101
|
Matusik K, Kamińska K, Sobiborowicz-Sadowska A, Borzuta H, Buczma K, Cudnoch-Jędrzejewska A. The significance of the apelinergic system in doxorubicin-induced cardiotoxicity. Heart Fail Rev 2024; 29:969-988. [PMID: 38990214 PMCID: PMC11306362 DOI: 10.1007/s10741-024-10414-w] [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] [Accepted: 06/27/2024] [Indexed: 07/12/2024]
Abstract
Cancer is the leading cause of death worldwide, and the number of cancer-related deaths is expected to increase. Common types of cancer include skin, breast, lung, prostate, and colorectal cancers. While clinical research has improved cancer therapies, these treatments often come with significant side effects such as chronic fatigue, hair loss, and nausea. In addition, cancer treatments can cause long-term cardiovascular complications. Doxorubicin (DOX) therapy is one example, which can lead to decreased left ventricle (LV) echocardiography (ECHO) parameters, increased oxidative stress in cellular level, and even cardiac fibrosis. The apelinergic system, specifically apelin and its receptor, together, has shown properties that could potentially protect the heart and mitigate the damages caused by DOX anti-cancer treatment. Studies have suggested that stimulating the apelinergic system may have therapeutic benefits for heart damage induced by DOX. Further research in chronic preclinical models is needed to confirm this hypothesis and understand the mechanism of action for the apelinergic system. This review aims to collect and present data on the effects of the apelinergic system on doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Katarzyna Matusik
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Katarzyna Kamińska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland.
| | - Aleksandra Sobiborowicz-Sadowska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Hubert Borzuta
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Kasper Buczma
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| | - Agnieszka Cudnoch-Jędrzejewska
- Department of Experimental and Clinical Physiology, Laboratory of Centre for Preclinical Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
102
|
Asar TO, Al-Abbasi FA, Sheikh RA, Zeyadi MAM, Nadeem MS, Naqvi S, Kumar V, Anwar F. Metformin's dual impact on Gut microbiota and cardiovascular health: A comprehensive analysis. Biomed Pharmacother 2024; 178:117128. [PMID: 39079259 DOI: 10.1016/j.biopha.2024.117128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 08/25/2024] Open
Abstract
Cardiovascular diseases (CVD) cause significant global morbidity, mortality and public health burden annually. CVD alters richness, diversity, and composition of Gut microbiota along with RAS and histopathological differences. Present study explores Metformin role in mitigating doxorubicin induced cardiovascular toxicity/remodeling. Animals were divided into 4 groups with n=6: Group I (N. Control) free access to diet and water; Group II (MET. Control) on oral Metformin (250 mg/kg) daily; Group III (DOX. Control) alternate day intraperitoneal Doxorubicin (3 mg/kg) totaling 18 mg/kg; Group IV (DOX. MET. Control) received both daily oral Metformin (250 mg/kg) and alternate day Doxorubicin (3 mg/kg). Gut microbial analysis was made from stool before animals were sacrificed for biochemical and histopathological analysis. Significant alterations were observed in ɑ and β-diversity with new genus from Firmicutes, specifically Clostridia_UCG-014, Eubacterium ruminantium, and Tunicibacter, were prevalent in both the DOX. Control and DOX.MET groups. Proteobacteria, represented by Succinivibrio, were absent in all groups. Additionally, Parabacteroides from the Bacteroidia phylum was absent in all groups except the N. control. In the DOX.MET Control group, levels of Angiotensin II ( 7.75± 0.49 nmol/min, p<0.01) and Renin (2.60±0.26 ng/ml/hr) were significantly reduced. Conversely, levels of CK-MB, Fibrinogen, Troponin, CRP ( p < 0.0001), and TNFɑ (p < 0.05) were elevated. Histopathological examination revealed substantial cardiac changes, including Fibrinogen and fat deposition and eosinophilic infiltration, as well as liver damage characterized by binucleated cells and damaged hepatocytes, along with altered renal tissues in the DOX.MET.Control group. The findings suggest that MET. significantly modifies gut microbiota, particularly impacting the Firmicutes and Proteobacteria phyla. The reduction in Angiotensin II levels, alongside increased inflammatory markers and myocardial damage, highlights the complex interactions and potential adverse effects associated with MET therapy on cardiovascular health.
Collapse
Affiliation(s)
- Turky Omar Asar
- Department of Biology, College of Science and Arts at Alkamil, University of Jeddah, Jeddah, Saudi Arabia.
| | - Fahad A Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Ryan Adnan Sheikh
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | | | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| | - Salma Naqvi
- Department of Biomedical Sciences, College of Medicine, Gulf Medical University, Ajman, United Arab Emirates.
| | - Vikas Kumar
- Natural Product Discovery Laboratory, Department of Pharmaceutical Sciences, Shalom Institute of Health and Allied Sciences, SHUATS, Prayagraj, India.
| | - Firoz Anwar
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
103
|
Belenkov YN, Ilgisonis IS, Khabarova NV, Kirichenko Yu Yu YY. Modern Instrumental Methods of Diagnostics and Risk Assessment of Developing Antitumor Therapy Cardiovasculotoxicity. KARDIOLOGIIA 2024; 64:3-12. [PMID: 39262348 DOI: 10.18087/cardio.2024.8.n2753] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 09/03/2024] [Indexed: 09/13/2024]
Abstract
The most important component of cardio-oncology is the assessment of the risk of development and diagnosis of cardiovascular toxicity of the antitumor therapy, the detection of which is largely based on visualization of the cardiovascular system. The article addresses up-to-date methods of non-invasive visualization of the heart and blood vessels, according to the 2022 European Society of Cardiology Clinical Guidelines on cardio-oncology. Also, the article discusses promising cardiovascular imaging techniques that are not yet included in the guidelines: assessment of coronary calcium using multislice computed tomography and positron emission computed tomography with 18F-labeled 2-deoxy-2-fluoro-d-glucose.
Collapse
Affiliation(s)
- Yu N Belenkov
- Sechenov First Moscow Medical University; Department of Hospital Therapy #1, Sklifosovsky Institute of Clinical Medicine, Moscow
| | - I S Ilgisonis
- Sechenov First Moscow Medical University; Department of Hospital Therapy #1, Sklifosovsky Institute of Clinical Medicine, Moscow
| | - N V Khabarova
- Sechenov First Moscow Medical University; Department of Hospital Therapy #1, Sklifosovsky Institute of Clinical Medicine, Moscow
| | - Yu Yu Kirichenko Yu Yu
- Sechenov First Moscow Medical University; Department of Hospital Therapy #1, Sklifosovsky Institute of Clinical Medicine, Moscow
| |
Collapse
|
104
|
Bertolini D, Pizzi C, Donal E, Galli E. Cancer and Heart Failure: Dangerous Liaisons. J Cardiovasc Dev Dis 2024; 11:263. [PMID: 39330321 PMCID: PMC11432566 DOI: 10.3390/jcdd11090263] [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: 06/09/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Cancer and heart failure (HF) are increasingly relevant worldwide, both from an epidemiologic and clinical point of view. This review aims to explore the relationship between cancer and HF by underscoring risk factors and disclosing the cardiotoxic effects of the current chemotherapy agents. We also deal with the current evidence on the diagnosis and management of HF related to cancer therapy. Finally, we will address the main gaps in knowledge and future perspectives in this field.
Collapse
Affiliation(s)
- Davide Bertolini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Carmine Pizzi
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Erwan Donal
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| | - Elena Galli
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| |
Collapse
|
105
|
Kim JH. Profiling the Cardiovascular Toxicities of CDK4/6 Inhibitors: A Real-World Pharmacovigilance Study. Cancers (Basel) 2024; 16:2869. [PMID: 39199640 PMCID: PMC11352810 DOI: 10.3390/cancers16162869] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/12/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Cyclin-dependent kinase 4 and 6 (CDK4/6) inhibitors are approved for the treatment of human epidermal growth factor receptor 2 (HER-2)-negative, hormone receptor-positive breast cancer. The cardiovascular toxicity of CDK4/6 inhibitors is not well understood. This study aims to profile the cardiac events associated with CDK4/6 inhibitors. Reports from 2015Q1 to 2024Q1 were obtained from the FDA Adverse Event Reporting System (FAERS). Reports identifying palbociclib, ribociclib, and abemaciclib as the primary suspect were examined for cardiovascular toxicity, including hypertension, cardiac failure, cardiomyopathy, arrhythmia, myocardial infarction, and myocarditis. Signal detection was performed using the proportional reporting ratio (PRR), reporting odds ratio (ROR), and information component (IC). A total of 69,139 reports were analyzed. The median time to adverse events was 69 days (interquartile range [IQR], 18-260 days). Of these, 2065 reports documented cardiac adverse events. Ribociclib and QT prolongation were re-confirmed as a signal (PRR 8.43, ROR 8.65, IC025 2.86). Hypertension and cardiac failure were the most frequently reported cardiovascular toxicities. This study demonstrates that the use of CDK4/6 inhibitors is associated with cardiovascular adverse events, such as heart failure and hypertension. Further research is needed to understand the mechanisms and risk factors contributing to the cardiovascular toxicity of CDK4/6 inhibitors.
Collapse
Affiliation(s)
- Jae Hyun Kim
- School of Pharmacy and Institute of New Drug Development, Jeonbuk National University, Jeonju 54896, Republic of Korea; ; Tel.: +82-63-219-5638
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju 54907, Republic of Korea
| |
Collapse
|
106
|
Lipe DN, Qdaisat A, Krishnamani PP, Nguyen TD, Chaftari P, El Messiri N, Srinivasan A, Galvis-Carvajal E, Reyes-Gibby CC, Wattana MK. Myocarditis, Myositis, and Myasthenia Gravis Overlap Syndrome Associated with Immune Checkpoint Inhibitors: A Systematic Review. Diagnostics (Basel) 2024; 14:1794. [PMID: 39202282 PMCID: PMC11353298 DOI: 10.3390/diagnostics14161794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/24/2024] [Accepted: 08/09/2024] [Indexed: 09/03/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have significantly transformed cancer treatment, but their use is linked to immune-related adverse events (irAEs), including the rare ICI-associated myocarditis, myositis, and myasthenia gravis (MMM) overlap syndrome. This systematic review aims to highlight MMM's clinical implications in emergency departments. PubMed and Embase were searched using a specific search strategy. Reports were eligible for inclusion if all three conditions were present and associated with the use of an ICI. Data were extracted by independent reviewers using the Rayyan web application for systematic reviews. Descriptive statistics and qualitative synthesis were used to summarize demographic, clinical, and treatment data for the reported cases. Among 50 cases, predominantly associated with melanoma, lung cancer, and renal cancer, the in-hospital mortality rate was 38.0%. The most commonly presenting symptoms were ptosis (58%), dyspnea (48%), diplopia (42%), or myalgia (36%). The median time from ICI initiation to MMM presentation was 21 days (interquartile range: 15-28 days). Corticosteroids were the primary treatment for the irAEs. MMM, a rare but potentially fatal complication of ICI therapy, requires prompt recognition in emergency settings. Corticosteroids should be initiated if suspected, without waiting for confirmation. Multidisciplinary collaboration is vital for diagnosis and treatment planning. Research on MMM's link to specific cancers and ICIs is imperative for better risk assessment and interventions.
Collapse
Affiliation(s)
| | - Aiham Qdaisat
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.Q.)
| | - Pavitra P. Krishnamani
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.Q.)
| | - Trung D. Nguyen
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.Q.)
| | - Patrick Chaftari
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.Q.)
| | - Nour El Messiri
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.Q.)
| | - Aswin Srinivasan
- Department of Cardiology, HCA Houston Kingwood, College of Medicine, University of Houston, Kingwood, TX 77339, USA
| | - Elkin Galvis-Carvajal
- Department of Emergency Medicine, Baylor College of Medicine, Houston, TX 77030, USA
| | - Cielito C. Reyes-Gibby
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.Q.)
| | - Monica K. Wattana
- Department of Emergency Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA; (A.Q.)
| |
Collapse
|
107
|
Yang C, Song X, Sun H, Chen X, Liu C, Chen M. Cardiovascular adverse events associated with PARP inhibitors for ovarian cancer: a real world study (RWS) with Bayesian disproportional analysis based on the FDA adverse event reporting system (FAERS). Expert Opin Drug Saf 2024:1-8. [PMID: 39132853 DOI: 10.1080/14740338.2024.2390640] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 06/27/2024] [Indexed: 08/13/2024]
Abstract
BACKGROUND To investigate the pharmacovigilance (PV) and make pairwise comparisons on reporting proportion, seriousness, and severity of outcomes of major adverse cardiovascular events (MACE) among poly(ADP-ribose) polymerase-inhibitors (PARPis) in treating ovarian cancer, fallopian tube carcinoma, and primary peritoneal cancer (collectively named EOC) from the US Food and Drug Administration Adverse Event Reporting System (FAERS). RESEARCH DESIGN AND METHODS Data on adverse cardiovascular events reports related to EOC treatment submitted to FAERS from the first quarter of 2015 to the second quarter of 2023 were harvested. Three PARPis were identified: olaparib, niraparib, and rucaparib. RESULTS Eventually, a total of 258,596 eligible records were enrolled with 12,331 reports including 5,292 reports of MACE and 7,039 reports of other cardiovascular events. For the primary composite endpoint, a PV signal associated with MACE was detected in niraparib (ROR = 1.12; IC025 = 0.03), whereas it was not detected in olaparib and rucaparib; For the secondary endpoint, PV signals associated with other cardiovascular events were detected in niraparib (ROR = 1.17;IC025 = 0.04), but not in olaparib and rucaparib. CONCLUSIONS For EOC patients, close monitoring of blood pressure, heart rate, and coagulation function should be conducted when selecting niraparib for treatment.
Collapse
Affiliation(s)
- Chenguang Yang
- Department of Gynaecology and Obstetrics, Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, Anhui Province, China
| | - Xuan Song
- Department of General Medicine, Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, Anhui Province, China
| | - Hongmei Sun
- Department of Gynaecology and Obstetrics, Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, Anhui Province, China
| | - Xi Chen
- Department of Epidemiology and Statistics, School of Public Health, Medical College, Zhejiang University, Zhejiang, China
| | - Chengjiang Liu
- Department of General Medicine, Affiliated Anqing First People's Hospital of Anhui Medical University, Hefei, China
| | - Min Chen
- Department of Gynaecology and Obstetrics, Affiliated Xuancheng Hospital of Wannan Medical College, Xuancheng, Anhui Province, China
| |
Collapse
|
108
|
Nielsen MØ, Ljoki A, Zerahn B, Jensen LT, Kristensen B. Reproducibility and Repeatability in Focus: Evaluating LVEF Measurements with 3D Echocardiography by Medical Technologists. Diagnostics (Basel) 2024; 14:1729. [PMID: 39202217 PMCID: PMC11353652 DOI: 10.3390/diagnostics14161729] [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: 06/28/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 09/03/2024] Open
Abstract
Three-dimensional echocardiography (3DE) is currently the preferred method for monitoring left ventricular ejection fraction (LVEF) in cancer patients receiving potentially cardiotoxic anti-neoplastic therapy. In Denmark, however, the traditional standard for LVEF monitoring has been rooted in nuclear medicine departments utilizing equilibrium radionuclide angiography (ERNA). Although ERNA remains a principal modality, there is an emerging trend towards the adoption of echocardiography for this purpose. Given this context, assessing the reproducibility of 3DE among non-specialized medical personnel is crucial for its clinical adoption in such departments. To assess the feasibility of 3DE for LVEF measurements by technologists, we evaluated the repeatability and reproducibility of two moderately experienced technologists. They performed 3DE on 12 volunteers over two sessions, with a collaborative review of the results from the first session before the second session. Two-way intraclass correlation values increased from 0.03 to 0.77 across the sessions. This increase in agreement was mainly due to the recognition of false low measurements. Our findings underscore the importance of incorporating reproducibility exercises in the context of 3DE, especially when operated by technologists. Additionally, routine control of the acquisitions by physicians is deemed necessary. Ensuring these hurdles are adequately managed enables the adoption of 3DE for LVEF measurements by technologists.
Collapse
Affiliation(s)
- Marc Østergaard Nielsen
- Department of Nuclear Medicine, Herlev University Hospital, 2730 Herlev, Denmark; (A.L.); (B.Z.); (L.T.J.); (B.K.)
| | | | | | | | | |
Collapse
|
109
|
Sun SJ, Jiao XD, Chen ZG, Cao Q, Zhu JH, Shen QR, Liu Y, Zhang Z, Xu FF, Shi Y, Tong J, Ouyang SX, Fu JT, Zhao Y, Ren J, Li DJ, Shen FM, Wang P. Gasdermin-E-mediated pyroptosis drives immune checkpoint inhibitor-associated myocarditis via cGAS-STING activation. Nat Commun 2024; 15:6640. [PMID: 39103324 PMCID: PMC11300882 DOI: 10.1038/s41467-024-50996-5] [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: 10/24/2023] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Immune checkpoint inhibitor (ICI)-induced myocarditis involves intensive immune/inflammation activation; however, its molecular basis is unclear. Here, we show that gasdermin-E (GSDME), a gasdermin family member, drives ICI-induced myocarditis. Pyroptosis mediated by GSDME, but not the canonical GSDMD, is activated in myocardial tissue of mice and cancer patients with ICI-induced myocarditis. Deficiency of GSDME in male mice alleviates ICI-induced cardiac infiltration of T cells, macrophages, and monocytes, as well as mitochondrial damage and inflammation. Restoration of GSDME expression specifically in cardiomyocytes, rather than myeloid cells, in GSDME-deficient mice reproduces ICI-induced myocarditis. Mechanistically, quantitative proteomics reveal that GSDME-dependent pyroptosis promotes cell death and mitochondrial DNA release, which in turn activates cGAS-STING signaling, triggering a robust interferon response and myocardial immune/inflammation activation. Pharmacological blockade of GSDME attenuates ICI-induced myocarditis and improves long-term survival in mice. Our findings may advance the understanding of ICI-induced myocarditis and suggest that targeting the GSDME-cGAS-STING-interferon axis may help prevent and manage ICI-associated myocarditis.
Collapse
Affiliation(s)
- Si-Jia Sun
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Xiao-Dong Jiao
- Department of Oncology, Changzheng Hospital, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Zhi-Gang Chen
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, Shanghai, China
| | - Qi Cao
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Jia-Hui Zhu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Qi-Rui Shen
- Department of Pharmacy, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yi Liu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Zhen Zhang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fang-Fang Xu
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Yu Shi
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jie Tong
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shen-Xi Ouyang
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jiang-Tao Fu
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China
| | - Yi Zhao
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Jun Ren
- Department of Cardiology and Shanghai Institute of Cardiovascular Disease, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Dong-Jie Li
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Fu-Ming Shen
- Department of Pharmacy, Shanghai Tenth People's Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Pei Wang
- The Center for Basic Research and Innovation of Medicine and Pharmacy (MOE), School of Pharmacy, Naval Medical University/Second Military Medical University, Shanghai, China.
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, Naval Medical University/Second Military Medical University, Shanghai, China.
- The National Demonstration Center for Experimental Pharmaceutical Education, Naval Medical University/Second Military Medical University, Shanghai, China.
| |
Collapse
|
110
|
Jacquemyn X, Zhan J, Van den Eynde J, Cordrey K, Long R, Rao S, Barnes BT, Thompson WR, Danford D, Kutty S. Time course of hypertension and myocardial dysfunction following anthracycline chemotherapy in pediatric patients. IJC HEART & VASCULATURE 2024; 53:101436. [PMID: 38872982 PMCID: PMC11169083 DOI: 10.1016/j.ijcha.2024.101436] [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: 12/28/2023] [Revised: 04/11/2024] [Accepted: 05/27/2024] [Indexed: 06/15/2024]
Abstract
Background Anthracyclines are associated with cardiac dysfunction. Little is known about the interplay of pre-existing hypertension and treatment response. We aimed to investigate the relationship between hypertension and the development of cancer therapy-related cardiac dysfunction (CTRCD) in pediatric patients treated with anthracycline chemotherapy. Methods Pediatric patients with cancer who received anthracycline chemotherapy from 2013 to 2021 were retrospectively included. Serial cardiac assessments were conducted during and after chemotherapy. The primary outcome was the development of CTRCD, classified as mild, moderate, or severe according to contemporary definitions. Results Among 190 patients undergoing anthracycline chemotherapy, 34 patients (17.9 %) had hypertension (24 patients Stage 1, and 10 patients Stage 2) at baseline evaluation. Patients underwent chemotherapy for a median of 234.4 days (interquartile range 127.8-690.3 days) and were subsequently followed up. Hypertension was frequent during follow-up 31.3 % (0-3 months), 15.8 % (3-6 months), 21.9 % (0.5-1 years), 24.7 % (1-2 years), 31.1 % (2-4 years) and 35.8 % (beyond 4 years) (P for trend < 0.001). Freedom from mild CTRCD at 5 years was 45.0 %, freedom from moderate CTRCD was 87.8 % at 5 years. Baseline hypertension did not increase the risk of mild (HR 0.77, 95 % CI: 0.41-1.42, P = 0.385) or moderate CTRCD (HR 0.62, 95 % CI: 0.14-2.72, P = 0.504). Patients with baseline hypertension showed different global longitudinal strain (P < 0.001) and LVEF (P < 0.001) patterns during follow-up. Conclusions Pediatric patients often develop CTRCD post-anthracycline chemotherapy. Those with pre-existing hypertension show a unique treatment response, despite no increased CTRCD risk, warranting further investigation.
Collapse
Affiliation(s)
- Xander Jacquemyn
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Junzhen Zhan
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Jef Van den Eynde
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
- Department of Cardiovascular Sciences, KU Leuven & Congenital and Structural Cardiology, UZ Leuven, Leuven, Belgium
| | - Kyla Cordrey
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Rita Long
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Sruti Rao
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Benjamin T. Barnes
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - W. Reid Thompson
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - David Danford
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| | - Shelby Kutty
- The Blalock-Taussig-Thomas Pediatric and Congenital Heart Center, Department of Pediatrics, Johns Hopkins School of Medicine, Johns Hopkins University, Baltimore, MD, USA
| |
Collapse
|
111
|
Lee S, Alsamarrai A, Xiao A, Wang TKM. Prevention of anthracycline and trastuzumab-induced decline in left ventricular ejection fraction with angiotensin-converting enzyme inhibitors or angiotensin receptor blocker: a narrative systematic review of randomised controlled trials. Intern Med J 2024; 54:1254-1263. [PMID: 38874281 DOI: 10.1111/imj.16437] [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] [Accepted: 05/13/2024] [Indexed: 06/15/2024]
Abstract
Cancer therapy-related cardiac dysfunction (CTRCD) is a complication of selected cancer therapy agents associated with decline in left ventricular ejection fraction (LVEF). Angiotensin-converting enzyme inhibitors (ACEIs) and angiotensin receptor blockers (ARBs) have established benefits in heart failure with reduced ejection fraction, but their efficacy for preventing CTRCD remains controversial. This narrative systematic review assessed the efficacy and safety of ACEI/ARB in the prevention of cancer therapy LVEF decline. We systematically searched PubMed, Embase and Cochrane from January 1980 to June 2022. Studies of interest were randomised controlled trials of patients with normal LVEF and active malignancy receiving cancer therapy, randomised to receive either an ACEI or ARB compared with a control group. The outcome was the change in LVEF from baseline to the end of the follow-up period. Death, clinical heart failure and adverse drug reactions were recorded. A total of 3731 search records were screened and 12 studies were included, comprising a total of 1645 participants. Nine studies assessed the prevention of anthracycline-induced LVEF decline, of which five showed a beneficial effect (1%-14% higher LVEF in treated groups), whereas four studies showed no effect. Three studies assessed the prevention of trastuzumab-induced LVEF decline, of which one showed a beneficial effect (4% higher LVEF) in a subset of participants. There are mixed data regarding the efficacy of ACEI/ARB in preventing the LVEF decline in patients undergoing anthracycline or trastuzumab therapy, with evidence suggesting no clinically meaningful benefit observed in recent studies.
Collapse
Affiliation(s)
- Simon Lee
- Cardiology Department, Middlemore Hospital, Auckland, New Zealand
- Cardiology Department, Tauranga Hospital, Tauranga, New Zealand
| | - Ammar Alsamarrai
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
| | - Amy Xiao
- Cardiology Department, Middlemore Hospital, Auckland, New Zealand
| | - Tom K M Wang
- Green Lane Cardiovascular Service, Auckland City Hospital, Auckland, New Zealand
- Department of Cardiovascular Medicine, Heart, Vascular, and Thoracic Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
112
|
Mirabel M, Eslami A, Thibault C, Oudard S, Mousseaux E, Wahbi K, Fabre E, Terrier B, Marijon E, Villefaillot A, Fayol A, Dragon-Durey MA, Le Louet AL, Bruno RM, Soulat G, Hulot JS. Adverse myocardial and vascular side effects of immune checkpoint inhibitors: a prospective multimodal cardiovascular assessment. Clin Res Cardiol 2024; 113:1263-1273. [PMID: 38806821 DOI: 10.1007/s00392-024-02462-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 05/15/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) can induce cardiovascular toxicities. OBJECTIVES To prospectively assess the incidence of major cardiovascular events (MACE) on ICIs in solid cancer patients: myocarditis, pericarditis, acute coronary syndrome, heart failure, high-degree conduction abnormalities or sustained ventricular arrhythmias, or cardiovascular death at 6 weeks (early MACE), including asymptomatic clinical changes by an independent adjudication committee using current recommended diagnostic criteria. The secondary objective was the incidence of the above-mentioned events adding atrial fibrillation (AF) at 6 months (late MACE). RESULTS Participants underwent pre-ICIs and repeated multimodality cardiac imaging (echocardiogram, cardiac magnetic resonance (CMR)), serum biomarkers (ultrasensitive troponin I), and rhythm surveillance (ambulatory ECG monitoring) at 6 weeks and 6 months. Forty-nine patients (38 (77.6%) male; mean age 64.3 (SD 11.0) years old) were included (June 2020-December 2021). Early MACE were observed in 9 (18.4%) patients at mean 40.1 (SD 5.9) days, with heart failure (HF) in 5 (10.2%), ventricular arrhythmias, or new conduction disorders in 4 (8.2%) patients. History of AF (HR 4.49 (CI 1.11-18.14), P = 0.035) predicted early MACE. At 6 months follow-up, 18 MACE were observed in 15/49 (31%) patients, with 6 (12.2%) HF events, 5 (10.2%) significant ventricular arrhythmias, or conduction disorders, and 4 (8.2%) AF. There was a significant decline in LVEF (P < 0.001) in patients with no MACE (P = 0.003) or HF (P = 0.0028). Higher creatinine at inclusion (HR 0.99 [0.98-1.00], P = 0.006) predicted HF on multivariate analysis. There were no significant T1 or T2 mapping changes in our study cohort on repeated CMR. CONCLUSIONS Cardiotoxicity on ICIs is more frequent than previously described when using a thorough detection strategy, consisting mainly in HF and asymptomatic rhythm disorders.
Collapse
Affiliation(s)
- Mariana Mirabel
- Cardiology, Institut Mutualiste Montsouris, 42 bd Jourdan, 75014, Paris, France.
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France.
| | - Assié Eslami
- Cardiology, DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Constance Thibault
- Oncology, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Stéphane Oudard
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France
- Oncology, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Elie Mousseaux
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France
- Radiology, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Karim Wahbi
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France
- Cardiology, DMU CARTE, AP-HP, Hôpital Cochin, F-75014, Paris, France
| | - Elizabeth Fabre
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France
- Thoracic Oncology, AP-HP, CARPEM Cancer Institute, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | | | - Eloi Marijon
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France
- Oncology, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Aurélie Villefaillot
- Unité de Recherche Clinique, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Antoine Fayol
- Cardiology, DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | | | - Agnès Lillo Le Louet
- Pharmacovigilance, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | | | - Gilles Soulat
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France
- Radiology, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| | - Jean Sébastien Hulot
- Université Paris Cité, INSERM, PARCC, F-75015, Paris, France
- CIC1418 and DMU CARTE, AP-HP, Hôpital Européen Georges-Pompidou, F-75015, Paris, France
| |
Collapse
|
113
|
Aznar MC, Bergler-Klein J, Boriani G, Cutter DJ, Hurkmans C, Levis M, López-Fernández T, Lyon AR, Maraldo MV. Cardiovascular toxicities of radiotherapy: From practical issues to new perspectives. Radiother Oncol 2024; 197:110336. [PMID: 38797493 DOI: 10.1016/j.radonc.2024.110336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Revised: 05/09/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024]
Affiliation(s)
- Marianne C Aznar
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, the United Kingdom of Great Britain and Northern Ireland.
| | | | - Giuseppe Boriani
- Cardiology Division, Department of Biomedical, Metabolic and Neural SciencesUniversity of Modena and Reggio Emilia, Policlinico di Modena, Modena, Italy
| | - David J Cutter
- Nuffield Department of Population Health, University of Oxford, Oxford, the United Kingdom of Great Britain and Northern Ireland; Oxford Cancer and Haematology Centre, Oxford University Hospitals NHS Foundation Trust, Churchill Hospital, Oxford, the United Kingdom of Great Britain and Northern Ireland
| | - Coen Hurkmans
- Dept of Radiation Therapy, Catharina Hospital Eindhoven, the Netherlands; Dept of Electrical Engineering and Dept of Applied Physics, Technical University Eindhoven, the Netherlands
| | - Mario Levis
- Department of Oncology, University of Turin, Turin, Italy
| | - Teresa López-Fernández
- Cardiology Department, Cardio-Oncology Unit, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain; Cardiology Department, Cardio-Oncology Unit, Quironsalud Madrid University Hospital, Madrid, Spain
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, London, the United Kingdom of Great Britain and Northern Ireland
| | - Maja V Maraldo
- Dept of Oncology, Copenhagen University Hospital - Rigshospitalet, Copenhagen, Denmark; Dept of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
114
|
Gent DG, Saif M, Dobson R, Wright DJ. Cardiovascular Disease After Hematopoietic Stem Cell Transplantation in Adults: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:475-495. [PMID: 39239331 PMCID: PMC11372032 DOI: 10.1016/j.jaccao.2024.06.004] [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: 02/01/2024] [Revised: 06/11/2024] [Accepted: 06/28/2024] [Indexed: 09/07/2024] Open
Abstract
The use of hematopoietic cell transplantation (HCT) has expanded in the last 4 decades to include an older and more comorbid population. These patients face an increased risk of cardiovascular disease after HCT. The risk varies depending on several factors, including the type of transplant (autologous or allogeneic). Many therapies used in HCT have the potential to be cardiotoxic. Cardiovascular complications after HCT include atrial arrhythmias, heart failure, myocardial infarction, and pericardial effusions. Before HCT, patients should undergo a comprehensive cardiovascular assessment, with ongoing surveillance tailored to their individual level of cardiovascular risk. In this review, we provide an overview of cardiotoxicity after HCT and outline our approach to risk assessment and ongoing care.
Collapse
Affiliation(s)
- David G Gent
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - Muhammad Saif
- The Clatterbridge Cancer Centre, Liverpool, United Kingdom
| | - Rebecca Dobson
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| | - David J Wright
- Liverpool Centre for Cardiovascular Science, University of Liverpool and Liverpool Heart and Chest Hospital, Liverpool, United Kingdom
| |
Collapse
|
115
|
Liu X, Ge S, Zhang A. Pediatric Cardio-Oncology: Screening, Risk Stratification, and Prevention of Cardiotoxicity Associated with Anthracyclines. CHILDREN (BASEL, SWITZERLAND) 2024; 11:884. [PMID: 39062333 PMCID: PMC11276082 DOI: 10.3390/children11070884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/02/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024]
Abstract
Anthracyclines have significantly improved the survival of children with malignant tumors, but the associated cardiotoxicity, an effect now under the purview of pediatric cardio-oncology, due to its cumulative and irreversible effects on the heart, limits their clinical application. A systematic screening and risk stratification approach provides the opportunity for early identification and intervention to mitigate, reverse, or prevent myocardial injury, remodeling, and dysfunction associated with anthracyclines. This review summarizes the risk factors, surveillance indexes, and preventive strategies of anthracycline-related cardiotoxicity to improve the safety and efficacy of anthracyclines.
Collapse
Affiliation(s)
- Xiaomeng Liu
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Shuping Ge
- Department of Pediatric and Adult Congenital Cardiology, Geisinger Clinic, Danville, PA 17822, USA
| | - Aijun Zhang
- Department of Pediatrics, Qilu Hospital of Shandong University, Jinan 250012, China
| |
Collapse
|
116
|
Kovoor JG, Nann SD, Chambers C, Mishra K, Goel S, Thompson I, Koh D, Litwin P, Bacchi S, Harford PJ, Stretton B, Gupta AK. Prehabilitation before general surgery: Worth the effort? J Perioper Pract 2024; 34:219-225. [PMID: 38149496 DOI: 10.1177/17504589231214395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2023]
Abstract
Prehabilitation, or interventions before surgery aimed at improving preoperative health and postoperative outcomes, has various forms. Although it may confer benefit to patients undergoing general surgery, this is not certain. Furthermore, although it may yield a net monetary gain, it is also likely to require substantial monetary and non-monetary investment. The impact of prehabilitation is highly variable and dependent on multiple factors. Physical function and pulmonary outcomes are likely to be improved by most forms of prehabilitation involving physical and multimodal exercise programmes. However, other surgical outcomes have demonstrated mixed results from prehabilitation. Within this issue, the measures used for evaluating baseline patient biopsychosocial health are important, and collecting sufficient data to accurately inform patient-centred prehabilitation programmes is only possible through thorough clinical and laboratory investigation and synthesised metrics such as cardiopulmonary exercise testing. Although a multimodal approach to prehabilitation is the current gold standard, societal factors may affect engagement with programmes that require a significant in-person activity. However, this is weighed against the substantial financial and non-financial investment that accompanies many programmes. The overall effectiveness and optimal mode of intervention across the discipline of general surgery remains unclear, and further research is needed to prove prehabilitation's full worth.
Collapse
Affiliation(s)
- Joshua G Kovoor
- Discipline of Surgery, The University of Adelaide, Adelaide, SA, Australia
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
- Health and Information, Adelaide, SA, Australia
| | - Silas D Nann
- Health and Information, Adelaide, SA, Australia
- Gold Coast University Hospital, Gold Coast, QLD, Australia
| | - Courtney Chambers
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Kritika Mishra
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Sahil Goel
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Isabella Thompson
- Gold Coast University Hospital, Gold Coast, QLD, Australia
- Bond University, Gold Coast, QLD, Australia
| | - Dong Koh
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Peter Litwin
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
- Health and Information, Adelaide, SA, Australia
| | - Stephen Bacchi
- Discipline of Surgery, The University of Adelaide, Adelaide, SA, Australia
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
- Health and Information, Adelaide, SA, Australia
| | - Philip J Harford
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
| | - Brandon Stretton
- Discipline of Surgery, The University of Adelaide, Adelaide, SA, Australia
- The Queen Elizabeth Hospital, Adelaide, SA, Australia
- Royal Adelaide Hospital, Adelaide, SA, Australia
- Health and Information, Adelaide, SA, Australia
| | - Aashray K Gupta
- Discipline of Surgery, The University of Adelaide, Adelaide, SA, Australia
- Health and Information, Adelaide, SA, Australia
- Gold Coast University Hospital, Gold Coast, QLD, Australia
| |
Collapse
|
117
|
Díaz-Balboa E, Peña-Gil C, Rodríguez-Romero B, Cuesta-Vargas AI, Lado-Baleato O, Martínez-Monzonís A, Pedreira-Pérez M, Palacios-Ozores P, López-López R, González-Juanatey JR, González-Salvado V. Exercise-based cardio-oncology rehabilitation for cardiotoxicity prevention during breast cancer chemotherapy: The ONCORE randomized controlled trial. Prog Cardiovasc Dis 2024; 85:74-81. [PMID: 38395212 DOI: 10.1016/j.pcad.2024.02.002] [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: 02/16/2024] [Accepted: 02/16/2024] [Indexed: 02/25/2024]
Abstract
BACKGROUND Breast cancer (BC) treatment with anthracyclines and/or anti-human epidermal growth factor receptor-2 (HER2) antibodies is associated with an increased risk of cardiovascular disease complications, including cancer therapy-related cardiac dysfunction (CTRCD). While Cardio-Oncology Rehabilitation (CORe) programs including exercise have emerged to minimize these risks, its role in preventing CTRCD is unclear. OBJECTIVES We investigated the effectiveness of an exercise-based CORe program in preventing CTRCD [left ventricular ejection fraction (LVEF) drop ≥10% to a value <53% or a decrease >15% in global longitudinal strain (GLS)]. Secondary outcomes examined changes in cardiac biomarkers, physical performance including peak oxygen consumption, psychometric and lifestyle outcomes. Safety, adherence, and patient satisfaction were also assessed. METHODS This is a randomized controlled trial including 122 early-stage BC women receiving anthracyclines and/or anti-HER2 antibodies, randomized to CORe (n = 60) or usual care with exercise recommendation (n = 62). Comprehensive assessments were performed at baseline and after cardiotoxic treatment completion. The average duration of the intervention was 5.8 months. RESULTS No cases of CTRCD were identified during the study. LVEF decreased in both groups, but was significantly attenuated in the CORe group [-1.5% (-2.9, -0.1); p = 0.006], with no changes detected in GLS or cardiac biomarkers. The CORe intervention led to significant body mass index (BMI) reduction (p = 0.037), especially in obese patients [3.1 kg/m2 (1.3, 4.8)]. Physical performance and quality-of-life remained stable, while physical activity level increased in both groups. No adverse events were detected. CONCLUSIONS This study suggests that CORe programs are safe and may help attenuate LVEF decline in BC women receiving cardiotoxic therapy and reduce BMI in obese patients.
Collapse
Affiliation(s)
- Estíbaliz Díaz-Balboa
- University of A Coruña, Department of Physiotherapy, Medicine and Biomedical Sciences, Campus de Oza, A Coruña 15071, Spain; Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Carlos Peña-Gil
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Beatriz Rodríguez-Romero
- University of A Coruña. Psychosocial Intervention and Functional Rehabilitation Research Group, Department of Physiotherapy, Medicine and Biomedical Sciences, Campus de Oza, A Coruña, Spain 15071.
| | - Antonio I Cuesta-Vargas
- Instituto de Investigación Biomédica de Málaga y Plataforma en Nanomedicina (IBIMA Plataforma BIONAND), Málaga 29010, Spain; Department of Physiotherapy, University of Málaga, Málaga 29071, Spain.
| | - Oscar Lado-Baleato
- Unit of Biostatistics, Department of Statistics, Mathematical Analysis, and Optimization, Universidade de Santiago de Compostela, Spain.
| | - Amparo Martínez-Monzonís
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain
| | - Milagros Pedreira-Pérez
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Patricia Palacios-Ozores
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain; Medical Oncology Department and Translational Medical Oncology Group, University Clinical Hospital of Santiago (SERGAS); Centro de Investigación Biomédica en Red de Cáncer (CIBERONC); Santiago de Compostela University School of Medicine, 15706 Santiago de Compostela, A Coruña, Spain..
| | - Rafael López-López
- Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain; Medical Oncology Department and Translational Medical Oncology Group, University Clinical Hospital of Santiago (SERGAS); Centro de Investigación Biomédica en Red de Cáncer (CIBERONC); Santiago de Compostela University School of Medicine, 15706 Santiago de Compostela, A Coruña, Spain..
| | - José R González-Juanatey
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| | - Violeta González-Salvado
- Cardiology Department, University Clinical Hospital of Santiago de Compostela (SERGAS); Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV), 15706 Santiago de Compostela, A Coruña, Spain; Health Research Institute of Santiago de Compostela (IDIS), 15706 Santiago de Compostela, A Coruña, Spain.
| |
Collapse
|
118
|
Gupta P, Canonico ME, Faaborg-Andersen C, Prabhu N, Kondapalli L, Quintana RA. Updates in the management of cancer therapy-related hypertension. Curr Opin Cardiol 2024; 39:235-243. [PMID: 38391284 DOI: 10.1097/hco.0000000000001127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/24/2024]
Abstract
PURPOSE OF REVIEW To provide a comprehensive review of hypertension among patients with cancer. Several cancer therapies cause hypertension which has resulted in a growing and vulnerable population of patients with difficult to control hypertension which has significant downstream effects. RECENT FINDINGS Hypertension affects up to 50% of cancer patients and higher comorbidity when compared to the general population. Many anticancer therapies can cause hypertension through their treatment effect. Antihypertensive treatment is crucial given cardiovascular mortality is a leading cause of death among cancer patients. It is already known that hypertension is poorly controlled in the general population, and there are additional challenges in management among patients with cancer. Patients with cancer suffer from multimorbidity, are on multiple medications creating concern for drug interactions, and often have blood pressure lability, which can worsen clinical inertia among patients and their providers. It is crucial to effectively treat hypertension in cancer patients to mitigate downstream adverse cardiovascular events. SUMMARY In recent years, there have been significant changes in management guidelines of hypertension and simultaneously as influx of new cancer therapeutics. We provide an update on hypertension treatment among patients with cancer on different chemotherapeutic agents.
Collapse
Affiliation(s)
- Prerna Gupta
- Department of Medicine, Division of Cardiology, University of Colorado
| | - Mario Enrico Canonico
- Department of Medicine, Division of Cardiology, University of Colorado
- CPC Clinical Research, Aurora, Colorado
| | - Christian Faaborg-Andersen
- Department of Medicine, Massachusetts General Hospital / Harvard Medical School, Boston, Massachusetts, USA
| | - Nicole Prabhu
- Department of Medicine, Division of Cardiology, University of Colorado
| | | | | |
Collapse
|
119
|
Scalia IG, Gheyath B, Tamarappoo BK, Moudgil R, Otton J, Pereyra M, Narayanasamy H, Larsen C, Herrmann J, Arsanjani R, Ayoub C. Chemotherapy Related Cardiotoxicity Evaluation-A Contemporary Review with a Focus on Cardiac Imaging. J Clin Med 2024; 13:3714. [PMID: 38999280 PMCID: PMC11242267 DOI: 10.3390/jcm13133714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/14/2024] Open
Abstract
The long-term survivorship of patients diagnosed with cancer has improved due to accelerated detection and rapidly evolving cancer treatment strategies. As such, the evaluation and management of cancer therapy related complications has become increasingly important, including cardiovascular complications. These have been captured under the umbrella term "cardiotoxicity" and include left ventricular dysfunction and heart failure, acute coronary syndromes, valvular abnormalities, pericardial disease, arrhythmia, myocarditis, and vascular complications. These complications add to the burden of cardiovascular disease (CVD) or are risk factors patients with cancer treatment are presenting with. Of note, both pre- and newly developing CVD is of prognostic significance, not only from a cardiovascular perspective but also overall, potentially impacting the level of cancer therapy that is possible. Currently, there are varying recommendations and practices regarding CVD risk assessment and mitigating strategies throughout the cancer continuum. This article provides an overview on this topic, in particular, the role of cardiac imaging in the care of the patient with cancer. Furthermore, it summarizes the current evidence on the spectrum, prevention, and management of chemotherapy-related adverse cardiac effects.
Collapse
Affiliation(s)
- Isabel G. Scalia
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Bashaer Gheyath
- Department of Imaging, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Balaji K. Tamarappoo
- Division of Cardiology, Banner University Medical Center, The University of Arizona College of Medicine, Phoenix, AZ 85004, USA
| | - Rohit Moudgil
- Department of Cardiology, Heart and Vascular Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - James Otton
- Clinical School, St. Vincent’s Hospital, UNSW, Sydney, NSW 2010, Australia
| | - Milagros Pereyra
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Hema Narayanasamy
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Carolyn Larsen
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Reza Arsanjani
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| | - Chadi Ayoub
- Department of Cardiovascular Medicine, Mayo Clinic, Phoenix, AZ 85054, USA; (I.G.S.)
| |
Collapse
|
120
|
Yan P, Liu Y, Zhang M, Liu N, Zheng Y, Zhang H, Zhou H, Sun M. Reconstitution of peripheral blood T cell receptor β immune repertoire in immune checkpoint inhibitors associated myocarditis. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:35. [PMID: 38863010 PMCID: PMC11165862 DOI: 10.1186/s40959-024-00230-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/15/2024] [Indexed: 06/13/2024]
Abstract
PURPOSE Immune checkpoint inhibitors (ICIs)-associated myocarditis was a rare yet severe complication observed in individuals undergoing immunotherapy. This study investigated the immune status and characteristics of patients diagnosed with ICIs- associated myocarditis. METHODS A total of seven patients diagnosed with ICIs-associated myocarditis were included in the study, while five tumor patients without myocarditis were recruited as reference controls. Additionally, 30 healthy individuals were recruited as blank controls. Biochemical indices, electrocardiogram, and echocardiography measurements were obtained both prior to and following the occurrence of myocarditis. High-throughput sequencing of T cell receptor (TCR) was employed to assess the diversity and distribution characteristics of TCR CDR3 length, as well as the diversity of variable (V) and joining (J) genes of T lymphocytes in peripheral blood. RESULTS In the seven patients with ICIs-associated myocarditis, Troponin T (TNT) levels exhibited a significant increase following myocarditis, while other parameters such as brain natriuretic peptide (BNP), QTc interval, and left ventricular ejection fraction (LVEF) did not show any significant differences. Through sequencing, it was observed that the diversity and uniformity of CDR3 in the ICIs-associated myocarditis patients were significantly diminished. Additionally, the distribution of CDR3 nucleotides deviated from normality, and variations in the utilization of V and J gene segments. CONCLUSION The reconstitution of the TCR immune repertoire may play a pivotal role in the recognition of antigens in patients with ICIs-associated myocarditis.
Collapse
Affiliation(s)
- Peng Yan
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Yanan Liu
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Mingyan Zhang
- Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ning Liu
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Yawen Zheng
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Haiqin Zhang
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China
| | - Hao Zhou
- Graduate School, Shandong First Medical University, Jinan, China
| | - Meili Sun
- Department of Oncology, Jinan Central Hospital, Shandong First Medical University, Jinan, China.
| |
Collapse
|
121
|
Fabiani I, Chianca M, Aimo A, Emdin M, Dent S, Fedele A, Cipolla CM, Cardinale DM. Use of new and emerging cancer drugs: what the cardiologist needs to know. Eur Heart J 2024; 45:1971-1987. [PMID: 38591670 DOI: 10.1093/eurheartj/ehae161] [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: 11/18/2022] [Revised: 02/28/2024] [Accepted: 03/04/2024] [Indexed: 04/10/2024] Open
Abstract
The last decade has witnessed a paradigm shift in cancer therapy, from non-specific cytotoxic chemotherapies to agents targeting specific molecular mechanisms. Nonetheless, cardiovascular toxicity of cancer therapies remains an important concern. This is particularly relevant given the significant improvement in survival of solid and haematological cancers achieved in the last decades. Cardio-oncology is a subspecialty of medicine focusing on the identification and prevention of cancer therapy-related cardiovascular toxicity (CTR-CVT). This review will examine the new definition of CTR-CVT and guiding principles for baseline cardiovascular assessment and risk stratification before cancer therapy, providing take-home messages for non-specialized cardiologists.
Collapse
Affiliation(s)
- Iacopo Fabiani
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
| | - Michela Chianca
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Alberto Aimo
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Michele Emdin
- Cardiology Division, Fondazione Toscana Gabriele Monasterio, Via Giuseppe Moruzzi 1, 56124 Pisa, Italy
- Interdisciplinary Center for Health Science, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Susan Dent
- Duke Cancer Institute, Duke University, Durham, NC, USA
| | - Antonella Fedele
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Carlo Maria Cipolla
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Daniela Maria Cardinale
- Cardioncology Unit, Cardioncology and Second Opinion Division, European Institute of Oncology, Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| |
Collapse
|
122
|
Ogilvie LM, Coyle-Asbil B, Brunt KR, Petrik J, Simpson JA. Therapy-naïve malignancy causes cardiovascular disease: a state-of-the-art cardio-oncology perspective. Am J Physiol Heart Circ Physiol 2024; 326:H1515-H1537. [PMID: 38639740 DOI: 10.1152/ajpheart.00795.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 04/15/2024] [Accepted: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Cardiovascular disease (CVD) and cancer are the leading causes of mortality worldwide. Although generally thought of as distinct diseases, the intersectional overlap between CVD and cancer is increasingly evident in both causal and mechanistic relationships. The field of cardio-oncology is largely focused on the cardiotoxic effects of cancer therapies (e.g., chemotherapy, radiation). Furthermore, the cumulative effects of cardiotoxic therapy exposure and the prevalence of CVD risk factors in patients with cancer lead to long-term morbidity and poor quality of life in this patient population, even when patients are cancer-free. Evidence from patients with cancer and animal models demonstrates that the presence of malignancy itself, independent of cardiotoxic therapy exposure or CVD risk factors, negatively impacts cardiac structure and function. As such, the primary focus of this review is the cardiac pathophysiological and molecular features of therapy-naïve cancer. We also summarize the strengths and limitations of preclinical cancer models for cardio-oncology research and discuss therapeutic strategies that have been tested experimentally for the treatment of cancer-induced cardiac atrophy and dysfunction. Finally, we explore an adjacent area of interest, called "reverse cardio-oncology," where the sequelae of heart failure augment cancer progression. Here, we emphasize the cross-disease communication between malignancy and the injured heart and discuss the importance of chronic low-grade inflammation and endocrine factors in the progression of both diseases.
Collapse
Affiliation(s)
- Leslie M Ogilvie
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Bridget Coyle-Asbil
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Keith R Brunt
- Department of Pharmacology, Dalhousie Medicine New Brunswick, Saint John, New Brunswick, Canada
- IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, Ontario, Canada
| | - Jeremy A Simpson
- Department of Human Health and Nutritional Sciences, University of Guelph, Guelph, Ontario, Canada
- IMPART Investigator Team Canada, Saint John, New Brunswick, Canada
| |
Collapse
|
123
|
Palaskas NL, Ali HJ, Koutroumpakis E, Ganatra S, Deswal A. Cardiovascular toxicity of immune therapies for cancer. BMJ 2024; 385:e075859. [PMID: 38749554 DOI: 10.1136/bmj-2023-075859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
In addition to conventional chemoradiation and targeted cancer therapy, the use of immune based therapies, specifically immune checkpoint inhibitors (ICIs) and chimeric antigen receptor T cell therapy (CAR-T), has increased exponentially across a wide spectrum of cancers. This has been paralleled by recognition of off-target immune related adverse events that can affect almost any organ system including the cardiovascular system. The use of ICIs has been associated with myocarditis, a less common but highly fatal adverse effect, pericarditis and pericardial effusions, vasculitis, thromboembolism, and potentially accelerated atherosclerosis. CAR-T resulting in a systemic cytokine release syndrome has been associated with myriad cardiovascular consequences including arrhythmias, myocardial infarction, and heart failure. This review summarizes the current state of knowledge regarding adverse cardiovascular effects associated with ICIs and CAR-T.
Collapse
Affiliation(s)
| | - Hyeon-Ju Ali
- University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Sarju Ganatra
- Lahey Hospital and Medical Center, Burlington, MA 01805
| | - Anita Deswal
- University of Texas MD Anderson Cancer Center, Houston, TX, USA 01805
| |
Collapse
|
124
|
Kong Y, Wei X, Zhang D, Lin H, Peng M, Shang H. Prevention and treatment of anthracycline-induced cardiotoxicity: A bibliometric analysis of the years 2000-2023. Heliyon 2024; 10:e29926. [PMID: 38698971 PMCID: PMC11064157 DOI: 10.1016/j.heliyon.2024.e29926] [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: 07/27/2023] [Revised: 03/20/2024] [Accepted: 04/17/2024] [Indexed: 05/05/2024] Open
Abstract
Aims This study aimed to evaluate the global research trend in the prevention and treatment of cardiotoxicity caused by anthracyclines from 2000 to 2023, and to explore international cooperation, research hotspots, and frontier trends. Methods The articles on the prevention and treatment of anthracycline-induced cardiotoxicity published from 2000 to 2023 were searched by Web of Science. The bibliometrics software CiteSpace was used for visual analysis of countries, institutions, journals, authors, cited authors, cited references, and keywords. Results This study analyzed the current status of global research on the prevention and treatment of cardiotoxicity caused by anthracyclines. A total of 3,669 papers were searched and 851 studies were included. The number of publications increased gradually throughout the years. Cardiovascular Toxicology (15) is the journal with the most publications. Circulation (547) ranked first among cited journals. In this field, the country with the most publications is the United States (229), and the institution with the most publications is Charles Univ Prague (18). In the analysis of the authors, Tomas S (10) ranked first. Cardinale D (262) ranked first among cited authors. In the ranking of cited literature frequency, the article ranked first is "Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy" (121). The keywords "heart failure" (215) and "oxidative stress" (212) were the most frequent. "Enalapril", "inflammation", "cell death", "NF-κB" and "Nrf2" were the advanced research contents in 2019-2023. Conclusions This study provided valuable information for cardio-oncology researchers to identify potential collaborators and institutions, discover hot topics, and explore new research directions. The prevention and treatment of anthracycline-induced cardiotoxicity focuses on early detection and timely treatment. The results of the current clinical studies on the treatment of anthracycline-induced cardiotoxicity are contradictory, and more studies are needed to provide more reliable clinical evidence in the future.
Collapse
Affiliation(s)
- Yifan Kong
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohong Wei
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Di Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyuan Lin
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | | | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education and Beijing, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
- Weifang Medical University, Weifang, China
| |
Collapse
|
125
|
Cronin M, Lowery A, Kerin M, Wijns W, Soliman O. Risk Prediction, Diagnosis and Management of a Breast Cancer Patient with Treatment-Related Cardiovascular Toxicity: An Essential Overview. Cancers (Basel) 2024; 16:1845. [PMID: 38791923 PMCID: PMC11120055 DOI: 10.3390/cancers16101845] [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: 04/12/2024] [Revised: 05/02/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024] Open
Abstract
Breast cancer is amongst the most common invasive cancers in adults. There are established relationships between anti-cancer treatments for breast cancer and cardiovascular side effects. In recent years, novel anti-cancer treatments have been established, as well as the availability of multi-modal cardiac imaging and the sophistication of treatment for cardiac disease. This review provides an in-depth overview regarding the interface of breast cancer and cancer therapy-related cardiovascular toxicity. Specifically, it reviews the pathophysiology of breast cancer, the method of action in therapy-related cardiovascular toxicity from anti-cancer treatment, the use of echocardiography, cardiac CT, MRI, or nuclear medicine as diagnostics, and the current evidence-based treatments available. It is intended to be an all-encompassing review for clinicians caring for patients in this situation.
Collapse
Affiliation(s)
- Michael Cronin
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
| | - Aoife Lowery
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Michael Kerin
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| | - William Wijns
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
| | - Osama Soliman
- School of Medicine, University of Galway, H91 V4AY Galway, Ireland
- Precision Cardio-Oncology Research Enterprise (P-CORE), H91 TK33 Galway, Ireland
- CURAM Centre for Medical Devices, H91 TK33 Galway, Ireland
- Discipline of Surgery, Lambe Institute for Translational Research, University of Galway, H91 V4AY Galway, Ireland
| |
Collapse
|
126
|
Kuang Z, Kong M, Yan N, Ma X, Wu M, Li J. Precision Cardio-oncology: Update on Omics-Based Diagnostic Methods. Curr Treat Options Oncol 2024; 25:679-701. [PMID: 38676836 PMCID: PMC11082000 DOI: 10.1007/s11864-024-01203-6] [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] [Accepted: 04/02/2024] [Indexed: 04/29/2024]
Abstract
OPINION STATEMENT Cardio-oncology is an emerging interdisciplinary field dedicated to the early detection and treatment of adverse cardiovascular events associated with anticancer treatment, and current clinical management of anticancer-treatment-related cardiovascular toxicity (CTR-CVT) remains limited by a lack of detailed phenotypic data. However, the promise of diagnosing CTR-CVT using deep phenotyping has emerged with the development of precision medicine, particularly the use of omics-based methodologies to discover sensitive biomarkers of the disease. In the future, combining information produced by a variety of omics methodologies could expand the clinical practice of cardio-oncology. In this review, we demonstrate how omics approaches can improve our comprehension of CTR-CVT deep phenotyping, discuss the positive and negative aspects of available omics approaches for CTR-CVT diagnosis, and outline how to integrate multiple sets of omics data into individualized monitoring and treatment. This will offer a reliable technical route for lowering cardiovascular morbidity and mortality in cancer patients and survivors.
Collapse
Affiliation(s)
- Ziyu Kuang
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Miao Kong
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ningzhe Yan
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Graduate School, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyi Ma
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Min Wu
- Cardiovascular Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Jie Li
- Oncology Department, Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
127
|
Geenty PD, Gregory AT, Nolan M, Denniss AR, Pepe S, Sverdlov AL, Thomas L. Cardio-Oncology-Beyond Anthracyclines and Ejection Fraction. Heart Lung Circ 2024; 33:547-552. [PMID: 38823894 DOI: 10.1016/j.hlc.2024.05.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Affiliation(s)
- Paul D Geenty
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Westmead Clinical School, The University of Sydney, NSW, Australia
| | | | - Mark Nolan
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia; Peter MacCallum Cancer Centre, Melbourne, Vic, Australia
| | - A Robert Denniss
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Heart, Lung and Circulation, Sydney, NSW, Australia; The University of Sydney, Sydney, NSW, Australia; Department of Cardiology, Blacktown Hospital, and Western Sydney University, Sydney, NSW, Australia
| | - Salvatore Pepe
- Heart, Lung and Circulation, Sydney, NSW, Australia; Heart Research Group, Murdoch Children's Research Institute, Royal Children's Hospital, Melbourne; Department of Paediatrics, The University of Melbourne, Melbourne, Vic, Australia
| | - Aaron L Sverdlov
- Newcastle Centre of Excellence in Cardio-Oncology, The University of Newcastle; Hunter Medical Research Institute; Calvary Mater Newcastle, Hunter New England Health, Newcastle, NSW, Australia; Cardiovascular Department, John Hunter Hospital, Newcastle, NSW, Australia.
| | - Liza Thomas
- Department of Cardiology, Westmead Hospital, Sydney, NSW, Australia; Westmead Clinical School, The University of Sydney, NSW, Australia; Southwest Clinical School, University of New South Wales, NSW, Australia.
| |
Collapse
|
128
|
Zheng Y, Chen Z, Song W, Xu Y, Zhao Z, Sun Y, Wang Y, Geng X, Zhao J, Zhang X, Xu Y, Chan JSK, Tse G, Li G, Hong L, Liu T. Cardiovascular adverse events associated with immune checkpoint inhibitors: A retrospective multicenter cohort study. Cancer Med 2024; 13:e7233. [PMID: 38752474 PMCID: PMC11097245 DOI: 10.1002/cam4.7233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 04/16/2024] [Accepted: 04/20/2024] [Indexed: 05/19/2024] Open
Abstract
BACKGROUND Over the past decade, immune checkpoint inhibitors (ICIs) have significantly transformed cancer treatment. However, ICIs inevitably may cause a spectrum of immune-related adverse events, among which cardiovascular toxicity, particularly myocarditis, while infrequent, has garnered increasing attention due to its high fatality rate. METHODS We conducted a multicenter retrospective study to characterize ICI-associated cardiovascular adverse events. Logistic regression was performed to explore the risk factors for the development of myocarditis and severe myocarditis. Receiver operating characteristic curves were conducted to assess the diagnostic abilities of cardiac biomarkers to distinguish different cardiovascular toxicities, and the performance and calibration were evaluated using Hosmer-Lemeshow test. RESULTS Forty-four patients were identified, including thirty-five myocarditis, five heart failure, three arrhythmias, and one myocardial infarction. Compared with other patients, myocarditis patients had higher cardiac troponin-I (cTnI) levels (p < 0.001), higher creatine kinase levels (p = 0.003), higher creatine kinase isoenzyme-MB (CK-MB) levels (p = 0.013), and shorter time to the incidence of adverse cardiovascular events (p = 0.022) after ICI treatment. Twenty-one patients (60%) were classified as severe myocarditis, and they presented higher cardiac troponin I (cTnI) levels (p = 0.013), higher N-terminal pro-B-type natriuretic peptide levels (p = 0.031), higher creatine kinase levels (p = 0.018), higher CK-MB levels (p = 0.026), and higher neutrophil to lymphocyte ratio (NLR) levels (p = 0.016) compared to non-severe myocarditis patients after ICI treatment. Multivariate logistic regression showed that CK-MB (adjusted odds ratio [OR]: 1.775, 95% confidence interval [CI]: 1.055-2.984, p = 0.031) was the independent risk factor of the development of ICI-associated myocarditis, and cTnI (adjusted OR: 1.021, 95% CI: 1.002-1.039, p = 0.03) and NLR (adjusted OR: 1.890, 95% CI: 1.026-3.483, p = 0.041) were the independent risk factors of ICI-associated severe myocarditis. The receiver operating characteristic curve showed an area under curve of 0.785 (95% CI: 0.642 to 0.928, p = 0.013) for CK-MB, 0.765 (95% CI: 0.601 to 0.929, p = 0.013) for cTnI, and 0.773 for NLR (95% CI: 0.597 to 0.948, p = 0.016). CONCLUSIONS Elevated CK-MB after ICI treatment is the independent risk factor for the incidence of ICI-associated myocarditis, and elevated cTnI and NLR after ICI treatment are the independent risk factors for the development of ICI-associated severe myocarditis. CK-MB, cTnI, and NLR demonstrated a promising predictive utility for the identification of ICI-associated myocarditis and severe myocarditis.
Collapse
Affiliation(s)
- Yi Zheng
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Ziliang Chen
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Wenhua Song
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yu Xu
- Department of OncologyTianjin Huanghe HospitalTianjinChina
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yihong Sun
- Department of CardiologyChina‐Japan Friendship HospitalBeijingChina
| | - Yuanyuan Wang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Xuhong Geng
- Department of FunctionFourth Hospital of Hebei Medical UniversityShijiazhuangHebeiChina
| | - Jun Zhao
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Xiaowei Zhang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Yanmin Xu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | | | - Gary Tse
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
- School of Nursing and Health StudiesHong Kong Metropolitan UniversityHong KongChina
- Cardiac Electrophysiology Unit, Cardiovascular Analytics GroupPowerHealth LimitedHong KongChina
| | - Guangping Li
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| | - Lili Hong
- Department of OncologyTianjin Huanghe HospitalTianjinChina
| | - Tong Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of CardiologyTianjin Institute of Cardiology, Second Hospital of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
129
|
Heemelaar JC, Louisa M, Neilan TG. Treatment of Immune Checkpoint Inhibitor-associated Myocarditis. J Cardiovasc Pharmacol 2024; 83:384-391. [PMID: 37506676 PMCID: PMC10830893 DOI: 10.1097/fjc.0000000000001456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 07/08/2023] [Indexed: 07/30/2023]
Abstract
ABSTRACT Immune checkpoint inhibitors (ICIs) are a form immunotherapy where the negative regulators of host immunity are targeted, thereby leveraging the own immune system. ICIs have significantly improved cancer survival in several advanced malignancies, and there are currently more than 90 different cancer indications for ICIs. Most patients develop immune-related adverse events during ICI therapy. Most are mild, but a small subset of patients will develop severe and potentially fatal immune-related adverse events. A serious cardiovascular complication of ICI therapy is myocarditis. Although the incidence of myocarditis is low, mortality rates of up to 50% have been reported. The mainstay of ICI-associated myocarditis treatment is high-dose corticosteroids. Unfortunately, half of patients with myocarditis do not show clinical improvement after corticosteroid treatment. Also, high doses of corticosteroids may adversely impact cancer outcomes. There is an evidence gap in the optimal second-line treatment strategy. Currently, there is a paradigm shift in second-line treatment taking place from empirical corticosteroid-only strategies to either intensified initial immunosuppression where corticosteroids are combined with another immunosuppressant or targeted therapies directed at the pathophysiology of ICI myocarditis. However, the available evidence to support these novel strategies is limited to observational studies and case reports. The aim of this review is to summarize the literature, guidelines, and future directions on the pharmacological treatment of ICI myocarditis.
Collapse
Affiliation(s)
- Julius C Heemelaar
- Cardiovascular Imaging Research Center (CIRC), Department of Cardiology and Radiology, Massachusetts General Hospital, Boston, MA; and
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Maria Louisa
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Tomas G Neilan
- Cardiovascular Imaging Research Center (CIRC), Department of Cardiology and Radiology, Massachusetts General Hospital, Boston, MA; and
| |
Collapse
|
130
|
Nolan MT, Pathan F, Nott L, Black A, Pointon O, Marwick TH. Comparison of Echocardiography and Multi-Planar Gated Acquisition Scans for Predicting Cancer-Treatment-Related Cardiovascular Dysfunction. Heart Lung Circ 2024; 33:693-703. [PMID: 38692983 DOI: 10.1016/j.hlc.2024.03.010] [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: 12/27/2023] [Revised: 03/07/2024] [Accepted: 03/14/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Current guidelines recommend using sequential cardiac imaging to monitor for cancer treatment-related cardiac dysfunction (CTRCD) in patients undergoing potentially cardiotoxic chemotherapy. Multiple different imaging cardiac modalities are available and there are few prospective head-to-head comparative studies to help guide treatment. OBJECTIVES To perform an exploratory prospective cohort study of "real-world" CTRCD comparing multigated acquisition nuclear ventriculography (MUGA) at the referring cancer specialist's discretion with a novel echocardiographic strategy at an Australian tertiary hospital. METHOD Patients were recruited from haematology and oncology outpatient clinics if they were scheduled for treatment with anthracyclines and/or trastuzumab. Patients underwent simultaneous MUGA-based cardiac imaging (conventional strategy) at a frequency according to evidenced-based guidelines in addition to researcher-conducted echocardiographic imaging. The echocardiographic imaging was performed in all patients at time points recommended by international society guidelines. Outcomes included adherence to guideline recommendations, concordance between MUGA and echocardiographic left ventricular ejection fraction (LVEF) measurements, and detection of cardiac dysfunction (defined as >5% LVEF decrement from baseline by three-dimensional [3D]-LVEF). A secondary end point was accuracy of global longitudinal strain in predicting cardiac dysfunction. RESULTS In total, 35 patients were recruited, including 15 with breast cancer, 19 with haematological malignancy, and one with gastric cancer. MUGA and echocardiographic LVEF measurements correlated poorly with limits of agreement of 30% between 3D-LVEF and MUGA-LVEF and 37% for 3D-LVEF and MUGA-LVEF. Only one case (2.9%) of CTRCD was diagnosed by MUGA, compared with 12 (34.2%) cases by echocardiography. Four (4) patients had >10% decrement in 3D-LVEF that was not detected by MUGA. Global longitudinal strain at 2 months displayed significant ability to predict CTRCD (area under the curve, 0.75, 95% confidence interval, 0.55-0.94). CONCLUSIONS The MUGA correlates poorly with echocardiographic assessment with substantial discrepancy between MUGA and echocardiography in CTRCD diagnosis. Echocardiographic and MUGA imaging strategies should not be considered equivalent for imaging cancer patients, and a single imaging modality should ideally be used per patient to prevent misdiagnosis by inter-modality variation These findings should be considered hypothesis-generating and require confirmation with larger studies.
Collapse
Affiliation(s)
- Mark T Nolan
- Baker Heart and Diabetes Institute, Melbourne, Vic, Australia.
| | - Faraz Pathan
- Department of Medicine, The University of Sydney Nepean Clinical School, Kingswood, NSW, Australia
| | - Louise Nott
- Department of Medical Oncology, Royal Hobart Hospital, Hobart, Tas, Australia
| | - Allison Black
- Department of Medical Oncology, Royal Hobart Hospital, Hobart, Tas, Australia
| | - Owen Pointon
- Department of Nuclear Medicine, Royal Hobart Hospital, Hobart, Tas, Australia
| | - Thomas H Marwick
- Department of Cardiovascular Imaging, Baker Heart and Diabetes Institute, Melbourne, Vic, Australia
| |
Collapse
|
131
|
Tan S, Kader Z, Day D, Chen D, Nicholls SJ, Ramkumar S. Cardiotoxicity in Oncology Guidelines: Discrepancies Do Matter. Heart Lung Circ 2024; 33:553-557. [PMID: 38453605 DOI: 10.1016/j.hlc.2024.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 01/29/2024] [Accepted: 02/06/2024] [Indexed: 03/09/2024]
Affiliation(s)
- Sean Tan
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia; Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia.
| | - Zainel Kader
- Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia
| | - Daphne Day
- School of Clinical Sciences at Monash Health, Monash University, Melbourne, Vic, Australia; Department of Oncology, Monash Health, Melbourne, Vic, Australia
| | - Daniel Chen
- Prince of Wales and St George Hospitals, South Eastern Sydney Local Health District, Sydney, NSW, Australia; Hatter Cardiovascular Institute, University College of London, London, UK
| | - Stephen J Nicholls
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia; Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia
| | - Satish Ramkumar
- Victorian Heart Institute, Monash University, Melbourne, Vic, Australia; Monash Heart, Victorian Heart Hospital, Melbourne, Vic, Australia
| |
Collapse
|
132
|
Lammers EMJ, Nijdam A, Zijlstra JM, Janus CPM, de Weijer RJ, Appelman Y, Manintveld OC, Teske AJ, van Leeuwen FE, Aleman BMP. Cardiovascular screening outcomes in the Dutch survivorship care program for Hodgkin lymphoma survivors. J Cancer Surviv 2024:10.1007/s11764-024-01561-y. [PMID: 38649650 DOI: 10.1007/s11764-024-01561-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Accepted: 03/02/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE Hodgkin lymphoma (HL) survivors are at increased risk of cardiovascular disease (CVD) due to former lymphoma treatment. In 2013, cardiovascular screening for 5-year HL survivors according to national guidelines was implemented in Dutch survivorship clinics. We aim to assess the following: (1) adherence to screening guidelines and (2) the yield of (risk factors for) CVD in the screening program. METHODS The study population consisted of 5-year HL survivors who received survivorship care at three University Medical Centers from 2013 to 2016 through 2021. Patient characteristics, cardiovascular screening procedures, and outcomes were collected from the medical records. RESULTS In 186 survivors eligible for cardiovascular screening (mean age 47.8 years, 60.8% female), the following diagnostics were performed: complete blood tests (81.0%, median frequency: yearly instead of advised 5-yearly evaluation), electrocardiogram (93.0%), echocardiography (94.6%). Fifty-five percent of survivors had at least one modifiable cardiovascular risk factor (i.e., current smoking, overweight, new/insufficiently controlled hypertension, dyslipidemia, or diabetes). Screening detected ≥ 1 CVD in 31.1% of survivors. Among survivors with available echocardiography report (n = 106), screening detected new aortic and/or mitral valve dysfunction(s) in 51.0% (with grades 3-4 in 4.9%) and impaired left ventricular ejection fraction in 10.3%. CONCLUSIONS Adherence to the screening guidelines in the Dutch HL survivorship care program was reasonable to good and a substantial number of actionable (risk factors for) CVD were diagnosed. IMPLICATIONS FOR CANCER SURVIVORS Our findings inform HL survivors at high risk of late cardiotoxicity about cardiovascular screening findings and demonstrate appropriate therapeutic actions after diagnosis of (risk factors for) CVD.
Collapse
Affiliation(s)
- Eline M J Lammers
- Department of Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Annelies Nijdam
- Department of Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Josée M Zijlstra
- Department of Hematology, Amsterdam UMC Location Vrije Universiteit, Cancer Centre Amsterdam, Amsterdam, The Netherlands
| | - Cécile P M Janus
- Department of Radiation Oncology, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Roel J de Weijer
- Department of Hematology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Yolande Appelman
- Department of Cardiology, Amsterdam UMC, Location Vrije Universiteit, Amsterdam Cardiovascular Sciences, Amsterdam, The Netherlands
| | | | - Arco J Teske
- Department of Cardiology, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Flora E van Leeuwen
- Department of Epidemiology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Berthe M P Aleman
- Department of Radiation Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
133
|
Peng L, Yang D, Weng C. Sintilimab and Cardiovascular Toxicity. JAMA 2024; 331:1333-1334. [PMID: 38512227 DOI: 10.1001/jama.2024.0673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2024]
Affiliation(s)
- Linfeng Peng
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Dandan Yang
- Department of Cardiology, Peking University People's Hospital, Beijing, China
| | - Cuilian Weng
- Department of Intensive Care Unit, Shengli Clinical Medical College of Fujian Medical University, Fuzhou, China
| |
Collapse
|
134
|
Glen C, Morrow A, Roditi G, Hopkins T, Macpherson I, Stewart P, Petrie MC, Berry C, Epstein F, Lang NN, Mangion K. Cardiovascular sequelae of trastuzumab and anthracycline in long-term survivors of breast cancer. Heart 2024; 110:650-656. [PMID: 38103912 DOI: 10.1136/heartjnl-2023-323437] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
OBJECTIVES Long-term follow-up of patients treated with trastuzumab largely focuses on those with reduced left ventricular ejection fraction (LVEF) on treatment completion. This study sought to evaluate the prevalence of cardiovascular risk factors, overt cardiovascular disease and cardiac imaging abnormalities using cardiac magnetic resonance (CMR), in participants with normal LVEF on completion of trastuzumab±anthracycline therapy at least 5 years previously. METHODS Participants with human epidermal growth factor receptor 2-positive breast cancer treated with trastuzumab±anthracycline ≥5 years previously were identified from a clinical database. All participants had normal LVEF prior to, and on completion of, treatment. Participants underwent clinical cardiovascular evaluation, ECG, cardiac biomarker evaluation and CMR. Left ventricular systolic dysfunction (LVSD) was defined as LVEF <50%. RESULTS Forty participants were recruited between 15 March 2021 and 19 July 2022. Median time since completion of trastuzumab was 7.8 years (range 5.9-10.8 years) and 90% received prior anthracycline. 25% of participants had LVSD; median LVEF was 55.2% (Q1-Q3, 51.3-61.2). 30% of participants had N-terminal pro-B-type natriuretic peptide >125 pg/mL and 8% had high-sensitivity cardiac troponin T >14 ng/L. 33% of participants had a new finding of hypertension. 58% had total cholesterol >5.0 mmol/L, 43% had triglycerides >1.7 mmol/L and 5% had a new diagnosis of diabetes. CONCLUSIONS The presence of asymptomatic LVSD, abnormal cardiac biomarkers and cardiac risk factors in participants treated with trastuzumab and anthracycline at least 5 years previously is common, even in those with normal LVEF on completion of treatment. Our findings reinforce the relevance of comprehensive evaluation of cardiovascular risk factors following completion of cancer therapy, in addition to LVEF assessment.
Collapse
Affiliation(s)
- Claire Glen
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Andrew Morrow
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Giles Roditi
- Clinical Research Imaging Facility, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Tracey Hopkins
- Clinical Research Imaging Facility, NHS Greater Glasgow and Clyde, Glasgow, UK
| | - Iain Macpherson
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Philip Stewart
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Mark C Petrie
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Regional Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, UK
| | - Colin Berry
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Cardiology, Golden Jubilee National Hospital, Clydebank, UK
| | - Fred Epstein
- School of Engineering and Applied Science, University of Virginia, Charlottesville, Virginia, USA
| | - Ninian N Lang
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
| | - Kenneth Mangion
- School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, UK
- Regional Heart and Lung Centre, Golden Jubilee National Hospital, Clydebank, UK
| |
Collapse
|
135
|
López-Fernández T, Farmakis D, Ameri P, Asteggiano R, de Azambuja E, Aznar M, Barac A, Bayes-Genis A, Bax JJ, Bergler-Klein J, Boriani G, Celutkiene J, Coats A, Cohen-Solal A, Córdoba R, Cosyns B, Filippatos G, Fox K, Gulati G, Inciardi RM, Lee G, Mamas MA, Novo G, Plummer C, Psyrri A, Rakisheva A, Suter T, Tini G, Tocchetti CG, Toutouzas K, Wilhelm M, Metra M, Lyon AR, Rosano GMC. European Society of Cardiology Core Curriculum for cardio-oncology. Eur J Heart Fail 2024; 26:754-771. [PMID: 38059343 DOI: 10.1002/ejhf.3102] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/06/2023] [Accepted: 12/01/2023] [Indexed: 12/08/2023] Open
Abstract
Cardio-oncology is a rapidly growing field of cardiovascular (CV) medicine that has resulted from the continuously increasing clinical demand for specialized CV evaluation, prevention and management of patients suffering or surviving from malignant diseases. Dealing with CV disease in patients with cancer requires special knowledge beyond that included in the general core curriculum for cardiology. Therefore, the European Society of Cardiology (ESC) has developed a special core curriculum for cardio-oncology, a consensus document that defines the level of experience and knowledge required for cardiologists in this particular field. It is structured into 8 chapters, including (i) principles of cancer biology and therapy; (ii) forms and definitions of cancer therapy-related cardiovascular toxicity (CTR-CVT); (iii) risk stratification, prevention and monitoring protocols for CTR-CVT; (iv) diagnosis and management of CV disease in patients with cancer; (v) long-term survivorship programmes and cardio-oncology rehabilitation; (vi) multidisciplinary team management of special populations; (vii) organization of cardio-oncology services; (viii) research in cardio-oncology. The core curriculum aims at promoting standardization and harmonization of training and evaluation in cardio-oncology, while it further provides the ground for an ESC certification programme designed to recognize the competencies of certified specialists.
Collapse
Affiliation(s)
- Teresa López-Fernández
- Cardiology Department, La Paz University Hospital, IdiPAZ Research Institute, Madrid, Spain
- Cardiology Department, Hospital Universitario Quirónsalud Madrid, Madrid, Spain
| | - Dimitrios Farmakis
- Department of Cardiology, Athens University Hospital Attikon, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Pietro Ameri
- IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Riccardo Asteggiano
- Faculty of Medicine, Insubria University, Varese, Italy
- LARC (Laboratorio Analisi e Ricerca Clinica), Turin, Italy
| | - Evandro de Azambuja
- Institut Jules Bordet and l'Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Marianne Aznar
- Division of Cancer Sciences, Faculty of Biology, Medicine and Health, The University of Manchester; The Christie NHS Foundation Trust, Manchester, UK
| | - Ana Barac
- Inova Schar Heart and Vascular and Inova Schar Cancer Institute, Falls Church, VA, USA
| | - Antoni Bayes-Genis
- Heart Institute, Hospital Universitari Germans Trias i Pujol, Badalona, CIBERCV, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Jeroen J Bax
- Department of Cardiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Jutta Bergler-Klein
- Department of Cardiology, University Clinic of Internal Medicine II, Medical University of Vienna, Vienna, Austria
| | - Giuseppe Boriani
- Cardiology Division, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Jelena Celutkiene
- Clinic of Cardiac and Vascular Diseases, Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, Vilnius, Lithuania
| | | | - Alain Cohen-Solal
- Department of Cardiology, Lariboisière Hospital, Paris Cite University, INSERM U-942, Paris, France
| | - Raúl Córdoba
- Fundacion Jimenez Diaz University Hospital, Health Research Institute, Instituto de Investigación Sanitaria-Fundación Jiménez-Díaz (IIS-FJD), Madrid, Spain
| | - Bernard Cosyns
- Cardiology, CHVZ (Centrum voor Hart en Vaatziekten), ICMI (In Vivo Cellular and Molecular Imaging) Laboratory, Universitair ziekenhuis Brussel, Brussels, Belgium
| | - Gerasimos Filippatos
- Department of Cardiology, Athens University Hospital Attikon, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Kevin Fox
- Imperial College Healthcare NHS Trust; NHS Orkney, Orkney, UK
| | - Geeta Gulati
- Department of Cardiology, Division of Medicine, Oslo University Hospital, Ullevål, Norway
- K.G. Jebsen Center for Cardiac Biomarkers, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Riccardo M Inciardi
- ASST Spedali Civili di Brescia and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Geraldine Lee
- Florence Nightingale Faculty of Nursing Midwifery & Palliative Care, King's College London, London, UK
| | - Mamas A Mamas
- Keele Cardiovascular Research Group, Keele University, UK
| | - Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (ProMISE), University of Palermo, Palermo, Italy
- Cardiology Unit, University Hospital Paolo Giaccone, Palermo, Italy
| | - Chris Plummer
- Department of Cardiology, Newcastle Upon Tyne Hospitals NHS Foundation Trust, Newcastle Upon Tyne, UK
| | - Amanda Psyrri
- Department of Internal Medicine, Section of Medical Oncology, Attikon University Hospital, Athens, Greece
| | - Amina Rakisheva
- City Cardiolocal Center, Almaty, Kazakhstan
- Qonaev city hospital, Almaty, Kazakhstan
| | - Thomas Suter
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Giacomo Tini
- Cardiology, Department of Clinical and Molecular Medicine, Sapienza University of Rome, Azienda Ospedaliera Universitaria Sant'Andrea, Rome, Italy
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences (DISMET), Center for Basic and Clinical Immunology Research (CISI), Interdepartmental Center for Clinical and Translational Research (CIRCET), Interdepartmental Hypertension Research Center (CIRIAPA), Federico II University, Naples, Italy
| | | | - Matthias Wilhelm
- Rehabilitation & Sports Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Marco Metra
- ASST Spedali Civili di Brescia and Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guys and St. Thomas NHS Foundation Trust, London, UK
| | | |
Collapse
|
136
|
Zheng Y, Liu Y, Chen Z, Zhang Y, Qi Z, Wu N, Zhao Z, Tse G, Wang Y, Hu H, Niu Y, Liu T. Cardiovascular disease burden in patients with urological cancers: The new discipline of uro-cardio-oncology. CANCER INNOVATION 2024; 3:e108. [PMID: 38946935 PMCID: PMC11212304 DOI: 10.1002/cai2.108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 11/07/2023] [Accepted: 11/28/2023] [Indexed: 07/02/2024]
Abstract
Cancer remains a major cause of mortality worldwide, and urological cancers are the most common cancers among men. Several therapeutic agents have been used to treat urological cancer, leading to improved survival for patients. However, this has been accompanied by an increase in the frequency of survivors with cardiovascular complications caused by anticancer medications. Here, we propose the novel discipline of uro-cardio-oncology, an evolving subspecialty focused on the complex interactions between cardiovascular disease and urological cancer. In this comprehensive review, we discuss the various cardiovascular toxicities induced by different classes of antineoplastic agents used to treat urological cancers, including androgen deprivation therapy, vascular endothelial growth factor receptor tyrosine kinase inhibitors, immune checkpoint inhibitors, and chemotherapeutics. In addition, we discuss possible mechanisms underlying the cardiovascular toxicity associated with anticancer therapy and outline strategies for the surveillance, diagnosis, and effective management of cardiovascular complications. Finally, we provide an analysis of future perspectives in this emerging specialty, identifying areas in need of further research.
Collapse
Affiliation(s)
- Yi Zheng
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Ying Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Ziliang Chen
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Yunpeng Zhang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Zuo Qi
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Ning Wu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Zhiqiang Zhao
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Gary Tse
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
- School of Nursing and Health StudiesHong Kong Metropolitan UniversityHong KongChina
| | - Yong Wang
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Hailong Hu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Yuanjie Niu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| | - Tong Liu
- Tianjin Key Laboratory of Ionic‐Molecular Function of Cardiovascular Disease, Department of Cardiology, Tianjin Institute of CardiologySecond Hospital of Tianjin Medical UniversityTianjinChina
| |
Collapse
|
137
|
Scott JM, Lee J, Michalski MG, Batch K, Simpson AL, Peoples J, Lee CP, Harrison JN, Yu AF, Sasso JP, Dang C, Moskowitz CS, Jones LW, Eves ND. Mechanisms of Exercise Intolerance Across the Breast Cancer Continuum: A Pooled Analysis of Individual Patient Data. Med Sci Sports Exerc 2024; 56:590-599. [PMID: 38485730 PMCID: PMC10948020 DOI: 10.1249/mss.0000000000003348] [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] [Indexed: 03/19/2024]
Abstract
PURPOSE The purpose of this study is to evaluate the prevalence of abnormal cardiopulmonary responses to exercise and pathophysiological mechanism(s) underpinning exercise intolerance across the continuum of breast cancer (BC) care from diagnosis to metastatic disease. METHODS Individual participant data from four randomized trials spanning the BC continuum ([1] prechemotherapy [n = 146], [2] immediately postchemotherapy [n = 48], [3] survivorship [n = 138], and [4] metastatic [n = 47]) were pooled and compared with women at high-risk of BC (BC risk; n = 64). Identical treadmill-based peak cardiopulmonary exercise testing protocols evaluated exercise intolerance (peak oxygen consumption; V̇O2peak) and other resting, submaximal, and peak cardiopulmonary responses. The prevalence of 12 abnormal exercise responses was evaluated. Graphical plots of exercise responses were used to identify oxygen delivery and/or uptake mechanisms contributing to exercise intolerance. Unsupervised, hierarchical cluster analysis was conducted to explore exercise response phenogroups. RESULTS Mean V̇O2peak was 2.78 ml O2.kg-1·min-1 (95% confidence interval [CI], -3.94, -1.62 mL O2.kg-1·min-1; P < 0.001) lower in the pooled BC cohort (52 ± 11 yr) than BC risk (55 ± 10 yr). Compared with BC risk, the pooled BC cohort had a 2.5-fold increased risk of any abnormal cardiopulmonary response (odds ratio, 2.5; 95% confidence interval, 1.2, 5.3; P = 0.014). Distinct exercise responses in BC reflected impaired oxygen delivery and uptake relative to control, although considerable inter-individual heterogeneity within cohorts was observed. In unsupervised, hierarchical cluster analysis, six phenogroups were identified with marked differences in cardiopulmonary response patterns and unique clinical characteristics. CONCLUSIONS Abnormal cardiopulmonary response to exercise is common in BC and is related to impairments in oxygen delivery and uptake. The identification of exercise response phenogroups could help improve cardiovascular risk stratification and guide investigation of targeted exercise interventions.
Collapse
Affiliation(s)
- Jessica M. Scott
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Jasme Lee
- Memorial Sloan Kettering Cancer Center, New York, NY
| | | | | | | | | | | | | | - Anthony F. Yu
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | | | - Chau Dang
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Chaya S. Moskowitz
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Lee W. Jones
- Memorial Sloan Kettering Cancer Center, New York, NY
- Weill Cornell Medical College, New York, NY
| | - Neil D. Eves
- University of British Columbia, Kelowna, BC, CANADA
| |
Collapse
|
138
|
Upshaw JN, Nelson J, Sweigart B, Rodday AM, Kumar AJ, Konstam MA, Wong JB, Ky B, Karmiy S, Friedberg JW, Evens AM, Kent DM, Parsons SK. Impact of Preexisting Heart Failure on Treatment and Outcomes in Older Patients With Hodgkin Lymphoma. JACC CardioOncol 2024; 6:200-213. [PMID: 38774008 PMCID: PMC11103040 DOI: 10.1016/j.jaccao.2024.02.003] [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: 09/20/2023] [Revised: 01/31/2024] [Accepted: 02/02/2024] [Indexed: 05/24/2024] Open
Abstract
Background Older patients with Hodgkin lymphoma (HL) often have comorbid cardiovascular disease; however, the impact of pre-existing heart failure (HF) on the management and outcomes of HL is unknown. Objectives The aim of this study was to assess the prevalence of pre-existing HF in older patients with HL and its impact on treatment and outcomes. Methods Linked Surveillance, Epidemiology, and End Results (SEER) and Medicare data from 1999 to 2016 were used to identify patients 65 years and older with newly diagnosed HL. Pre-existing HF, comorbidities, and cancer treatment were ascertained from billing codes and cause-specific mortality from SEER. The associations between pre-existing HF and cancer treatment were estimated using multivariable logistic regression. Cause-specific Cox proportional hazards models adjusted for comorbidities and cancer treatment were used to estimate the association between pre-existing HF and cause-specific mortality. Results Among 3,348 patients (mean age 76 ± 7 years, 48.6% women) with newly diagnosed HL, pre-existing HF was present in 437 (13.1%). Pre-existing HF was associated with a lower likelihood of using anthracycline-based chemotherapy regimens (OR: 0.42; 95% CI: 0.29-0.60) and a higher likelihood of lymphoma mortality (HR: 1.25; 95% CI: 1.06-1.46) and cardiovascular mortality (HR: 2.57; 95% CI: 1.96-3.36) in models adjusted for comorbidities. One-year lymphoma mortality cumulative incidence was 37.4% (95% CI: 35.5%-39.5%) with pre-existing HF and 26.3% (95% CI: 25.0%-27.6%) without pre-existing HF. The cardioprotective medications dexrazoxane and liposomal doxorubicin were used in only 4.2% of patients. Conclusions Pre-existing HF in older patients with newly diagnosed HL is common and associated with higher 1-year mortality. Strategies are needed to improve lymphoma and cardiovascular outcomes in this high-risk population.
Collapse
Affiliation(s)
- Jenica N. Upshaw
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, USA
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jason Nelson
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Benjamin Sweigart
- Clinical and Translational Science Institute, Tufts University, Boston, Massachusetts, USA
| | - Angie Mae Rodday
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Anita J. Kumar
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| | - Marvin A. Konstam
- Division of Cardiology, Tufts Medical Center, Boston, Massachusetts, USA
| | - John B. Wong
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
- Division of Clinical Decision Making, Tufts Medical Center, Boston, Massachusetts, USA
| | - Bonnie Ky
- Division of Cardiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Samuel Karmiy
- Department of Medicine, Tufts Medical Center, Boston, Massachusetts, USA
| | - Jonathan W. Friedberg
- Wilmot Cancer Institute, University of Rochester Medical Center, Rochester, New York, USA
| | | | - David M. Kent
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
| | - Susan K. Parsons
- Institute of Clinical Research and Health Policy Studies, Tufts Medical Center, Boston, Massachusetts, USA
- Division of Hematology-Oncology, Tufts Medical Center, Boston, Massachusetts, USA
| |
Collapse
|
139
|
Jacobs JEJ, Greason G, Mangold KE, Wildiers H, Willems R, Janssens S, Noseworthy P, Lopez-Jimenez F, Voigt JU, Friedman P, Van Aelst L, Vandenberk B, Attia ZI, Herrmann J. Artificial intelligence electrocardiogram as a novel screening tool to detect a newly abnormal left ventricular ejection fraction after anthracycline-based cancer therapy. Eur J Prev Cardiol 2024; 31:560-566. [PMID: 37943680 PMCID: PMC10972628 DOI: 10.1093/eurjpc/zwad348] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 10/15/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023]
Abstract
AIMS Cardiotoxicity is a serious side effect of anthracycline treatment, most commonly manifesting as a reduction in left ventricular ejection fraction (EF). Early recognition and treatment have been advocated, but robust, convenient, and cost-effective alternatives to cardiac imaging are missing. Recent developments in artificial intelligence (AI) techniques applied to electrocardiograms (ECGs) may fill this gap, but no study so far has demonstrated its merit for the detection of an abnormal EF after anthracycline therapy. METHODS AND RESULTS Single centre consecutive cohort study of all breast cancer patients with ECG and transthoracic echocardiography (TTE) evaluation before and after (neo)adjuvant anthracycline chemotherapy. Patients with HER2-directed therapy, metastatic disease, second primary malignancy, or pre-existing cardiovascular disease were excluded from the analyses as were patients with EF decline for reasons other than anthracycline-induced cardiotoxicity. Primary readout was the diagnostic performance of AI-ECG by area under the curve (AUC) for EFs < 50%. Of 989 consecutive female breast cancer patients, 22 developed a decline in EF attributed to anthracycline therapy over a follow-up time of 9.8 ± 4.2 years. After exclusion of patients who did not have ECGs within 90 days of a TTE, 20 cases and 683 controls remained. The AI-ECG model detected an EF < 50% and ≤ 35% after anthracycline therapy with an AUC of 0.93 and 0.94, respectively. CONCLUSION These data support the use of AI-ECG for cardiotoxicity screening after anthracycline-based chemotherapy. This technology could serve as a gatekeeper to more costly cardiac imaging and could enable patients to monitor themselves over long periods of time.
Collapse
Affiliation(s)
- Johanna E J Jacobs
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Grace Greason
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Kathryn E Mangold
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Hans Wildiers
- Department of Oncology, University Hospitals Leuven,
Leuven, Belgium
| | - Rik Willems
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Stefan Janssens
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Peter Noseworthy
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Francisco Lopez-Jimenez
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Jens-Uwe Voigt
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Paul Friedman
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Lucas Van Aelst
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Bert Vandenberk
- Department of Cardiovascular Sciences, KU Leuven,
Leuven, Belgium
| | - Zachi Itzhak Attia
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic,
200 First St. SW, Rochester, MN 55905, USA
| |
Collapse
|
140
|
Oikonomou EK, Sangha V, Dhingra LS, Aminorroaya A, Coppi A, Krumholz HM, Baldassarre LA, Khera R. Artificial intelligence-enhanced risk stratification of cancer therapeutics-related cardiac dysfunction using electrocardiographic images. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.03.12.24304047. [PMID: 38562897 PMCID: PMC10984033 DOI: 10.1101/2024.03.12.24304047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Background Risk stratification strategies for cancer therapeutics-related cardiac dysfunction (CTRCD) rely on serial monitoring by specialized imaging, limiting their scalability. Objectives To examine an artificial intelligence (AI)-enhanced electrocardiographic (AI-ECG) surrogate for imaging risk biomarkers, and its association with CTRCD. Methods Across a five-hospital U.S.-based health system (2013-2023), we identified patients with breast cancer or non-Hodgkin lymphoma (NHL) who received anthracyclines (AC) and/or trastuzumab (TZM), and a control cohort receiving immune checkpoint inhibitors (ICI). We deployed a validated AI model of left ventricular systolic dysfunction (LVSD) to ECG images (≥0.1, positive screen) and explored its association with i) global longitudinal strain (GLS) measured within 15 days (n=7,271 pairs); ii) future CTRCD (new cardiomyopathy, heart failure, or left ventricular ejection fraction [LVEF]<50%), and LVEF<40%. In the ICI cohort we correlated baseline AI-ECG-LVSD predictions with downstream myocarditis. Results Higher AI-ECG LVSD predictions were associated with worse GLS (-18% [IQR:-20 to -17%] for predictions<0.1, to -12% [IQR:-15 to -9%] for ≥0.5 (p<0.001)). In 1,308 patients receiving AC/TZM (age 59 [IQR:49-67] years, 999 [76.4%] women, 80 [IQR:42-115] follow-up months) a positive baseline AI-ECG LVSD screen was associated with ~2-fold and ~4.8-fold increase in the incidence of the composite CTRCD endpoint (adj.HR 2.22 [95%CI:1.63-3.02]), and LVEF<40% (adj.HR 4.76 [95%CI:2.62-8.66]), respectively. Among 2,056 patients receiving ICI (age 65 [IQR:57-73] years, 913 [44.4%] women, follow-up 63 [IQR:28-99] months) AI-ECG predictions were not associated with ICI myocarditis (adj.HR 1.36 [95%CI:0.47-3.93]). Conclusion AI applied to baseline ECG images can stratify the risk of CTRCD associated with anthracycline or trastuzumab exposure.
Collapse
Affiliation(s)
- Evangelos K. Oikonomou
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Veer Sangha
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Department of Engineering Science, University of Oxford, Oxford, UK
| | - Lovedeep S. Dhingra
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Arya Aminorroaya
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Andreas Coppi
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT
| | - Harlan M. Krumholz
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT
| | - Lauren A. Baldassarre
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
| | - Rohan Khera
- Section of Cardiovascular Medicine, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, CT
- Section of Biomedical Informatics and Data Science, Yale School of Medicine, New Haven, CT
- Section of Health Informatics, Department of Biostatistics, Yale School of Public Health, New Haven, CT
| |
Collapse
|
141
|
Senechal I, Andres MS, Tong J, Perone Y, Ramalingam S, Nazir MS, Rosen SD, Turner N, Ring A, Lyon AR. Late cardiotoxicity related to HER2-targeted cancer therapy. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:14. [PMID: 38454509 PMCID: PMC10921578 DOI: 10.1186/s40959-024-00215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 02/25/2024] [Indexed: 03/09/2024]
Abstract
Long-term anti-HER2 therapy in metastatic HER2 + cancers is increasing, but data about the incidence and risk factors for developing late Cancer therapy-related cardiac dysfunction (CTRCD) are missing. We conducted a single-centre, retrospective analysis of a cohort of late anti-HER2 related cardiac dysfunction referred to our Cardio-Oncology service. We include seventeen patients with metastatic disease who developed CTRCD after at least five years of continuous anti-HER2 therapy. Events occurred after a median time of 6.5 years (IQR 5.3-9.0) on anti-HER2 therapy. The lowest (median) LVEF and GLS were 49% (IQR 45-55) and - 15.4% (IQR - 14.9 - -16.3) respectively. All our patients continued or restarted, after a brief interruption, their anti-HER2 therapy. Most (16/17) were started on heart failure medical therapy and normalized their left ventricular ejection fraction at a follow-up. Our study has demonstrated that CTRCD can occur after many years of stability on anti-HER2 therapy and reinforces the importance of continuing cardiovascular surveillance in this population.
Collapse
Affiliation(s)
- Isabelle Senechal
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK.
- Centre hospitalier universitaire de Québec, Québec, Canada.
| | - Maria Sol Andres
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Jieli Tong
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | | | - Sivatharshini Ramalingam
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| | - Muhummad Sohaib Nazir
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Stuart D Rosen
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
- National Heart and Lung Institute, Imperial College London, London, UK
| | | | | | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Guy's and St. Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
142
|
Zhou J, Liu T, Roever L, Zhang Q. Editorial: Interpretable predictive analytics for precision cardio-oncology preventive care. Front Cardiovasc Med 2024; 11:1377749. [PMID: 38510197 PMCID: PMC10951633 DOI: 10.3389/fcvm.2024.1377749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/14/2024] [Indexed: 03/22/2024] Open
Affiliation(s)
- Jiandong Zhou
- Department of Family Medicine and Primary Care, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Division of Health Science, Warwick Medical School, University of Warwick, Coventry, United Kingdom
| | - 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, China
| | - Leonardo Roever
- Lebanese American University, Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut, Lebanon
| | - Qingpeng Zhang
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Musketeers Foundation Institute of Data Science, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| |
Collapse
|
143
|
Michel L, Rassaf T. [Heart and cancer: cardio-oncology]. Herz 2024; 49:111-117. [PMID: 38289420 DOI: 10.1007/s00059-024-05232-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/08/2024] [Indexed: 03/07/2024]
Abstract
Cardiovascular diseases and cancer have a complex relationship and show a reciprocal linkage and influence. Mutual risk factors, demographic changes and increasing multimorbidity result in an increase in the incidence of both diseases. Advances in oncological and cardiological treatment lead to a further increase in patients with cured or chronic diseases as a relevant comorbidity. The induction of cardiovascular side effects by cancer therapies leads to increased cardiovascular morbidity and mortality in cancer patients. Recent data also show that cardiovascular disease, through various factors, can also promote the development and progression of cancer. An understanding of the interrelationship between cardiovascular diseases and cancer can be seen as a major medical challenge for the future. To this end, scientific, structural, clinical and educational interfaces between cardiology and oncology are essential. This article outlines the complex relationships between cardiovascular diseases and cancer and defines current and future challenges for the best possible care of affected patients.
Collapse
Affiliation(s)
- Lars Michel
- Klinik für Kardiologie und Angiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Hufelandstraße 55, 45147, Essen, Deutschland
| | - Tienush Rassaf
- Klinik für Kardiologie und Angiologie, Westdeutsches Herz- und Gefäßzentrum, Universitätsklinikum Essen, Hufelandstraße 55, 45147, Essen, Deutschland.
| |
Collapse
|
144
|
Leiva O, Beaty W, Soo S, Agarwal MA, Yang EH. Cancer Therapy-Associated Pulmonary Hypertension and Right Ventricular Dysfunction: Etiologies and Prognostic Implications. Rev Cardiovasc Med 2024; 25:87. [PMID: 39076943 PMCID: PMC11263834 DOI: 10.31083/j.rcm2503087] [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: 10/31/2023] [Revised: 12/22/2023] [Accepted: 01/04/2024] [Indexed: 07/31/2024] Open
Abstract
Advances in cancer therapies have improved oncologic outcomes but can potentially expose patients to risk of cardiovascular toxicity. While left ventricular (LV) dysfunction is a well-known cardiotoxicity of cancer therapy. Pulmonary hypertension (PH) and right ventricular (RV) dysfunction are seen with several cancer therapies, including alkylating agents, tyrosine kinase inhibitors (TKIs), and immunotherapy, and are associated with significant morbidity and mortality. Awareness and recognition of cancer therapy-associated PH and RV dysfunction is critical to identify underlying etiologies and institute the appropriate therapy. However, gaps exist in the current literature on the epidemiology of PH and RV dysfunction in cancer, underlying pathophysiology and optimal management strategies.
Collapse
Affiliation(s)
- Orly Leiva
- Division of Cardiology, Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - William Beaty
- Department of Medicine, New York University Grossman School of Medicine, New York, NY 10016, USA
| | - Steven Soo
- Department of Medicine, New York University Grossman Long Island School of Medicine, Mineola, NY 11501, USA
| | - Manyoo A. Agarwal
- Cardio-Oncology Program, Heart, Vascular and Thoracic Institute, Cleveland Clinic Abu Dhabi, 00000 Abu Dhabi, United Arab Emirates
| | - Eric H. Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
145
|
Vafa RG, Sabahizadeh A, Mofarrah R. Guarding the heart: How SGLT-2 inhibitors protect against chemotherapy-induced cardiotoxicity: SGLT-2 inhibitors and chemotherapy-induced cardiotoxicity. Curr Probl Cardiol 2024; 49:102350. [PMID: 38128634 DOI: 10.1016/j.cpcardiol.2023.102350] [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: 12/13/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
The introduction of chemotherapy agents has significantly transformed cancer treatment, with anthracyclines being one of the most commonly used drugs. While these agents have proven to be highly effective against various types of cancers, they come with complications, including neurotoxicity, nephrotoxicity, and cardiotoxicity. Among these side effects, cardiotoxicity is the leading cause of morbidity and mortality, with anthracyclines being the primary culprit. Chemotherapy medications have various mechanisms that can lead to cardiac injury. Hence, numerous studies have been conducted to decrease the cardiotoxicity of these treatments. Combination therapy with beta-blockers, Angiotensin-converting enzyme inhibitors, and angiotensin receptor blockers have effectively reduced such outcomes. However, a definitive preventive strategy is yet to be established. Meanwhile, sodium-glucose co-transporter-2 (SGLT-2) inhibitors lower blood glucose levels in type 2 diabetes by reducing its re-absorption in the kidneys. They are thus considered potent drugs for glycemic control and reduction of cardiovascular risks. Recent studies have shown that SGLT-2 inhibitors are crucial in preventing chemotherapy-induced cardiotoxicity. They enhance heart cell viability, prevent degenerative changes, stimulate autophagy, and reduce cell death. This drug class also reduces inflammation by inhibiting reactive oxygen species and inflammatory cytokine production. Moreover, it can not only reverse the harmful effects of anticancer agents on the heart structure but also enhance the effectiveness of chemotherapy by minimizing potential consequences on the heart. In conclusion, SGLT-2 inhibitors hold promise as a therapeutic strategy for protecting cancer patients from chemotherapy-induced heart damage and improving cardiovascular outcomes.
Collapse
|
146
|
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
|
147
|
Talty A, Morris R, Deighan C. Home-based self-management multimodal cancer interventions & cardiotoxicity: a scoping review. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:12. [PMID: 38424647 PMCID: PMC10903028 DOI: 10.1186/s40959-024-00204-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 01/17/2024] [Indexed: 03/02/2024]
Abstract
BACKGROUND Due to advancements in methods of cancer treatment, the population of people living with and beyond cancer is dramatically growing. The number of cancer survivors developing cardiovascular diseases and heart failure is also rising, due in part to the cardiotoxic nature of many cancer treatments. Guidelines are being increasingly released, emphasising the need for interdisciplinary action to address this gap in survivorship care. However, the extent to which interventions exist, incorporating the recommendations of cardio-oncology research, remains undetermined. OBJECTIVE The aim of this scoping review is to assess the nature, extent and remit of existing cancer care interventions and their integration of cardio-oncology principles. METHODS The review was conducted in accordance with the PRISMA Extension for Scoping Reviews Guidelines. Databases were independently searched for articles from 2010 to 2022, by two members of the research team. Data were charted and synthesised using the following criteria: (a) the focus of the intervention (b) the medium of delivery (c) the duration (d) the modalities included in the interventions (e) the research articles associated with each intervention (f) the type of studies conducted (g) key measures used (h) outcomes reported. RESULTS Interventions encompassed six key modalities: Psychological Support, Physical Activity, Nutrition, Patient Education, Lifestyle and Caregiver Support. The focus, medium of delivery and duration of interventions varied significantly. While a considerable number of study protocols and pilot studies exist documenting HSMIs, only 25% appear to have progressed beyond this stage of development. Of those that have, the present review did not identify any 'feasible' interventions that covered each of the six modalities, while being generalisable to all cancer survivors and incorporating the recommendations from cardio-oncology research. CONCLUSION Despite the substantial volume of research and evidence from the field of cardio-oncology, the findings of this scoping review suggest that the recommendations from guidelines have yet to be successfully translated from theory to practice. There is an opportunity, if not necessity, for cardiac rehabilitation to expand to meet the needs of those living with and beyond cancer.
Collapse
Affiliation(s)
- Anna Talty
- The Heart Manual Department, Astley Ainslie Hospital, Grange Loan, Edinburgh, Scotland, UK, EH9 2HL
| | - Roseanne Morris
- The Heart Manual Department, Astley Ainslie Hospital, Grange Loan, Edinburgh, Scotland, UK, EH9 2HL
| | - Carolyn Deighan
- The Heart Manual Department, Astley Ainslie Hospital, Grange Loan, Edinburgh, Scotland, UK, EH9 2HL.
| |
Collapse
|
148
|
Puła B, Kępski J, Misiewicz-Krzemińska I, Szmit S. Left and right ventricular global longitudinal strain assessment together with biomarker evaluation may have a predictive and prognostic role in patients qualified for hematopoietic stem cell transplantation due to hematopoietic and lymphoid malignancies - a pilot study description. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:9. [PMID: 38368369 PMCID: PMC10873966 DOI: 10.1186/s40959-024-00210-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
The hematopoietic stem cell transplantation (HSCT) procedure is considered a cardiovascular burden. This is due to the potentially cardiotoxic cytostatic agents used before and the risks associated with peri-transplant procedures. We designed a pilot study to determine the clinical utility of the new ST2 marker; furthermore, we routinely assessed cardiac parameters in HSCT recipients. Based on previous cardio-oncology experience in lung and prostate cancer, we can confirm the prognostic and predictive value of classic cardiac biomarkers and modern echocardiography parameters such as global longitudinal strain of the left and right ventricle. After conducting this pilot study we can create a predictive and prognostic model for patients undergoing HSCT. This will greatly enrich our clinical practice, especially in treating older people.
Collapse
Affiliation(s)
- Bartosz Puła
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Jarosław Kępski
- Department of Cardio-Oncology, Chair of Hematology and Transfusion Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland
| | | | - Sebastian Szmit
- Department of Cardio-Oncology, Chair of Hematology and Transfusion Medicine, Centre of Postgraduate Medical Education, Warsaw, Poland.
| |
Collapse
|
149
|
Murtagh G, deFilippi C, Zhao Q, Barac A. Circulating biomarkers in the diagnosis and prognosis of immune checkpoint inhibitor-related myocarditis: time for a risk-based approach. Front Cardiovasc Med 2024; 11:1350585. [PMID: 38410245 PMCID: PMC10894940 DOI: 10.3389/fcvm.2024.1350585] [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: 12/05/2023] [Accepted: 01/16/2024] [Indexed: 02/28/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are monoclonal antibodies that block immune checkpoints and therefore activate immune cells, allowing them to recognize and attack cancer cells. ICIs have revolutionized oncology practice, but their use has been complicated by immune-related adverse events (irAEs). Of cardiovascular (CV) irAEs, ICI-related myocarditis has received significant attention due to high mortality rates, ranging from 25% to 50%, despite its overall low incidence. Establishing the early diagnosis of ICI-myocarditis is important for early initiation of steroids and consideration of hospitalization in patients who are at risk for hemodynamic compromise and need high acuity care in a tertiary setting. In this review, we summarize the diagnostic and prognostic tools for ICI-myocarditis, including electrocardiography, echocardiography, cardiac magnetic resonance imaging, with emphasis on circulating biomarkers. Cardiac troponins (cTns) are an essential component of the diagnosis of ICI-myocarditis, and we provide a summary of the recent studies that utilized different assays (cTnI vs. cTnT) and outcomes (diagnosis vs. prognosis including major adverse cardiac outcomes). With the exponential increase in ICI use across different oncology indications, there is a major need to include biomarkers in risk stratification to guide diagnosis and treatment. Our review proposes a framework for future multisite registries, including cTn evaluation at baseline and at the time of irAE suspicion, with development of central biobanking to allow head-to-head evaluation and clinical validation of different biomarker assays in ICI-myocarditis. This approach, with the inclusion of CV biomarkers into clinical and pragmatic oncology trials, holds promise to improve the early recognition and management of ICI-myocarditis and CV irAEs, thus leading to better outcomes.
Collapse
Affiliation(s)
- Gillian Murtagh
- Core Diagnostics, Abbott Laboratories, Abbott Park, IL, United States
| | | | - Qiong Zhao
- Inova Schar Heart and Vascular, Falls Church, VA, United States
| | - Ana Barac
- Inova Schar Heart and Vascular, Falls Church, VA, United States
| |
Collapse
|
150
|
Ou W, Jiang T, Zhang N, Lu K, Weng Y, Zhou X, Wang D, Dong Q, Tang X. Role of HDL cholesterol in anthracycline-induced subclinical cardiotoxicity: a prospective observational study in patients with diffuse large B-cell lymphoma treated with R-CHOP. BMJ Open 2024; 14:e074541. [PMID: 38341200 PMCID: PMC10862278 DOI: 10.1136/bmjopen-2023-074541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
OBJECTIVES Anthracycline-induced cardiotoxicity is a debilitating cardiac dysfunction for which there are no effective treatments, making early prevention of anthracycline-induced subclinical cardiotoxicity (AISC) crucial. High-density lipoprotein cholesterol (HDL-C) plays a role in cardioprotection, but its impact on AISC remains unclear. Our study aims to elucidate the protective capacity of HDL-C in AISC in patients with diffuse large B-cell lymphoma (DLBCL) treated with R-CHOP (cyclophosphamide, vincristine, doxorubicin, prednisone and rituximab). DESIGN Prospective observational study. SETTING Conducted in China from September 2020 to September 2022. PARTICIPANTS 70 chemotherapy-naïve patients newly diagnosed with DLBCL who were scheduled to receive the standard dose of R-CHOP; 60 participants included in a case-control study (DOI: 10.1186/s12885-022-10085-6). PRIMARY OUTCOME MEASURES Serum biomarkers, 2D speckle tracking echocardiography and conventional echocardiography were measured at baseline, at the end of the third and sixth cycles of R-CHOP and 6 and 12 months after chemotherapy. RESULTS 24 patients experienced AISC, while 10 did not. 36 patients were lost to follow-up and death. Cox regression analysis showed that higher levels of HDL-C were associated with a significantly lower risk of AISC (unadjusted HR=0.24, 95% CI 0.09 to 0.67, p=0.006; adjusted HR=0.27, 95% CI 0.09 to 0.79, p=0.017). Patients without AISC had a more stable and higher HDL-C level during the follow-up period. HDL-C levels significantly decreased from the end of the third cycle of chemotherapy to the end of the sixth cycle of chemotherapy in all patients (p=0.034), and particularly in the AISC group (p=0.003). The highest level of HDL-C was significantly higher in patients without AISC than in those with AISC (1.52±0.49 vs 1.22±0.29, p=0.034). CONCLUSIONS Our study suggests that higher HDL-C levels may associate with lower AISC risk in patients with DLBCL treated with R-CHOP. HDL-C could be a cardioprotective target, but further research is needed to confirm its benefits and limitations. STUDY REGISTRATION NUMBER Study registration number: ChiCTR2100054721.
Collapse
Affiliation(s)
- Wenxin Ou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Tiantian Jiang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Nan Zhang
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Kai Lu
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yue Weng
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xi Zhou
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dong Wang
- Department of Ultrasound, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Qian Dong
- Department of Cardiology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xiaoqiong Tang
- Department of Hematology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|