1
|
Rankin S, Fountain C, Gemmell AJ, Quinn D, Henderson A, McClure J, Small S, Venugopal B, McKay P, Slomka PJ, Colville D, Petrie MC, Meléndez GC, Lang NN. Arterial effects of anthracycline: structural and inflammatory assessments in non-human primates and lymphoma patients. Clin Sci (Lond) 2025; 139:29-41. [PMID: 39680089 DOI: 10.1042/cs20241529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 12/04/2024] [Accepted: 12/16/2024] [Indexed: 12/17/2024]
Abstract
Anthracyclines, such as doxorubicin, are important anti-cancer therapies but are associated with arterial injury. Histopathological insights have been limited to small animal models, and the role of inflammation in the arterial toxic effects of anthracycline is unclear in humans. Our aims were (1) to evaluate aortic media fibrosis and injury in non-human primates treated with anthracyclines; (2) to assess the effect of anthracycline on aortic inflammation in patients treated for lymphoma. African Green monkeys (AGMs) received doxorubicin (30-60 mg/m2/biweekly intravenously, cumulative dose: 240 mg/m2). Blinded histopathologic analyses of the ascending aorta were performed 15 weeks after the last doxorubicin dose and compared to five age- and gender-matched healthy, untreated AGMs. Analysis of the thoracic aorta of patients with diffuse large B-cell lymphoma (DLBCL), at baseline and after doxorubicin exposure, was performed using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in this observational study by maximal tissue-to-background ratio (TBRmax). In AGMs, doxorubicin exposure was associated with greater aortic fibrosis (collagen deposition: doxorubicin 6.23 ± 0.88% vs. controls 4.67 ± 0.54%; P=0.01) and intracellular vacuolization (doxorubicin 66.3 ± 10.1 vs. controls 11.5 ± 4.2 vacuoles/field, P<0.0001) than untreated controls. In 101 patients with DLBCL, there was no change in aortic TBRmax after anthracycline exposure (TBRmax 1.46 ± 0.16 vs. 1.44 ± 0.14, respectively, P=0.14). Univariate analyses yielded similar results. In a large animal model, anthracycline exposure was associated with aortic fibrosis. In patients with lymphoma, anthracycline exposure was not associated with aortic inflammation. Further research is required to elucidate the mechanisms of anthracycline-related vascular harm.
Collapse
MESH Headings
- Animals
- Humans
- Female
- Doxorubicin/adverse effects
- Male
- Middle Aged
- Fibrosis/chemically induced
- Antibiotics, Antineoplastic/adverse effects
- Antibiotics, Antineoplastic/toxicity
- Lymphoma, Large B-Cell, Diffuse/drug therapy
- Lymphoma, Large B-Cell, Diffuse/pathology
- Lymphoma, Large B-Cell, Diffuse/diagnostic imaging
- Positron Emission Tomography Computed Tomography
- Aged
- Anthracyclines/adverse effects
- Inflammation/pathology
- Inflammation/chemically induced
- Adult
- Aorta, Thoracic/pathology
- Aorta, Thoracic/diagnostic imaging
- Aorta, Thoracic/drug effects
- Aorta/pathology
- Aorta/diagnostic imaging
- Aorta/drug effects
Collapse
Affiliation(s)
- Stephen Rankin
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Caitlin Fountain
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, U.S.A
| | - Alastair J Gemmell
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Daire Quinn
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Alasdair Henderson
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - John McClure
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Sandy Small
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Balaji Venugopal
- School of Medicine, Dentistry and Nursing, University of Glasgow
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Pamela McKay
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Piotr J Slomka
- Cedars-Sinai, Division of Artificial Intelligence in Medicine, Department of Medicine, Los Angeles, U.S.A
| | - David Colville
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Mark C Petrie
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| | - Giselle C Meléndez
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine, Wake Forest University School of Medicine, Winston-Salem, U.S.A
| | - Ninian N Lang
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow, U.K
| |
Collapse
|
2
|
Krüger DN, Bosman M, Van Craenenbroeck EM, De Meyer GRY, Franssen C, Guns PJ. Dexrazoxane prevents vascular toxicity in doxorubicin-treated mice. CARDIO-ONCOLOGY (LONDON, ENGLAND) 2024; 10:65. [PMID: 39367508 PMCID: PMC11451066 DOI: 10.1186/s40959-024-00270-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Accepted: 09/25/2024] [Indexed: 10/06/2024]
Abstract
BACKGROUND Doxorubicin (DOX) is used for breast cancer and lymphoma, but can cause cardiotoxicity, arterial stiffness, and endothelial dysfunction. We recently reported SERPINA3N as biomarker of cardiovascular toxicity in patients and mice. Dexrazoxane (DEXRA) is an FDA-approved drug that prevents DOX-induced cardiac toxicity in high-risk patients. However, the effect of DEXRA on vascular dysfunction during DOX treatment has not been documented. Therefore, here we investigated whether DEXRA protects against DOX-induced arterial stiffness, endothelial dysfunction, and SERPINA3N upregulation in tissue and plasma from mice. METHODS Male C57BL6/J mice were treated with DOX (4 mg/kg), DEXRA (40 mg/kg), a combination (DEXRA + DOX), or VEHICLE (0.9% NaCl) weekly i.p. for 6 weeks (n = 8 per group). Cardiovascular function was measured in vivo by ultrasound imaging at baseline, weeks 2 and 6. Vascular reactivity was analyzed ex vivo in the thoracic aorta at week 6 and molecular analysis was performed. RESULTS DEXRA prevented left ventricular ejection fraction decline by DOX (DEXRA + DOX: 62 ± 2% vs DOX: 51 ± 2%). Moreover, DEXRA prevented the increase in pulse wave velocity by DOX (DEXRA + DOX: 2.1 ± 0.2 m/s vs DOX: 4.5 ± 0.3 m/s) and preserved endothelium-dependent relaxation (DEXRA + DOX: 82 ± 3% vs DOX: 62 ± 3%). In contrast to DOX-treated mice, SERPINA3N did not increase in the DEXRA + DOX group. CONCLUSION Our results not only confirm the cardioprotective effects of DEXRA against DOX-induced cardiotoxicity but also add preservation of vascular endothelial cell function as an important mechanism. Moreover, the study demonstrates the potential of SERPINA3N as a biomarker for monitoring cardiovascular complications of DOX in high-risk patients.
Collapse
Affiliation(s)
- Dustin N Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium.
| | - Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, University of Antwerp, Antwerp, B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem, B-2650, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp, B-2610, Belgium
| |
Collapse
|
3
|
Dillon HT, Foulkes SJ, Baik AH, Scott JM, Touyz RM, Herrmann J, Haykowsky MJ, La Gerche A, Howden EJ. Cancer Therapy and Exercise Intolerance: The Heart Is But a Part: JACC: CardioOncology State-of-the-Art Review. JACC CardioOncol 2024; 6:496-513. [PMID: 39239327 PMCID: PMC11372306 DOI: 10.1016/j.jaccao.2024.04.006] [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: 02/08/2024] [Revised: 04/03/2024] [Accepted: 04/08/2024] [Indexed: 09/07/2024] Open
Abstract
The landscape of cancer therapeutics is continually evolving, with successes in improved survivorship and reduced disease progression for many patients with cancer. Improved cancer outcomes expose competing comorbidities, some of which may be exacerbated by cancer therapies. The leading cause of disability and death for many early-stage cancers is cardiovascular disease (CVD), which is often attributed to direct or indirect cardiac injury from cancer therapy. In this review, the authors propose that toxicities related to conventional and novel cancer therapeutics should be considered beyond the heart. The authors provide a framework using the oxygen pathway to understand the impact of cancer treatment on peak oxygen uptake, a marker of integrative cardiopulmonary function and CVD risk. Peripheral toxicities and the impact on oxygen transport are discussed. Consideration for the broad effects of cancer therapies will improve the prediction and identification of cancer survivors at risk for CVD, functional disability, and premature mortality and those who would benefit from therapeutic intervention, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Hayley T. Dillon
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Institute for Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia
| | - Stephen J. Foulkes
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
- Heart Exercise and Research Trials (HEART) Lab, St Vincent’s Institute, Fitzroy, Victoria, Australia
| | - Alan H. Baik
- Division of Cardiovascular Medicine, Department of Medicine, University of California, San Francisco, San Francisco, California, USA
| | - Jessica M. Scott
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Rhian M. Touyz
- Research Institute of McGill University Health Centre, Department of Medicine, McGill University, Montreal, Quebec, Canada
| | - Joerg Herrmann
- Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Mark J. Haykowsky
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Faculty of Nursing, College of Health Sciences, University of Alberta, Edmonton, Alberta, Canada
| | - André La Gerche
- Heart Exercise and Research Trials (HEART) Lab, St Vincent’s Institute, Fitzroy, Victoria, Australia
- Cardiology Department, St. Vincent’s Hospital Melbourne, Fitzroy, Australia
- HEART Lab, Victor Chang Cardiovascular Research Institute, Darlinghurst, NSW, Australia
| | - Erin J. Howden
- Baker Heart and Diabetes Institute, Melbourne, Australia
- Department of Cardiometabolic Health, University of Melbourne, Melbourne, Australia
| |
Collapse
|
4
|
Rankin S, Fountain C, Gemmell AJ, Quinn D, Henderson A, McClure J, Small S, Venugopal B, McKay P, Slomka PJ, Colville D, Petrie MC, Meléndez GC, Lang NN. Arterial effects of anthracycline: structural and inflammatory assessments in non-human primates and lymphoma patients using 18F-FDG positron emission tomography. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.30.596741. [PMID: 38895275 PMCID: PMC11185566 DOI: 10.1101/2024.05.30.596741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2024]
Abstract
Background Anthracyclines, such as doxorubicin, are important anti-cancer therapies but are associated with arterial injury. Histopathological insights have been limited to small animal models and the role of inflammation in the arterial toxic effects of anthracycline is unclear in humans. Our aims were: 1) To evaluate aortic media fibrosis and injury in non-human primates treated with anthracyclines; 2) To assess the effect of anthracycline on aortic inflammation in patients treated for lymphoma. Methods 1) African Green monkeys (AGM) received doxorubicin (30-60 mg/m2/biweekly IV, cumulative dose: 240 mg/m2). Blinded histopathologic analyses of collagen deposition and cell vacuolization in the ascending aorta were performed 15 weeks after the last doxorubicin dose and compared to 5 age- and gender-matched healthy, untreated AGMs. 2) Analysis of the thoracic aorta of patients with diffuse large B-cell lymphoma (DLBCL), at baseline and after doxorubicin exposure, was performed using 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) in this observational study. The primary outcome was change in maximal tissue-to-background ratio (TBRmax) of the thoracic aorta from baseline to their end-of-treatment clinical PET/CT. Results In AGMs, doxorubicin exposure was associated with greater aortic fibrosis (collagen deposition: doxorubicin cohort 6.23±0.88% vs. controls 4.67±0.54%; p=0.01) and increased intracellular vacuolization (doxorubicin 66.3 ± 10.1 vs controls 11.5 ± 4.2 vacuoles/field, p<0.0001) than untreated controls.In 101 patients with DLBCL, there was no change in aortic TBRmax after anthracycline exposure (pre-doxorubicin TBRmax 1.46±0.16 vs post-doxorubicin TBRmax 1.44±0.14, p=0.14). The absence of change in TBRmax was consistent across all univariate analyses. Conclusions In a large animal model, anthracycline exposure was associated with aortic fibrosis. In patients with lymphoma, anthracycline exposure was not associated with aortic inflammation.Further research is required to elucidate the mechanisms of anthracycline-related vascular harm.
Collapse
Affiliation(s)
- Stephen Rankin
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - Caitlin Fountain
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine. Wake Forest University School of Medicine, Winston-Salem, USA
| | - Alastair J Gemmell
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Daire Quinn
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Alasdair Henderson
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - John McClure
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - Sandy Small
- Department of Clinical Physics & Bioengineering, NHS Greater Glasgow & Clyde, Glasgow
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Balaji Venugopal
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Pamela McKay
- The Beatson West of Scotland Cancer Centre, Glasgow, UK
| | - Piotr J Slomka
- Cedars-Sinai, Division of Artificial Intelligence in Medicine, Department of Medicine, Los Angeles, USA
| | - David Colville
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
- School of Medicine, Dentistry and Nursing, University of Glasgow
| | - Mark C Petrie
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| | - Giselle C. Meléndez
- Departments of Internal Medicine, Section on Cardiology and Pathology, Section on Comparative Medicine. Wake Forest University School of Medicine, Winston-Salem, USA
| | - Ninian N Lang
- BHF Glasgow Cardiovascular Research Centre, School of Cardiovascular and Metabolic Health, University of Glasgow, Glasgow UK
| |
Collapse
|
5
|
Shen H, Zhou W, ChunrongTu, Peng Y, Li X, Liu D, Wang X, Zhang X, Zeng X, Zhang J. Thoracic aorta injury detected by 4D flow MRI predicts subsequent main adverse cardiovascular events in breast cancer patients receiving anthracyclines: A longitudinal study. Magn Reson Imaging 2024; 109:67-73. [PMID: 38484947 DOI: 10.1016/j.mri.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: 01/20/2024] [Revised: 03/02/2024] [Accepted: 03/08/2024] [Indexed: 04/09/2024]
Abstract
PURPOSE To investigate longitudinal thoracic aorta injury using 3-dimensional phase-contrast magnetic resonance imaging (4D flow MRI) parameters and to evaluate their value for predicting the subsequent main adverse cardiovascular events (MACEs) in breast cancer patients receiving anthracyclines. METHODS Between July 2020 and July 2021, eighty-eight female participants with breast cancer scheduled to receive anthracyclines with or without trastuzumab prospectively enrolled. Each subjects underwent 4D flow MRI at baseline, 3 and 6 months in relation to baseline. The diameter, peak velocity (Vpeak), wall shear stress (WSS), pulse wave velocity (PWV), energy loss (EL) and pressure gradient (PG) of thoracic aorta were measured. The association between these parameters and subsequent MACEs was performed by Cox proportional hazard models. RESULTS Ten participants had subsequently MACEs. The Vpeak and PG gradually decreased and the WSS, PWV and EL progressively increased at 3 and 6 months compared with baseline. Adjusted multivariable analysis showed that the WSS of the proximal, mid- and distal ascending aorta [HR, 1.314 (95% confidence interval (CI): 1.003, 1.898)], [HR, 1.320 (95% CI: 1.002, 1.801)] and [HR, 1.322 (95% CI: 1.001, 1.805)] and PWV of ascending aorta [HR, 2.223 (95% CI: 1.010, 4.653)] at 3 months were associated with subsequent MACEs. Combined WSS and PWV of ascending aorta at 3 months yielded the highest AUC (0.912) for predicting subsequent MACEs. CONCLUSION Combined WSS and PWV of ascending aorta at 3 months is helpful for predicting the subsequent MACEs in breast cancer patients treated by anthracyclines.
Collapse
Affiliation(s)
- Hesong Shen
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Wenqi Zhou
- Department of Breast Cancer Center, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - ChunrongTu
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Yangling Peng
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Xiaoqin Li
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Daihong Liu
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China
| | - Xiaoyong Zhang
- Clinical Science, Philips Healthcare, 1268 Tianfu Avenue, Hitech Zone, Chengdu, China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital & Chongqing Cancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China.
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital & ChongqingCancer Institute & Chongqing Cancer Hospital, 181 Hanyu Road, Shapingba District, Chongqing, China.
| |
Collapse
|
6
|
Benkhedda S, Bengherbi N, Cherifi Y, Ouabdesselam S, Waheed N, Harris CM. Arterial Stiffness Changes in Adult Cancer Patients Receiving Anticancer Chemotherapy: A Real-World Bicentric Experience. Cureus 2024; 16:e56647. [PMID: 38646338 PMCID: PMC11032169 DOI: 10.7759/cureus.56647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/21/2024] [Indexed: 04/23/2024] Open
Abstract
Background Chemotherapy correlates to acute and long-term cardiotoxicity, is reflected clinically by myocardial and vascular endothelial dysfunction, and can cause cardiovascular complications. Thus, early diagnosis of cardiovascular disease in cancer patients undergoing anti-cancer treatment is necessary to enhance long-term survival. Our principal objective in this study was to discern the impact of specific anti-cancer chemotherapeutics and biologics on arterial stiffness alterations before and after the administration. Methods Conducted at Mustafa Bacha University Hospital, Algeria, the study focused on arterial stiffness in anti-cancer chemotherapy patients. Assessments included blood pressure, diabetes, and dyslipidemia, with precise measurements using validated systems, particularly pulse wave velocity (PWV). Various chemotherapy protocols were applied, and statistical analysis with R software (R Foundation for Statistical Computing, Vienna, Austria) maintained a significance level of p=0.05. Key outcomes centered on carotid-femoral PWV and secondary endpoints such as central and peripheral pressures and pulse pressure (PP). Univariate and bivariate analyses were conducted using appropriate statistical tests. Results A comparative prospective observational study was completed on 58 patients (34 women and 24 men; mean age: 52.64 +/- 12.12 years) treated with anti-cancer chemotherapy agents. Our evaluation included a complete clinical exam, electrocardiogram, Doppler echocardiography, and applanation tonometry with arterial stiffness measurement using PWV. Patients presented significantly higher levels of carotid-femoral PWV, regardless of the chosen chemotherapy protocol, with no return to the initial level after one year of stopping treatment (p-value < 0.01). Moreover, this increase was more significant in patients with diabetes and hypertension and patients treated with monoclonal antibodies or intercalants. Conclusion This prospective study shows that chemotherapy patients have elevated arterial stiffness, emphasizing the need to assess PWV and monitor cardiovascular risk factors. PP measurement with PWV could improve risk management.
Collapse
Affiliation(s)
- Salim Benkhedda
- Cardiology, Cardiology Oncology Collaborative Research Group, Faculty of Family Medicine, University of Algiers Benyoucef Benkhedda, Algiers, DZA
| | - Nacera Bengherbi
- Cardiology, Cardiology Oncology Collaborative Research Group, Faculty of Family Medicine, University of Algiers Benyoucef Benkhedda, Algiers, DZA
| | - Yahia Cherifi
- Cardiology, Cardiology Oncology Collaborative Research Group, Faculty of Family Medicine, University of Algiers Benyoucef Benkhedda, Algiers, DZA
| | - Souhila Ouabdesselam
- Cardiology, Cardiology Oncology Collaborative Research Group, Faculty of Family Medicine, University of Algiers Benyoucef Benkhedda, Algiers, DZA
| | - Nabila Waheed
- Radiation Oncology, The Center for Cancer & Blood Disorders, Fort Worth, USA
| | - Clara M Harris
- Internal Medicine, Baylor Scott & White All Saints Medical Center - Fort Worth, Fort Worth, USA
| |
Collapse
|
7
|
Ray M, Butel-Simoes LE, Lombard JM, Nordman IIC, Van der Westhuizen A, Collins NJ, Ngo DTM, Sverdlov AL. Women's cardiovascular health - the cardio-oncologic jigsaw. Climacteric 2024; 27:60-67. [PMID: 38073542 DOI: 10.1080/13697137.2023.2286382] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 11/15/2023] [Indexed: 01/16/2024]
Abstract
Improvements in cancer care have led to an exponential increase in cancer survival. This is particularly the case for breast cancer, where 5-year survival in Australia exceeds 90%. Cardiovascular disease (CVD) has emerged as one of the competing causes of morbidity and mortality among cancer survivors, both as a complication of cancer therapies and because the risk factors for cancer are shared with those for CVD. In this review we cover the key aspects of cardiovascular care for women throughout their cancer journey: the need for baseline cardiovascular risk assessment and management, a crucial component of the cardiovascular care; the importance of long-term surveillance for ongoing maintenance of cardiovascular health; and strong evidence for the beneficial effects of physical exercise to improve both cancer and cardiovascular outcomes. There is general disparity in cardiovascular outcomes for women, which is further exacerbated when both CVD and cancer co-exist. Collaboration between oncology and cardiac services, with an emergence of the whole field of cardio-oncology, allows for expedited investigation and treatment for these patients. This collaboration as well as a holistic approach to patient care and key role of patients' general practitioners are essential to ensure long-term health of people living with, during and beyond cancer.
Collapse
Affiliation(s)
- M Ray
- Cardiovascular Department, John Hunter Hospital, Hunter New England Local Health District, Newcastle, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - L E Butel-Simoes
- Cardiovascular Department, John Hunter Hospital, Hunter New England Local Health District, Newcastle, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
| | - J M Lombard
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, NSW, Australia
| | - I I C Nordman
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, NSW, Australia
| | - A Van der Westhuizen
- Department of Medical Oncology, Calvary Mater Newcastle, Waratah, NSW, Australia
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - N J Collins
- Cardiovascular Department, John Hunter Hospital, Hunter New England Local Health District, Newcastle, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - D T M Ngo
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| | - A L Sverdlov
- Cardiovascular Department, John Hunter Hospital, Hunter New England Local Health District, Newcastle, NSW, Australia
- Newcastle Centre of Excellence in Cardio-Oncology, Hunter Medical Research Institute, Hunter New England Local Health District, University of Newcastle and Calvary Mater Newcastle, Newcastle, NSW, Australia
- College of Health, Medicine and Wellbeing, University of Newcastle, Callaghan, NSW, Australia
| |
Collapse
|
8
|
Thomas A, O'Connell NS, Douglas E, Hatcher S, Park CJ, Dent S, Ansley K, Klem I, Bansal R, Westbrook K, Hundley WG, Bottinor W, Hackney MH, Richardson KM, Sirkisoon SR, D'Agostino RB, Jordan JH. Cardiovascular impact of near complete estrogen deprivation in premenopausal women with breast cancer: The CROWN study. Am Heart J 2024; 267:33-43. [PMID: 37890547 PMCID: PMC10976295 DOI: 10.1016/j.ahj.2023.10.007] [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: 07/21/2023] [Revised: 10/18/2023] [Accepted: 10/21/2023] [Indexed: 10/29/2023]
Abstract
Survival with operable breast cancer has improved markedly in recent decades, however, treatment-related cardiovascular toxicities threaten to offset these gains. Ovarian function suppression paired with aromatase inhibition, for premenopausal women with hormone receptor (HR)-positive breast cancer, is a newer widely adopted therapy with the potential for significant long-term cardiovascular toxicity. Abrupt estrogen deprivation for non-cancer reasons is associated with accelerated coronary artery disease. Women with breast cancer treated with aromatase inhibition in addition to ovarian function suppression experience a dual hit with regards to estrogen exposure. The CaRdiac Outcomes With Near-complete estrogen deprivation (CROWN) study seeks to understand the early, subclinical natural history of cardiovascular compromise in young women undergoing near-complete estrogen deprivation (NCED) therapy. It is critical to understand the early subclinical development of cardiovascular disease to identify a window for therapeutic intervention before overt cardiovascular events occur. This three-site regional study (Atrium Health Wake Forest, Duke, and Virginia Commonwealth University) uses serial stress cardiac magnetic resonance (CMR) imaging and cardiac computed tomography angiography (CCTA) obtained during the initial two years of NCED therapy to study myocardial prefusion reserve (MPR), large cardiovascular vessel changes, left ventricular function, and other cardiovascular parameters. The CROWN cohort will consist of 90 premenopausal women with breast cancer, 67 with HR-positive disease receiving NCED and 23 comparators with HR-negative disease. Participants will undergo three annual CMR scans and 2 CCTA scans during the 2-year study period. After initial activation hurdles, accrual has been brisk, and the study is expected to complete accrual in December 2024. Efforts are in place to encourage participant retention with the study primary outcome, change in MPR between the two groups, to be reported in 2026 to 2027. The results of this study will enable premenopausal women with breast cancer to balance the health burdens of cancer at a young age and treatment-related cardiovascular morbidity. Finally, the tools developed here can be utilized to study cardiovascular risk across a range of cancer types and cancer therapies with the ultimate goals of both developing generalizable risk stratification tools as well as validating interventions which prevent overt cardiovascular compromise.
Collapse
Affiliation(s)
- Alexandra Thomas
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | | | - Emily Douglas
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Sarah Hatcher
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Carolyn J Park
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Susan Dent
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC
| | - Katherine Ansley
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC; Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Igor Klem
- Duke Cardiovascular Magnetic Resonance Center, Duke University Medical Center, Durham, NC
| | - Rani Bansal
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC
| | - Kelly Westbrook
- Duke Cancer Institute, Department of Medicine, Duke University, Durham, NC
| | - W Gregory Hundley
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Wendy Bottinor
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA
| | - Mary Helen Hackney
- Division of Hematology, Oncology and Palliative Care, Massey Comprehensive Cancer Center, Virginia Commonwealth University, Richmond, VA
| | - Karl M Richardson
- Department of Internal Medicine, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Sherona R Sirkisoon
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Ralph B D'Agostino
- Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC
| | - Jennifer H Jordan
- Division of Cardiology, Pauley Heart Center, Virginia Commonwealth University, Richmond, VA; Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, VA.
| |
Collapse
|
9
|
Bosman M, Krüger DN, Favere K, De Meyer GRY, Franssen C, Van Craenenbroeck EM, Guns PJ. Dexrazoxane does not mitigate early vascular toxicity induced by doxorubicin in mice. PLoS One 2023; 18:e0294848. [PMID: 38015959 PMCID: PMC10684076 DOI: 10.1371/journal.pone.0294848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Accepted: 11/08/2023] [Indexed: 11/30/2023] Open
Abstract
Apart from cardiotoxicity, the chemotherapeutic agent doxorubicin (DOX) provokes acute and long-term vascular toxicity. Dexrazoxane (DEXRA) is an effective drug for treatment of DOX-induced cardiotoxicity, yet it remains currently unknown whether DEXRA prevents vascular toxicity associated with DOX. Accordingly, the present study aimed to evaluate the protective potential of DEXRA against DOX-related vascular toxicity in a previously-established in vivo and ex vivo model of vascular dysfunction induced by 16 hour (h) DOX exposure. Vascular function was evaluated in the thoracic aorta in organ baths, 16h after administration of DOX (4 mg/kg) or DOX with DEXRA (40 mg/kg) to male C57BL6/J mice. In parallel, vascular reactivity was evaluated after ex vivo incubation (16h) of murine aortic segments with DOX (1 μM) or DOX with DEXRA (10 μM). In both in vivo and ex vivo experiments, DOX impaired acetylcholine-stimulated endothelium-dependent vasodilation. In the ex vivo setting, DOX additionally attenuated phenylephrine-elicited vascular smooth muscle cell (VSMC) contraction. Importantly, DEXRA failed to prevent DOX-induced endothelial dysfunction and hypocontraction. Furthermore, RT-qPCR and Western blotting showed that DOX decreased the protein levels of topoisomerase-IIβ (TOP-IIβ), a key target of DEXRA, in the heart, but not in the aorta. Additionally, the effect of N-acetylcysteine (NAC, 10 μM), a reactive oxygen species (ROS) scavenger, was evaluated ex vivo. NAC did not prevent DOX-induced impairment of acetylcholine-stimulated vasodilation. In conclusion, our results show that DEXRA fails to prevent vascular toxicity resulting from 16h DOX treatment. This may relate to DOX provoking vascular toxicity in a ROS- and TOP-IIβ-independent way, at least in the evaluated acute setting. However, it is important to mention that these findings only apply to the acute (16h) treatment period, and further research is warranted to delineate the therapeutic potential of DEXRA against vascular toxicity associated with longer-term repetitive DOX dosing.
Collapse
Affiliation(s)
- Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| | - Dustin N. Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| | - Kasper Favere
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Guido R. Y. De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Emeline M. Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Edegem, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
10
|
Bosman M, Krüger D, Van Assche C, Boen H, Neutel C, Favere K, Franssen C, Martinet W, Roth L, De Meyer GRY, Cillero-Pastor B, Delrue L, Heggermont W, Van Craenenbroeck EM, Guns PJ. Doxorubicin-induced cardiovascular toxicity: a longitudinal evaluation of functional and molecular markers. Cardiovasc Res 2023; 119:2579-2590. [PMID: 37625456 PMCID: PMC10676457 DOI: 10.1093/cvr/cvad136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 06/19/2023] [Accepted: 07/27/2023] [Indexed: 08/27/2023] Open
Abstract
AIMS Apart from cardiotoxicity, the chemotherapeutic doxorubicin (DOX) induces vascular toxicity, represented by arterial stiffness and endothelial dysfunction. Both parameters are of interest for cardiovascular risk stratification as they are independent predictors of future cardiovascular events in the general population. However, the time course of DOX-induced cardiovascular toxicity remains unclear. Moreover, current biomarkers for cardiovascular toxicity prove insufficient. Here, we longitudinally evaluated functional and molecular markers of DOX-induced cardiovascular toxicity in a murine model. Molecular markers were further validated in patient plasma. METHODS AND RESULTS DOX (4 mg/kg) or saline (vehicle) was administered intra-peritoneally to young, male mice weekly for 6 weeks. In vivo cardiovascular function and ex vivo arterial stiffness and vascular reactivity were evaluated at baseline, during DOX therapy (Weeks 2 and 4) and after therapy cessation (Weeks 6, 9, and 15). Left ventricular ejection fraction (LVEF) declined from Week 4 in the DOX group. DOX increased arterial stiffness in vivo and ex vivo at Week 2, which reverted thereafter. Importantly, DOX-induced arterial stiffness preceded reduced LVEF. Further, DOX impaired endothelium-dependent vasodilation at Weeks 2 and 6, which recovered at Weeks 9 and 15. Conversely, contraction with phenylephrine was consistently higher in the DOX-treated group. Furthermore, proteomic analysis on aortic tissue identified increased thrombospondin-1 (THBS1) and alpha-1-antichymotrypsin (SERPINA3) at Weeks 2 and 6. Up-regulated THBS1 and SERPINA3 persisted during follow-up. Finally, THBS1 and SERPINA3 were quantified in plasma of patients. Cancer survivors with anthracycline-induced cardiotoxicity (AICT; LVEF < 50%) showed elevated THBS1 and SERPINA3 levels compared with age-matched control patients (LVEF ≥ 60%). CONCLUSIONS DOX increased arterial stiffness and impaired endothelial function, which both preceded reduced LVEF. Vascular dysfunction restored after DOX therapy cessation, whereas cardiac dysfunction persisted. Further, we identified SERPINA3 and THBS1 as promising biomarkers of DOX-induced cardiovascular toxicity, which were confirmed in AICT patients.
Collapse
Affiliation(s)
- Matthias Bosman
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Dustin Krüger
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Charles Van Assche
- Research Group M4I—Imaging Mass Spectrometry (IMS); Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
| | - Hanne Boen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Cédric Neutel
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Kasper Favere
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Constantijn Franssen
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Wim Martinet
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Lynn Roth
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Guido R Y De Meyer
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| | - Berta Cillero-Pastor
- Research Group M4I—Imaging Mass Spectrometry (IMS); Faculty of Health, Medicine and Life Sciences, Maastricht MultiModal Molecular Imaging Institute, Maastricht University, Universiteitssingel 50, 6229 ER, Maastricht, The Netherlands
- Department of Cell Biology-Inspired Tissue Engineering, Institute for Technology-Inspired Regenerative Medicine, Universiteitssingel 40, 6229 ER Maastricht/Room C3.577, PO Box 616, Maastricht 6200 MD, The Netherlands
| | - Leen Delrue
- Department of Cardiology, Cardiovascular Center OLV Hospital Aalst, Moorselbaan 164, Aalst B-9300, Belgium
| | - Ward Heggermont
- Department of Cardiology, Cardiovascular Center OLV Hospital Aalst, Moorselbaan 164, Aalst B-9300, Belgium
- Department of Cardiology, Center for Molecular and Vascular Biology, KU Leuven, Herestraat 49, Leuven B-3000, Belgium
| | - Emeline M Van Craenenbroeck
- Research Group Cardiovascular Diseases, GENCOR, University of Antwerp, Antwerp B-2610, Belgium
- Department of Cardiology, Antwerp University Hospital (UZA), Drie Eikenstraat 655, Edegem B-2650, Belgium
| | - Pieter-Jan Guns
- Laboratory of Physiopharmacology, Faculty of Medicine and Health Sciences, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Campus Drie Eiken, University of Antwerp, Universiteitsplein 1, Antwerp B-2610, Belgium
| |
Collapse
|
11
|
Anastasiou M, Oikonomou E, Theofilis P, Papamikroulis GA, Gazouli M, Kalogeras K, Lygkoni S, Pesiridis T, Goliopoulou A, Papatheodoridi A, Psyrri A, Zagouri F, Siasos G, Tousoulis D. Prolonged impact of anti-cancer therapy on endothelial function and arterial stiffness in breast cancer patients. Vascul Pharmacol 2023; 152:107195. [PMID: 37455009 DOI: 10.1016/j.vph.2023.107195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 07/09/2023] [Accepted: 07/13/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND Cardiotoxicity restricts anthracycline and trastuzumab treatment of Human Epidermal Growth Factor Receptor 2 positive early breast cancer. Endothelial dysfunction and arteriosclerosis are significant cardiovascular risk factors. OBJECTIVES We studied the effect of anthracycline-based chemotherapy, with or without trastuzumab, on endothelium and arteriosclerosis in patients with breast cancer. METHODS In this case-control study, 52 women with breast cancer and 104 women without breast cancer were examined longitudinally up to 15 months following (in the breast cancer group) initiation of chemotherapy. Arterial stiffness was evaluated through pulse wave velocity (PWV), while endothelial function via flow-mediated dilatation (FMD) at baseline (T0), 3 (T1), 6 (T2), and 15 (T3) months later. RESULTS There was no difference between subjects with breast cancer and control in PWV and FMD at baseline. Longitudinally, participants with breast cancer exhibited considerable impairment of PWV and FMD compared to the control group (p for interaction <0.001 for both parameters). In breast cancer patients, there was a significant increase from T0 to T3 in PWV (7.43 ± 1.68 m/s vs. 8.18 ± 2.00 m/s, p = 0.01) and decrease in FMD (6.95 ± 2.86% vs. 5.03 ± 2.83%, p = 0.006). The addition of trastuzumab in the treatment did not have any effect on PWV (p = 0.74) or FMD (p = 0.91). CONCLUSIONS In patients with breast cancer, there is progression of endothelial dysfunction and arteriosclerosis up to 15 months following initiation of anthracycline-based chemotherapy. Trastuzumab has no additive effect on endothelial function or arterial stiffness.
Collapse
Affiliation(s)
- Maria Anastasiou
- Section of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Evangelos Oikonomou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece.
| | - Panagiotis Theofilis
- 1st Cardiology Department, "Hippokration" General Hospital, Medical School, University of Athens, 11527 Athens, Greece
| | - George Angelos Papamikroulis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Maria Gazouli
- Laboratory of Biology, Department of Basic Medical Sciences, Medical School, National and Kapodistrian University of Athens, Greece
| | - Konstantinos Kalogeras
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Stavroula Lygkoni
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Theodoros Pesiridis
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Athina Goliopoulou
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Alkistis Papatheodoridi
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, 11528 Athens, Greece
| | - Amanda Psyrri
- Section of Medical Oncology, Department of Internal Medicine, Faculty of Medicine, National and Kapodistrian University of Athens, Attikon University Hospital, Athens, Greece
| | - Flora Zagouri
- Department of Clinical Therapeutics, Alexandra Hospital, National and Kapodistrian University of Athens School of Medicine, 11528 Athens, Greece
| | - Gerasimos Siasos
- 3rd Department of Cardiology, Sotiria Chest Disease Hospital, National and Kapodistrian University of Athens, Medical School, Athens, Greece
| | - Dimitris Tousoulis
- 1st Cardiology Department, "Hippokration" General Hospital, Medical School, University of Athens, 11527 Athens, Greece
| |
Collapse
|
12
|
Suddala S, O’Connell N, D’Agostino R, Ladd A, Lucas AR, Jordan JH, Salloum FN, Ky B, Hackney MH, Hundley WG. Reduced Thoracic Aortic Distensibility Upon Breast Cancer Diagnosis (WF-97415 and WF-98213). JACC CardioOncol 2023; 5:704-706. [PMID: 37969653 PMCID: PMC10635869 DOI: 10.1016/j.jaccao.2022.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2023] Open
Affiliation(s)
| | | | | | | | | | | | | | | | | | - W. Gregory Hundley
- Virginia Commonwealth University School of Medicine, PO Box 980335, Richmond, Virginia 23298-0335, USA @VCUHealthHeart@oscardneufville@greghundleymd
| |
Collapse
|
13
|
Climie RE, Dillon HT, Horne-Okano Y, Wallace I, Avery S, Kingwell BA, La Gerche A, Howden EJ. Vascular Aging Is Accelerated in Hematological Cancer Survivors Who Undergo Allogeneic Stem Cell Transplant. Hypertension 2023; 80:1881-1889. [PMID: 37476996 DOI: 10.1161/hypertensionaha.123.21115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 07/05/2023] [Indexed: 07/22/2023]
Abstract
BACKGROUND Survivors of allogeneic stem cell transplant (SCT) receive intensive cancer treatments that are associated with cardiovascular dysfunction. Markers of vascular age can indicate early signs of adverse (cardio)vascular changes; however, the impact of SCT on these makers is unknown. We aimed to determine the short (3 months) and longer-term (≥2 years) effect of SCT on markers of vascular age in hematologic cancer survivors compared with an age-matched noncancer control group. METHODS The short-term effects of SCT, markers of vascular age (aortic compliance, arterial elastance, and ventricular-vascular coupling) were assessed via cardiac magnetic resonance imaging (cardiac and aortic volumes) before and ≈3 months post-SCT in 13 short-term survivors and compared with 11 controls. The longer-term impact was assessed by comparing 14 long-term survivors (6.5 [2-20] years post-SCT) to the short-term survivors (post-SCT) and controls (n=16). RESULTS The groups were similar for age and body mass index. In the short-term survivors, no significant group-by-time interactions were observed for any markers of vascular aging from pretransplant to posttransplant (net difference for change in compliance between groups -0.07 [95% CI, -1.49 to 1.35]). For the time-course analysis, aortic compliance was significantly lower in both SCT groups (overall P=0.007) compared with controls, whereas ventricular-vascular coupling was higher in both survivor groups as was arterial elastance in long-term SCT survivors (ie, worse; P<0.01 for all). CONCLUSION This study provides evidence of an accelerated vascular aging phenotype in allogeneic SCT survivors and provides insight into the increased burden of cardiovascular disease among hematologic cancer survivors.
Collapse
Affiliation(s)
- Rachel E Climie
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Australia (R.E.C.)
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Australia (R.E.C., H.T.S., Y.H.-O., I.W., A.L.G., E.J.H.)
| | - Hayley T Dillon
- Institute of Physical Activity and Nutrition, School of Exercise and Nutrition Sciences, Deakin University, Geelong, Australia (H.T.D.)
| | - Yuki Horne-Okano
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Australia (R.E.C., H.T.S., Y.H.-O., I.W., A.L.G., E.J.H.)
| | - Imogen Wallace
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Australia (R.E.C., H.T.S., Y.H.-O., I.W., A.L.G., E.J.H.)
| | - Sharon Avery
- Malignant Haematology and Stem Cell Transplantation Service, Alfred Hospital, Melbourne, Australia (S.A.)
| | | | - Andre La Gerche
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Australia (R.E.C., H.T.S., Y.H.-O., I.W., A.L.G., E.J.H.)
- Cardiology Department, St Vincent's Hospital Melbourne, Fitzroy, Australia (A.L.G.)
| | - Erin J Howden
- Clinical Research Domain, Baker Heart and Diabetes Institute, Melbourne, Australia (R.E.C., H.T.S., Y.H.-O., I.W., A.L.G., E.J.H.)
| |
Collapse
|
14
|
Maurer GS, Clayton ZS. Anthracycline chemotherapy, vascular dysfunction and cognitive impairment: burgeoning topics and future directions. Future Cardiol 2023; 19:547-566. [PMID: 36354315 PMCID: PMC10599408 DOI: 10.2217/fca-2022-0086] [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: 09/09/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Anthracyclines, chemotherapeutic agents used to treat common forms of cancer, increase cardiovascular (CV) complications, thereby necessitating research regarding interventions to improve the health of cancer survivors. Vascular dysfunction, which is induced by anthracycline chemotherapy, is an established antecedent to overt CV diseases. Potential treatment options for ameliorating vascular dysfunction have largely been understudied. Furthermore, patients treated with anthracyclines have impaired cognitive function and vascular dysfunction is an independent risk factor for the development of mild cognitive impairment. Here, we will focus on: anthracycline chemotherapy associated CV diseases risk; how targeting mechanisms underlying vascular dysfunction may be a means to improve both CV and cognitive health; and research gaps and potential future directions for the field of cardio-oncology.
Collapse
Affiliation(s)
- Grace S Maurer
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
15
|
Lucas AR, D’Agostino R, Jordan JH, Reding KW, O’Connell NS, Melendez GC, Salloum FN, Weaver KE, Lesser GJ, Hundley WG. Atorvastatin Does Not Attenuate Aortic Stiffening After Doxorubicin for Breast Cancer and Lymphoma (PREVENT-WF-98213). JACC CardioOncol 2023; 5:412-414. [PMID: 37397089 PMCID: PMC10308033 DOI: 10.1016/j.jaccao.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/04/2023] Open
Affiliation(s)
- Alexander R. Lucas
- VCU Pauley Heart Center, Division of Cardiology, Virginia Commonwealth University, 830 East Main Street, Richmond, Virginia 23235, USA @oscardnfville
| | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Bottinor W, Hundley WG. Does Physical Activity During Cancer Treatment Preserve Exercise Capacity? Circulation 2023; 147:546-548. [PMID: 36780389 DOI: 10.1161/circulationaha.122.063192] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Affiliation(s)
- Wendy Bottinor
- Department of Internal Medicine, Division of Cardiovascular Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond
| | - W Gregory Hundley
- Department of Internal Medicine, Division of Cardiovascular Medicine, Pauley Heart Center, Virginia Commonwealth University, Richmond
| |
Collapse
|
17
|
Clayton ZS, Ade CJ, Dieli-Conwright CM, Mathelier HM. A bench to bedside perspective on anthracycline chemotherapy-mediated cardiovascular dysfunction: challenges and opportunities. A symposium review. J Appl Physiol (1985) 2022; 133:1415-1429. [PMID: 36302155 PMCID: PMC9762976 DOI: 10.1152/japplphysiol.00471.2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 01/11/2023] Open
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide and the risk of developing CVD is markedly increased following anthracycline chemotherapy treatment. Anthracyclines are an essential component of the cancer treatment regimen used for common forms of cancer in male and female children, adolescents, young adults, and older adults. Increased CVD risk with anthracyclines occurs, in part, due to vascular dysfunction-impaired endothelial function and arterial stiffening. These features of vascular dysfunction also play a major role in other common disorders observed following anthracycline treatment, including chronic kidney disease, dementia, and exercise intolerance. However, the mechanisms by which anthracycline chemotherapy induces and sustains vascular dysfunction are incompletely understood. This budding area of biomedical research is termed cardio-oncology, which presents the unique opportunity for collaboration between physicians and basic scientists. This symposium, presented at Experimental Biology 2022, provided a timely update on this important biomedical research topic. The speakers presented observations made at levels from cells to mice to humans treated with anthracycline chemotherapeutic agents using an array of translational research approaches. The speaker panel included a diverse mix of female and male investigators and unique insight from a cardio-oncology physician-scientist. Particular emphasis was placed on challenges and opportunities in this field as well as mechanisms that could be viewed as therapeutic targets leading to novel treatment strategies.
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, Colorado
| | - Carl J Ade
- Department of Kinesiology, Kansas State University, Manhattan, Kansas
| | - Christina M Dieli-Conwright
- Division of Population Sciences, Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Hansie M Mathelier
- Penn Medicine, University of Pennsylvania Health System, Philadelphia, Pennsylvania
| |
Collapse
|
18
|
Harries I, Biglino G, Ford K, Nelson M, Rego G, Srivastava P, Williams M, Berlot B, De Garate E, Baritussio A, Liang K, Baquedano M, Chavda N, Lawton C, Shearn A, Otton S, Lowry L, Nightingale AK, Carlos Plana J, Marks D, Emanueli C, Bucciarelli-Ducci C. Prospective multiparametric CMR characterization and MicroRNA profiling of anthracycline cardiotoxicity: A pilot translational study. IJC HEART & VASCULATURE 2022; 43:101134. [PMID: 36389268 PMCID: PMC9647504 DOI: 10.1016/j.ijcha.2022.101134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/14/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022]
Abstract
Background Anthracycline cardiotoxicity is a significant clinical challenge. Biomarkers to improve risk stratification and identify early cardiac injury are required. Objectives The purpose of this pilot study was to prospectively characterize anthracycline cardiotoxicity using cardiovascular magnetic resonance (CMR), echocardiography and MicroRNAs (MiRNAs), and identify baseline predictors of LVEF recovery. Methods Twenty-four patients (age 56 range 18-75 years; 42 % female) with haematological malignancy scheduled to receive anthracycline chemotherapy (median dose 272 mg/m2 doxorubicin equivalent) were recruited and evaluated at three timepoints (baseline, completion of chemotherapy, and 6 months after completion of chemotherapy) with multiparametric 1.5 T CMR, echocardiography and circulating miRNAs sequencing. Results Seventeen complete datasets were obtained. CMR left ventricular ejection fraction (LVEF) fell significantly between baseline and completion of chemotherapy (61 ± 3 vs 53 ± 3 %, p < 0.001), before recovering significantly at 6-month follow-up (55 ± 3 %, p = 0.018). Similar results were observed for 3D echocardiography-derived LVEF and CMR-derived longitudinal, circumferential and radial feature-tracking strain. Patients were divided into tertiles according to LVEF recovery (poor recovery, partial recovery, good recovery). CMR-derived mitral annular plane systolic excursion (MAPSE) was significantly different at baseline in patients exhibiting poor LVEF recovery (11.7 ± 1.5 mm) in comparison to partial recovery (13.7 ± 2.7 mm), and good recovery (15.7 ± 3.1 mm; p = 0.028). Furthermore, baseline miRNA-181-5p and miRNA-221-3p expression were significantly higher in this group. T2 mapping increased significantly on completion of chemotherapy compared to baseline (54.0 ± 4.6 to 57.8 ± 4.9 ms, p = 0.001), but was not predictive of LVEF recovery. No changes to LV mass, extracellular volume fraction, T1 mapping or late gadolinium enhancement were observed. Conclusions Baseline CMR-derived MAPSE, circulating miRNA-181-5p, and miRNA-221-3p were associated with poor recovery of LVEF 6 months after completion of anthracycline chemotherapy, suggesting their potential predictive role in this context. T2 mapping increased significantly on completion of chemotherapy but was not predictive of LVEF recovery.
Collapse
Key Words
- CMR, cardiovascular magnetic resonance
- Cancer therapeutics-related cardiac dysfunction
- Cardio-oncology
- Cardiovascular magnetic resonance
- ECV, extracellular volume
- LAVi, left atrial volume indexed
- LGE, late gadolinium enhancement
- LV, left ventricle
- LVEF, left ventricular ejection fraction
- MAPSE, mitral annular plane systolic excursion
- MiRNAs, MicroRNAs
- iLVEDV, left ventricular end-diastolic volume indexed
- iLVESV, indexed left ventricular end-systolic volume indexed
Collapse
Affiliation(s)
- Iwan Harries
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Giovanni Biglino
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
- Myocardial Function – National Heart and Lung Institute, Imperial College London, London, UK
- NIHR Bristol Biomedical Research Centre, Bristol Heart Institute, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Kerrie Ford
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Martin Nelson
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Gui Rego
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Prashant Srivastava
- Myocardial Function – National Heart and Lung Institute, Imperial College London, London, UK
| | - Matthew Williams
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Bostjan Berlot
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Estefania De Garate
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Anna Baritussio
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Kate Liang
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Mai Baquedano
- NIHR Bristol Biomedical Research Centre, Bristol Heart Institute, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Nikesh Chavda
- Bristol Heamatology and Oncology Centre, University Hospitals Bristol NHS Trust, Bristol United Kingdom, UK
| | - Christopher Lawton
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | - Andrew Shearn
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | | | | | - Angus K. Nightingale
- Bristol Heart Institute, Bristol Medical School, University Hospitals Bristol, Bristol, UK
| | | | - David Marks
- Bristol Heamatology and Oncology Centre, University Hospitals Bristol NHS Trust, Bristol United Kingdom, UK
| | - Costanza Emanueli
- Myocardial Function – National Heart and Lung Institute, Imperial College London, London, UK
- NIHR Bristol Biomedical Research Centre, Bristol Heart Institute, University Hospitals Bristol NHS Foundation Trust, Bristol, UK
| | - Chiara Bucciarelli-Ducci
- Royal Brompton and Harefield Hospitals, Guys’ and St Thomas NHS Foundation Trust, London
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, Kings College, London
| |
Collapse
|
19
|
Schneider C, González-Jaramillo N, Marcin T, Campbell KL, Suter T, Bano A, Wilhelm M, Eser P. Time-Dependent Effect of Anthracycline-Based Chemotherapy on Central Arterial Stiffness: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2022; 9:873898. [PMID: 35865379 PMCID: PMC9295862 DOI: 10.3389/fcvm.2022.873898] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/13/2022] [Indexed: 11/23/2022] Open
Abstract
Background and Aims Anthracycline-based chemotherapy (ANTH-BC) has been proposed to increase arterial stiffness, however, the time-dependency of these effects remain unclear. This systematic review and meta-analysis aimed to investigate the time-dependent effect of ANTH-BC on markers of central aortic stiffness, namely aortic distensibility (AD) and pulse-wave-velocity (PWV) in cancer patients. Methods An extensive literature search without language restrictions was performed to identify all studies presenting longitudinal data on the effect of ANTH-BC on either AD and/or central PWV in cancer patients of all ages. An inverse-variance weighted random-effect model was performed with differences from before to after chemotherapy, as well as for short vs. mid-term effects. Results Of 2,130 articles identified, 9 observational studies with a total of 535 patients (mean age 52 ± 11; 73% women) were included, of which four studies measured AD and seven PWV. Short-term (2–4 months), there was a clinically meaningful increase in arterial stiffness, namely an increase in PWV of 2.05 m/s (95% CI 0.68–3.43) and a decrease in AD (albeit non-significant) of −1.49 mmHg-1 (−3.25 to 0.27) but a smaller effect was observed mid-term (6–12 months) for PWV of 0.88 m/s (−0.25 to 2.02) and AD of −0.37 mmHg-1 (−1.13 to 0.39). There was considerable heterogeneity among the studies. Conclusions Results from this analysis suggest that in the short-term, ANTH-BC increases arterial stiffness, but that these changes may partly be reversible after therapy termination. Future studies need to elucidate the long-term consequences of ANTH-BC on arterial stiffness, by performing repeated follow-up measurements after ANTH-BC termination. Systematic Review Registration [www.crd.york.ac.uk/prospero/], identifier [CRD42019141837].
Collapse
Affiliation(s)
- Caroline Schneider
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
| | - Nathalia González-Jaramillo
- Graduate School for Health Sciences, University of Bern, Bern, Switzerland
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Thimo Marcin
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | | | - Thomas Suter
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Arjola Bano
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Institute of Social and Preventive Medicine (ISPM), University of Bern, Bern, Switzerland
| | - Matthias Wilhelm
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Prisca Eser
- Department of Cardiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- *Correspondence: Prisca Eser,
| |
Collapse
|
20
|
Rahimi O, Melo AC, Westwood B, Grier RDM, Tallant EA, Gallagher PE. Angiotensin-(1-7) reduces doxorubicin-induced aortic arch dysfunction in male and female juvenile Sprague Dawley rats through pleiotropic mechanisms. Peptides 2022; 152:170784. [PMID: 35288251 DOI: 10.1016/j.peptides.2022.170784] [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: 11/30/2021] [Revised: 03/08/2022] [Accepted: 03/09/2022] [Indexed: 11/28/2022]
Abstract
Doxorubicin (Dox), an effective chemotherapeutic, can cause cumulative dose-dependent cardiovascular toxicity, which may manifest as vascular dysfunction leading to long-term end-organ damage. Currently, there are no effective treatments to mitigate Dox-induced vascular damage in cancer patients, particularly pediatric patients. We showed that angiotensin-(1-7) [Ang-(1-7)], an endogenous peptide hormone, mitigated cardiac damage in Dox-treated juvenile rats. In this study assessing aortic stiffness, juvenile male and female rats were administered a clinically equivalent dose of Dox (21-24 mg/kg) over 6 weeks, in the presence and absence of Ang-(1-7) [24 µg/kg/h]. Aortic function was measured using echocardiography. Ang-(1-7) reduced the Dox-mediated increase in pulse wave velocity, a measure of arterial stiffness (males: p < 0.05; females: p < 0.001) as compared in control animals. Dox decreased aortic lumen diameter (p < 0.0001) and increased wall thickness (p < 0.01) in males, which was attenuated by Ang-(1-7). In male but not female aortic arches, Dox increased media hypertrophy (p < 0.05) and reduced elastin content (p < 0.001), which were prevented by Ang-(1-7). Conversely, Dox increased fibrosis (p < 0.0001) in juvenile female rats, which was reduced by Ang-(1-7). Adjunct Ang-(1-7) prevented the Dox-induced increase in total cell and nuclear pERK1/2 in the aortic intima and media of male rats and nuclear pSMAD2 in the intimal and medial regions of the aortic arches of both sexes. These results demonstrate that Ang-(1-7) attenuated Dox-induced aortic dysfunction in both sexes of juvenile rats, albeit through different mechanisms, suggesting that Ang-(1-7) may serve as an effective adjuvant to ameliorate cardiovascular and long-term end-organ damage in pediatric patients produced by anthracyclines.
Collapse
Affiliation(s)
- Omeed Rahimi
- Surgery/Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Ana Clara Melo
- Surgery/Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Brian Westwood
- Surgery/Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Rui D M Grier
- Surgery/Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - E Ann Tallant
- Surgery/Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Patricia E Gallagher
- Surgery/Hypertension and Vascular Research, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| |
Collapse
|
21
|
Diao Y, Liu Z, Chen L, Zhang W, Sun D. The Relationship Between Cancer and Functional and Structural Markers of Subclinical Atherosclerosis: A Systematic Review and Meta-Analysis. Front Cardiovasc Med 2022; 9:849538. [PMID: 35600484 PMCID: PMC9115552 DOI: 10.3389/fcvm.2022.849538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2022] [Accepted: 04/11/2022] [Indexed: 12/04/2022] Open
Abstract
Objectives The relationship between cancer and subclinical atherosclerosis has always been the focus of people's attention. We conducted a systematic review and meta-analysis by evaluating the effects of cancer on functional and structural markers of subclinical atherosclerosis:intima-media thickness (IMT), pulse wave velocity (PWV), and flow-mediated vasodilation (FMD). Methods A comprehensive and systematic literature search was conducted on the internet. Sensitivity analysis, publication bias, standard mean difference (SMD), corresponding 95% confidence interval (95% CI), and subgroup analysis were performed for all relevant research indicators in the retrieved literature. Results Forty-six studies were included, including 3,729 cancer patients and 2,404 healthy controls. Cancer patients had significantly thicker IMT [SMD (95%CI) = 0.290 (0.069 to 0.511), P = 0.010] and higher PWV [SMD (95%CI) = 0.392 (0.136 to 0.647), P = 0.003] compared with healthy controls. There was no significant difference in FMD [SMD (95% CI) = −0.192 (−0.527 to 0.144), P > 0.05). After subgrouping by age, male proportion, and treatment, the analysis results of IMT ≥ 50 years old, PWV and FMD < 50 years old, male proportion ≥50%, chemotherapy group, IMT and PWV radiotherapy group, and PWV endocrine therapy group were statistically significant (P < 0.05). There were no significant differences in other subgroup analyses, overall sensitivity analysis, and publication bias (p < 0.05). Conclusions Cancer may promote subclinical atherosclerosis, and change the functional and structural markers of subclinical atherosclerosis such as IMT and PWV. Early intervention and prevention should be pursued.
Collapse
|
22
|
Wang X, Tan Y, Liu D, Shen H, Deng Y, Tan Y, Wang L, Zhang Y, Ma X, Zeng X, Zhang J. Chemotherapy-associated steatohepatitis was concomitant with epicardial adipose tissue volume increasing in breast cancer patients who received neoadjuvant chemotherapy. Eur Radiol 2022; 32:4898-4908. [PMID: 35394181 DOI: 10.1007/s00330-022-08581-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/08/2022] [Accepted: 01/12/2022] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To investigate the prevalence of chemotherapy-associated steatohepatitis, quantitate the epicardial adipose tissue (EAT) volume in breast cancer patients, and explore the mediating effect of liver fat content on EAT volume in breast cancer patients who received neoadjuvant chemotherapy (NAC). METHODS From October 2018 to April 2020, patients were retrospectively reviewed and divided into breast cancer non-NAC and NAC groups. The prevalence of chemotherapy-associated steatohepatitis was evaluated through quantitative MRI mDIXON-Quant examinations by using defined proton density fat fraction cutoffs of liver fat. The EAT volume was quantified on chest CT by semi-automatic volume analysis software. Bootstrap analysis was used in the breast cancer NAC group to test the significance of the mediating effect of liver fat content on EAT volume. RESULTS A total of 662 breast cancer patients (non-NAC group: 445 patients; NAC group: 217 patients) were included. The prevalence of chemotherapy-associated steatohepatitis in the NAC group was significantly higher than the prevalence of hepatic steatosis in the non-NAC group (42.8% vs. 33.3%, p < 0.001). EAT volume was measured in 561 of 662 breast cancer patients, and was significantly higher in the NAC group than in the non-NAC group (137.26 ± 53.48 mL vs. 125.14 ± 58.77 mL, p = 0.020). In the breast cancer NAC group, the indirect effect of liver fat content on EAT volume was 2.545 (p < 0.001), and the contribution rate to the effect was 69.1%. CONCLUSIONS EAT volume was significantly higher in the BC-NAC group than in the BC-non-NAC group. KEY POINTS • The prevalence of CASH was as high as 42.8% in BC patients. • NAC significantly increased the EAT volume in BC patients. • The liver fat content caused the change of EAT volume through mediating effect.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Yuchuan Tan
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Daihong Liu
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Hesong Shen
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Yongchun Deng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400030, People's Republic of China
| | - Yong Tan
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Lei Wang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Yipeng Zhang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Xin Ma
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China
| | - Xiaohua Zeng
- Department of Breast Cancer Center, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, Chongqing, 400030, People's Republic of China.
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, School of Medicine, Chongqing University, No.181 Hanyu Road, Shapingba District, Chongqing, 400030, People's Republic of China.
| |
Collapse
|
23
|
Cadour F, Thuny F, Sourdon J. New Insights in Early Detection of Anticancer Drug-Related Cardiotoxicity Using Perfusion and Metabolic Imaging. Front Cardiovasc Med 2022; 9:813883. [PMID: 35198613 PMCID: PMC8858802 DOI: 10.3389/fcvm.2022.813883] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/06/2022] [Indexed: 12/16/2022] Open
Abstract
Cardio-oncology requires a good knowledge of the cardiotoxicity of anticancer drugs, their mechanisms, and their diagnosis for better management. Anthracyclines, anti-vascular endothelial growth factor (VEGF), alkylating agents, antimetabolites, anti-human epidermal growth factor receptor (HER), and receptor tyrosine kinase inhibitors (RTKi) are therapeutics whose cardiotoxicity involves several mechanisms at the cellular and subcellular levels. Current guidelines for anticancer drugs cardiotoxicity are essentially based on monitoring left ventricle ejection fraction (LVEF). However, knowledge of microvascular and metabolic dysfunction allows for better imaging assessment before overt LVEF impairment. Early detection of anticancer drug-related cardiotoxicity would therefore advance the prevention and patient care. In this review, we provide a comprehensive overview of the cardiotoxic effects of anticancer drugs and describe myocardial perfusion, metabolic, and mitochondrial function imaging approaches to detect them before over LVEF impairment.
Collapse
Affiliation(s)
- Farah Cadour
- Aix-Marseille Université, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
| | - Franck Thuny
- Aix-Marseille University, University Mediterranean Center of Cardio-Oncology, Unit of Heart Failure and Valvular Heart Diseases, Department of Cardiology, North Hospital, Assistance Publique - Hôpitaux de Marseille, Centre for CardioVascular and Nutrition Research (C2VN), Inserm 1263, Inrae 1260, Marseille, France
| | - Joevin Sourdon
- Aix-Marseille Université, CNRS, CRMBM, Marseille, France
- APHM, Hôpital Universitaire Timone, CEMEREM, Marseille, France
- *Correspondence: Joevin Sourdon
| |
Collapse
|
24
|
Endothelial Dysfunction in Childhood Cancer Survivors: A Narrative Review. Life (Basel) 2021; 12:life12010045. [PMID: 35054438 PMCID: PMC8780257 DOI: 10.3390/life12010045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/08/2021] [Accepted: 12/16/2021] [Indexed: 12/14/2022] Open
Abstract
Assessment of endothelial dysfunction in cancer survivors may have a role in the early identification of non-communicable diseases and cardiovascular late effects. Oncological therapies may impair endothelial function. Therefore, in patients such as childhood cancer survivors who could benefit from early cardioprotective pharmacological interventions, it is essential to monitor endothelial function, even if the optimal methodology for investigating the multifaceted aspects of endothelial dysfunction is still under debate. Biochemical markers, as well as invasive and non-invasive tools with and without pharmacological stimuli have been studied. Human clinical studies that have examined lifestyle or cancer treatment protocols have yielded evidence showing the involvement of lipid and lipoprotein levels, glycemic control, blood pressure, adiposity, inflammation, and oxidative stress markers on the state of endothelial health and its role as an early indicator of cardiometabolic risk. However, with regards to pharmacological interventions, cautious interpretation of the result attained whilst monitoring the endothelial function is warranted due to methodological limitations and substantial heterogeneity of the results reported in the published studies. In this narrative review, an overview of evidence from human clinical trials examining the effects of cancer therapies on endothelial disease is provided together with a discussion of endothelial function assessment using the different non-invasive techniques available for researchers and clinicians, in recent years.
Collapse
|
25
|
Doxorubicin Impairs Smooth Muscle Cell Contraction: Novel Insights in Vascular Toxicity. Int J Mol Sci 2021; 22:ijms222312812. [PMID: 34884612 PMCID: PMC8657832 DOI: 10.3390/ijms222312812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 11/19/2021] [Accepted: 11/23/2021] [Indexed: 01/12/2023] Open
Abstract
Clinical and animal studies have demonstrated that chemotherapeutic doxorubicin (DOX) increases arterial stiffness, a predictor of cardiovascular risk. Despite consensus about DOX-impaired endothelium-dependent vasodilation as a contributing mechanism, some studies have reported conflicting results on vascular smooth muscle cell (VSMC) function after DOX treatment. The present study aimed to investigate the effects of DOX on VSMC function. To this end, mice received a single injection of 4 mg DOX/kg, or mouse aortic segments were treated ex vivo with 1 μM DOX, followed by vascular reactivity evaluation 16 h later. Phenylephrine (PE)-induced VSMC contraction was decreased after DOX treatment. DOX did not affect the transient PE contraction dependent on Ca2+ release from the sarcoplasmic reticulum (0 mM Ca2+), but it reduced the subsequent tonic phase characterised by Ca2+ influx. These findings were supported by similar angiotensin II and attenuated endothelin-1 contractions. The involvement of voltage-gated Ca2+ channels in DOX-decreased contraction was excluded by using levcromakalim and diltiazem in PE-induced contraction and corroborated by similar K+ and serotonin contractions. Despite the evaluation of multiple blockers of transient receptor potential channels, the exact mechanism for DOX-decreased VSMC contraction remains elusive. Surprisingly, DOX reduced ex vivo but not in vivo arterial stiffness, highlighting the importance of appropriate timing for evaluating arterial stiffness in DOX-treated patients.
Collapse
|
26
|
Kotla S, Zhang A, Imanishi M, Ko KA, Lin SH, Gi YJ, Moczygemba M, Isgandarova S, Schadler KL, Chung C, Milgrom SA, Banchs J, Yusuf SW, Amaya DN, Guo H, Thomas TN, Shen YH, Deswal A, Herrmann J, Kleinerman ES, Entman ML, Cooke JP, Schifitto G, Maggirwar SB, McBeath E, Gupte AA, Krishnan S, Patel ZS, Yoon Y, Burks JK, Fujiwara K, Brookes PS, Le NT, Hamilton DJ, Abe JI. Nucleus-mitochondria positive feedback loop formed by ERK5 S496 phosphorylation-mediated poly (ADP-ribose) polymerase activation provokes persistent pro-inflammatory senescent phenotype and accelerates coronary atherosclerosis after chemo-radiation. Redox Biol 2021; 47:102132. [PMID: 34619528 PMCID: PMC8502954 DOI: 10.1016/j.redox.2021.102132] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/08/2021] [Accepted: 09/11/2021] [Indexed: 02/08/2023] Open
Abstract
The incidence of cardiovascular disease (CVD) is higher in cancer survivors than in the general population. Several cancer treatments are recognized as risk factors for CVD, but specific therapies are unavailable. Many cancer treatments activate shared signaling events, which reprogram myeloid cells (MCs) towards persistent senescence-associated secretory phenotype (SASP) and consequently CVD, but the exact mechanisms remain unclear. This study aimed to provide mechanistic insights and potential treatments by investigating how chemo-radiation can induce persistent SASP. We generated ERK5 S496A knock-in mice and determined SASP in myeloid cells (MCs) by evaluating their efferocytotic ability, antioxidation-related molecule expression, telomere length, and inflammatory gene expression. Candidate SASP inducers were identified by high-throughput screening, using the ERK5 transcriptional activity reporter cell system. Various chemotherapy agents and ionizing radiation (IR) up-regulated p90RSK-mediated ERK5 S496 phosphorylation. Doxorubicin and IR caused metabolic changes with nicotinamide adenine dinucleotide depletion and ensuing mitochondrial stunning (reversible mitochondria dysfunction without showing any cell death under ATP depletion) via p90RSK-ERK5 modulation and poly (ADP-ribose) polymerase (PARP) activation, which formed a nucleus-mitochondria positive feedback loop. This feedback loop reprogramed MCs to induce a sustained SASP state, and ultimately primed MCs to be more sensitive to reactive oxygen species. This priming was also detected in circulating monocytes from cancer patients after IR. When PARP activity was transiently inhibited at the time of IR, mitochondrial stunning, priming, macrophage infiltration, and coronary atherosclerosis were all eradicated. The p90RSK-ERK5 module plays a crucial role in SASP-mediated mitochondrial stunning via regulating PARP activation. Our data show for the first time that the nucleus-mitochondria positive feedback loop formed by p90RSK-ERK5 S496 phosphorylation-mediated PARP activation plays a crucial role of persistent SASP state, and also provide preclinical evidence supporting that transient inhibition of PARP activation only at the time of radiation therapy can prevent future CVD in cancer survivors.
Collapse
Affiliation(s)
- Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Aijun Zhang
- Department of Medicine, Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Masaki Imanishi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kyung Ae Ko
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Young Jin Gi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Margie Moczygemba
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Sevinj Isgandarova
- Center for Infectious and Inflammatory Diseases, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Keri L Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caroline Chung
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sarah A Milgrom
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Radiation Oncology, University of Colorado Cancer Center, Aurora, CO, 80045, USA
| | - Jose Banchs
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Diana N Amaya
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Huifang Guo
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tamlyn N Thomas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ying H Shen
- Division of Cardiothoracic Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, MN, USA
| | - Eugenie S Kleinerman
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mark L Entman
- Division of Cardiovascular Sciences, Baylor College of Medicine, Houston, TX, USA
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | | | - Sanjay B Maggirwar
- Department of Microbiology, Immunology, and Tropical Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Elena McBeath
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Endocrine Neoplasia and Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Anisha A Gupte
- Department of Medicine, Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Sunil Krishnan
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Radiation Oncology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | | | - Yisang Yoon
- Department of Physiology, Medical College of Georgia, Augusta, GA, USA
| | - Jared K Burks
- Department of Leukemia, Division of Center Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Keigi Fujiwara
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul S Brookes
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, Rochester, NY, USA
| | - Nhat-Tu Le
- Division of Cardiovascular Sciences, Baylor College of Medicine, Houston, TX, USA
| | - Dale J Hamilton
- Department of Medicine, Center for Bioenergetics, Houston Methodist Research Institute, Houston, TX, USA
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
27
|
Perturbations of Adjuvant Chemotherapy on Cardiovascular Responses and Exercise Tolerance in Patients with Early-Stage Breast Cancer. BIOLOGY 2021; 10:biology10090910. [PMID: 34571786 PMCID: PMC8472454 DOI: 10.3390/biology10090910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 09/06/2021] [Accepted: 09/13/2021] [Indexed: 11/29/2022]
Abstract
Simple Summary The present study aimed to assess and compare the effects of receiving CAF (cyclophosphamide/doxorubicin/fluorouracil) and AC-T (doxorubicin/cyclophosphamide→taxanes) on exercise tolerance and cardiovascular responses in patients with early-stage breast cancer. We herein demonstrated that AC-T chemotherapy increased resting heart rate (RHR) and induced a greater reduction in exercise tolerance at the end of chemotherapy compared with CAF. Moreover, AC-T also lowered myocardial perfusion more than CAF, and it appeared that myocardial impairment occurred before the development of arterial stiffening after chemotherapy. We, therefore, suggest that AC-T chemotherapy might further limit the exercise capacity of patients with early-stage breast cancer. This study provides fundamental information regarding the variety of cardiovascular responses to exercise after chemotherapy in patients with early-stage breast cancer. This information will help clinical professionals in the fields of oncological and rehabilitation medicine to precisely prescribe post-chemotherapy exercise programs when patients are receiving different chemotherapies. Abstract Background: Adjuvant chemotherapies are commonly used for treating early-stage breast cancer. However, whether chemotherapeutic regimens affect exercise tolerance and cardiovascular responses remains unclear. Therefore, we investigated the effects of receiving CAF and AC-T on exercise tolerance and cardiovascular responses in patients with early-stage breast cancer. Methods: Thirty-four patients with breast cancer (age: 44 ± 1 years; stage I-II) received either CAF (n = 15) or AC-T (n = 19), depending on clinical decisions. Their step-exercise tolerance and cardiovascular responses were assessed before and after chemotherapy. Results: After chemotherapy, there were no differences in baseline measurements between patients receiving CAF or AC-T. The increases in resting heart rate (RHR) of those receiving AC-T was significantly greater than that of those receiving CAF. CAF and AC-T did not result in increased pulse wave velocity (PWV), yet the subendocardial viability ratio (SEVR) in patients receiving AC-T was significantly lower than the baseline. Greater change in post-exercise heart rate recovery (recovery HR) after chemotherapy was observed in those who had received AC-T; the Recovery HR in AC-T patients was significantly higher during post-exercise period than that in CAF patients. Conclusions: AC-T chemotherapy increases RHR and impairs exercise tolerance after chemotherapy more than CAF. Moreover, AC-T also lowers myocardial perfusion more than CAF after chemotherapy.
Collapse
|
28
|
Sawicki KT, Sala V, Prever L, Hirsch E, Ardehali H, Ghigo A. Preventing and Treating Anthracycline Cardiotoxicity: New Insights. Annu Rev Pharmacol Toxicol 2021; 61:309-332. [PMID: 33022184 DOI: 10.1146/annurev-pharmtox-030620-104842] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Anthracyclines are the cornerstone of many chemotherapy regimens for a variety of cancers. Unfortunately, their use is limited by a cumulative dose-dependent cardiotoxicity. Despite more than five decades of research, the biological mechanisms underlying anthracycline cardiotoxicity are not completely understood. In this review, we discuss the incidence, risk factors, types, and pathophysiology of anthracycline cardiotoxicity, as well as methods to prevent and treat this condition. We also summarize and discuss advances made in the last decade in the comprehension of the molecular mechanisms underlying the pathology.
Collapse
Affiliation(s)
- Konrad Teodor Sawicki
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Valentina Sala
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Lorenzo Prever
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| | - Hossein Ardehali
- Division of Cardiology, Feinberg Cardiovascular and Renal Research Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, USA;
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy;
| |
Collapse
|
29
|
de Souza TF, Silva TQ, Antunes-Correa L, Drobni ZD, Costa FO, Dertkigil SSJ, Nadruz W, Brenelli F, Sposito AC, Matos-Souza JR, Coelho OR, Neilan TG, Jerosch-Herold M, Coelho-Filho OR. Cardiac magnetic resonance assessment of right ventricular remodeling after anthracycline therapy. Sci Rep 2021; 11:17132. [PMID: 34429493 PMCID: PMC8385101 DOI: 10.1038/s41598-021-96630-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 08/11/2021] [Indexed: 11/13/2022] Open
Abstract
There are limited data on the effects of anthracyclines on right ventricular (RV) structure, function, and tissue characteristics. The goal of this study was to investigate the effects of anthracyclines on the RV using cardiac magnetic resonance (CMR). This was a post-hoc analysis of a prospective study of 27 breast cancer (BC) patients (51.8 ± 8.9 years) using CMR prior, and up to 3-times after anthracyclines (240 mg/m2) to measure RV volumes and mass, RV extracellular volume (ECV) and cardiomyocyte mass (CM). Before anthracyclines, LVEF (69.4 ± 3.6%) and RVEF (55.6 ± 9%) were normal. The median follow-up after anthracyclines was 399 days (IQR 310–517). The RVEF reached its nadir (46.3 ± 6.8%) after 9-months (P < 0.001). RV mass-index and RV CM decreased to 13 ± 2.8 g/m2 and 8.13 ± 2 g/m2, respectively, at 16-months after anthracyclines. The RV ECV expanded from 0.26 ± 0.07 by 0.14 (53%) to 0.40 ± 0.1 (P < 0.001). The RV ECV expansion correlated with a decrease in RV mass-index (r = −0.46; P < 0.001) and the increase in CK-MB. An RV ESV index at baseline above its median predicted an increased risk of LV dysfunction post-anthracyclines. In BC patients treated with anthracyclines, RV atrophy, systolic dysfunction, and a parallel increase of diffuse interstitial fibrosis indicate a cardiotoxic response on a similar scale as previously seen in the systemic left ventricle.
Collapse
Affiliation(s)
- Thiago Ferreira de Souza
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Thiago Quinaglia Silva
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Lígia Antunes-Correa
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Zsofia D Drobni
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Felipe Osório Costa
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Sergio San Juan Dertkigil
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Wilson Nadruz
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Fabrício Brenelli
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Andrei C Sposito
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - José Roberto Matos-Souza
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Otávio Rizzi Coelho
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil
| | - Tomas G Neilan
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Michael Jerosch-Herold
- Noninvasive Cardiovascular Imaging Program and Department of Radiology, Brigham and Women's Hospital, Boston, MA, USA
| | - Otávio Rizzi Coelho-Filho
- Division of Cardiology, Department of Medicine, Faculdade de Ciências Médicas - Universidade Estadual de Campinas (UNICAMP), Rua Tessália Viera de Camargo, 126, Campinas, SP, CEP 13083-887, Brazil.
| |
Collapse
|
30
|
Burt MG, Mangelsdorf BL, Drake SM, Swan M, Padman S, Vatandoust S, Koczwara B. Insulin sensitivity, cardiovascular function and bone health in women with early stage breast cancer before and after cancer treatment. Intern Med J 2021; 52:1917-1924. [PMID: 34343400 DOI: 10.1111/imj.15469] [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: 04/13/2021] [Revised: 07/06/2021] [Accepted: 07/11/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Cardiovascular disease is a leading cause of death in breast cancer survivors, but the underlying cause is not fully characterised. AIMS To determine whether insulin sensitivity, cardiovascular risk markers and body composition were perturbed in women treated with chemotherapy for early stage breast cancer and whether perturbations occurred before or after cancer treatment. METHODS Sixteen women with breast cancer and 17 control subjects were studied. Twelve breast cancer patients returned for a second visit following cancer treatment comprising chemotherapy (n=2), or chemotherapy and radiotherapy (n=10)). Matsuda index to estimate insulin sensitivity, fasting lipids, pulse wave velocity (PWV), reactive hyperaemia index (RHI), and body composition by dual energy X-ray absorptiometry were measured. RESULTS There were no significant differences in age (53±9 vs 54±11 years, p=0.82) or body mass index (28±7 vs 28±6, p=0.97) between patients with breast cancer and controls. Patients with breast cancer had higher triglycerides than controls (1.2±0.1 vs 0.8±0.1 mmol/L, p=0.03), but there were no significant differences in Matsuda index, PWV and RHI. Following cancer treatment there was a lower Matsuda index (6.3±1.2 vs 5.2±1.0, p=0.01), but this was not associated with a significant change in vascular function. Bone mass fell by 3% from 2.27±0.11 to 2.20±0.10 kg after cancer treatment (p=0.03). CONCLUSIONS Patients with breast cancer had higher triglycerides before treatment and a reduction in insulin sensitivity and bone mass following cancer treatment. Future larger and longer-term studies should characterise the effect of reduced insulin sensitivity on rates of diabetes, cardiovascular disease, cancer outcomes and fracture. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Morton G Burt
- Southern Adelaide Diabetes and Endocrine Services, Flinders Medical Centre, Adelaide, Australia.,College of Medicine and Public Health, Flinders University, Adelaide, Australia
| | - Brenda L Mangelsdorf
- Southern Adelaide Diabetes and Endocrine Services, Flinders Medical Centre, Adelaide, Australia
| | - Sophie M Drake
- Southern Adelaide Diabetes and Endocrine Services, Flinders Medical Centre, Adelaide, Australia
| | - Monique Swan
- Southern Adelaide Health Service, Flinders Medical Centre, Adelaide, Australia
| | - Sunita Padman
- Southern Adelaide Health Service, Flinders Medical Centre, Adelaide, Australia
| | - Sina Vatandoust
- College of Medicine and Public Health, Flinders University, Adelaide, Australia.,Southern Adelaide Health Service, Flinders Medical Centre, Adelaide, Australia
| | - Bogda Koczwara
- College of Medicine and Public Health, Flinders University, Adelaide, Australia.,Southern Adelaide Health Service, Flinders Medical Centre, Adelaide, Australia
| |
Collapse
|
31
|
Takeuchi S, Takayama N, Soejima K, Yoshino H. A penetrating atherosclerotic ulcer rapidly growing into a saccular aortic aneurysm during treatment of leukaemia: a case report. Eur Heart J Case Rep 2021; 5:ytab196. [PMID: 34514296 PMCID: PMC8422330 DOI: 10.1093/ehjcr/ytab196] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Revised: 10/27/2020] [Accepted: 03/08/2021] [Indexed: 12/03/2022]
Abstract
Background The clinical course of penetrating atherosclerotic ulcers is variable and can be complicated with intramural haematomas, dissection, pseudoaneurysms, or aortic rupture. Because it can lead to life-threatening conditions, it needs to be managed carefully. Case summary A 68-year-old woman, who was treated for acute myeloid leukaemia (subtype: M0-FAB) approximately 1 year before presentation, visited the hospital with complaints of a headache and lumbar pain. After hospitalization, investigations revealed miliary tuberculosis. On the same day, she developed a Stanford type A acute aortic dissection (AAD) with cardiac tamponade; during the course of the previous leukaemia treatment, a small ulcerative lesion at the distal aortic arch grew into a small saccular aortic aneurysm (SAA) that expanded rapidly and finally developed into a Stanford type A AAD. However, the relationship between the SAA and aortic dissection could not be confirmed. Discussion The chronological changes in the atherosclerotic lesion at the distal aortic arch could be clearly observed because computed tomography scans were repeatedly obtained until just before the onset of AAD. The rapid progression of atherosclerotic lesions in the unique context of leukaemia treatment and miliary tuberculosis was considered to be a pathological characteristic, and the mechanism underlying this process was investigated. Clinicians should be aware of the aortic complications that may progress under special circumstances, such as anthracycline use or immunodeficiency. Careful observation is mandatory for patients with aortic disease.
Collapse
Affiliation(s)
- Shinsuke Takeuchi
- Department of Cardiology, Kyorin University School of Medicine , 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Nobuyuki Takayama
- Department of Hematology, Kyorin University School of Medicine , 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Kyoko Soejima
- Department of Cardiology, Kyorin University School of Medicine , 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| | - Hideaki Yoshino
- Department of Cardiology, Kyorin University School of Medicine , 6-20-2 Shinkawa, Mitaka-shi, Tokyo 181-8611, Japan
| |
Collapse
|
32
|
Clayton ZS, Hutton DA, Mahoney SA, Seals DR. Anthracycline chemotherapy-mediated vascular dysfunction as a model of accelerated vascular aging. ACTA ACUST UNITED AC 2021; 2:45-69. [PMID: 34212156 DOI: 10.1002/aac2.12033] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiovascular diseases (CVD) are the leading cause of death worldwide, and age is by far the greatest risk factor for developing CVD. Vascular dysfunction, including endothelial dysfunction and arterial stiffening, is responsible for much of the increase in CVD risk with aging. A key mechanism involved in vascular dysfunction with aging is oxidative stress, which reduces the bioavailability of nitric oxide (NO) and induces adverse changes to the extracellular matrix of the arterial wall (e.g., elastin fragmentation/degradation, collagen deposition) and an increase in advanced glycation end products, which form crosslinks in arterial wall structural proteins. Although vascular dysfunction and CVD are most prevalent in older adults, several conditions can "accelerate" these events at any age. One such factor is chemotherapy with anthracyclines, such as doxorubicin (DOXO), to combat common forms of cancer. Children, adolescents and young adults treated with these chemotherapeutic agents demonstrate impaired vascular function and an increased risk of future CVD development compared with healthy age-matched controls. Anthracycline treatment also worsens vascular dysfunction in mid-life (50-64 years of age) and older (65 and older) adults such that endothelial dysfunction and arterial stiffness are greater compared to age-matched controls. Collectively, these observations indicate that use of anthracycline chemotherapeutic agents induce a vascular aging-like phenotype and that the latter contributes to premature CVD in cancer survivors exposed to these agents. Here, we review the existing literature supporting these ideas, discuss potential mechanisms as well as interventions that may protect arteries from these adverse effects, identify research gaps and make recommendations for future research.
Collapse
|
33
|
Parr SK, Steele CC, Hammond ST, Turpin VRG, Ade CJ. Arterial stiffness is associated with cardiovascular and cancer mortality in cancer patients: Insight from NHANESIII. Int J Cardiol Hypertens 2021; 9:100085. [PMID: 34095811 PMCID: PMC8167280 DOI: 10.1016/j.ijchy.2021.100085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/14/2021] [Accepted: 04/23/2021] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND Cancer survivors are at greater risk for cardiovascular disease (CVD) than second malignancy, resulting in a decreased quality of life and increased cost of care. Additional knowledge of CVD prevention by identifying possible risk factors has clinical relevance. Our main objective was to determine the relevance of a clinical index of arterial stiffness, pulse pressure, in predicting CVD mortality in cancer patients, with a second objective to examine its relationship with cancer mortality. METHODS We retrospectively analyzed 781 cancer patients from Third National Health and Nutrition Examination Survey and Linked Mortality File, including demographic, anthropometric, blood pressure, and cause of death. Kaplan-Meier survival curve and Cox hazard regression analyses were performed to assess the relationship between pulse pressure and cardiovascular, cancer, and all-cause mortality. RESULTS During a mean follow-up time of 8.1 years, 603 deaths, 257 cancer and 151 CVD, occurred. In unadjusted models, the risk of CVD, cancer, and all-cause mortality were 3.8-fold, 5.3-fold, and 1.6-fold higher, respectively, for pulse pressure ≥70 mmHg compared to <50 mmHg. Adjusted analyses revealed a higher CVD mortality in cancer patients <65 years with a pulse pressure 60-70 mmHg (adjusted hazard ratio, 5.26; 95%CI, 1.12-24.78) when compared to pulse pressure of <50 mmHg. Pulse pressure was not associated with risk of all-cause, CVD, or cancer in those ≥65 years. CONCLUSION Pulse pressure, an index of arterial stiffness, is predictive of CVD mortality in cancer patients. Our findings support non-invasive office-setting measurements of arterial stiffness to identify high risk patients.
Collapse
Affiliation(s)
- Shannon K. Parr
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, USA
| | - Catherine C. Steele
- Department of Psychology and Communication, College of Arts and Sciences, Texas A&M International University, USA
| | - Stephen T. Hammond
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, USA
| | - Vanessa Rose G. Turpin
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, USA
| | - Carl J. Ade
- Department of Kinesiology, College of Health and Human Sciences, Kansas State University, USA
- Physician Assistant Studies, College of Health and Human Sciences, Kansas State University, USA
- Johnson Cancer Center, Kansas State University, USA
| |
Collapse
|
34
|
Narezkina A, Narayan HK, Zemljic-Harpf AE. Molecular mechanisms of anthracycline cardiovascular toxicity. Clin Sci (Lond) 2021; 135:1311-1332. [PMID: 34047339 PMCID: PMC10866014 DOI: 10.1042/cs20200301] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 05/07/2021] [Accepted: 05/11/2021] [Indexed: 12/21/2022]
Abstract
Anthracyclines are effective chemotherapeutic agents, commonly used in the treatment of a variety of hematologic malignancies and solid tumors. However, their use is associated with a significant risk of cardiovascular toxicities and may result in cardiomyopathy and heart failure. Cardiomyocyte toxicity occurs via multiple molecular mechanisms, including topoisomerase II-mediated DNA double-strand breaks and reactive oxygen species (ROS) formation via effects on the mitochondrial electron transport chain, NADPH oxidases (NOXs), and nitric oxide synthases (NOSs). Excess ROS may cause mitochondrial dysfunction, endoplasmic reticulum stress, calcium release, and DNA damage, which may result in cardiomyocyte dysfunction or cell death. These pathophysiologic mechanisms cause tissue-level manifestations, including characteristic histopathologic changes (myocyte vacuolization, myofibrillar loss, and cell death), atrophy and fibrosis, and organ-level manifestations including cardiac contractile dysfunction and vascular dysfunction. In addition, these mechanisms are relevant to current and emerging strategies to diagnose, prevent, and treat anthracycline-induced cardiomyopathy. This review details the established and emerging data regarding the molecular mechanisms of anthracycline-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Anna Narezkina
- Department of Medicine, Division of Cardiovascular Medicine, UCSD Cardiovascular Institute, University of California, San Diego
| | - Hari K. Narayan
- Department of Pediatrics, Division of Cardiology, University of California, San Diego
| | - Alice E. Zemljic-Harpf
- Veterans Affairs San Diego Healthcare System, San Diego, USA
- Department of Anesthesiology, University of California San Diego, La Jolla, California, USA
| |
Collapse
|
35
|
Novo G, Di Lisi D, Manganaro R, Manno G, Lazzara S, Immordino FA, Madaudo C, Carerj S, Russo A, Incorvaia L, Zito C. Arterial Stiffness: Effects of Anticancer Drugs Used for Breast Cancer Women. Front Physiol 2021; 12:661464. [PMID: 34054578 PMCID: PMC8161497 DOI: 10.3389/fphys.2021.661464] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 04/07/2021] [Indexed: 01/10/2023] Open
Abstract
Purpose: It is well known that anticancer drugs used for treating breast cancer can cause cardiac toxicity, and less is known about vascular toxicity. The aim of this study was to assess subclinical vascular effects of anthracyclines and trastuzumab (TRZ) in women treated for breast cancer. Methods: We enrolled 133 female patients with breast cancer undergoing adjuvant treatment with anthracycline-containing chemotherapy (CT) followed by taxane (paclitaxel/docetaxel) + TRZ. Patients underwent a standard echocardiography including measurement of left ventricular ejection fraction and global longitudinal strain at baseline and at follow-up. Vascular toxicity was evaluated by measuring brachial blood pressure (BP) and arterial stiffness indices (pulse wave velocity and Beta stiffness index) at T0 (baseline), T1 (3 months), T2 (6 months), and T3 (12 months). Results: Arterial stiffness indices were significantly increased at T1 in patients treated with anthracycline-containing CT (PWV 5.5 m/s IQR 5.15-6.4 at T0 vs. PWV 6.7 m/s IQR 5.6-7.2 at T1, p < 0.05; Beta index PWV 6.7 IQR 5.25-6.65 at T0, PWV 8.35 IQR 6.5-10.15 at T1, p < 0.05) but not at T2 and T3, when treatment with anthracyclines was stopped and patients were under treatment with taxane and TRZ. Blood pressure values did not significantly change during follow-up. Conclusion: Changes in arterial stiffness parameters occur early after starting treatment with anthracyclines, and they seem to be reversible if anthracycline treatment is stopped. These changes are not influenced by blood pressure values modifications. Therefore, in breast cancer women, anthracyclines seem to cause early reversible subclinical vascular injury.
Collapse
Affiliation(s)
- Giuseppina Novo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - Daniela Di Lisi
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - Roberta Manganaro
- Department of Clinical and Experimental Medicine, Section of Cardiology, University of Messina, University Hospital "Policlinico G. Martino", Messina, Italy
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - Simone Lazzara
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - Federico Angelo Immordino
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - Cristina Madaudo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Cardiology Unit, University Hospital P. Giaccone, Palermo, Italy
| | - Scipione Carerj
- Department of Clinical and Experimental Medicine, Section of Cardiology, University of Messina, University Hospital "Policlinico G. Martino", Messina, Italy
| | - Antonio Russo
- Department of Surgical, Oncological and Oral Sciences, Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Lorena Incorvaia
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (Bi.N.D.), Section of Medical Oncology, University of Palermo, Palermo, Italy
| | - Concetta Zito
- Department of Clinical and Experimental Medicine, Section of Cardiology, University of Messina, University Hospital "Policlinico G. Martino", Messina, Italy
| |
Collapse
|
36
|
Role of cardiovascular magnetic resonance in early detection and treatment of cardiac dysfunction in oncology patients. Int J Cardiovasc Imaging 2021; 37:3003-3017. [PMID: 33982196 DOI: 10.1007/s10554-021-02271-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 05/02/2021] [Indexed: 12/26/2022]
Abstract
The purpose of this review is to provide an overview of the essential role that cardiovascular magnetic resonance (CMR) has in the field of cardio-oncology. Recent findings: CMR has been increasingly used for early identification of cancer therapy related cardiac dysfunction (CTRCD) due to its precision in detecting subtle changes in cardiac function and for myocardial tissue characterization. Summary: CMR is able to identify subclinical CTRCD in patients receiving potentially cardiotoxic chemotherapy and guide initiation of cardio protective therapy. Multiparametric analysis with myocardial strain, tissue characterization play a critical role in understanding important clinical questions in cardio-oncology.
Collapse
|
37
|
Bosman M, Favere K, Neutel CHG, Jacobs G, De Meyer GRY, Martinet W, Van Craenenbroeck EM, Guns PJDF. Doxorubicin induces arterial stiffness: A comprehensive in vivo and ex vivo evaluation of vascular toxicity in mice. Toxicol Lett 2021; 346:23-33. [PMID: 33895255 DOI: 10.1016/j.toxlet.2021.04.015] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 03/29/2021] [Accepted: 04/20/2021] [Indexed: 01/09/2023]
Abstract
Arterial stiffness is an important predictor of cardiovascular risk. Clinical studies have demonstrated that arterial stiffness increases in cancer patients treated with the chemotherapeutic doxorubicin (DOX). However, the mechanisms of DOX-induced arterial stiffness remain largely unknown. This study aimed to evaluate artery stiffening in DOX-treated mice using in vivo and ex vivo techniques. Male C57BL/6J mice were treated for 2 weeks with 2 mg/kg (low dose) or 4 mg/kg (high dose) of DOX weekly. Arterial stiffness was assessed in vivo with ultrasound imaging (abdominal aorta pulse wave velocity (aaPWV)) and applanation tonometry (carotid-femoral PWV) combined with ex vivo vascular stiffness and reactivity evaluation. The high dose increased aaPWV, while cfPWV did not reach statistical significance. Phenylephrine (PE)-contracted aortic segments showed a higher Peterson's modulus (Ep) in the high dose group, while Ep did not differ when vascular smooth muscle cells (VSMCs) were relaxed by a NO donor (DEANO). In addition, aortic rings of DOX-treated mice showed increased PE contraction, decreased basal nitric oxide (NO) index and impaired acetylcholine-induced endothelium-dependent relaxation. DOX treatment contributed to endothelial cell loss and reduced endothelial nitric oxide synthase (eNOS) expression in the aorta. In conclusion, we have replicated DOX-induced arterial stiffness in a murine model and this aortic stiffness is driven by impaired endothelial function, contributing to increased vascular tone.
Collapse
Affiliation(s)
- Matthias Bosman
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium.
| | - Kasper Favere
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium; University of Antwerp, Research Group Cardiovascular Diseases, GENCOR, Antwerp, Belgium; Ghent University, Faculty of Medicine and Health Sciences, Department of Internal Medicine, C. Heymanslaan 10, B-9000, Ghent, Belgium; Antwerp University Hospital (UZA), Department of Cardiology, Drie Eikenstraat 655, B-2650, Edegem, Belgium
| | - Cédric H G Neutel
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Griet Jacobs
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Guido R Y De Meyer
- University of Antwerp, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Wim Martinet
- University of Antwerp, Faculty of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| | - Emeline M Van Craenenbroeck
- University of Antwerp, Research Group Cardiovascular Diseases, GENCOR, Antwerp, Belgium; Antwerp University Hospital (UZA), Department of Cardiology, Drie Eikenstraat 655, B-2650, Edegem, Belgium
| | - Pieter-Jan D F Guns
- University of Antwerp, Faculty of Medicine and Health Sciences, Laboratory of Physiopharmacology, Campus Drie Eiken, Universiteitsplein 1, B-2610, Antwerp, Belgium
| |
Collapse
|
38
|
Saunderson CED, Plein S, Manisty CH. Role of cardiovascular magnetic resonance imaging in cardio-oncology. Eur Heart J Cardiovasc Imaging 2021; 22:383-396. [PMID: 33404058 DOI: 10.1093/ehjci/jeaa345] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/10/2020] [Indexed: 12/28/2022] Open
Abstract
Advances in cancer therapy have led to significantly longer cancer-free survival times over the last 40 years. Improved survivorship coupled with increasing recognition of an expanding range of adverse cardiovascular effects of many established and novel cancer therapies has highlighted the impact of cardiovascular disease in this population. This has led to the emergence of dedicated cardio-oncology services that can provide pre-treatment risk stratification, surveillance, diagnosis, and monitoring of cardiotoxicity during cancer therapies, and late effects screening following completion of treatment. Cardiovascular imaging and the development of imaging biomarkers that can accurately and reliably detect pre-clinical disease and enhance our understanding of the underlying pathophysiology of cancer treatment-related cardiotoxicity are becoming increasingly important. Multi-parametric cardiovascular magnetic resonance (CMR) is able to assess cardiac structure, function, and provide myocardial tissue characterization, and hence can be used to address a variety of important clinical questions in the emerging field of cardio-oncology. In this review, we discuss the current and potential future applications of CMR in the investigation and management of cancer patients.
Collapse
Affiliation(s)
- Christopher E D Saunderson
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sven Plein
- Department of Biomedical Imaging Science, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Charlotte H Manisty
- Department of Cardio-Oncology, Barts Heart Centre, Barts Health NHS Trust, West Smithfield, London EC1A 7BE, UK
| |
Collapse
|
39
|
Demissei BG, Fan Y, Qian Y, Cheng HG, Smith AM, Shimamoto K, Vedage N, Narayan HK, Scherrer-Crosbie M, Davatzikos C, Ky B. Left ventricular segmental strain and the prediction of cancer therapy-related cardiac dysfunction. Eur Heart J Cardiovasc Imaging 2021; 22:418-426. [PMID: 33206976 PMCID: PMC7984733 DOI: 10.1093/ehjci/jeaa288] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/12/2020] [Indexed: 01/02/2023] Open
Abstract
AIMS We aimed to determine the early changes and predictive value of left ventricular (LV) segmental strain measures in women with breast cancer receiving doxorubicin. METHODS AND RESULTS In a cohort of 237 women with breast cancer receiving doxorubicin with or without trastuzumab, 1151 echocardiograms were prospectively acquired over a median (Q1-Q3) of 7 (2-24) months. LV ejection fraction (LVEF) and 36 segmental strain measures were core lab quantified. A supervised machine learning (ML) model was then developed using random forest regression to identify segmental strain measures predictive of nadir LVEF post-doxorubicin completion. Cancer therapy-related cardiac dysfunction (CTRCD) was defined as a ≥10% absolute LVEF decline pre-treatment to a value <50%. Median (Q1-Q3) baseline age was 48 (41-57) years. Thirty-five women developed CTRCD, and eight of these developed symptomatic heart failure. From pre-treatment to doxorubicin completion, longitudinal strain worsened across the basal and mid-LV segments but not in the apical segments; circumferential strain worsened primarily in the septum; radial strain worsened uniformly and transverse strain remained unchanged across all LV segments. In the ML model, anterolateral and inferoseptal circumferential strain were the most predictive features; longitudinal and transverse strain in the basal inferoseptal, anterior, basal anterolateral, and apical lateral segments were also top predictive features. The addition of predictive segmental strain measures to a model including age, cancer therapy regimen, hypertension, and LVEF increased the area under the curve (AUC) from 0.70 (95% confidence interval (CI) 0.60-0.80) to 0.87 (95% CI 0.81-0.92), ΔAUC = 0.18 (95% CI 0.08-0.27) for the prediction of CTRCD. CONCLUSION Our findings suggest that segmental strain measures can enhance cardiotoxicity risk prediction in women with breast cancer receiving doxorubicin.
Collapse
Affiliation(s)
- Biniyam G Demissei
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yong Fan
- Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yiwen Qian
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Henry G Cheng
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Amanda M Smith
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Kelsey Shimamoto
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Natasha Vedage
- Department of Internal Medicine, Temple University Hospital, Philadelphia, PA, USA
| | - Hari K Narayan
- Department of Pediatrics, Division of Cardiology, Rady Children’s Hospital San Diego, The University of California San Diego, San Diego, CA, USA
| | - Marielle Scherrer-Crosbie
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Christos Davatzikos
- Department of Radiology, Center for Biomedical Image Computing and Analytics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Biostatistics, Epidemiology & Informatics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
40
|
Clayton ZS, Brunt VE, Hutton DA, Casso AG, Ziemba BP, Melov S, Campisi J, Seals DR. Tumor Necrosis Factor Alpha-Mediated Inflammation and Remodeling of the Extracellular Matrix Underlies Aortic Stiffening Induced by the Common Chemotherapeutic Agent Doxorubicin. Hypertension 2021; 77:1581-1590. [PMID: 33719511 DOI: 10.1161/hypertensionaha.120.16759] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Zachary S Clayton
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Vienna E Brunt
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - David A Hutton
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Abigail G Casso
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Brian P Ziemba
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| | - Simon Melov
- Buck Institute for Research on Aging, Novato, CA (S.M., J.C.)
| | - Judith Campisi
- Buck Institute for Research on Aging, Novato, CA (S.M., J.C.).,Lawrence Berkeley National Laboratory, CA (J.C.)
| | - Douglas R Seals
- Department of Integrative Physiology, University of Colorado Boulder, Colorado (Z.S.C., V.E.B., D.A.H., A.G.C., B.P.Z., D.R.S.)
| |
Collapse
|
41
|
Mizrachi A, Ben-Aharon I, Li H, Bar-Joseph H, Bodden C, Hikri E, Popovtzer A, Shalgi R, Haimovitz-Friedman A. Chemotherapy-induced acute vascular injury involves intracellular generation of ROS via activation of the acid sphingomyelinase pathway. Cell Signal 2021; 82:109969. [PMID: 33647448 PMCID: PMC10402763 DOI: 10.1016/j.cellsig.2021.109969] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 02/07/2023]
Abstract
Several categories of chemotherapy confer substantial risk for late-term vascular morbidity and mortality. In the present study, we aimed to investigate the mechanism of acute chemotherapy-induced vascular injury in normal tissues. Specifically, we looked at activation of the acid sphingomyelinase (ASMase)/ceramide pathway, which leads to generation of reactive oxygen species (ROS) and induction of oxidative stress that may result in vascular injury. In particular, we focused on two distinct drugs, doxorubicin (DOX) and cisplatin (CIS) and their effects on normal endothelial cells. In vitro, DOX resulted in increased ASMase activity, intra-cellular ROS production and induction of apoptosis. CIS treatment generated significantly reduced effects in endothelial cells. In-vivo, murine femoral arterial blood flow was measured in real-time, during and after DOX or CIS administration, using fluorescence optical imaging system. While DOX caused constriction of small vessels and disintegration of large vessels' wall, CIS induced minor vascular changes in arterial blood flow, correlating with the in vitro findings. These results demonstrate that DOX induces acute vascular injury by increased ROS production, via activation of ASMase/ceramide pathway, while CIS increases ROS production and its immediate extracellular translocation, without causing detectable acute vascular injury. Our findings may potentially lead to the development of new strategies to prevent long-term cardiovascular morbidity in cancer survivors.
Collapse
Affiliation(s)
- Aviram Mizrachi
- Head and Neck Service, Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY, USA; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Otorhinolaryngology Head and Neck Surgery and Center for Translational Research in Head and Neck Cancer, Rabin Medical Center, Petah Tikva, Israel
| | - Irit Ben-Aharon
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Hongyan Li
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Hadas Bar-Joseph
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Chloe Bodden
- Department of Radiation Oncology, Memorial Sloan-Kettering Cancer Center, New York, NY, USA
| | - Elad Hikri
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Otorhinolaryngology Head and Neck Surgery and Center for Translational Research in Head and Neck Cancer, Rabin Medical Center, Petah Tikva, Israel
| | - Aron Popovtzer
- Division of Oncology, Rambam Health Care Campus, Haifa, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel; Department of Otorhinolaryngology Head and Neck Surgery and Center for Translational Research in Head and Neck Cancer, Rabin Medical Center, Petah Tikva, Israel
| | - Ruth Shalgi
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | |
Collapse
|
42
|
Galán-Arriola C, Vílchez-Tschischke JP, Lobo M, López GJ, de Molina-Iracheta A, Pérez-Martínez C, Villena-Gutiérrez R, Macías Á, Díaz-Rengifo IA, Oliver E, Fuster V, Sánchez-González J, Ibanez B. Coronary microcirculation damage in anthracycline cardiotoxicity. Cardiovasc Res 2021; 118:531-541. [PMID: 33605403 PMCID: PMC8803079 DOI: 10.1093/cvr/cvab053] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Revised: 12/28/2020] [Accepted: 02/17/2021] [Indexed: 01/19/2023] Open
Abstract
AIMS The aim of this study was to study changes in coronary microcirculation status during and after several cycles of anthracycline treatment. METHODS AND RESULTS Large-White male pigs (n = 40) were included in different experimental protocols (ExPr.) according to anthracycline cumulative exposure (0.45 mg/kg intracoronary (IC) doxorubicin per injection) and follow-up: Control (no doxorubicin); Single injection and sacrifice either at 48 hours (ExPr. 1) or 2 weeks (ExPr. 2); Three injections two weeks apart (low cumulative dose) and sacrifice either 2 weeks (ExPr. 3) or 12 weeks (ExPr. 4) after third injection; Five injections two weeks apart (high cumulative dose) and sacrifice 8 weeks after fifth injection (ExPr. 5). All groups were assessed by serial cardiac magnetic resonance (CMR) to quantify perfusion and invasive measurement of coronary flow reserve (CFR). At the end of each protocol, animals were sacrificed for ex vivo analyses. Vascular function was further evaluated by myography in explanted coronary arteries of pigs undergoing ExPr. 3 and controls.A single doxorubicin injection had no impact on microcirculation status, excluding a direct chemical toxicity. A series of five fortnightly doxorubicin injections (high cumulative dose) triggered a progressive decline in microcirculation status, evidenced by reduced CMR-based myocardial perfusion and CFR-measured impaired functional microcirculation. In the high cumulative dose regime (ExPr. 5), microcirculation changes appeared long before any contractile defect became apparent. Low cumulative doxorubicin dose (3 biweekly injections) was not associated with any contractile defect across long-term follow-up, but provoked persistent microcirculation damage, evident soon after third dose injection. Histological and myograph evaluations confirmed structural damage to arteries of all calibers even in animals undergoing low cumulative dose regimes. Conversely, arteriole damage and capillary bed alteration occurred only after high cumulative dose regime. CONCLUSION Serial in vivo evaluations of microcirculation status using state-of-the-art CMR and invasive CFR show that anthracyclines treatment is associated with progressive and irreversible damage to the microcirculation. This long-persisting damage is present even in low cumulative dose regimes, which are not associated with cardiac contractile deficits. Microcirculation damage might explain some of the increased incidence of cardiovascular events in cancer survivors who received anthracyclines without showing cardiac contractile defects.
Collapse
Affiliation(s)
- Carlos Galán-Arriola
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Jean Paul Vílchez-Tschischke
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Complejo Hospitalario Ruber Juan Bravo, Madrid, Spain
| | - Manuel Lobo
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Complejo Hospitalario Ruber Juan Bravo, Madrid, Spain
| | - Gonzalo J López
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | | | | | - Rocio Villena-Gutiérrez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | | | | | - Eduardo Oliver
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York
| | | | - Borja Ibanez
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain.,Cardiology Department, IIS-Fundación Jiménez Díaz Hospital, Madrid
| |
Collapse
|
43
|
Kirkham AA, Paterson DI, Haykowsky MJ, Beaudry RI, Mackey JR, Pituskin E, Grenier JG, Thompson RB. Aerobic Fitness Is Related to Myocardial Fibrosis Post-Anthracycline Therapy. Med Sci Sports Exerc 2021; 53:267-274. [PMID: 32826630 DOI: 10.1249/mss.0000000000002469] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
PURPOSE We evaluated the impact of anthracyclines on left ventricular function and myocardial tissue characteristics using cardiovascular magnetic resonance (CMR) imaging to determine their relationship with V˙O2peak. METHODS Women with breast cancer who had not yet received treatment (No-AT, n = 16) and had received anthracycline treatment ~1 yr earlier (Post-AT, n = 16) and controls without cancer (CON, n = 16) performed a maximal exercise test and a comprehensive 3T CMR examination, including native myocardial T1 mapping, where elevated T1 times are indicative of myocardial fibrosis. ANOVA and linear regression were used to compare CMR variables between groups and to determine associations with V˙O2peak. Subgroup analysis was performed by categorizing participants as "fit" or "unfit" based on whether their V˙O2peak value was greater or less than 100% of reference value for age, respectively. RESULTS Left ventricular end-diastolic volume, ejection fraction, and mass were similar between groups. Post-AT, T1 times were elevated (1534 ± 32 vs 1503 ± 28 ms, P < 0.01), and V˙O2peak was reduced (23.1 ± 7.5 vs 29.5 ± 7.7 mL·kg-1⋅min-1, P = 0.02) compared with CON. In No-AT, T1 times and V˙O2peak were similar to CON. In the Post-AT group, T1 time was associated with V˙O2peak (R2 = 64%), whereas in the absence of anthracyclines (i.e., No-AT and CON groups), T1 time was not associated with V˙O2peak. Regardless of group, all fit women had similar T1 times, whereas unfit women Post-AT had higher T1 than unfit CON (1546 ± 22 vs 1500 ± 33 ms, P < 0.01). CONCLUSIONS After anthracycline chemotherapy, an elevated T1 time suggesting greater extent of myocardial fibrosis, was associated with lower V˙O2peak. However, those who were fit did not have evidence of myocardial fibrosis after anthracycline treatment.
Collapse
Affiliation(s)
- Amy Ashley Kirkham
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, CANADA
| | - D Ian Paterson
- Division of Cardiology, University of Alberta, Edmonton, AB, CANADA
| | | | - Rhys I Beaudry
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, TX
| | - John R Mackey
- Department of Oncology, University of Alberta, Edmonton, AB, CANADA
| | - Edith Pituskin
- Faculty of Nursing, University of Alberta, Edmonton, AB, CANADA
| | - Justin G Grenier
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, CANADA
| | - Richard B Thompson
- Department of Biomedical Engineering, University of Alberta, Edmonton, AB, CANADA
| |
Collapse
|
44
|
Morel S, Delvin E, Marcil V, Levy E. Intestinal Dysbiosis and Development of Cardiometabolic Disorders in Childhood Cancer Survivors: A Critical Review. Antioxid Redox Signal 2021; 34:223-251. [PMID: 32390455 DOI: 10.1089/ars.2020.8102] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Significance: Survivors of pediatric cancers have a high risk of developing side effects after the end of their treatments. Many potential factors have been associated with the onset of cardiometabolic disorders (CMD), including cancer disease itself, chemotherapy, hormonal treatment, radiotherapy, and genetics. However, the precise etiology and underlying mechanisms of these long-term complications are poorly understood. Recent Advances: Greater awareness is currently paid to the role of microbiota in the emergence of cancers and modulation of cancer therapies in both children and adults. Alterations in the composition and diversity of intestinal microbiota can clearly influence tumor development and progression as well as immune responses and clinical output. As dysbiosis is closely linked to the development of host metabolic diseases, including obesity, metabolic syndrome, type 2 diabetes, and non-alcoholic fatty liver disease, it may increase the risk of CMD in cancer populations. Critical Issues: Only limited studies targeting the profile of intestinal dysbiosis before and after cancer treatment have been conducted. Further, the exact contribution of intestinal dysbiosis to the development of CMD in cancer survivors is poorly appreciated. This review intends to clarify the influence of gut microbiota on CMD in childhood cancer survivors, elucidate the potential mechanisms, and evaluate the latest research on the interplay between diet/food supplement, microbiota, and cancer-related CMD. Future Directions: The implication of intestinal dysbiosis in late metabolic complications of childhood cancer survivors should be clarified. Intervention strategies could be developed to reduce the risk of survivors to CMD. Antioxid. Redox Signal. 34, 223-251.
Collapse
Affiliation(s)
- Sophia Morel
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, Canada.,Department of Nutrition and Université de Montréal, Montreal, Canada
| | - Edgard Delvin
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, Canada
| | - Valérie Marcil
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, Canada.,Department of Nutrition and Université de Montréal, Montreal, Canada
| | - Emile Levy
- Research Centre, Sainte-Justine University Hospital Health Center, Université de Montréal, Montreal, Canada.,Department of Nutrition and Université de Montréal, Montreal, Canada.,Department of Pediatrics, Université de Montréal, Montreal, Canada
| |
Collapse
|
45
|
Lawal IO, Orunmuyi AT, Popoola GO, Lengana T, Mokoala KM, Ankrah AO, Sathekge MM. FDG PET/CT for evaluating systemic arterial inflammation induced by anthracycline-based chemotherapy of Hodgkin lymphoma: A retrospective cohort study. Medicine (Baltimore) 2020; 99:e23259. [PMID: 33235083 PMCID: PMC7710260 DOI: 10.1097/md.0000000000023259] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/22/2020] [Accepted: 10/20/2020] [Indexed: 12/05/2022] Open
Abstract
To evaluate arterial fluorodeoxyglucose (FDG) uptake as a marker of arterial inflammation in multiple vascular beds in patients treated with anthracycline-based chemotherapy for Hodgkin lymphoma (HL).We used maximum standardized uptake value (SUVmax) and target-to-background ratio (TBR) to quantify arterial FDG uptake in the carotid artery, ascending aorta, abdominal aorta, and femoral artery obtained on positron emission tomography/computed tomography (PET/CT) imaging performed at baseline before chemotherapy and after completion of chemotherapy in patients with HL treated with an anthracycline-containing regimen. We compared the SUVmax and TBR obtained at baseline with that obtained post-chemotherapy for each arterial bed to evaluate the effect of anthracycline-based chemotherapy. We evaluated the effect of cardiovascular risk factors such as human immunodeficiency virus (HIV) infection, smoking, hypertension, and diabetes on the changes in SUVmax and TBR seen in the different arterial beds after anthracycline-based chemotherapy.Fifty-two patients were included with a mean age of 34.56 ± 10.19 years. There were 33 males, and 18 patients were HIV-infected. The mean interval between completion of chemotherapy and follow-up flourine-18 fluorodeoxyglucose positron emission tomography/computed tomography (FDG PET/CT) scan was 65 weeks. We found no significant difference in arterial FDG uptake measured by SUVmax and TBR in all arterial beds between the pre- and post-chemotherapy FDG PET/CT. There was no significant impact of HIV infection, smoking, and hypertension on the changes in arterial FDG uptake following treatment with anthracycline-based chemotherapy.In patients with HL who were treated with anthracycline-based chemotherapy, we found no significant increase in arterial inflammation measured by FDG PET/CT after an average follow-up period of about 65 weeks since completion of chemotherapy.
Collapse
Affiliation(s)
- Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
| | | | - Gbenga O. Popoola
- Department of Epidemiology and Community Health, University of Ilorin, Ilorin, Nigeria
| | - Thabo Lengana
- Department of Nuclear Medicine, University of Pretoria
- KVNR Nuclear and Molecular Imaging, South Africa
| | | | - Alfred O. Ankrah
- Department of Nuclear Medicine, University of Pretoria
- Department of Nuclear Medicine and Molecular Imaging, University of Groningen, Groningen, The Netherlands
| | - Mike M. Sathekge
- Department of Nuclear Medicine, University of Pretoria
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria, South Africa
| |
Collapse
|
46
|
Jafari F, Safaei AM, Hosseini L, Asadian S, Kamangar TM, Zadehbagheri F, Rezaeian N. The role of cardiac magnetic resonance imaging in the detection and monitoring of cardiotoxicity in patients with breast cancer after treatment: a comprehensive review. Heart Fail Rev 2020; 26:679-697. [PMID: 33029698 DOI: 10.1007/s10741-020-10028-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/15/2020] [Indexed: 01/04/2023]
Abstract
The use of chemotherapy medicines for breast cancer (BC) has been associated with an increased risk of cardiotoxicity. In recent years, there have been growing interests regarding the application of cardiovascular magnetic resonance (CMR) imaging, a safe and noninvasive modality, with the potential to identify subtle morphological and functional changes in the myocardium. In this investigation, we aimed to review the performance of various CMR methods in diagnosing cardiotoxicity in BC, induced by chemotherapy or radiotherapy. For this purpose, we reviewed the literature available in PubMed, MEDLINE, Cochrane, Google Scholar, and Scopus databases. Our literature review showed that CMR is a valuable modality for identifying and predicting subclinical cardiotoxicity induced by chemotherapy. The novel T1, T2, and extracellular volume mapping techniques may provide critical information about cardiotoxicity, in addition to other CMR features such as functional and structural changes. However, further research is needed to verify the exact role of these methods in identifying cardiotoxicity and patient management. Since multiple studies have reported the improvement of left ventricular performance following the termination of chemotherapy regimens, CMR remains an essential imaging tool for the prediction of cardiotoxicity and, consequently, decreases the mortality rate of BC due to heart failure.
Collapse
Affiliation(s)
- Fatemeh Jafari
- Department of Radiation Oncology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.,Radiation Oncology Research Center (RORC), Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Afsane Maddah Safaei
- Radiation Oncology Research Center (RORC), Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Hosseini
- North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Sanaz Asadian
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Tara Molanaie Kamangar
- Radiation Oncology Research Center (RORC), Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Nahid Rezaeian
- Rajaie Cardiovascular Medical and Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
47
|
Parr SK, Liang J, Schadler KL, Gilchrist SC, Steele CC, Ade CJ. Anticancer Therapy-Related Increases in Arterial Stiffness: A Systematic Review and Meta-Analysis. J Am Heart Assoc 2020; 9:e015598. [PMID: 32648507 PMCID: PMC7660726 DOI: 10.1161/jaha.119.015598] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Background Cardio‐oncology is a clinical discipline focused primarily on the early detection of anticancer therapy–related cardiomyopathy. However, there is growing evidence that the direct adverse consequences extend beyond the myocardium to affect the vasculature, but this evidence remains limited. In addition, there remains a paucity of clinically based strategies for monitoring vascular toxicity in these patients. Importantly, arterial stiffness is increasingly recognized as a surrogate end point for cardiovascular disease and may be an important vascular outcome to consider. Therefore, the aim of this systematic review and meta‐analysis was to summarize evidence of increased arterial stiffening with anticancer therapy and evaluate the effect of treatment modifiers. Methods and Results A total of 19 longitudinal and cross‐sectional studies that evaluated arterial stiffness both during and following anticancer therapy were identified using multiple databases. Two separate analyses were performed: baseline to follow‐up (12 studies) and control versus patient groups (10 studies). Subgroup analysis evaluated whether stiffness differed as a function of treatment type and follow‐up time. Standard mean differences and mean differences were calculated using random effect models. Significant increases in arterial stiffness were identified from baseline to follow‐up (standard mean difference, 0.890; 95% CI, 0.448–1.332; P<0.0001; mean difference, 1.505; 95% CI, 0.789–2.221; P≤0.0001) and in patient versus control groups (standard mean difference, 0.860; 95% CI, 0.402–1.318; P=0.0002; mean difference, 1.437; 95% CI, 0.426–2.448; P=0.0052). Subgroup analysis indicated differences in arterial stiffness between anthracycline‐based and non‐anthracycline‐based therapies (standard mean difference, 0.20; 95% CI, 0.001–0.41; P=0.048), but not follow‐up time. Conclusions Significant arterial stiffening occurs following anticancer therapy. Our findings support the use of arterial stiffness as part of a targeted vascular imaging strategy for the identification of early cardiovascular injury during treatment and for the detection of long‐term cardiovascular injury into survivorship.
Collapse
Affiliation(s)
- Shannon K Parr
- Department of Kinesiology College of Health and Human Sciences Kansas State University Manhattan KS
| | - Jia Liang
- Department of Statistics Kansas State University Manhattan KS
| | - Keri L Schadler
- Division of Pediatrics Department of Pediatrics The University of Texas MD Anderson Cancer Center Houston TX
| | - Susan C Gilchrist
- Department of Clinical Cancer Prevention and Department of Cardiology The University of Texas MD Anderson Cancer Center Houston TX
| | - Catherine C Steele
- Department of Food, Nutrition, Dietetics, Health Kansas State University Manhattan KS
| | - Carl J Ade
- Department of Kinesiology College of Health and Human Sciences Kansas State University Manhattan KS
| |
Collapse
|
48
|
Sabater-Molina M, Navarro-Peñalver M, Muñoz-Esparza C, Esteban-Gil Á, Santos-Mateo JJ, Gimeno JR. Genetic Factors Involved in Cardiomyopathies and in Cancer. J Clin Med 2020; 9:E1702. [PMID: 32498335 PMCID: PMC7356401 DOI: 10.3390/jcm9061702] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Revised: 05/20/2020] [Accepted: 05/22/2020] [Indexed: 01/05/2023] Open
Abstract
Cancer therapy-induced cardiomyopathy (CCM) manifests as left ventricular (LV) dysfunction and heart failure (HF). It is associated withparticular pharmacological agents and it is typically dose dependent, but significant individual variability has been observed. History of prior cardiac disease, abuse of toxics, cardiac overload conditions, age, and genetic predisposing factors modulate the degree of the cardiac reserve and the response to the injury. Genetic/familial cardiomyopathies (CMY) are increasingly recognized in general populations with an estimated prevalence of 1:250. Association between cardiac and oncologic diseases regarding genetics involves not only the toxicity process, but pathogenicity. Genetic variants in germinal cells that cause CMY (LMNA, RAS/MAPK) can increase susceptibility for certain types of cancer. The study of mutations found in cancer cells (somatic) has revealed the implication of genes commonly associated with the development of CMY. In particular, desmosomal mutations have been related to increased undifferentiation and invasiveness of cancer. In this article, the authors review the knowledge on the relevance of environmental and genetic background in CCM and give insights into the shared genetic role in the pathogenicity of the cancer process and development of CMY.
Collapse
Affiliation(s)
- María Sabater-Molina
- Unidad de Cardiopatías Hereditarias, Servicio de Cardiología, Hospital Universitario Virgen dela Arrixaca, El Palmar, 30120 Murcia, Spain; (M.S.-M.); (M.N.-P.); (C.M.-E.); (J.R.G.)
- Universidad de Murcia, El Palmar, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), El Palmar, 30120 Murcia, Spain
- European Reference Networks (Guard-Heart), European Commission, 30120 Murcia, Spain
- Red de investigación Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Marina Navarro-Peñalver
- Unidad de Cardiopatías Hereditarias, Servicio de Cardiología, Hospital Universitario Virgen dela Arrixaca, El Palmar, 30120 Murcia, Spain; (M.S.-M.); (M.N.-P.); (C.M.-E.); (J.R.G.)
- Universidad de Murcia, El Palmar, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), El Palmar, 30120 Murcia, Spain
- European Reference Networks (Guard-Heart), European Commission, 30120 Murcia, Spain
| | - Carmen Muñoz-Esparza
- Unidad de Cardiopatías Hereditarias, Servicio de Cardiología, Hospital Universitario Virgen dela Arrixaca, El Palmar, 30120 Murcia, Spain; (M.S.-M.); (M.N.-P.); (C.M.-E.); (J.R.G.)
- Universidad de Murcia, El Palmar, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), El Palmar, 30120 Murcia, Spain
- European Reference Networks (Guard-Heart), European Commission, 30120 Murcia, Spain
- Red de investigación Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Ángel Esteban-Gil
- Biomedical Informatics & Bioinformatics Platform, Institute for Biomedical Research of Murcia (IMIB)/Foundation for Healthcare Training & Research of the Region of Murcia (FFIS), 30003 Murcia, Spain;
| | - Juan Jose Santos-Mateo
- Unidad de Cardiopatías Hereditarias, Servicio de Cardiología, Hospital Universitario Virgen dela Arrixaca, El Palmar, 30120 Murcia, Spain; (M.S.-M.); (M.N.-P.); (C.M.-E.); (J.R.G.)
- Universidad de Murcia, El Palmar, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), El Palmar, 30120 Murcia, Spain
- European Reference Networks (Guard-Heart), European Commission, 30120 Murcia, Spain
| | - Juan R. Gimeno
- Unidad de Cardiopatías Hereditarias, Servicio de Cardiología, Hospital Universitario Virgen dela Arrixaca, El Palmar, 30120 Murcia, Spain; (M.S.-M.); (M.N.-P.); (C.M.-E.); (J.R.G.)
- Universidad de Murcia, El Palmar, 30120 Murcia, Spain
- Instituto Murciano de Investigación Biosanitaria (IMIB), El Palmar, 30120 Murcia, Spain
- European Reference Networks (Guard-Heart), European Commission, 30120 Murcia, Spain
- Red de investigación Cardiovascular (CIBERCV), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
49
|
Harries I, Liang K, Williams M, Berlot B, Biglino G, Lancellotti P, Plana JC, Bucciarelli-Ducci C. Magnetic Resonance Imaging to Detect Cardiovascular Effects of Cancer Therapy: JACC CardioOncology State-of-the-Art Review. JACC CardioOncol 2020; 2:270-292. [PMID: 34396235 PMCID: PMC8352317 DOI: 10.1016/j.jaccao.2020.04.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 04/12/2020] [Accepted: 04/15/2020] [Indexed: 01/06/2023] Open
Abstract
This paper aims to empower and inform cardio-oncologists by providing a practical guide to the clinical application of cardiac magnetic resonance (CMR) in the rapidly evolving field of cardio-oncology. Specifically, we describe how CMR can be used to assess the cardiovascular effects of cancer therapy. The CMR literature, relevant societal guidelines, indication-specific imaging protocols, and methods to overcome some of the challenges encountered in performing and accessing CMR are reviewed.
Collapse
Affiliation(s)
- Iwan Harries
- Bristol Heart Institute, Bristol National Institute of Health Research (NIHR) Biomedical Research Centre, University Hospitals Bristol NHS Trust and University of Bristol. Bristol, United Kingdom
| | - Kate Liang
- Bristol Heart Institute, Bristol National Institute of Health Research (NIHR) Biomedical Research Centre, University Hospitals Bristol NHS Trust and University of Bristol. Bristol, United Kingdom
| | - Matthew Williams
- Bristol Heart Institute, Bristol National Institute of Health Research (NIHR) Biomedical Research Centre, University Hospitals Bristol NHS Trust and University of Bristol. Bristol, United Kingdom
| | - Bostjan Berlot
- Bristol Heart Institute, Bristol National Institute of Health Research (NIHR) Biomedical Research Centre, University Hospitals Bristol NHS Trust and University of Bristol. Bristol, United Kingdom
- Department of Cardiology, University Medical Centre Ljubljana, Slovenia
| | - Giovanni Biglino
- Bristol Heart Institute, Bristol National Institute of Health Research (NIHR) Biomedical Research Centre, University Hospitals Bristol NHS Trust and University of Bristol. Bristol, United Kingdom
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Patrizio Lancellotti
- University of Liège Hospital, GIGA Cardiovascular Sciences, Departments of Cardiology, Heart Valve Clinic, CHU Sart Tilman, Liège, Belgium
- Gruppo Villa Maria Care and Research, Anthea Hospital, Bari, Italy
| | - Juan Carlos Plana
- Texas Heart Institute at Baylor St. Luke’s Medical Center, Baylor College of Medicine, Houston, Texas, USA
| | - Chiara Bucciarelli-Ducci
- Bristol Heart Institute, Bristol National Institute of Health Research (NIHR) Biomedical Research Centre, University Hospitals Bristol NHS Trust and University of Bristol. Bristol, United Kingdom
| |
Collapse
|
50
|
Lv H, Tan R, Liao J, Hao Z, Yang X, Liu Y, Xia Y. Doxorubicin contributes to thrombus formation and vascular injury by interfering with platelet function. Am J Physiol Heart Circ Physiol 2020; 319:H133-H143. [PMID: 32469636 DOI: 10.1152/ajpheart.00456.2019] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In clinical studies, platelet aggregation and risk of thrombosis are increased in patients after doxorubicin treatment. However, the exact role of doxorubicin in platelet functions and thrombus formation in vivo remain unclear. The present study is to investigate the role of doxorubicin in platelet function in relation to thrombus formation and vascular toxicity, as well as the efficacy of antiplatelet therapy. Mice were treated with doxorubicin or vehicle (5 mg/kg iv, 4 wk), and the following parameters were determined: platelet count and size, platelet surface adhesive receptors by flow cytometry, density of granules by electron microscopy, platelet aggregation and degranulation at resting or agonist-stimulated state, platelet adhesion on fibrinogen or endothelial cells, and thrombus formation on collagen matrix. The efficacy of clopidogrel (15 mg·kg-1·day-1, followed by 5 mg·kg-1·day-1) on doxorubicin-induced changes in the aforementioned parameters as well as vascular injury were also determined. Whereas platelet count and size were similar between doxorubicin-treated and vehicle-treated mice, doxorubicin promoted thrombus formation evidenced by greater platelet aggregation, degranulation, and adhesion to endothelial cells evoked by agonists. Clopidogrel treatment attenuated the enhanced platelet activity and thrombus formation by doxorubicin, as well as vascular platelet infiltration and reactive oxygen species generation. Collectively, this study demonstrates that platelet functions are enhanced after long-term doxorubicin administration, which leads to thrombus formation and vascular toxicity, and that doxorubicin-induced changes in the functionality of platelets can be effectively inhibited by antiplatelet drugs.NEW & NOTEWORTHY Doxorubicin therapy in mice (antitumor dosage) markedly enhanced platelet functions measured as agonist-induced platelet aggregation, degranulation, and adhesion to endothelial cells, actions leading to thrombus formation and thrombosis-independent vascular injury. Clopidogrel treatment ameliorated thrombus formation and vascular toxicity induced by doxorubicin via inhibiting platelet activity.
Collapse
Affiliation(s)
- Haichen Lv
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ruopeng Tan
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Jiawei Liao
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhujing Hao
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Xiaolei Yang
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yang Liu
- Institute of Cardiovascular Diseases, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Yunlong Xia
- Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|