgroups. RESULTS Our study showed that PLD and epirubicin had similar efficacy (overall survival (OS) p = 0.776; progression-free survival (PFS) p = 0.959). RCDOP (30-45 mg/m2 PLD) group had a higher complete remission (CR) rate of 75.9% compared with RCdOP (20-30 mg/m2 PLD) group (P D vs. d = 0.018). In the overall population, there was no significant difference in survival between RCDOP and RCdOP groups (OS P D vs. d = 0.661; PFS P D vs. d = 0.212). In patients with underlying cardiovascular diseases, the PFS of the RCDOP group was significantly better than the RCdOP group (p = 0.043). Meanwhile, patients in the RCDOP group tended to have a better prognosis than those in the RCEOP group (OS: RCDOP vs. RCEOP p = 0.054, PFS: RCDOP vs. RCEOP p = 0.053). There was no significant difference in the incidence of cardiotoxicity and other adverse events among the three groups. For the low-risk (age-adjusted-International Prognostic Index = 0/1) old patients without cardiovascular disease, RCdOP was considered a better strategy in OS (p = 0.020). CONCLUSION In the general population, the CR rate in the RCDOP group was significantly higher than that in the RCdOP group (p = 0.018). For elderly DLBCL patients with cardiovascular disease, the effect benefit brought by the PLD dose was more obvious, and the PFS of the RCDOP group was significantly better than that of the RCdOP group (p = 0.043). Full dose of PLD is an efficient alternative in the treatment of patients with preexisting cardiovascular diseases.
Collapse
Affiliation(s)
- Li Li
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Rongrong Chen
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Program in Clinical Medicine, School of Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - De Zhou
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jianai Sun
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lulu Wang
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Program in Clinical Medicine, School of Medicine of Zhejiang University, Hangzhou, Zhejiang, China
| | - Lixia Zhu
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Huafei Shen
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wanzhuo Xie
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiujin Ye
- Department of Hematology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
24
|
Narayan HK, Sheline K, Wong V, Kuo D, Choo S, Yoon J, Leger K, Kutty S, Fradley M, Tremoulet A, Ky B, Armenian S, Guha A. Cardiovascular toxicities with pediatric tyrosine kinase inhibitor therapy: An analysis of adverse events reported to the Food and Drug Administration. Pediatr Blood Cancer 2023; 70:e30059. [PMID: 36385736 DOI: 10.1002/pbc.30059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 09/21/2022] [Accepted: 09/26/2022] [Indexed: 11/18/2022]
Abstract
We sought to examine cardiovascular toxicities associated with tyrosine kinase inhibitors in pediatrics. We examined 1624 pediatric adverse events with imatinib, dasatinib, sorafenib, pazopanib, crizotinib, and ruxolitinib reported to the Food and Drug Administration between January 1, 2015, and August 14, 2020. There were 102 cardiovascular event reports. Hypertension was the most commonly reported cardiovascular event and was most frequently associated with sorafenib and pazopanib. The presence of infection increased the reporting odds of cardiovascular events overall and specifically cardiac arrest, heart failure, and hypertension. These data provide early insight into cardiovascular toxicities with tyrosine kinase inhibitor use in pediatrics.
Collapse
Affiliation(s)
- Hari K Narayan
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Karyn Sheline
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Victor Wong
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Dennis Kuo
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Sun Choo
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Janet Yoon
- Department of Pediatrics, City of Hope, Duarte, California, USA
| | - Kasey Leger
- Department of Pediatrics, University of Washington, Seattle, Washington, USA
| | - Shelby Kutty
- Department of Pediatrics, Johns Hopkins University, Baltimore, Maryland, USA
| | - Michael Fradley
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Adriana Tremoulet
- Department of Pediatrics, University of California San Diego, La Jolla, California, USA
| | - Bonnie Ky
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Saro Armenian
- Department of Pediatrics, City of Hope, Duarte, California, USA
| | - Avirup Guha
- Cardio-Oncology Program, Georgia Cancer Center, Medical College of Georgia at Augusta University, Augusta, Georgia, USA.,Division of Cardiology, Department of Medicine, Medical College of Georgia at Augusta University, Augusta, Georgia, USA
| |
Collapse
|
25
|
Mihalcea D, Memis H, Mihaila S, Vinereanu D. Cardiovascular Toxicity Induced by Vascular Endothelial Growth Factor Inhibitors. Life (Basel) 2023; 13:life13020366. [PMID: 36836722 PMCID: PMC9965690 DOI: 10.3390/life13020366] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 01/26/2023] [Accepted: 01/27/2023] [Indexed: 01/31/2023] Open
Abstract
Cardiotoxicity is an important side effect of vascular endothelial growth factor (VEGF) inhibitors therapy used in the treatment of various malignancies, leading to increased morbidity and mortality. Arterial hypertension, cardiac ischemia with the acceleration of atherosclerosis, arrhythmias, myocardial dysfunction and thromboembolic disease are the most feared cardiovascular adverse reactions due to VEGF inhibitors. Susceptibility for the occurrence of VEGF inhibitors-induced cardiotoxicity has multifactorial determinants, with a significant inter-individual variation. Baseline cardiovascular risk assessment of the patient, type and stage of cancer, dose and duration of VEGF inhibitors treatment and adjuvant chemotherapy or radiotherapy are the main predictors for cardiotoxicity. The role of the cardio-oncology team becomes essential for achieving maximum therapeutic anti-angiogenic effects with minimum cardiovascular side effects. This review will summarize the incidence, risk factors, mechanisms, management and treatment of VEGF inhibitors-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Diana Mihalcea
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
- Cardiology Department, University and Emergency Hospital, 050098 Bucharest, Romania
| | - Hayat Memis
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
| | - Sorina Mihaila
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
- Cardiology Department, University and Emergency Hospital, 050098 Bucharest, Romania
| | - Dragos Vinereanu
- Cardiology and Cardiovascular Surgery Department, University of Medicine and Pharmacy Carol Davila, Splaiul Independentei 169, 050098 Bucharest, Romania
- Cardiology Department, University and Emergency Hospital, 050098 Bucharest, Romania
- Correspondence: ; Tel./Fax: +40-21-3180576
| |
Collapse
|
26
|
Zhang J, Li X, Liu J, Shang Y, Tan L, Guo Y. Early and dynamic detection of doxorubicin induced cardiotoxicity by myocardial contrast echocardiography combined with two-dimensional speckle tracking echocardiography in rats. Front Cardiovasc Med 2023; 9:1063499. [PMID: 36712239 PMCID: PMC9880177 DOI: 10.3389/fcvm.2022.1063499] [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: 10/07/2022] [Accepted: 12/27/2022] [Indexed: 01/15/2023] Open
Abstract
Background Anthracycline-induced cardiotoxicity is well-known as a side effect of chemotherapy. Currently, clinical imaging techniques are not capable to detect doxorubicin (DOX)-induced cardiotoxicity before a functional decline. The purpose of this study was to evaluate whether myocardial contrast echocardiography (MCE) can dynamically monitor the cardiac changes in the early stage in the DOX-induced rat model of cardiotoxicity. Methods A weekly injection of 2.5 mg/kg of DOX was used to generate a rat model of cardiotoxicity. All groups underwent ultrasonic examinations including standard echocardiography, 2D speckle tracking echocardiography (2D-STE), and MCE. Then all rats were sacrificed immediately for histopathological evaluation. Results A total of eight control rats and 32 DOX-treated rats were included in the study and grouped according to their treatment period. Decreased quantitative parameters of myocardial blood flow (MBF) (control vs. group 1: 133.31 ± 20.23 dB/s vs. 103.35 ± 21.60 dB/s, P = 0.048) and β (control vs. group 2: 11.17 ± 1.48/s vs. 7.15 ± 1.23/s, P < 0.001) were observed after 2 and 4 weeks of treatment, respectively, while left ventricular global strain (control vs. group 3: -23.67 ± 3.92% vs. -16.01 ± 3.40%, P = 0.002) decreased after 6 weeks of treatment and left ventricular ejection fraction (LVEF) (control vs. group 4: 82.41 ± 3.20% vs. 70.89 ± 9.30%, P = 0.008) decreased after 8 weeks of treatment. The main histopathological features are increased myocardial vacuolization and interstitial fibrosis and decreased myocardial microvessel density. Conclusion Compared with standard echocardiography and 2D-STE, MCE can accurately and non-invasively detect changes in early myocardial perfusion, demonstrating the clinical potential of continuous and dynamic monitoring of DOX-induced cardiotoxicity.
Collapse
|
27
|
Hegazy M, Ghaleb S, Das BB. Diagnosis and Management of Cancer Treatment-Related Cardiac Dysfunction and Heart Failure in Children. CHILDREN (BASEL, SWITZERLAND) 2023; 10:children10010149. [PMID: 36670699 PMCID: PMC9856743 DOI: 10.3390/children10010149] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/05/2023] [Accepted: 01/09/2023] [Indexed: 01/13/2023]
Abstract
It is disheartening for parents to discover that their children have long-term cardiac dysfunction after being cured of life-threatening childhood cancers. As the number of childhood cancer survivors increases, early and late oncology-therapy-related cardiovascular complications continues to rise. It is essential to understand that cardiotoxicity in childhood cancer survivors is persistent and progressive. A child's cancer experience extends throughout his lifetime, and ongoing care for long-term survivors is recognized as an essential part of the cancer care continuum. Initially, there was a lack of recognition of late cardiotoxicities related to cancer therapy. About 38 years ago, in 1984, pioneers like Dr. Lipshultz and others published anecdotal case reports of late cardiotoxicities in children and adolescents exposed to chemotherapy, including some who ended up with heart transplantation. At that time, cardiac tests for cancer survivors were denied by insurance companies because they did not meet appropriate use criteria. Since then, cardio-oncology has been an emerging field of cardiology that focuses on the early detection of cancer therapy-related cardiac dysfunction occurring during and after oncological treatment. The passionate pursuit of many healthcare professionals to make life better for childhood cancer survivors led to more than 10,000 peer-reviewed publications in the last 40 years. We synthesized the existing evidence-based practice and described our experiences in this review to share our current method of surveillance and management of cardiac dysfunction related to cancer therapy. This review aims to discuss the pathological basis of cancer therapy-related cardiac dysfunction and heart failure, how to stratify patients prone to cardiotoxicity by identifying modifiable risk factors, early detection of cardiac dysfunction, and prevention and management of heart failure during and after cancer therapy in children. We emphasize serial longitudinal follow-ups of childhood cancer survivors and targeted intervention for high-risk patients. We describe our experience with the new paradigm of cardio-oncology care, and collaboration between cardiologist and oncologist is needed to maximize cancer survival while minimizing late cardiotoxicity.
Collapse
Affiliation(s)
- Mohamed Hegazy
- University of Mississippi Medical Center Program, Jackson, MS 39216, USA
| | - Stephanie Ghaleb
- Division of Pediatric Cardiology, Department of Pediatrics, Children’s of Mississippi Heart Center, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Bibhuti B Das
- Division of Pediatric Cardiology, Department of Pediatrics, McLane Children’s Baylor Scott and White Medical Center, Baylor College of Medicine-Temple, Temple, TX 76502, USA
- Correspondence: ; Tel.: +1-254-935-4980
| |
Collapse
|
28
|
Franczyk B, Rysz J, Ławiński J, Ciałkowska-Rysz A, Gluba-Brzózka A. Cardiotoxicity of Selected Vascular Endothelial Growth Factor Receptor Tyrosine Kinase Inhibitors in Patients with Renal Cell Carcinoma. Biomedicines 2023; 11:181. [PMID: 36672689 PMCID: PMC9855533 DOI: 10.3390/biomedicines11010181] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/28/2022] [Accepted: 01/03/2023] [Indexed: 01/13/2023] Open
Abstract
Renal cell carcinoma (RCC) is one of the most frequent malignant neoplasms of the kidney. The therapeutic options available for the treatment of advanced or metastatic RCC include vascular endothelial growth factor receptor (VEGFR)-targeted molecules, for example, tyrosine kinase inhibitors (TKI). Various VEGFR-TKIs proved to be effective in the treatment of patients with solid tumours. The combination of two drugs may prove most beneficial in the treatment of metastatic RCC; however, it also enhances the risk of toxicity compared to monotherapy. Specific VEGFR-TKIs (e.g., sunitinib, sorafenib or pazopanib) may increase the rate of cardiotoxicity in metastatic settings. VEGF inhibitors modulate multiple signalling pathways; thus, the identification of the mechanism underlying cardiotoxicity appears challenging. VEGF signalling is vital for the maintenance of cardiomyocyte homeostasis and cardiac function; therefore, its inhibition can be responsible for the reported adverse effects. Disturbed growth factor signalling pathways may be associated with endothelial dysfunction, impaired revascularization, the development of dilated cardiomyopathy, cardiac hypertrophies and altered peripheral vascular load. Patients at high cardiovascular risk at baseline could benefit from clinical follow-up in the first 2-4 weeks after the introduction of targeted molecular therapy; however, there is no consensus concerning the surveillance strategy.
Collapse
Affiliation(s)
- Beata Franczyk
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| | - Jacek Rysz
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| | - Janusz Ławiński
- Department of Urology, Institute of Medical Sciences, Medical College of Rzeszow University, 35-055 Rzeszow, Poland
| | | | - Anna Gluba-Brzózka
- Department of Nephrology, Hypertension and Family Medicine, Medical University of Lodz, 113 Żeromskiego Street, 90-549 Lodz, Poland
| |
Collapse
|
29
|
Kang JJ, Lee H, Park BH, Song YK, Park SE, Kim R, Lee KA. Efficacy of a 4-Week Nurse-Led Exercise Rehabilitation Program in Improving the Quality of Life in Women Receiving a Post-Mastectomy Reconstruction Using the Motiva Ergonomix TM Round SilkSurface. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2022; 20:16. [PMID: 36612340 PMCID: PMC9819378 DOI: 10.3390/ijerph20010016] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/30/2022] [Revised: 12/13/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
We assessed the efficacy of a 4-week nurse-led exercise rehabilitation (ER) program in improving the quality of life (QOL) of breast cancer survivors (BCS) receiving an implant-based breast reconstruction. The eligible patients were equally randomized to either of both groups: the intervention group (n = 30; a 4-week nurse-led ER program) and the control group (n = 30; a 4-week physical therapist-supervised one). Both after a 4-week ER program and at baseline, the patients were evaluated for the European Organization for Research and Treatment of Cancer Quality of Life Questionnaire Core 30 (EORTC QLQ-C30) and Fatigue Severity Scale (FSS) scores. There was a significantly higher degree of increase in global health status/QOL scores, physical functioning scores, role functioning scores, and emotional functioning scores at 4 weeks from baseline in the intervention group as compared with the control group (p = 0.001). However, there was a significantly higher degree of decrease in fatigue scores, nausea/vomiting scores, pain scores, dyspnea scores, and FSS scores in the intervention group as compared with the control group (p = 0.001). In conclusion, our results indicate that a 4-week nurse-led ER program might be effective in the QOL in BCS receiving a post-mastectomy implant-based reconstruction using the Motiva ErgonomixTM Round SilkSurface.
Collapse
Affiliation(s)
- Jung Joong Kang
- Department of Physical Medicine and Rehabilitation, Booboo Medical Healthcare Hospital, Mokpo 58655, Republic of Korea
| | - Hyunho Lee
- Department of Anesthesiology and Pain Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Bom Hui Park
- Department of Medical Device Management and Research, SAIHST, Sungkyunkwan University, Seoul 06355, Republic of Korea
| | - Yu Kwan Song
- Department of Plastic and Reconstructive Surgery, Chung Ju Mirae Hospital, Chungju 27361, Republic of Korea
| | - Soon Eun Park
- Department of Anesthesiology and Pain Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea
| | - Robert Kim
- Department of Medical and Pharmaceutical Affairs, Doctor CONSULT, Seoul 06296, Republic of Korea
| | - Kyung Ah Lee
- Department of Plastic and Reconstructive Surgery, Inje University Haeundae Paik Hospital, 875 Haeun-daero, Busan 48108, Republic of Korea
| |
Collapse
|
30
|
An efficient human stem cells derived cardiotoxicity testing platform for testing oncotherapeutic analogues of quercetin and cinnamic acid. Sci Rep 2022; 12:21362. [PMID: 36494370 PMCID: PMC9734143 DOI: 10.1038/s41598-022-21721-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Accepted: 09/30/2022] [Indexed: 12/13/2022] Open
Abstract
Oncotherapeutics research is progressing at a rapid pace, however, not many drugs complete the successful clinical trial because of severe off-target toxicity to cardiomyocytes which ultimately leads to cardiac dysfunction. It is thus important to emphasize the need for early testing for possible cardiotoxicity of emerging oncotherapeutics. In this study, we assessed a novel stem cell-derived cardiac model for testing for cardiotoxicity of novel oncotherapeutics. We evaluated the cardiotoxic effect of synthesized derivatives of oncotherapeutics, quercetin (QMJ-2, -5, and -6) and cinnamic acid (NMJ-1, -2, and -3) using human Wharton's jelly mesenchymal stem cells-derived cardiomyocytes (WJCM) against known cardiotoxic oncologic drugs, doxorubicin, 5-fluorouracil, cisplatin. QMJ-6, NMJ-2, and NMJ-3 were not cardiotoxic and had minimum cardiac side effects. They did not show any effect on cardiomyocyte viability, caused low LDH release, and intracellular ROS production kept the calcium flux minimal and protected the active mitochondrial status in cardiomyocytes. They persevered cardiac-specific gene expression as well. However, compounds QMJ-2, QMJ-5, and NMJ-1 were cardiotoxic and the concentration needs to be reduced to prevent toxic effects on cardiomyocytes. Significantly, we were able to demonstrate that WJCM is an efficient cardiac testing model to analyze the cardiotoxicity of drugs in a human context.
Collapse
|
31
|
Cherukuri SP, Chikatimalla R, Dasaradhan T, Koneti J, Gadde S, Kalluru R. Breast Cancer and the Cardiovascular Disease: A Narrative Review. Cureus 2022; 14:e27917. [PMID: 36110451 PMCID: PMC9464354 DOI: 10.7759/cureus.27917] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2022] [Indexed: 11/05/2022] Open
Abstract
Breast cancer is the most common malignancy affecting females worldwide and is also among the top causes of all cancer-related deaths. Cardiovascular disease (CVD) is known to have the highest rate of mortality in women. There are several risk factors for both CVD and breast cancer that overlap, such as diet, smoking, and obesity, and also the current breast cancer treatment has a significant detrimental effect on cardiovascular health in general. Patients with exposure to potentially cardiotoxic treatments, including anthracyclines, trastuzumab, and radiation therapy, are more likely to develop CVD than non-cancer controls. Early detection and treatment may reduce the risk of the development of cardiac morbidity and mortality and would increase the number of breast cancer survivors. This article provides a comprehensive overview of breast cancer, identifies shared risk factors among breast cancer and CVD, and the cardiotoxic effects of therapy. It also reviews possible prevention and treatment of CVD in breast cancer patients and reviews literature about chemoprevention of cardiac disease in the setting of breast cancer treatment.
Collapse
|
32
|
Matesun DA, Mensah KB, Yamoah P, Bangalee V, Padayachee N. Adverse drug reactions associated with doxorubicin and epirubicin: A descriptive analysis from VigiBase. J Oncol Pharm Pract 2022:10781552221113578. [PMID: 35833221 DOI: 10.1177/10781552221113578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Cancer is one of the leading causes of death globally. Owing to high toxicity, patients using chemotherapy drugs have a higher risk for developing adverse drug reactions (ADRs). Pharmacovigilance studies are essential in oncology to evaluate ADRs caused by anticancer drugs and improve patient safety. This study aimed to analyze serious ADRs associated with the use of doxorubicin and epirubicin reported to VigiBase. METHOD All anonymized data on suspected ADRs for doxorubicin and epirubicin as 'serious' and 'suspected' or 'interacting' drugs between 1968 and 30 August 2021, were extracted from VigiBase. Descriptive statistics were conducted in Microsoft Excel, and data were summarized using frequencies and percentages. RESULTS A total of 35,620 serious individual case safety reports was analyzed. The majority of reports were from females (Dox = 61.41%; Epi = 86.56%), while the predominant age group was 45-64 years (Dox = 42.06%; Epi = 57.39%). Physicians were the more likely group to report serious ADRs (Dox = 50.03%; Epi = 34.11%). In general, Europe reported the highest for doxorubicin (38.08%), while Asia recorded the highest reports for epirubicin (53.28%). Oceania reported the least for both drugs (Dox = 0.45%; Epi = 0.04%), followed by Africa (Dox = 0.72%; Epi = 0.29%). Blood and lymphatic system disorders were the most reported serious category (Dox = 11053 [44.47%]; Epi = 6659 [61.84%]). The most common manifestations were febrile neutropenia (Dox = 10.52%) and bone marrow failure (Epi = 23.89%). CONCLUSION This study provides relevant global insights into serious ADRs for doxorubicin and epirubicin. This knowledge may assist in minimizing and proactively managing ADRs. It can also inform policies to improve patients' quality of life.
Collapse
Affiliation(s)
- Deborah A Matesun
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, 37707University of the Witwatersrand, Johannesburg, South Africa
| | - Kofi Boamah Mensah
- Department of Pharmacy Practice, Faculty of Pharmacy and Pharmaceutical Science, College of Health Science, 98763Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
- Discipline of Pharmaceutical Sciences, 72753College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Peter Yamoah
- School of Pharmacy, 549574University of Health and Allied Sciences, Ho, Ghana
- College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Varsha Bangalee
- Discipline of Pharmaceutical Sciences, 72753College of Health Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Neelaveni Padayachee
- Department of Pharmacy and Pharmacology, Faculty of Health Sciences, 37707University of the Witwatersrand, Johannesburg, South Africa
| |
Collapse
|
33
|
Keeping Cell Death Alive: An Introduction into the French Cell Death Research Network. Biomolecules 2022; 12:biom12070901. [PMID: 35883457 PMCID: PMC9313292 DOI: 10.3390/biom12070901] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 06/16/2022] [Accepted: 06/20/2022] [Indexed: 02/01/2023] Open
Abstract
Since the Nobel Prize award more than twenty years ago for discovering the core apoptotic pathway in C. elegans, apoptosis and various other forms of regulated cell death have been thoroughly characterized by researchers around the world. Although many aspects of regulated cell death still remain to be elucidated in specific cell subtypes and disease conditions, many predicted that research into cell death was inexorably reaching a plateau. However, this was not the case since the last decade saw a multitude of cell death modalities being described, while harnessing their therapeutic potential reached clinical use in certain cases. In line with keeping research into cell death alive, francophone researchers from several institutions in France and Belgium established the French Cell Death Research Network (FCDRN). The research conducted by FCDRN is at the leading edge of emerging topics such as non-apoptotic functions of apoptotic effectors, paracrine effects of cell death, novel canonical and non-canonical mechanisms to induce apoptosis in cell death-resistant cancer cells or regulated forms of necrosis and the associated immunogenic response. Collectively, these various lines of research all emerged from the study of apoptosis and in the next few years will increase the mechanistic knowledge into regulated cell death and how to harness it for therapy.
Collapse
|
34
|
Terada CI, Onoue K, Fujii T, Itami H, Morita K, Uchiyama T, Takeda M, Nakagawa H, Nakano T, Baba Y, Amemiya K, Saito Y, Hatakeyama K, Ohbayashi C. Histopathological and epigenetic changes in myocardium associated with cancer therapy-related cardiac dysfunction. ESC Heart Fail 2022; 9:3031-3043. [PMID: 35747987 DOI: 10.1002/ehf2.14034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 04/26/2022] [Accepted: 06/09/2022] [Indexed: 11/10/2022] Open
Abstract
AIMS Cancer therapy-related cardiac dysfunction (CTRCD) is commonly reported, but its histopathology, mechanisms, and risk factors are not known. We aimed to clarify the histopathology and mechanisms of CTRCD to identify risk factors. METHODS AND RESULTS We performed myocardial histopathological studies on 13 endomyocardial biopsies from CTRCD patients, 35 autopsied cancer cases with or without cardiac dysfunction, and controls without cancer (10 biopsies and 9 autopsies). Cardiotoxicity risk scores were calculated based on medication; and patient-related risk factors, fibrosis, and cardiomyocyte changes were scored; and p53 and H3K27ac histone modification were evaluated by histological score (H-score). In the biopsy cases, all histopathological changes and the p53 evaluation were significantly higher in the CTRCD group than in the controls [p53 H-score; 63 (9.109) vs. 33 (5.099), P < 0.05]. In patients with a short time between drug and disease onset (<4.2 years), fibrosis and p53 positively correlated (r = 0.76, P < 0.05), and in those with late onset disease (>4.2 years), cellular abnormalities and p53 trended to a positive correlation and cardiotoxicity risk scores and p53 positively correlated (r = 0.95, P < 0.05). A year after biopsy, the short-term group had significant recovery of ejection fraction compared with the long-term group (P < 0.05). The CTRCD group had a significantly worse overall survival prognosis than the control group [hazard ratio 7.61 (95% confidence interval 1.30-44.6), P < 0.05]. Autopsy cases with cancer treatment also had a high grade of histopathological changes, with even more severe changes in patients with cardiac dysfunction, and had increased p53 and H3K27ac expression levels, compared with controls. H-scores of p53 and H3K27ac showed a positive correlation in the CTRCD group in biopsy cases (r = 0.62, P < 0.05) and a positive correlation in autopsy cases. CONCLUSIONS Our results indicate distinct morphological characteristics in myocardial histopathology associated with CTRCD. p53 and H3K27ac histone modification could be sensitive markers of CTRCD and suggest a mechanistic involvement of epigenetic changes.
Collapse
Affiliation(s)
- Chiyoko-Ikeda Terada
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan
| | - Kenji Onoue
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Tomomi Fujii
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan
| | - Hiroe Itami
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan
| | - Kohei Morita
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan
| | - Tomoko Uchiyama
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan
| | - Maiko Takeda
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan
| | - Hitoshi Nakagawa
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Tomoya Nakano
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Youichirou Baba
- Department of Pathology, Suzuka General Hospital, 1275-53 Yasuduka, Suzuka, Mie, 513-8630, Japan
| | - Kisaki Amemiya
- Department of Pathology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Yoshihiko Saito
- Department of Cardiovascular Medicine, Nara Medical University, 840 Shijo, Kashihara, Nara, 634-8522, Japan
| | - Kinta Hatakeyama
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan.,Department of Pathology, National Cerebral and Cardiovascular Center, 6-1 Kishibe-Shimmachi, Suita, Osaka, 564-8565, Japan
| | - Chiho Ohbayashi
- Department of Diagnostic Pathology, Nara Medical University, 840 Shijo, Kashihara, Nara, 643-8522, Japan
| |
Collapse
|
35
|
Krishnarao K, Bruno KA, Di Florio DN, Edenfield BH, Whelan ER, Macomb LP, McGuire MM, Hill AR, Ray JC, Cornell LF, Tan W, Geiger XJ, Salomon GR, Douglass EJ, Fairweather D, Yamani MH. Upregulation of Endothelin-1 May Predict Chemotherapy-Induced Cardiotoxicity in Women with Breast Cancer. J Clin Med 2022; 11:jcm11123547. [PMID: 35743613 PMCID: PMC9224558 DOI: 10.3390/jcm11123547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 06/10/2022] [Accepted: 06/11/2022] [Indexed: 02/04/2023] Open
Abstract
As survival in breast cancer patients from newer therapies increases, concerns for chemotherapy-induced cardiotoxicity (CIC) have offset some of these benefits, manifesting as a decline in left ventricular ejection fraction (LVEF). Patients receiving anthracycline-based chemotherapy followed by trastuzumab are at risk for CIC. Previous research evaluating whether clinical biomarkers predict cardiotoxicity has been inconsistent. Recently, angiotensin II type 1 receptor (ATR1) and endothelin 1 (ET1) have been shown to play a role in breast tumor growth. We evaluated ATR1 and ET1 expression in breast cancer tissue and its association with CIC. A total of 33 paraffin-embedded breast tissue specimens from women with breast cancer treated with anthracycline-based chemotherapy and trastuzumab were analyzed by immunohistochemistry (IHC) and qRT-PCR. We found that ET1 expression was increased in patients with an LVEF ≤ 50% (p = 0.032) with a lower LVEF correlating with higher ET1 expression (r = 0.377, p = 0.031). In patients with a change in LVEF of greater than 10%, greater ET1 expression was noted compared to those without a change in LVEF (p = 0.017). Increased ET1 expression in breast tumor tissue is associated with reduced LVEF. Future studies need to examine whether ET1 may be a tissue biomarker that helps predict the risk of developing CIC in women with breast cancer.
Collapse
Affiliation(s)
- Krithika Krishnarao
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Department of Cardiovascular Medicine, Ochsner Health, New Orleans, LA 70121, USA
- Correspondence: ; Tel.: +1-504-842-9780
| | - Katelyn A. Bruno
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Damian N. Di Florio
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL 32224, USA
| | | | - Emily R. Whelan
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Logan P. Macomb
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Molly M. McGuire
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Anneliese R. Hill
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Jordan C. Ray
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Lauren F. Cornell
- Department of Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (L.F.C.); (W.T.)
| | - Winston Tan
- Department of Oncology, Mayo Clinic, Jacksonville, FL 32224, USA; (L.F.C.); (W.T.)
| | | | - Gary R. Salomon
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - Erika J. Douglass
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| | - DeLisa Fairweather
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
- Center for Clinical and Translational Science, Mayo Clinic, Jacksonville, FL 32224, USA
- Department of Environmental Health Sciences, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, MD 21205, USA
| | - Mohamad H. Yamani
- Department of Cardiovascular Medicine, Mayo Clinic, Jacksonville, FL 32224, USA; (K.A.B.); (D.N.D.F.); (E.R.W.); (L.P.M.); (M.M.M.); (A.R.H.); (J.C.R.); (G.R.S.); (E.J.D.); (D.F.); (M.H.Y.)
| |
Collapse
|
36
|
New Insights on the Toxicity on Heart and Vessels of Breast Cancer Therapies. Med Sci (Basel) 2022; 10:medsci10020027. [PMID: 35736347 PMCID: PMC9229896 DOI: 10.3390/medsci10020027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 11/17/2022] Open
Abstract
Cardiovascular diseases are largely represented in patients with cancer and appear to be important side effects of cancer treatments, heavily affecting quality of life and leading to premature morbidity and death among cancer survivors. In particular, treatments for breast cancer have been shown to potentially play serious detrimental effects on cardiovascular health. This review aims to explore the available literature on breast cancer therapy-induced side effects on heart and vessels, illustrating the molecular mechanisms of cardiotoxicity known so far. Moreover, principles of cardiovascular risk assessment and management of cardiotoxicity in clinical practice will also be elucidated. Chemotherapy (anthracycline, taxanes, cyclophosphamide and 5-fluorouracil), hormonal therapy (estrogen receptor modulator and gonadotropin or luteinizing releasing hormone agonists) and targeted therapy (epidermal growth factor receptor 2 and Cyclin-dependent kinases 4 and 6 inhibitors) adverse events include arterial and pulmonary hypertension, supraventricular and ventricular arrhythmias, systolic and diastolic cardiac dysfunction and coronary artery diseases due to different and still not well-dissected molecular pathways. Therefore, cardiovascular prevention programs and treatment of cardiotoxicity appear to be crucial to improve morbidity and mortality of cancer survivors.
Collapse
|
37
|
Tranchita E, Murri A, Grazioli E, Cerulli C, Emerenziani GP, Ceci R, Caporossi D, Dimauro I, Parisi A. The Beneficial Role of Physical Exercise on Anthracyclines Induced Cardiotoxicity in Breast Cancer Patients. Cancers (Basel) 2022; 14:cancers14092288. [PMID: 35565417 PMCID: PMC9104319 DOI: 10.3390/cancers14092288] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Revised: 04/15/2022] [Accepted: 04/29/2022] [Indexed: 12/24/2022] Open
Abstract
The increase in breast cancer (BC) survival has determined a growing survivor population that seems to develop several comorbidities and, specifically, treatment-induced cardiovascular disease (CVD), especially those patients treated with anthracyclines. Indeed, it is known that these compounds act through the induction of supraphysiological production of reactive oxygen species (ROS), which appear to be central mediators of numerous direct and indirect cardiac adverse consequences. Evidence suggests that physical exercise (PE) practised before, during or after BC treatments could represent a viable non-pharmacological strategy as it increases heart tolerance against many cardiotoxic agents, and therefore improves several functional, subclinical, and clinical parameters. At molecular level, the cardioprotective effects are mainly associated with an exercise-induced increase of stress response proteins (HSP60 and HSP70) and antioxidant (SOD activity, GSH), as well as a decrease in lipid peroxidation, and pro-apoptotic proteins such as Bax, Bax-to-Bcl-2 ratio. Moreover, this protection can potentially be explained by a preservation of myosin heavy chain (MHC) isoform distribution. Despite this knowledge, it is not clear which type of exercise should be suggested in BC patient undergoing anthracycline treatment. This highlights the lack of special guidelines on how affected patients should be managed more efficiently. This review offers a general framework for the role of anthracyclines in the physio-pathological mechanisms of cardiotoxicity and the potential protective role of PE. Finally, potential exercise-based strategies are discussed on the basis of scientific findings.
Collapse
Affiliation(s)
- Eliana Tranchita
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy; (E.T.); (A.M.); (C.C.); (A.P.)
| | - Arianna Murri
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy; (E.T.); (A.M.); (C.C.); (A.P.)
| | - Elisa Grazioli
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy; (E.T.); (A.M.); (C.C.); (A.P.)
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy;
- Correspondence: ; Tel.: +39-06-3673-3532
| | - Claudia Cerulli
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy; (E.T.); (A.M.); (C.C.); (A.P.)
| | - Gian Pietro Emerenziani
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy;
| | - Roberta Ceci
- Laboratory of Biochemistry and Molecular Biology, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, 00135 Rome, Italy;
| | - Daniela Caporossi
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy; (D.C.); (I.D.)
| | - Ivan Dimauro
- Unit of Biology and Genetics of Movement, Department of Movement, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy; (D.C.); (I.D.)
| | - Attilio Parisi
- Laboratory of Physical Exercise and Sport Science, Department of Exercise, Human and Health Sciences, University of Rome Foro Italico, Piazza Lauro de Bosis 15, 00135 Rome, Italy; (E.T.); (A.M.); (C.C.); (A.P.)
| |
Collapse
|
38
|
Carrabba N, Pontone G, Andreini D, Buffa V, Cademartiri F, Carbone I, Clemente A, Guaricci AI, Guglielmo M, Indolfi C, La Grutta L, Ligabue G, Liguori C, Mercuro G, Mushtaq S, Neglia D, Palmisano A, Sciagrà R, Seitun S, Vignale D, Francone M, Esposito A. Appropriateness criteria for the use of cardiac computed tomography, SIC-SIRM part 2: acute chest pain evaluation; stent and coronary artery bypass graft patency evaluation; planning of coronary revascularization and transcatheter valve procedures; cardiomyopathies, electrophysiological applications, cardiac masses, cardio-oncology and pericardial diseases evaluation. J Cardiovasc Med (Hagerstown) 2022; 23:290-303. [PMID: 35486680 DOI: 10.2459/jcm.0000000000001303] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
In the past 20 years, cardiac computed tomography (CCT) has become a pivotal technique for the noninvasive diagnostic workup of coronary and cardiac diseases. Continuous technical and methodological improvements, combined with fast growing scientific evidence, have progressively expanded the clinical role of CCT. Randomized clinical trials documented the value of CCT in increasing the cost-effectiveness of the management of patients with acute chest pain presenting in the emergency department, also during the pandemic. Beyond the evaluation of stents and surgical graft patency, the anatomical and functional coronary imaging have the potential to guide treatment decision-making and planning for complex left main and three-vessel coronary disease. Furthermore, there has been an increasing demand to use CCT for preinterventional planning in minimally invasive procedures, such as transcatheter valve implantation and mitral valve repair. Yet, the use of CCT as a roadmap for tailored electrophysiological procedures has gained increasing importance to assure maximum success. In the meantime, innovations and advanced postprocessing tools have generated new potential applications of CCT from the simple coronary anatomy to the complete assessment of structural, functional and pathophysiological biomarkers of cardiac disease. In this complex and revolutionary scenario, it is urgently needed to provide an updated guide for the appropriate use of CCT in different clinical settings. This manuscript, endorsed by the Italian Society of Cardiology (SIC) and the Italian Society of Medical and Interventional Radiology (SIRM), represents the second of two consensus documents collecting the expert opinion of cardiologists and radiologists about current appropriate use of CCT.
Collapse
Affiliation(s)
- Nazario Carrabba
- Department of Cardiothoracovascular Medicine, Azienda Ospedaliero-Universitaria Careggi, Florence
| | | | - Daniele Andreini
- Centro Cardiologico Monzino IRCCS.,Department of Clinical Sciences and Community Health, University of Milan, Milan
| | - Vitaliano Buffa
- Department of Radiology, Azienda Ospedaliera San Camillo Forlanini, Rome
| | | | - Iacopo Carbone
- Department of Radiological, Oncological and Pathological Sciences, 'Sapienza' University of Rome, Rome
| | - Alberto Clemente
- Department of Radiology, CNR (National Council of Research)/Tuscany Region 'Gabriele Monasterio' Foundation (FTGM), Massa
| | - Andrea Igoren Guaricci
- University Cardiology Unit, Cardiothoracic Department, Policlinic University Hospital, Bari
| | | | - Ciro Indolfi
- Department of Medical and Surgical Sciences, Magna Grecia University, Catanzaro
| | - Ludovico La Grutta
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties-ProMISE, University of Palermo
| | - Guido Ligabue
- Department of Medical and Surgical Sciences, Modena and Raggio Emilia University.,Radiology Department, AOU of Modena, Modena
| | - Carlo Liguori
- Radiology Unit, Ospedale del Mare -A.S.L Na1- Centro, Naples
| | - Giuseppe Mercuro
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari
| | | | - Danilo Neglia
- Cardiovascular Department, CNR (National Council of Research)/Tuscany Region 'Gabriele Monasterio' Foundation (FTGM), Pisa
| | - Anna Palmisano
- Clinical and Experimental Radiology Unit, Experimental Imaging Center, IRCCS Ospedale San Raffaele.,Vita-Salute San Raffaele University, Milan
| | - Roberto Sciagrà
- Nuclear Medicine Unit, Department of Experimental and Clinical Biomedical Sciences 'Mario Serio', University of Florence, Florence
| | - Sara Seitun
- Radiology Department, Ospedale Policlinico San Martino, IRCCS Per L'Oncologia e le Neuroscienze, Genoa, Italy
| | - Davide Vignale
- Clinical and Experimental Radiology Unit, Experimental Imaging Center, IRCCS Ospedale San Raffaele.,Vita-Salute San Raffaele University, Milan
| | - Marco Francone
- Department of Radiological, Oncological and Pathological Sciences, 'Sapienza' University of Rome, Rome
| | - Antonio Esposito
- Clinical and Experimental Radiology Unit, Experimental Imaging Center, IRCCS Ospedale San Raffaele.,Vita-Salute San Raffaele University, Milan
| |
Collapse
|
39
|
Morelli MB, Bongiovanni C, Da Pra S, Miano C, Sacchi F, Lauriola M, D’Uva G. Cardiotoxicity of Anticancer Drugs: Molecular Mechanisms and Strategies for Cardioprotection. Front Cardiovasc Med 2022; 9:847012. [PMID: 35497981 PMCID: PMC9051244 DOI: 10.3389/fcvm.2022.847012] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/03/2022] [Indexed: 12/13/2022] Open
Abstract
Chemotherapy and targeted therapies have significantly improved the prognosis of oncology patients. However, these antineoplastic treatments may also induce adverse cardiovascular effects, which may lead to acute or delayed onset of cardiac dysfunction. These common cardiovascular complications, commonly referred to as cardiotoxicity, not only may require the modification, suspension, or withdrawal of life-saving antineoplastic therapies, with the risk of reducing their efficacy, but can also strongly impact the quality of life and overall survival, regardless of the oncological prognosis. The onset of cardiotoxicity may depend on the class, dose, route, and duration of administration of anticancer drugs, as well as on individual risk factors. Importantly, the cardiotoxic side effects may be reversible, if cardiac function is restored upon discontinuation of the therapy, or irreversible, characterized by injury and loss of cardiac muscle cells. Subclinical myocardial dysfunction induced by anticancer therapies may also subsequently evolve in symptomatic congestive heart failure. Hence, there is an urgent need for cardioprotective therapies to reduce the clinical and subclinical cardiotoxicity onset and progression and to limit the acute or chronic manifestation of cardiac damages. In this review, we summarize the knowledge regarding the cellular and molecular mechanisms contributing to the onset of cardiotoxicity associated with common classes of chemotherapy and targeted therapy drugs. Furthermore, we describe and discuss current and potential strategies to cope with the cardiotoxic side effects as well as cardioprotective preventive approaches that may be useful to flank anticancer therapies.
Collapse
Affiliation(s)
| | - Chiara Bongiovanni
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Silvia Da Pra
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Carmen Miano
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
| | - Francesca Sacchi
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Mattia Lauriola
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Gabriele D’Uva
- National Laboratory of Molecular Biology and Stem Cell Engineering, National Institute of Biostructures and Biosystems (INBB), Bologna, Italy
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
- *Correspondence: Gabriele D’Uva,
| |
Collapse
|
40
|
Das A, Dasgupta S, Gong Y, Shah UA, Fradley MG, Cheng RK, Roy B, Guha A. Cardiotoxicity as an adverse effect of immunomodulatory drugs and proteasome inhibitors in multiple myeloma: A network meta-analysis of randomized clinical trials. Hematol Oncol 2022; 40:233-242. [PMID: 34940983 PMCID: PMC9423942 DOI: 10.1002/hon.2959] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/01/2021] [Accepted: 12/12/2021] [Indexed: 01/21/2023]
Abstract
We aim to determine the cumulative and comparative risk of cardiovascular events associated with different Immunomodulatory Drugs (iMiDs) and Proteasome Inhibitor (PIs) in Multiple Myeloma (MM) patients through pairwise and network meta-analysis. Electronic searches were conducted using Ovid MEDLINE, EMBASE, CINAHL, Web of Science, and Clinical Trial Registry (Clinical Trials.gov) up to May 2021. Phase 3 randomized clinical trials (RCTs) reporting cardiotoxicity in MM patients (newly diagnoses and/or relapsed) treated with iMiD and/or PI. Studies, where iMiD or PI was used alongside the chemotherapy versus placebo or no additional drugs (control) in the other arm were included. The primary outcome was the presence of cardiotoxicity after follow-up. Pairwise meta-analysis and network meta-analysis were performed using the frequentist's approach to estimate the odds ratio (OR). Twenty RCTs with 10,373 MM patients were included in this analysis. Eleven studies compared iMiDs with control, seven studies compared PIs with control, and two studies compared bortezomib against carfilzomib. CTACE high-grade (≥grade 3) cardiotoxic events were increased with iMiDs compared to their control counterpart (OR 2.05; 95% CI 1.30-3.26). Similar high-grade cardiotoxicity was also noted more frequently with PI use when compared to the control group (OR 1.67; 95% CI 1.17-2.40). Among the PIs, carfilzomib was associated with a maximum risk of cardiotoxicity (OR 2.68; 95% CI 1.63-4.40). There was no evidence of publication bias among studies. iMiDs and PIs, particularly carfilzomib, appear to be associated with increased risk of high-grade cardiovascular events in MM patients.
Collapse
Affiliation(s)
- Avash Das
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Subhajit Dasgupta
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Yan Gong
- Department of Pharmacotherapy and Translational Research, Center for Pharmacogenomics, College of Pharmacy, University of Florida, Gainesville, FL, USA
| | - Urvi A Shah
- Myeloma Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michael G Fradley
- Division of Cardiology, Department of Medicine, University of Pennsylvania, PA, USA
| | - Richard K Cheng
- Cardiology Division, University of Washington, Seattle, WA, USA
| | - Bhaskar Roy
- Department of Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Avirup Guha
- Harrington Heart and Vascular Institute, UH Cleveland Medical Center, Cleveland, OH, USA
- Division of Cardiology, Department of Medicine, Augusta University, Augusta, GA, USA
| |
Collapse
|
41
|
Yaros K, Eksi B, Chandra A, Agusala K, Lehmann LH, Zaha Vlad G. Cardio-oncology imaging tools at the translational interface. J Mol Cell Cardiol 2022; 168:24-32. [DOI: 10.1016/j.yjmcc.2022.03.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 02/03/2022] [Accepted: 03/27/2022] [Indexed: 10/18/2022]
|
42
|
Kim JS, Arango AS, Shah S, Arnold WR, Tajkhorshid E, Das A. Anthracycline derivatives inhibit cardiac CYP2J2. J Inorg Biochem 2022; 229:111722. [PMID: 35078036 PMCID: PMC8860876 DOI: 10.1016/j.jinorgbio.2022.111722] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 11/20/2022]
Abstract
Anthracycline chemotherapeutics are highly effective, but their clinical usefulness is hampered by adverse side effects such as cardiotoxicity. Cytochrome P450 2J2 (CYP2J2) is a cytochrome P450 epoxygenase in human cardiomyocytes that converts arachidonic acid (AA) to cardioprotective epoxyeicosatrienoic acid (EET) regioisomers. Herein, we performed biochemical studies to understand the interaction of anthracycline derivatives (daunorubicin, doxorubicin, epirubicin, idarubicin, 5-iminodaunorubicin, zorubicin, valrubicin, and aclarubicin) with CYP2J2. We utilized fluorescence polarization (FP) to assess whether anthracyclines bind to CYP2J2. We found that aclarubicin bound the strongest to CYP2J2 despite it having large bulky groups. We determined that ebastine competitively inhibits anthracycline binding, suggesting that ebastine and anthracyclines may share the same binding site. Molecular dynamics and ensemble docking revealed electrostatic interactions between the anthracyclines and CYP2J2, contributing to binding stability. In particular, the glycosamine groups in anthracyclines are stabilized by binding to glutamate and aspartate residues in CYP2J2 forming salt bridge interactions. Furthermore, we used iterative ensemble docking schemes to gauge anthracycline influence on EET regioisomer production and anthracycline inhibition on AA metabolism. This was followed by experimental validation of CYP2J2-mediated metabolism of anthracycline derivatives using liquid chromatography tandem mass spectrometry fragmentation analysis and inhibition of CYP2J2-mediated AA metabolism by these derivatives. Taken together, we use both experimental and theoretical methodologies to unveil the interactions of anthracycline derivatives with CYP2J2. These studies will help identify alternative mechanisms of how anthracycline cardiotoxicity may be mediated through the inhibition of cardiac P450, which will aid in the design of new anthracycline derivatives with lower toxicity.
Collapse
Affiliation(s)
- Justin S Kim
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Andres S Arango
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Swapnil Shah
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - William R Arnold
- Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Emad Tajkhorshid
- Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Biochemistry, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America
| | - Aditi Das
- Division of Nutritional Sciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Center for Biophysics and Quantitative Biology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Beckman Institute for Advanced Science and Technology, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Bioengineering, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America; Department of Comparative Biosciences, University of Illinois Urbana-Champaign, Urbana, IL 61801, United States of America.
| |
Collapse
|
43
|
Grela-Wojewoda A, Pacholczak-Madej R, Adamczyk A, Korman M, Püsküllüoğlu M. Cardiotoxicity Induced by Protein Kinase Inhibitors in Patients with Cancer. Int J Mol Sci 2022; 23:ijms23052815. [PMID: 35269958 PMCID: PMC8910876 DOI: 10.3390/ijms23052815] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/01/2022] [Accepted: 03/02/2022] [Indexed: 12/24/2022] Open
Abstract
Kinase inhibitors (KIs) represent a growing class of drugs directed at various protein kinases and used in the treatment of both solid tumors and hematologic malignancies. It is a heterogeneous group of compounds that are widely applied not only in different types of tumors but also in tumors that are positive for a specific predictive factor. This review summarizes common cardiotoxic effects of KIs, including hypertension, arrhythmias with bradycardia and QTc prolongation, and cardiomyopathy that can lead to heart failure, as well as less common effects such as fluid retention, ischemic heart disease, and elevated risk of thromboembolic events. The guidelines for cardiac monitoring and management of the most common cardiotoxic effects of protein KIs are discussed. Potential signaling pathways affected by KIs and likely contributing to cardiac damage are also described. Finally, the need for further research into the molecular mechanisms underlying the cardiovascular toxicity of these drugs is indicated.
Collapse
Affiliation(s)
- Aleksandra Grela-Wojewoda
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Correspondence: ; Tel.: +48-1263-48350
| | - Renata Pacholczak-Madej
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
- Department of Anatomy, Jagiellonian University Medical College, 31-008 Kraków, Poland
| | - Agnieszka Adamczyk
- Department of Tumour Pathology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland;
| | - Michał Korman
- Faculty of Medicine, Jagiellonian University Medical College, 31-008 Kraków, Poland;
| | - Mirosława Püsküllüoğlu
- Department of Clinical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, Kraków Branch, Garncarska 11, 31-115 Kraków, Poland; (R.P.-M.); (M.P.)
| |
Collapse
|
44
|
Martinez DSL, Noseworthy PA, Akbilgic O, Herrmann J, Ruddy KJ, Hamid A, Maddula R, Singh A, Davis R, Gunturkun F, Jefferies JL, Brown SA. Artificial intelligence opportunities in cardio-oncology: Overview with spotlight on electrocardiography. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 15:100129. [PMID: 35721662 PMCID: PMC9202996 DOI: 10.1016/j.ahjo.2022.100129] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 01/21/2023]
Abstract
Cardiovascular disease is a leading cause of death among cancer survivors, second only to cancer recurrence or development of new tumors. Cardio-oncology has therefore emerged as a relatively new specialty focused on prevention and management of cardiovascular consequences of cancer therapies. Yet challenges remain regarding precision and accuracy with predicting individuals at highest risk for cardiotoxicity. Barriers such as access to care also limit screening and early diagnosis to improve prognosis. Thus, developing innovative approaches for prediction and early detection of cardiovascular illness in this population is critical. In this review, we provide an overview of the present state of machine learning applications in cardio-oncology. We begin by outlining some factors that should be considered while utilizing machine learning algorithms. We then examine research in which machine learning has been applied to improve prediction of cardiac dysfunction in cancer survivors. We also highlight the use of artificial intelligence (AI) in conjunction with electrocardiogram (ECG) to predict cardiac malfunction and also atrial fibrillation (AF), and we discuss the potential role of wearables. Additionally, the article summarizes future prospects and critical takeaways for the application of machine learning in cardio-oncology. This study is the first in a series on artificial intelligence in cardio-oncology, and complements our manuscript on echocardiography and other forms of imaging relevant to cancer survivors cared for in cardiology clinical practice.
Collapse
Affiliation(s)
- Daniel Sierra-Lara Martinez
- Coronary Care Unit, National Institute of Cardiology/Instituto Nacional de Cardiologia, Ciudad de Mexico, Mexico
| | | | - Oguz Akbilgic
- Department of Health Informatics and Data Science, Parkinson School of Health Sciences and Public Health, Loyola University Chicago, Maywood, IL, USA
- Section of Cardiovascular Medicine, Department of Internal Medicine, Wake Forest School of Medicine, Wake Forest, NC, USA
| | - Joerg Herrmann
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
| | | | | | | | - Ashima Singh
- Institute of Health and Equity, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Robert Davis
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, USA
| | - Fatma Gunturkun
- Center for Biomedical Informatics, University of Tennessee Health Sciences Center, USA
| | - John L. Jefferies
- Division of Cardiovascular Diseases, University of Tennessee Health Sciences Center, USA
- Department of Epidemiology, St. Jude Children's Research Hospital, USA
| | - Sherry-Ann Brown
- Department of Cardiovascular Diseases, Mayo Clinic, Rochester, MN, USA
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
45
|
Madan N, Lucas J, Akhter N, Collier P, Cheng F, Guha A, Zhang L, Sharma A, Hamid A, Ndiokho I, Wen E, Garster NC, Scherrer-Crosbie M, Brown SA. Artificial intelligence and imaging: Opportunities in cardio-oncology. AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2022; 15:100126. [PMID: 35693323 PMCID: PMC9187287 DOI: 10.1016/j.ahjo.2022.100126] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 12/29/2022]
Abstract
Cardiovascular disease is a leading cause of death in cancer survivors. It is critical to apply new predictive and early diagnostic methods in this population, as this can potentially inform cardiovascular treatment and surveillance decision-making. We discuss the application of artificial intelligence (AI) technologies to cardiovascular imaging in cardio-oncology, with a particular emphasis on prevention and targeted treatment of a variety of cardiovascular conditions in cancer patients. Recently, the use of AI-augmented cardiac imaging in cardio-oncology is gaining traction. A large proportion of cardio-oncology patients are screened and followed using left ventricular ejection fraction (LVEF) and global longitudinal strain (GLS), currently obtained using echocardiography. This use will continue to increase with new cardiotoxic cancer treatments. AI is being tested to increase precision, throughput, and accuracy of LVEF and GLS, guide point-of-care image acquisition, and integrate imaging and clinical data to optimize the prediction and detection of cardiac dysfunction. The application of AI to cardiovascular magnetic resonance imaging (CMR), computed tomography (CT; especially coronary artery calcium or CAC scans), single proton emission computed tomography (SPECT) and positron emission tomography (PET) imaging acquisition is also in early stages of analysis for prediction and assessment of cardiac tumors and cardiovascular adverse events in patients treated for childhood or adult cancer. The opportunities for application of AI in cardio-oncology imaging are promising, and if availed, will improve clinical practice and benefit patient care.
Collapse
Affiliation(s)
- Nidhi Madan
- Division of Cardiology, Rush University Medical Center, Chicago, IL, USA
| | | | - Nausheen Akhter
- Division of Cardiology, Northwestern University, Chicago, IL, USA
| | - Patrick Collier
- Robert and Suzanne Tomsich Department of Cardiovascular Medicine, Sydell and Arnold Miller Family Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Avirup Guha
- Harrington Heart and Vascular Institute, Cleveland, OH, USA
| | - Lili Zhang
- Cardio-Oncology Program, Division of Cardiology, Department of Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Abhinav Sharma
- Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Imeh Ndiokho
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - Ethan Wen
- Medical College of Wisconsin, Milwaukee, WI, USA
| | - Noelle C. Garster
- Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| | | | - Sherry-Ann Brown
- Cardio-Oncology Program, Division of Cardiovascular Medicine, Medical College of Wisconsin, Milwaukee, WI, USA
| |
Collapse
|
46
|
Reynolds HR, Merz CNB, Berry C, Samuel R, Saw J, Smilowitz NR, de Souza ACDA, Sykes R, Taqueti VR, Wei J. Coronary Arterial Function and Disease in Women With No Obstructive Coronary Arteries. Circ Res 2022; 130:529-551. [PMID: 35175840 PMCID: PMC8911308 DOI: 10.1161/circresaha.121.319892] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Ischemic heart disease (IHD) is the leading cause of mortality in women. While traditional cardiovascular risk factors play an important role in the development of IHD in women, women may experience sex-specific IHD risk factors and pathophysiology, and thus female-specific risk stratification is needed for IHD prevention, diagnosis, and treatment. Emerging data from the past 2 decades have significantly improved the understanding of IHD in women, including mechanisms of ischemia with no obstructive coronary arteries and myocardial infarction with no obstructive coronary arteries. Despite this progress, sex differences in IHD outcomes persist, particularly in young women. This review highlights the contemporary understanding of coronary arterial function and disease in women with no obstructive coronary arteries, including coronary anatomy and physiology, mechanisms of ischemia with no obstructive coronary arteries and myocardial infarction with no obstructive coronary arteries, noninvasive and invasive diagnostic strategies, and management of IHD.
Collapse
Affiliation(s)
- Harmony R Reynolds
- Sarah Ross Soter Center for Women’s Cardiovascular Research, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - C. Noel Bairey Merz
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Colin Berry
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, G12 8TA, UK, West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK, Department of Cardiology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde Health Board, Glasgow, UK
| | - Rohit Samuel
- Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jacqueline Saw
- Vancouver General Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Nathaniel R Smilowitz
- Sarah Ross Soter Center for Women’s Cardiovascular Research, Leon H. Charney Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, New York, USA
| | - Ana Carolina do A.H. de Souza
- Cardiovascular Imaging Program, Departments of Radiology and Medicine (Cardiology), Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Robert Sykes
- British Heart Foundation Glasgow Cardiovascular Research Centre, University of Glasgow, G12 8TA, UK, West of Scotland Heart and Lung Centre, Golden Jubilee National Hospital, Glasgow, UK
| | - Viviany R. Taqueti
- Cardiovascular Imaging Program, Departments of Radiology and Medicine (Cardiology), Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Janet Wei
- Barbra Streisand Women’s Heart Center, Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
47
|
Tamargo J, Caballero R, Delpón E. Cancer Chemotherapy-Induced Sinus Bradycardia: A Narrative Review of a Forgotten Adverse Effect of Cardiotoxicity. Drug Saf 2022; 45:101-126. [PMID: 35025085 DOI: 10.1007/s40264-021-01132-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/20/2021] [Indexed: 12/20/2022]
Abstract
Cardiotoxicity is a common adverse effect of anticancer drugs (ACDs), including the so-called targeted drugs, and increases morbidity and mortality in patients with cancer. Attention has focused mainly on ACD-induced heart failure, myocardial ischemia, hypertension, thromboembolism, QT prolongation, and tachyarrhythmias. Yet, although an increasing number of ACDs can produce sinus bradycardia (SB), this proarrhythmic effect remains an underappreciated complication, probably because of its low incidence and severity since most patients are asymptomatic. However, SB merits our interest because its incidence increases with the aging of the population and cancer is an age-related disease and because SB represents a risk factor for QT prolongation. Indeed, several ACDs that produce SB also prolong the QT interval. We reviewed published reports on ACD-induced SB from January 1971 to November 2020 using the PubMed and EMBASE databases. Published reports from clinical trials, case reports, and recent reviews were considered. This review describes the associations between ACDs and SB, their clinical relevance, risk factors, and possible mechanisms of onset and treatment.
Collapse
Affiliation(s)
- Juan Tamargo
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain.
| | - Ricardo Caballero
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| | - Eva Delpón
- Department of Pharmacology, School of Medicine, Universidad Complutense, Institute of Health Gregorio Marañón, CIBERCV, 28040, Madrid, Spain
| |
Collapse
|
48
|
Caru M, Curnier D. The pediatric oncology exercise field speeds up to address important issues regarding chemotherapy-related cardiotoxicity. Front Pediatr 2022; 10:998337. [PMID: 36313894 PMCID: PMC9614334 DOI: 10.3389/fped.2022.998337] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 09/28/2022] [Indexed: 11/22/2022] Open
Affiliation(s)
- Maxime Caru
- Department of Pediatric, Division of Hematology and Oncology, Penn State College of Medicine, Hershey, PA, United States.,Department of Public Health Sciences, Penn State College of Medicine, Hershey, PA, United States
| | - Daniel Curnier
- School of Kinesiology and Physical Activity Sciences, Faculty of Medicine, University of Montreal, Montreal, QC, Canada.,Division of Pediatric Hematology and Oncology, Sainte-Justine University Health Center, Research Center, Montreal, QC, Canada
| |
Collapse
|
49
|
He X, Dai X, Ji J, Liu H, Shi G, Yeung SCJ. Nine-Year Median Follow-up of Cardiotoxicity and Efficacy of Trastuzumab Concurrently With Anthracycline-Based and Anthracycline-Free Neoadjuvant Chemotherapy in HER2-Positive Breast Cancer Patients. Clin Breast Cancer 2022; 22:e80-e90. [PMID: 34312098 DOI: 10.1016/j.clbc.2021.05.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 03/13/2021] [Accepted: 05/14/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The combination of trastuzumab with anthracycline chemotherapy drugs is associated with synergistic cardiotoxicity. The aim of this study is to compare the efficacy and late-onset cardiac toxicity of neoadjuvant chemotherapy regimens, trastuzumab plus paclitaxel followed by 5-fluorouracil, epirubicin, and cyclophosphamide (PH-FECH) versus trastuzumab plus docetaxel and carboplatin (TCH), for human epidermal growth factor receptor 2-positive (HER2+) breast cancer (BC). METHODS Patients with HER2+ BC who received neoadjuvant chemotherapy with PH-FECH or TCH between 2002 and 2009 at MD Anderson Cancer Center were included. The primary endpoint was progression-free survival (PFS). Secondary endpoints included pathological complete response (pCR), overall survival, cardiac events, breast cancer-specific survival, noncardiac toxicities, and chemotherapy interruption. RESULTS We identified 249 consecutive patients (184 who received PH-FECH and 65 who received TCH). The 10-year PFS was higher in the PH-FECH group than in the TCH group (83.6% vs. 72.2%; P = .044). The pCR rate was significantly higher in the PH-FECH group (58.2% vs. 41.5%; P = .021). The rate of cardiac events was higher in the PH-FECH group, but the difference was not significant (13.0% vs. 7.7%; P = .352). More patients developed late-onset cardiotoxicity in the PH-FECH group (3.8%) than in the TCH group (1.5%). Hypertension (odds ratio, 4.402 [95% confidence interval, 1.020-18.998]; P = .047) was an independent predictor of late-onset cardiotoxicity. CONCLUSIONS Both neoadjuvant regimens are effective and tolerable in patients with HER2+ BC. The PH-FECH regimen offers a higher pCR rate and higher PFS but no difference in overall survival or breast cancer-specific survival. Higher frequency of cardiac toxicity with PH-FECH was noted.
Collapse
Affiliation(s)
- Xuexin He
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Xiaolan Dai
- Department of Pharmacology, School of Medicine, Shantou University, Shantou, China
| | - Jiali Ji
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Hong Liu
- Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University, Hangzhou, China
| | - Ganggang Shi
- Department of Pharmacology, School of Medicine, Shantou University, Shantou, China
| | - Sai-Ching Jim Yeung
- Division of Internal Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
50
|
Li W, Liu M, Yu F, Jiang T, Zhu W, Liu H. Changes in epicardial adipose tissue among women treated with trastuzumab for breast cancer. Int J Cardiol 2021; 348:163-168. [PMID: 34890762 DOI: 10.1016/j.ijcard.2021.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/05/2021] [Accepted: 12/03/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Epicardial adipose tissue (EAT) as an endocrine organ, secreting hormones, and inflammatory cytokines is associated with adverse cardiovascular outcomes and may have a role in trastuzumab-induced cardiotoxicity (TIC). We sought to assess changes in EAT volume and radiodensity after receiving trastuzumab and if they are associated with TIC. METHODS A total of 185 women treated with trastuzumab for human epidermal growth factor receptor 2-positive breast cancer were retrospectively recruited. All patients underwent echocardiography and CT before and during trastuzumab therapy. The time interval between CT and echocardiography was <10 days. EAT volume and density were quantified by CT. TIC was defined as a left ventricular ejection fraction (LVEF) decrease of >10% and < 53%. RESULTS Of the 185 patients, 18 (9.7%) experienced TIC. After receiving trastuzumab, EAT volume and radiodensity were increased, despite similar BMI. TIC group showed a significantly higher increment of EAT volume (21.2 ± 6.3 vs. 11.7 ± 10.5 ml, p < 0.001) and radiodensity (2.7 ± 1.8 vs. 1.5 ± 2.0HU, p < 0.05) than no TIC group. There was a high negative correlation between changes in EAT volume and LVEF (r = -0.70; p < 0.001) and a moderately negative correlation between changes in EAT radiodensity and LVEF (r = -0.50; p < 0.001). Increased EAT volume, but not radiodensity appeared to be a good imaging biomarker for TIC (AUC: 0.79 vs. 0.65, p < 0.05). CONCLUSIONS EAT volume and radiodensity were increased after receiving trastuzumab particularly in the TIC patients despite similar BMI. Notably, the increased EAT volume rather than radiodensity was strongly negatively associated with LVEF and appeared to be a good imaging biomarker of TIC.
Collapse
Affiliation(s)
- Wenhuan Li
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China.
| | - Mingxi Liu
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China
| | - Fangfang Yu
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China
| | - Tao Jiang
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China
| | - Weiwei Zhu
- Department of Echocardiography, Heart Center, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China
| | - He Liu
- Department of Breast Surgery, Beijing Chao-Yang Hospital, Capital Medical University, 8 Gongren Tiyuchang Nanlu, Chaoyang District, Beijing 100020, China
| |
Collapse
|