1
|
Abe J, Chau K, Mojiri A, Wang G, Oikawa M, Samanthapudi VSK, Osborn AM, Ostos-Mendoza KC, Mariscal-Reyes KN, Mathur T, Jain A, Herrmann J, Yusuf SW, Krishnan S, Deswal A, Lin SH, Kotla S, Cooke JP, Le NT. Impacts of Radiation on Metabolism and Vascular Cell Senescence. Antioxid Redox Signal 2025. [PMID: 40233257 DOI: 10.1089/ars.2024.0741] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Significance: This review investigates how radiation therapy (RT) increases the risk of delayed cardiovascular disease (CVD) in cancer survivors. Understanding the mechanisms underlying radiation-induced CVD is essential for developing targeted therapies to mitigate these effects and improve long-term outcomes for patients with cancer. Recent Advances: Recent studies have primarily focused on metabolic alterations induced by irradiation in various cancer cell types. However, there remains a significant knowledge gap regarding the role of chronic metabolic alterations in normal cells, particularly vascular cells, in the progression of CVD after RT. Critical Issues: This review centers on RT-induced metabolic alterations in vascular cells and their contribution to senescence accumulation and chronic inflammation across the vasculature post-RT. We discuss key metabolic pathways, including glycolysis, the tricarboxylic acid cycle, lipid metabolism, glutamine metabolism, and redox metabolism (nicotinamide adenine dinucleotide/Nicotinamide adenine dinucleotide (NADH) and nicotinamide adenine dinucleotide phosphate (NADP+)/NADPH). We further explore the roles of regulatory proteins such as p53, adenosine monophosphate-activated protein kinase, and mammalian target of rapamycin in driving these metabolic dysregulations. The review emphasizes the impact of immune-vascular crosstalk mediated by the senescence-associated secretory phenotype, which perpetuates metabolic dysfunction, enhances chronic inflammation, drives senescence accumulation, and causes vascular damage, ultimately contributing to cardiovascular pathogenesis. Future Directions: Future research should prioritize identifying therapeutic targets within these metabolic pathways or the immune-vascular interactions influenced by RT. Correcting metabolic dysfunction and reducing chronic inflammation through targeted therapies could significantly improve cardiovascular outcomes in cancer survivors. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Junichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Khanh Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Anahita Mojiri
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Masayoshi Oikawa
- Department of Cardiovascular Medicine, Fukushima Medical University, Fukushima, Japan
| | - Venkata S K Samanthapudi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abigail M Osborn
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | | | - Tammay Mathur
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas, USA
| | - Abhishek Jain
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
- Department of Biomedical Engineering, College of Engineering, Texas A&M University, College Station, Texas, USA
- Department of Medical Physiology, School of Medicine, Texas A&M Health Science Center, College Station, Texas, USA
| | - Joerg Herrmann
- Cardio Oncology Clinic, Division of Preventive Cardiology, Department of Cardiovascular Medicine, Mayo Clinic, Rochester, Minnesota, USA
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Steven H Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - John P Cooke
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, Texas, USA
| |
Collapse
|
2
|
Quagliariello V, Berretta M, Bisceglia I, Giacobbe I, Iovine M, Barbato M, Maurea C, Canale ML, Paccone A, Inno A, Scherillo M, Oliva S, Cadeddu Dessalvi C, Mauriello A, Fonderico C, Maratea AC, Gabrielli D, Maurea N. In the Era of Cardiovascular-Kidney-Metabolic Syndrome in Cardio-Oncology: From Pathogenesis to Prevention and Therapy. Cancers (Basel) 2025; 17:1169. [PMID: 40227756 PMCID: PMC11988012 DOI: 10.3390/cancers17071169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/15/2025] Open
Abstract
Cardiovascular-kidney-metabolic (CKM) syndrome represents a complex interplay between cardiovascular disease (CVD), chronic kidney disease (CKD), and metabolic disorders, significantly impacting cancer patients. The presence of CKM syndrome in cancer patients not only worsens their prognosis but also increases the risk of major adverse cardiovascular events (MACE), reduces quality of life (QoL), and affects overall survival (OS). Furthermore, several anticancer therapies, including anthracyclines, tyrosine kinase inhibitors, immune checkpoint inhibitors, and hormonal treatments, can exacerbate CKM syndrome by inducing cardiotoxicity, nephrotoxicity, and metabolic dysregulation. This review explores the pathophysiology of CKM syndrome in cancer patients and highlights emerging therapeutic strategies to mitigate its impact. We discuss the role of novel pharmacological interventions, including sodium-glucose cotransporter-2 inhibitors (SGLT2i), proprotein convertase subtilisin/kexin type 9 inhibitors (PCSK9i), and soluble guanylate cyclase (sGC) activators, as well as dietary and lifestyle interventions. Optimizing the management of CKM syndrome in cancer patients is crucial to improving OS, enhancing QoL, and reducing MACE. By integrating cardiometabolic therapies into oncologic care, we can create a more comprehensive treatment approach that reduces the burden of cardiovascular and renal complications in this vulnerable population. Further research is needed to establish personalized strategies for CKM syndrome prevention and treatment in cancer patients.
Collapse
Affiliation(s)
- Vincenzo Quagliariello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Massimiliano Berretta
- Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| | - Irma Bisceglia
- Servizi Cardiologici Integrati, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00148 Rome, Italy
| | - Ilaria Giacobbe
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Martina Iovine
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Matteo Barbato
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Carlo Maurea
- ASL NA1, UOC Neurology and Stroke Unit, Ospedale del Mare, 23807 Naples, Italy
| | | | - Andrea Paccone
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Alessandro Inno
- Medical Oncology, IRCCS Ospedale Sacro Cuore Don Calabria, 37024 Negrar di Valpolicella, Italy
| | - Marino Scherillo
- Cardiologia Interventistica e UTIC, A.O. San Pio, Presidio Ospedaliero Gaetano Rummo, 82100 Benevento, Italy
| | - Stefano Oliva
- Cardio-Oncology Unit, IRCCS Istituto Tumori, “Giovanni Paolo II”, 70124 Bari, Italy
| | | | - Alfredo Mauriello
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Celeste Fonderico
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Anna Chiara Maratea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| | - Domenico Gabrielli
- U.O.C. Cardiologia, Dipartimento Cardio-Toraco-Vascolare, Azienda Ospedaliera San Camillo Forlanini, 00152 Rome, Italy
| | - Nicola Maurea
- Division of Cardiology, Istituto Nazionale Tumori-IRCCS-Fondazione G. Pascale, 80131 Napoli, Italy
| |
Collapse
|
3
|
Vaitiekiene A, Kulboke M, Bieseviciene M, Kaunaite A, Markeviciute A, Bartnykaite A, Kireilis B, Rinkuniene D, Jankauskas A, Gaidamavicius I, Gerbutavicius R, Vaitiekus D, Sakalyte G, Vaskelyte JJ. Evaluation of Subclinical Cancer Therapy-Related Cardiac Dysfunction in Patients Undergoing Hematopoietic Stem Cell Transplantation: An Echocardiography Study. Cancers (Basel) 2024; 16:3808. [PMID: 39594762 PMCID: PMC11592334 DOI: 10.3390/cancers16223808] [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: 09/23/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 11/28/2024] Open
Abstract
Background: Hematopoietic stem cell transplantation (HSCT) is a potentially curative procedure that is used in various hematological malignancies. However, among an increasing number of HSCT, the amount of cancer therapy-related cardiac dysfunction (CTRCD) is increasing as well. This study aimed to determine the prevalence of subclinical CTRCD in HSCT patients 12 months after HSCT and to assess the impact of clinical factors on the development of CTRCD. Material and Methods: We included 55 patients who underwent autologous or allogeneic HSCT. The patients were assessed using an echocardiography method before and 12 months after a HSCT procedure. Results: Our study revealed that during the 12-month follow-up period, asymptomatic CTRCD was observed in 15 patients (27.3%), 6 experienced moderate CTRCD, and 9 experienced mild CTRCD. Patients with previous use of anthracyclines tended to have CTRCD more often: nine patients (60%) in the CTRCD group and nine patients (22.5%) in non-CTRCD group. Patients who received the BEAM regimen for conditioning also experienced CTRCD more often: five patients (33.3%) in CTRCD group vs. two patients (5%) in the non-CTRCD group. Conclusions: Our study showed that asymptomatic CTRCD was found in 27.3% of the patients 12 months after HSCT. The BEAM chemotherapy conditioning protocol following prior anthracycline use were identified as factors contributing to the development of CTRCD.
Collapse
Affiliation(s)
- Audrone Vaitiekiene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Migle Kulboke
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Monika Bieseviciene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Austeja Kaunaite
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Auste Markeviciute
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Agne Bartnykaite
- Oncology Research Laboratory, Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Benas Kireilis
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Diana Rinkuniene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Antanas Jankauskas
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| | - Ignas Gaidamavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Rolandas Gerbutavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Domas Vaitiekus
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Gintare Sakalyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| | - Jolanta Justina Vaskelyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| |
Collapse
|
4
|
Garg R, D’Agostino RB, O’Connell N, Lesser GJ, Salloum FN, Hines AL, Meléndez GC, Jordan JH, Ky B, Wagner LI, Sutton AL, Bottinor W, Olson K, Ladd AC, Hundley WG, The UPBEAT Study Team. Hypertension Severity and Declines in Left Ventricular Ejection Fraction Among Women Receiving Adjuvant Chemotherapy for Breast Cancer (WF-97415 UPBEAT). Hypertension 2024; 81:1365-1373. [PMID: 38634292 PMCID: PMC11304519 DOI: 10.1161/hypertensionaha.123.21817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 04/02/2024] [Indexed: 04/19/2024]
Abstract
BACKGROUND Hypertension is a risk factor for experiencing left ventricular ejection fraction (LVEF) declines during receipt of potentially cardiotoxic breast cancer (BC) treatment. We sought to determine whether the hypertension stage is associated with LVEF decline during BC treatment. METHODS Across 24 centers, cardiac magnetic resonance measures of LVEF and brachial arterial blood pressure (BP) measurements were performed in women with stages I to III BC before and 3 months after initiating potentially cardiotoxic chemotherapy. Using multivariable analysis, we assessed in a blinded fashion the association between 3-month ΔLVEF and precancer treatment American Heart Association/American College of Cardiology stages of hypertension. RESULTS Among 204 women, age averaged 56±1 years with 75% being White and 20% of Black race. Participants received anthracycline (45.6%), trastuzumab (22.5%), cyclophosphamide (52.9%), or paclitaxel (50%). After accounting for pretreatment LVEF, diabetes status, tobacco use, age, the number of antihypertensive medications, and body mass index, those with stage II hypertension experienced an LVEF decline of -2.89% ([95% CI, -0.69% to -5.19%]; P=0.01) relative to individuals with normal BP. Other stages saw nonsignificant declines relative to normal BP to elevated BP (-1.63% [95% CI, -0.62% to 3.88%]; P=0.16) and stage I hypertension (-0.94% [95% CI, -0.90% to 2.78%]; P=0.32). CONCLUSIONS Compared with women receiving treatment for BC with normal BP, there is a stronger association of decline in LVEF in women with stage II hypertension relative to women with other hypertension stages. This raises the possibility that stage along with hypertension presence may be associated with an increased risk for the LVEF decline among women receiving potentially cardiotoxic chemotherapy for BC. REGISTRATION URL: https://www.clinicaltrials.gov; Unique identifier: NCT02791581 and NCT01719562.
Collapse
Affiliation(s)
- Riya Garg
- VCU Pauley Heart Center, Division of Cardiology at Virginia Commonwealth University (VCU), Richmond, Virginia
| | - Ralph B. D’Agostino
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Nathaniel O’Connell
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Glenn J. Lesser
- Section on Hematology and Oncology, Department of Internal Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Fadi N. Salloum
- VCU Pauley Heart Center, Division of Cardiology at Virginia Commonwealth University (VCU), Richmond, Virginia
| | - Anika L. Hines
- Department of Health Behavior and Policy, Virginia Commonwealth University, Richmond, Virginia
| | - Giselle C. Meléndez
- Department of Internal Medicine, Section on Cardiovascular Medicine, Department of Pathology, Section on Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jennifer H. Jordan
- VCU Pauley Heart Center, Division of Cardiology at Virginia Commonwealth University (VCU), Richmond, Virginia
- Department of Biomedical Engineering, Virginia Commonwealth University, Richmond, Virginia
| | - Bonnie Ky
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania
| | - Lynne I. Wagner
- Department of Social Sciences and Health Policy, Division of Public Health Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Arnethea L. Sutton
- Department of Kinesiology and Health Sciences, Virginia Commonwealth University, Richmond, Virginia
| | - Wendy Bottinor
- VCU Pauley Heart Center, Division of Cardiology at Virginia Commonwealth University (VCU), Richmond, Virginia
| | - Kristine Olson
- VCU Pauley Heart Center, Division of Cardiology at Virginia Commonwealth University (VCU), Richmond, Virginia
| | - Amy C. Ladd
- VCU Pauley Heart Center, Division of Cardiology at Virginia Commonwealth University (VCU), Richmond, Virginia
| | - W. Gregory Hundley
- VCU Pauley Heart Center, Division of Cardiology at Virginia Commonwealth University (VCU), Richmond, Virginia
| | | |
Collapse
|
5
|
Vaitiekiene A, Kulboke M, Bieseviciene M, Jankauskas A, Bartnykaite A, Rinkuniene D, Strazdiene I, Lidziute E, Jankauskaite D, Gaidamavicius I, Bucius P, Lapinskas T, Gerbutavicius R, Juozaityte E, Vaskelyte JJ, Vaitiekus D, Sakalyte G. T1 Mapping in Cardiovascular Magnetic Resonance-A Marker of Diffuse Myocardial Fibrosis in Patients Undergoing Hematopoietic Stem Cell Transplantation. J Pers Med 2024; 14:412. [PMID: 38673039 PMCID: PMC11051481 DOI: 10.3390/jpm14040412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/03/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Introduction: Hematopoietic stem cell transplantation (HSCT) recipients are at increased risk of cardiovascular diseases. In our study, we aimed to find subclinical changes in myocardial tissue after HSCT with the help of cardiovascular magnetic resonance (CMR) tissue imaging techniques. Methods: The data of 44 patients undergoing autologous and allogeneic HSCT in the Hospital of Lithuanian University of Health Sciences Kaunas Clinics from October 2021 to February 2023 were analyzed. Bioethics approval for the prospective study was obtained (No BE-2-96). CMR was performed two times: before enrolling for the HSCT procedure (before starting mobilization chemotherapy for autologous HSCT and before starting the conditioning regimen for allogeneic HSCT) and 12 ± 1 months after HSCT. LV end-diastolic volume, LV end-systolic volume, LV mass and values indexed to body surface area (BSA), and LV ejection fraction were calculated. T1 and T2 mapping values were measured. Results: There was a statistically significant change in T1 mapping values. Before HSCT, mean T1 mapping was 1226.13 ± 39.74 ms, and after HSCT, it was 1248.70 ± 41.07 ms (p = 0.01). The other parameters did not differ significantly. Conclusions: Increases in T1 mapping values following HSCT can show the progress of diffuse myocardial fibrosis and may reflect subclinical injury. T2 mapping values remain the same and do not show edema and active inflammation processes at 12 months after HSCT.
Collapse
Affiliation(s)
- Audrone Vaitiekiene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Migle Kulboke
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Monika Bieseviciene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Antanas Jankauskas
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Agne Bartnykaite
- Oncology Research Laboratory, Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Diana Rinkuniene
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Igne Strazdiene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Emilija Lidziute
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Darija Jankauskaite
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Ignas Gaidamavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Paulius Bucius
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Tomas Lapinskas
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Rolandas Gerbutavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Elona Juozaityte
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Jolanta Justina Vaskelyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Domas Vaitiekus
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Gintare Sakalyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| |
Collapse
|
6
|
Belger C, Abrahams C, Imamdin A, Lecour S. Doxorubicin-induced cardiotoxicity and risk factors. IJC HEART & VASCULATURE 2024; 50:101332. [PMID: 38222069 PMCID: PMC10784684 DOI: 10.1016/j.ijcha.2023.101332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2023] [Revised: 12/14/2023] [Accepted: 12/16/2023] [Indexed: 01/16/2024]
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic widely used as a chemotherapeutic agent to treat solid tumours and hematologic malignancies. Although useful in the treatment of cancers, the benefit of DOX is limited due to its cardiotoxic effect that is observed in a large number of patients. In the literature, there is evidence that the presence of various factors may increase the risk of developing DOX-induced cardiotoxicity. A better understanding of the role of these different factors in DOX-induced cardiotoxicity may facilitate the choice of the therapeutic approach in cancer patients suffering from various cardiovascular risk factors. In this review, we therefore discuss the latest findings in both preclinical and clinical research suggesting a link between DOX-induced cardiotoxicity and various risk factors including sex, age, ethnicity, diabetes, dyslipidaemia, obesity, hypertension, cardiovascular disease and co-medications.
Collapse
Affiliation(s)
| | | | - Aqeela Imamdin
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Sandrine Lecour
- Cardioprotection Group, Cape Heart Institute, Department of Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| |
Collapse
|
7
|
Vaitiekiene A, Kulboke M, Bieseviciene M, Bartnykaite A, Kireilis B, Rinkuniene D, Jankauskas A, Zemaitis J, Gaidamavicius I, Gerbutavicius R, Vaitiekus D, Vaskelyte JJ, Sakalyte G. Early Impact of Mobilization Process on Cardiac Function and Size in Patients Undergoing Autologous Hematopoietic Stem Cell Transplantation. J Clin Med 2024; 13:773. [PMID: 38337467 PMCID: PMC10856069 DOI: 10.3390/jcm13030773] [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: 12/14/2023] [Revised: 01/17/2024] [Accepted: 01/26/2024] [Indexed: 02/12/2024] Open
Abstract
Background: The hematopoietic stem cell transplantation (HSCT) process is known to cause cardiac toxicity of different grades. In this paper, we aimed to evaluate the impact of mobilization procedure of hematopoietic stem cells for autologous HSCT process for left and right ventricle sizes and functions. Material and Methods: The data of 47 patients undergoing autologous HSCT were analyzed. All patients underwent hematopoietic stem cell mobilization with chemotherapy and filgrastim at 10 µg/kg/d. Echocardiography was performed two times: before enrolling in the transplantation process and after mobilization before the conditioning regimen for transplantation. Changes in left and right ventricle (RV) diameter and systolic and diastolic function of the left ventricle and systolic function of the RV were measured. Results: A statistically significant difference was observed in the change of right ventricular function (S')-it slightly decreased. Mean S' before mobilization was 13.93 ± 2.85 cm/s, and after mobilization it was 12.19 ± 2.64 cm/s (p = 0.003). No statistically significant change in left ventricular diameter and systolic and diastolic function and RV diameter was observed. Conclusions: The mobilization procedure in patients undergoing autologous HSCT is associated with reduced RV systolic function. S' could be used as a reliable tool to evaluate early cardiotoxicity in HSCT patients and guide further follow-up.
Collapse
Affiliation(s)
- Audrone Vaitiekiene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
| | - Migle Kulboke
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Monika Bieseviciene
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
| | - Agne Bartnykaite
- Oncology Research Laboratory, Oncology Institute, Lithuanian University of Health Sciences, 50161 Kaunas, Lithuania
| | - Benas Kireilis
- Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Diana Rinkuniene
- Institute of Physiology and Pharmacology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Antanas Jankauskas
- Department of Radiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| | - Justinas Zemaitis
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
| | - Ignas Gaidamavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Rolandas Gerbutavicius
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Domas Vaitiekus
- Department of Oncology and Hematology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania
| | - Jolanta Justina Vaskelyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| | - Gintare Sakalyte
- Department of Cardiology, Medical Academy, Lithuanian University of Health Sciences, 44307 Kaunas, Lithuania (J.J.V.)
- Institute of Cardiology, Lithuanian University of Health Sciences, 47181 Kaunas, Lithuania
| |
Collapse
|
8
|
Liao Y, Meng Q. Protection against cancer therapy-induced cardiovascular injury by planed-derived polyphenols and nanomaterials. ENVIRONMENTAL RESEARCH 2023; 238:116896. [PMID: 37586453 DOI: 10.1016/j.envres.2023.116896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 08/18/2023]
Abstract
Cancer therapy-induced heart injury is a significant concern for cancer patients undergoing chemotherapy, radiotherapy, immunotherapy, and also targeted molecular therapy. The use of these treatments can lead to oxidative stress and cardiomyocyte damage in the heart, which can result in heart failure and other cardiac complications. Experimental studies have revealed that chemotherapy drugs such as doxorubicin and cyclophosphamide can cause severe side effects such as cardiac fibrosis, electrophysiological remodeling, chronic oxidative stress and inflammation, etc., which may increase risk of cardiac disorders and attacks for patients that underwent chemotherapy. Similar consequences may also be observed for patients that undergo radiotherapy for left breast or lung malignancies. Polyphenols, a group of natural compounds with antioxidant and anti-inflammatory properties, have shown the potential in protecting against cancer therapy-induced heart injury. These compounds have been found to reduce oxidative stress, necrosis and apoptosis in the heart, thereby preserving cardiac function. In recent years, nanoparticles loaded with polyphenols have also provided for the delivery of these compounds and increasing their efficacy in different organs. These nanoparticles can improve the bioavailability and efficacy of polyphenols while minimizing their toxicity. This review article summarizes the current understanding of the protective effects of polyphenols and nanoparticles loaded with polyphenols against cancer therapy-induced heart injury. The article discusses the mechanisms by which polyphenols protect the heart, including antioxidant and anti-inflammation abilities. The article also highlights the potential benefits of using nanoparticles for the delivery of polyphenols.
Collapse
Affiliation(s)
- Yunshu Liao
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China
| | - Qinghua Meng
- Department of Cardiac Surgery, The First Hospital Affiliated to the Army Medical University, Chongqing, 400038, China.
| |
Collapse
|
9
|
Balaji S, Antony AK, Tonchev H, Scichilone G, Morsy M, Deen H, Mirza I, Ali MM, Mahmoud AM. Racial Disparity in Anthracycline-induced Cardiotoxicity in Breast Cancer Patients. Biomedicines 2023; 11:2286. [PMID: 37626782 PMCID: PMC10452913 DOI: 10.3390/biomedicines11082286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/13/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Breast cancer has become the most common cancer in the US and worldwide. While advances in early detection and treatment have resulted in a 40% reduction in breast cancer mortality, this reduction has not been achieved uniformly among racial groups. A large percentage of non-metastatic breast cancer mortality is related to the cardiovascular effects of breast cancer therapies. These effects appear to be more prevalent among patients from historically marginalized racial/ethnic backgrounds, such as African American and Hispanic individuals. Anthracyclines, particularly doxorubicin and daunorubicin, are the first-line treatments for breast cancer patients. However, their use is limited by their dose-dependent and cumulative cardiotoxicity, manifested by cardiomyopathy, ischemic heart disease, arrhythmias, hypertension, thromboembolic disorders, and heart failure. Cardiotoxicity risk factors, such as genetic predisposition and preexisting obesity, diabetes, hypertension, and heart diseases, are more prevalent in racial/ethnic minorities and undoubtedly contribute to the risk. Yet, beyond these risk factors, racial/ethnic minorities also face unique challenges that contribute to disparities in the emerging field of cardio-oncology, including socioeconomic factors, food insecurity, and the inability to access healthcare providers, among others. The current review will address genetic, clinical, and social determinants that potentially contribute to this disparity.
Collapse
Affiliation(s)
- Swetha Balaji
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Antu K. Antony
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Harry Tonchev
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Giorgia Scichilone
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Mohammed Morsy
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Hania Deen
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Imaduddin Mirza
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Mohamed M. Ali
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
| | - Abeer M. Mahmoud
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (S.B.); (A.K.A.); (H.T.); (G.S.); (M.M.); (H.D.); (I.M.); (M.M.A.)
- Department of Kinesiology, College of Applied Health Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
10
|
Le NT. Metabolic regulation of endothelial senescence. Front Cardiovasc Med 2023; 10:1232681. [PMID: 37649668 PMCID: PMC10464912 DOI: 10.3389/fcvm.2023.1232681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/18/2023] [Indexed: 09/01/2023] Open
Abstract
Endothelial cell (EC) senescence is increasingly recognized as a significant contributor to the development of vascular dysfunction and age-related disorders and diseases, including cancer and cardiovascular diseases (CVD). The regulation of cellular senescence is known to be influenced by cellular metabolism. While extensive research has been conducted on the metabolic regulation of senescence in other cells such as cancer cells and fibroblasts, our understanding of the metabolic regulation of EC senescence remains limited. The specific metabolic changes that drive EC senescence are yet to be fully elucidated. The objective of this review is to provide an overview of the intricate interplay between cellular metabolism and senescence, with a particular emphasis on recent advancements in understanding the metabolic changes preceding cellular senescence. I will summarize the current knowledge on the metabolic regulation of EC senescence, aiming to offer insights into the underlying mechanisms and future research directions.
Collapse
Affiliation(s)
- Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
11
|
Banerjee P, Rosales JE, Chau K, Nguyen MTH, Kotla S, Lin SH, Deswal A, Dantzer R, Olmsted-Davis EA, Nguyen H, Wang G, Cooke JP, Abe JI, Le NT. Possible molecular mechanisms underlying the development of atherosclerosis in cancer survivors. Front Cardiovasc Med 2023; 10:1186679. [PMID: 37332576 PMCID: PMC10272458 DOI: 10.3389/fcvm.2023.1186679] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/17/2023] [Indexed: 06/20/2023] Open
Abstract
Cancer survivors undergone treatment face an increased risk of developing atherosclerotic cardiovascular disease (CVD), yet the underlying mechanisms remain elusive. Recent studies have revealed that chemotherapy can drive senescent cancer cells to acquire a proliferative phenotype known as senescence-associated stemness (SAS). These SAS cells exhibit enhanced growth and resistance to cancer treatment, thereby contributing to disease progression. Endothelial cell (EC) senescence has been implicated in atherosclerosis and cancer, including among cancer survivors. Treatment modalities for cancer can induce EC senescence, leading to the development of SAS phenotype and subsequent atherosclerosis in cancer survivors. Consequently, targeting senescent ECs displaying the SAS phenotype hold promise as a therapeutic approach for managing atherosclerotic CVD in this population. This review aims to provide a mechanistic understanding of SAS induction in ECs and its contribution to atherosclerosis among cancer survivors. We delve into the mechanisms underlying EC senescence in response to disturbed flow and ionizing radiation, which play pivotal role in atherosclerosis and cancer. Key pathways, including p90RSK/TERF2IP, TGFβR1/SMAD, and BH4 signaling are explored as potential targets for cancer treatment. By comprehending the similarities and distinctions between different types of senescence and the associated pathways, we can pave the way for targeted interventions aim at enhancing the cardiovascular health of this vulnerable population. The insights gained from this review may facilitate the development of novel therapeutic strategies for managing atherosclerotic CVD in cancer survivors.
Collapse
Affiliation(s)
- Priyanka Banerjee
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Julia Enterría Rosales
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- School of Medicine, Instituto Tecnológico de Monterrey, Guadalajara, Mexico
| | - Khanh Chau
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Minh T. H. Nguyen
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
- Department of Life Science, University of Science and Technology of Hanoi, Vietnam Academy of Science and Technology, Hanoi, Vietnam
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Steven H. Lin
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Robert Dantzer
- Department of Symptom Research, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Elizabeth A. Olmsted-Davis
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Hung Nguyen
- Cancer Division, Burnett School of Biomedical Science, College of Medicine, University of Central Florida, Orlando, FL, United States
| | - Guangyu Wang
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - John P. Cooke
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| | - Jun-ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Nhat-Tu Le
- Center for Cardiovascular Regeneration, Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, United States
| |
Collapse
|
12
|
Neves MF. Renin-Angiotensin System Inhibition and Beta Blockade Adrenergic May Be Useful to Attenuate Cardiotoxicity by Anthracyclines. Arq Bras Cardiol 2023; 120:e20230280. [PMID: 37341298 PMCID: PMC10263401 DOI: 10.36660/abc.20230280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/22/2023] Open
Affiliation(s)
- Mario Fritsch Neves
- Departamento de Clínica MédicaUniversidade do Estado do Rio de JaneiroRio de JaneiroRJBrasilDepartamento de Clínica Médica – Universidade do Estado do Rio de Janeiro, Rio de Janeiro, RJ – Brasil
| |
Collapse
|
13
|
Jin H, Xu J, Sui Z, Wang L. Risk factors from Framingham risk score for anthracyclines cardiotoxicity in breast cancer: A systematic review and meta-analysis. Front Cardiovasc Med 2023; 10:1101585. [PMID: 36742068 PMCID: PMC9892715 DOI: 10.3389/fcvm.2023.1101585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 01/05/2023] [Indexed: 01/20/2023] Open
Abstract
Background Framingham risk score (FRS) is an effective tool for evaluating the 10-year risk of cardiovascular diseases. However, the sensitivity of FRS for anthracycline-induced cardiotoxicity is unclear. This meta-analysis aims to evaluate the correlation between risk factors (hypertension, hyperlipidemia, diabetes, smoking, and obesity) in FRS and anthracycline-induced cardiotoxicity in breast cancer. Methods We searched PubMed, EMBASE, and Cochrane Library for studies published from inception to January 2022 which reported cardiotoxicity due to anthracycline. Cardiotoxicity defined as any cardiac events were used as the primary endpoint. A total of 33 studies involving 55,708 breast cancer patients treated with anthracyclines were included in this meta-analysis. Results At least one risk factor was identified at baseline for the 55,708 breast cancer patients treated with anthracycline. Hypertension [I 2 = 45%, Fixed, RR (95% CI) = 1.40 (1.22, 1.60), p < 0.00001], hyperlipidemia [I 2 = 0%, Fixed, RR (95% CI): 1.35 (1.12, 1.62), p = 0.002], diabetes [I 2 = 0%, Fixed, RR (95% CI): 1.29 (1.05, 1.57), p = 0.01], and obesity [I 2 = 0%, Fixed, RR (95% CI): 1.32 (1.05, 1.67), p = 0.02] were associated with increased risks of cardiac events. In addition, smoking was also associated with reduced left ventricular ejection fraction (LVEF) during anthracycline chemotherapy [I 2 = 0%, Fixed, OR (95% CI): 1.91 (1.24, 2.95), p = 0.003] in studies that recorded only the odds ratio (OR). Conclusion Hypertension, hyperlipidemia, diabetes, smoking, and obesity are associated with increased risks of anthracycline-induced cardiotoxicity. Therefore, corresponding measures should be used to manage cardiovascular risk factors in breast cancer during and after anthracycline treatment.
Collapse
Affiliation(s)
- Hao Jin
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jianfeng Xu
- Department of Orthopedics, Affiliated Zhongshan Hospital of Dalian University, Dalian, China
| | - Zheng Sui
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Lili Wang
- Department of Cardiology, Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|
14
|
Philip LJ, Findlay SG, Gill JH. Baseline blood pressure and development of cardiotoxicity in patients treated with anthracyclines: A systematic review. INTERNATIONAL JOURNAL OF CARDIOLOGY. CARDIOVASCULAR RISK AND PREVENTION 2022; 15:200153. [PMID: 36573186 PMCID: PMC9789356 DOI: 10.1016/j.ijcrp.2022.200153] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 09/29/2022] [Accepted: 10/03/2022] [Indexed: 11/07/2022]
Abstract
Aims Anthracyclines, a mainstay of cancer treatment, are associated with significant life-threatening cardiotoxicity. As cancer survivorship improves, there is a growing need to identify patients most at risk and strategies to mitigate anthracycline-associated cardiotoxicity. Elevated baseline blood pressure (bBP) is a possible risk factor for cardiotoxicity. The aim of this systematic review was to summarise the literature and evaluate relationships between bBP and anthracycline-associated cardiotoxicity. Methods and results Systematic searches were conducted, limited to English language but without restrictions on study type or country of origin. All studies fulfilled the PRISMA statement and relevant studies reviewed and narratively synthesised. A total of 1330 papers were screened, with 12 included in the qualitative synthesis. Eight papers indicated elevated bBP was associated with significantly higher risk of developing cardiotoxicity. Four papers noted significant relationships between left ventricular ejection fraction (LVEF) decline and elevated bBP. Of the four papers that failed to show an association, one noted increased risk of developing chronic heart failure. A relationship between baseline diastolic and systolic BP and anthracycline-associated cardiotoxicity is also noted. Conclusions This study indicates adult patients with elevated bBP have increased vulnerability to anthracycline-associated cardiotoxicity, with those with pre-hypertension or raised systolic versus diastolic pressure potentially an overlooked population. Recommendations for inclusion of bBP, incorporating individual systolic versus diastolic pressures, in cardio-oncology risk prediction models to guide clinical decision-making are thus warranted.
Collapse
Affiliation(s)
- Laura J. Philip
- School of Pharmacy, Faculty of Medical Sciences, Newcastle University, UK
| | - Simon G. Findlay
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK,Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle University, UK
| | - Jason H. Gill
- School of Pharmacy, Faculty of Medical Sciences, Newcastle University, UK,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, UK,Newcastle University Centre for Cancer, Faculty of Medical Sciences, Newcastle University, UK,Corresponding author. School of Pharmacy, Faculty of Medical Sciences, Newcastle University, King George VI Building, Newcastle upon Tyne, NE1 7RU, UK.
| |
Collapse
|
15
|
Zhang M, Yang H, Xu C, Jin F, Zheng A. Risk Factors for Anthracycline-Induced Cardiotoxicity in Breast Cancer Treatment: A Meta-Analysis. Front Oncol 2022; 12:899782. [PMID: 35785172 PMCID: PMC9248259 DOI: 10.3389/fonc.2022.899782] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/11/2022] [Indexed: 01/05/2023] Open
Abstract
Background Anthracyclines play an important role in the treatment of breast cancer (BC) and other malignant tumors. However, accompanied side-effects are non-ignorable. The purpose of this meta-analysis is to determine the risk factors for anthracycline-induced cardiotoxicity (ACT), so as to identify high-risk patients. Methods The search for literature was conducted in PubMed, The Cochrane Library, Embase and Web of science. Records were selected with inclusion criteria and exclusion criteria. The newcastle-ottawa scale (NOS) was used to assess the quality of literature, and Review Manager 5.3 software was used for meta-analysis. Results Thirteen studies met the inclusion criteria. Meta-analysis indicated that risk factors for ACT were use of trastuzumab (odds ratio [OR]: 2.84, 95% confidence interval [CI]: 2.49-3.22, p < 0.00001), cumulative dose of anthracyclines (OR: 1.45, 95%CI: 1.28-1.65, p < 0.00001), hypertension (OR: 2.95, 95%CI: 1.75-4.97, p < 0.0001), diabetes mellitus (DM) (OR: 1.39, 95%CI: 1.20-1.61, p < 0.0001), tumor metastasis (OR: 1.91, 95%CI: 1.17-3.11, p = 0.009) and coronary heart disease (CAD) (OR: 2.17, 95%CI: 1.50-3.15, p < 0.0001). In addition, our analysis revealed that body mass index (BMI) had no effect on ACT (OR: 1.18, 95%CI: 0.98-1.43, p = 0.08). Conclusions Patients with high risk for ACT can be identified by these factors. For such patients, a higher level of monitoring and protection for the cardiac function should be performed by clinicians. Systematic Review Registration INPLASY, identifier INPLASY202250140.
Collapse
Affiliation(s)
- Meilin Zhang
- Department of Burn Plastic Surgery, Chaoyang Central Hospital, Chaoyang, China
| | - Hongguang Yang
- Department of Burn Plastic Surgery, Chaoyang Central Hospital, Chaoyang, China
| | - Changcun Xu
- Department of Cardiology, Chaoyang Central Hospital, Chaoyang, China
| | - Feng Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Feng Jin, ; Ang Zheng,
| | - Ang Zheng
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Feng Jin, ; Ang Zheng,
| |
Collapse
|
16
|
Kwan ML, Cheng RK, Iribarren C, Neugebauer R, Rana JS, Nguyen-Huynh M, Shi Z, Laurent CA, Lee VS, Roh JM, Shen H, Rillamas-Sun E, Santiago-Torres M, Hershman DL, Kushi LH, Greenlee H. Risk of Cardiometabolic Risk Factors in Women With and Without a History of Breast Cancer: The Pathways Heart Study. J Clin Oncol 2022; 40:1635-1646. [PMID: 35025627 PMCID: PMC9113213 DOI: 10.1200/jco.21.01738] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE The incidence of cardiometabolic risk factors in breast cancer (BC) survivors has not been well described. Thus, we compared risk of hypertension, diabetes, and dyslipidemia in women with and without BC. METHODS Women with invasive BC diagnosed from 2005 to 2013 at Kaiser Permanente Northern California (KPNC) were identified and matched 1:5 to noncancer controls on birth year, race, and ethnicity. Cumulative incidence rates of hypertension, diabetes, and dyslipidemia were estimated with competing risk of overall death. Subdistribution hazard ratios (sHRs) were estimated by Fine and Gray regression, adjusted for cardiovascular disease-related risk factors, and stratified by treatment and body mass index (BMI). RESULTS A total of 14,942 BC cases and 74,702 matched controls were identified with mean age 61.2 years and 65% non-Hispanic White. Compared with controls, BC cases had higher cumulative incidence rates of hypertension (10.9% v 8.9%) and diabetes (2.1% v 1.7%) after 2 years, with higher diabetes incidence persisting after 10 years (9.3% v 8.8%). In multivariable models, cases had higher risk of diabetes (sHR, 1.16; 95% CI, 1.07 to 1.26) versus controls. Cases treated with chemotherapy (sHR, 1.23; 95% CI, 1.11 to 1.38), left-sided radiation (sHR, 1.29; 95% CI, 1.13 to 1.48), or endocrine therapy (sHR, 1.23; 95% CI, 1.12 to 1.34) continued to have higher diabetes risk. Hypertension risk was higher for cases receiving left-sided radiation (sHR, 1.11; 95% CI, 1.02 to 1.21) or endocrine therapy (sHR, 1.10; 95% CI, 1.03 to 1.16). Normal-weight (BMI < 24.9 kg/m2) cases had higher risks overall and within treatment subgroups versus controls. CONCLUSION BC survivors at KPNC experienced elevated risks of diabetes and hypertension compared with women without BC depending on treatments received and BMI. Future studies should examine strategies for cardiometabolic risk factor prevention in BC survivors.
Collapse
Affiliation(s)
- Marilyn L Kwan
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Richard K Cheng
- University of Washington School of Medicine, Seattle, WA.,Seattle Cancer Care Alliance, Seattle, WA, US
| | - Carlos Iribarren
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Romain Neugebauer
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Jamal S Rana
- Division of Research, Kaiser Permanente Northern California, Oakland, CA.,Oakland Medical Center, Kaiser Permanente Northern California, Oakland, CA
| | - Mai Nguyen-Huynh
- Division of Research, Kaiser Permanente Northern California, Oakland, CA.,Walnut Creek Medical Center, Kaiser Permanente Northern California, Walnut Creek, CA
| | - Zaixing Shi
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics and Key Laboratory of Health Technology Assessment of Fujian Province, School of Public Health, Xiamen University, Xiamen, China.,Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Cecile A Laurent
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Valerie S Lee
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Janise M Roh
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Hanjie Shen
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Eileen Rillamas-Sun
- Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| | | | - Dawn L Hershman
- Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY
| | - Lawrence H Kushi
- Division of Research, Kaiser Permanente Northern California, Oakland, CA
| | - Heather Greenlee
- University of Washington School of Medicine, Seattle, WA.,Seattle Cancer Care Alliance, Seattle, WA, US.,Division of Public Health Sciences, Fred Hutchinson Cancer Research Center, Seattle, WA
| |
Collapse
|
17
|
Xiu W, Huang Y, Li Y, Yu M, Gong Y. Comorbidities and mortality risk among extensive-stage small-cell lung cancer patients in mainland China: impacts of hypertension, type 2 diabetes mellitus, and chronic hepatitis B virus infection. Anticancer Drugs 2022; 33:80-90. [PMID: 34183497 PMCID: PMC8670332 DOI: 10.1097/cad.0000000000001133] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 06/08/2021] [Indexed: 02/05/2023]
Abstract
The present study investigated the impact of major comorbidities, including hypertension, type 2 diabetes mellitus (T2DM), and chronic hepatitis B virus (HBV) infection, on the progression-free survival (PFS) and overall survival (OS) of extensive-stage small-cell lung cancer (ES-SCLC) patients in China. Patients having a pathologic diagnosis of ES-SCLC between 2009 and 2017 were enrolled and grouped according to their specific comorbidities. The PFS and OS for each group were evaluated using the Kaplan-Meier method and Cox proportional hazard models. In total, 632 patients were analyzed. The median PFS (mPFS) of these patients was 9 months [95% confidence interval (CI), 6-12 months]. The mPFS of patients without hypertension or T2DM was 9 months; conversely, it was significantly reduced for patients with hypertension [7 months (P < 0.0001)] or T2DM [5 months (P < 0.0001)]. However, mPFS was not significantly different between patients with and without HBV infection (P = 0.2936). A similar trend was observed for OS as well. Further multivariate analyses showed that the OS of patients with hypertension [hazard ratio (HR), 1.344; 95% CI, 1.073-1.683; P = 0.010] or T2DM (HR, 1.455; 95% CI, 1.134-1.868; P = 0.003) was significantly shorter than that of patients without these comorbidities. Accordingly, mortality risk was the highest in patients with concurrent hypertension and T2DM (HR, 1.665; 95% CI, 1.037-2.672; P = 0.00058). Our study found that hypertension and T2DM may be associated with a worse prognosis in ES-SCLC patients. Considerable attention should be paid to the accompanying anti-comorbidity therapies available for patients with ES-SCLC.
Collapse
Affiliation(s)
- Weigang Xiu
- Department of Thoracic Oncology and State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yin Huang
- Department of Thoracic Oncology and State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Yanying Li
- Department of Thoracic Oncology and State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Min Yu
- Department of Thoracic Oncology and State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| | - Youling Gong
- Department of Thoracic Oncology and State Key Laboratory of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, PR China
| |
Collapse
|
18
|
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
|
19
|
Qiu S, Zhou T, Qiu B, Zhang Y, Zhou Y, Yu H, Zhang J, Liu L, Yuan L, Yang G, Duan Y, Xing C. Risk Factors for Anthracycline-Induced Cardiotoxicity. Front Cardiovasc Med 2021; 8:736854. [PMID: 34660739 PMCID: PMC8511483 DOI: 10.3389/fcvm.2021.736854] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022] Open
Abstract
Background: Several cardiovascular risk factors have been suggested to be associated with anthracycline-induced cardiotoxicity, but their quantitative effects have not reached a consensus. Methods: We searched PubMed, EMBASE, and Cochrane Library databases for manuscripts published from inception to February 2021, which reported the results of cardiotoxicity due to anthracycline chemotherapy without trastuzumab. Cardiotoxicity defined by any reduction of left ventricular eject fraction (LVEF) to below 50% or a >10% reduction from baseline was defined as the primary endpoint. Odd ratios (OR) with 95% confidence intervals (CI) were calculated using a random-effects model meta-analysis. Results: A total of 7,488 patients receiving anthracycline chemotherapy without trastuzumab were included, who had at least one risk factor at baseline. Hypertension (OR: 1.99; 95% CI: 1.43–2.76), diabetes mellitus (OR: 1.74; 95% CI: 1.11–2.74), and obesity (OR: 1.72; 95% CI: 1.13–2.61) were associated with increased risk of cardiotoxicity. In addition, the relative reduction of global longitudinal strain (GLS) from baseline after anthracycline treatment could significantly improve the detection ability of cardiotoxicity (28.5%, 95% CI: 22.1–35.8% vs. 16.4%, 95% CI: 13.4–19.9%) compared with LVEF. The early detection rate of anthracycline-induced cardiotoxicity (3 months after chemotherapy) by GLS was 30.2% (95% CI: 24.9–36.1%), which is similar with the overall result of GLS. Conclusions: Hypertension, diabetes mellitus, and obesity are associated with increased risk of anthracycline-induced cardiotoxicity, which indicates that corresponding protective strategies should be used during and after anthracycline treatment. The findings of higher detection rate and better early detection ability for cardiotoxicity than LVEF added new proofs for the advantages of GLS in detection of AIC.
Collapse
Affiliation(s)
- Shuo Qiu
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Tian Zhou
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Bo Qiu
- Department of General Surgery, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yuxin Zhang
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Yonggang Zhou
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Huihui Yu
- School of Nursing, Air Force Medical University, Xi'an, China
| | - Jingyi Zhang
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Li Liu
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Lijun Yuan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Guodong Yang
- Department of Biochemistry and Molecular Biology, Air Force Medical University, Xi'an, China
| | - Yunyou Duan
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Changyang Xing
- Department of Ultrasound Diagnostics, Tangdu Hospital, Air Force Medical University, Xi'an, China
| |
Collapse
|
20
|
Hsu PY, Mammadova A, Benkirane-Jessel N, Désaubry L, Nebigil CG. Updates on Anticancer Therapy-Mediated Vascular Toxicity and New Horizons in Therapeutic Strategies. Front Cardiovasc Med 2021; 8:694711. [PMID: 34386529 PMCID: PMC8353082 DOI: 10.3389/fcvm.2021.694711] [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: 04/13/2021] [Accepted: 06/18/2021] [Indexed: 12/11/2022] Open
Abstract
Vascular toxicity is a frequent adverse effect of current anticancer chemotherapies and often results from endothelial dysfunction. Vascular endothelial growth factor inhibitors (VEGFi), anthracyclines, plant alkaloids, alkylating agents, antimetabolites, and radiation therapy evoke vascular toxicity. These anticancer treatments not only affect tumor vascularization in a beneficial manner, they also damage ECs in the heart. Cardiac ECs have a vital role in cardiovascular functions including hemostasis, inflammatory and coagulation responses, vasculogenesis, and angiogenesis. EC damage can be resulted from capturing angiogenic factors, inhibiting EC proliferation, survival and signal transduction, or altering vascular tone. EC dysfunction accounts for the pathogenesis of myocardial infarction, atherothrombosis, microangiopathies, and hypertension. In this review, we provide a comprehensive overview of the effects of chemotherapeutic agents on vascular toxicity leading to hypertension, microvascular rarefaction thrombosis and atherosclerosis, and affecting drug delivery. We also describe the potential therapeutic approaches such as vascular endothelial growth factor (VEGF)-B and prokineticin receptor-1 agonists to maintain endothelial function during or following treatments with chemotherapeutic agents, without affecting anti-tumor effectiveness.
Collapse
Affiliation(s)
| | | | | | | | - Canan G. Nebigil
- INSERM UMR 1260, Regenerative Nanomedicine, University of Strasbourg, FMTS (Fédération de Médecine Translationnelle de l'Université de Strasbourg), Strasbourg, France
| |
Collapse
|
21
|
Alzahrani AM, Rajendran P, Veeraraghavan VP, Hanieh H. Cardiac Protective Effect of Kirenol against Doxorubicin-Induced Cardiac Hypertrophy in H9c2 Cells through Nrf2 Signaling via PI3K/AKT Pathways. Int J Mol Sci 2021; 22:ijms22063269. [PMID: 33806909 PMCID: PMC8004766 DOI: 10.3390/ijms22063269] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 12/14/2022] Open
Abstract
Kirenol (KRL) is a biologically active substance extracted from Herba Siegesbeckiae. This natural type of diterpenoid has been widely adopted for its important anti-inflammatory and anti-rheumatic properties. Despite several studies claiming the benefits of KRL, its cardiac effects have not yet been clarified. Cardiotoxicity remains a key concern associated with the long-term administration of doxorubicin (DOX). The generation of reactive oxygen species (ROS) causes oxidative stress, significantly contributing to DOX-induced cardiac damage. The purpose of the current study is to investigate the cardio-protective effects of KRL against apoptosis in H9c2 cells induced by DOX. The analysis of cellular apoptosis was performed using the terminal deoxynucleotidyl transferase dUTP nick-end labeling (TUNEL) staining assay and measuring the modulation in the expression levels of proteins involved in apoptosis and Nrf2 signaling, the oxidative stress markers. Furthermore, Western blotting was used to determine cell survival. KRL treatment, with Nrf2 upregulation and activation, accompanied by activation of PI3K/AKT, could prevent the administration of DOX to induce cardiac oxidative stress, remodeling, and other effects. Additionally, the diterpenoid enhanced the activation of Bcl2 and Bcl-xL, while suppressing apoptosis marker proteins. As a result, KRL is considered a potential agent against hypertrophy resulting from cardiac deterioration. The study results show that KRL not only activates the IGF-IR-dependent p-PI3K/p-AKT and Nrf2 signaling pathway, but also suppresses caspase-dependent apoptosis.
Collapse
Affiliation(s)
- Abdullah M. Alzahrani
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia;
| | - Peramaiyan Rajendran
- Department of Biological Sciences, College of Science, King Faisal University, Al Ahsa 31982, Saudi Arabia;
- Correspondence: ; Tel.: +97-0135899543
| | - Vishnu Priya Veeraraghavan
- Department of Biochemistry, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India;
| | - Hamza Hanieh
- Department of Medical Analysis, Al-Hussein Bin Talal University, Ma’an 71111, Jordan;
| |
Collapse
|
22
|
Tantawy M, Pamittan FG, Singh S, Gong Y. Epigenetic Changes Associated With Anthracycline-Induced Cardiotoxicity. Clin Transl Sci 2021; 14:36-46. [PMID: 32770710 PMCID: PMC7877852 DOI: 10.1111/cts.12857] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Accepted: 07/11/2020] [Indexed: 12/24/2022] Open
Abstract
Advances in cancer treatment have significantly improved the survival of patients with cancer, but, unfortunately, many of these treatments also have long-term complications. Cancer treatment-related cardiotoxicities are becoming a significant clinical problem that a new discipline, Cardio-Oncology, was established to advance the cardiovascular care of patients with growing cancer populations. Anthracyclines are a class of chemotherapeutic agents used to treat many cancers in adults and children. Their clinical use is limited by anthracycline-induced cardiotoxicity (AIC), which can lead to heart failure. Early-onset cardiotoxicity appears within a year of treatment, whereas late-onset cardiotoxicity occurs > 1 year and even up to decades after treatment completion. The pathophysiology of AIC was hypothesized to be caused by generation of reactive oxygen species that lead to lipid peroxidation, defective mitochondrial biogenesis, and DNA damage of the cardiomyocytes. The accumulation of anthracycline metabolites was also proposed to cause mitochondrial damage and the induction of cardiac cell apoptosis, which induces arrhythmias, contractile dysfunction, and cardiomyocyte death. This paper will provide a general overview of cardiotoxicity focusing on the effect of anthracyclines and their epigenetic molecular mechanisms on cardiotoxicity.
Collapse
Affiliation(s)
- Marwa Tantawy
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | - Frances G. Pamittan
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
| | | | - Yan Gong
- Department of Pharmacotherapy and Translational ResearchCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- Center for Pharmacogenomics and Precision MedicineCollege of PharmacyUniversity of FloridaGainesvilleFloridaUSA
- UF Health Cancer CenterGainesvilleFloridaUSA
| |
Collapse
|
23
|
Wang N, Zhang Y, Liu H, Wang A, Ren T, Gou J, Zhang Y, Yin T, He H, Tang X. Toxicity Reduction and Efficacy Promotion of Doxorubicin in the Treatment of Breast Tumors Assisted by Enhanced Oral Absorption of Curcumin-Loaded Lipid-Polyester Mixed Nanoparticles. Mol Pharm 2020; 17:4533-4547. [PMID: 33201717 DOI: 10.1021/acs.molpharmaceut.0c00718] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Curcumin (CUR), a polyphenol derived from turmeric, exhibits anticancer and anti-inflammatory properties. However, it has poor water solubility, stability, and oral bioavailability. To overcome these limitations, lipid-polyester mixed nanoparticles (NPs) embedded in enteric polymer-EudragitL100-55(Eu) were formulated (CUR-NPs-Eu). NPs composed of mPEG-b-PCL have a hybrid core made up of middle chain triglyceride (MCT) and poly(ε-caprolactone) (PCL) for enhancing drug loading. The CUR-NPs with MCT content of 10% had a particle size of 121.2 ± 16.8 nm, ζ potential of -16.25 ± 1.38 mV, drug loading of 9.8%, and encapsulation efficiency of 87.4%. The transport of the CUR-NPs-Eu across Caco-2 monolayers is enhanced compared with CUR alone (1.98 ± 0.94 × 10-6 of curcumin versus 55.43 ± 6.06 × 10-6 cm/s of curcumin-loaded NPs) because of the non-disassociated nanostructure during absorption. The absolute bioavailability of CUR-NPs-Eu was 7.14%, which was drastically improved from 1.08% of the CUR suspension (CUR-Sus). Therefore, in the xenograft 4T1 tumor-bearing mice, increased drug accumulation in heart and tumor was noticed because of enhanced oral bioavailability of CUR. The chemosensitizing effect of CUR was attributed to its NF-κB reduction effect (148 ± 11.83 of DOX alone versus 104 ± 8.71 of combined therapy, ng/g tissue). The cardioprotective effect of CUR was associated with maintenance of cardiac antioxidant enzyme activity and down-regulation of NF-κB. This study provided a partial illustration of the mechanisms of chemosensitizing and cardioprotective effects of CUR utilizing the oral availability promotion effect brought by the NPs-Eu formulation. And these results further demonstrated that the capability of this NPs-Eu system in oral delivery of poorly soluble and poorly permeable drugs.
Collapse
Affiliation(s)
- Na Wang
- School of Traditional Chinese Medicine, Shenyang Pharmaceutical University, No. 103 Wenhua Road, Shenyang 110016, China
| | | | | | - Andong Wang
- School of Pharmacy, Nantong University, Nantong 226001, China
| | - Tianyang Ren
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | | | | | | | | | | |
Collapse
|
24
|
HFE Gene Variants' Impact on Anthracycline-Based Chemotherapy-Induced Subclinical Cardiotoxicity. Cardiovasc Toxicol 2020; 21:59-66. [PMID: 32748118 DOI: 10.1007/s12012-020-09595-1] [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] [Received: 05/04/2020] [Accepted: 07/24/2020] [Indexed: 02/08/2023]
Abstract
Progress in oncology has allowed to improve outcomes in many breast cancer patients. The core stone of breast cancer chemotherapy is anthracycline-based chemotherapy. Unfortunately, anthracyclines cause cardiotoxicity which is a limiting factor of its use and lifetime cumulative dose of anthracyclines is the major risk factor for cardiotoxicity. With evolution of echocardiography subclinical damage is identified, and more sensitive evaluation can be performed. This leads to understanding the heart damage beyond cumulative dose in early phase and importance of other risk factors. There are many risk factors for anthracycline-based chemotherapy cardiotoxicity (ABCC) like arterial hypertension, obesity, diabetes, genetic predisposition, etc. One of possible pathophysiological pathways is iron metabolism, especially HFE gene-regulated iron metabolism pathway. Pre-existing genetic iron metabolism dysregulation increases risk for ABCC. Clinical studies and experimental models in mice have shown potential impact of HFE gene SNP on ABCC. The main objective of our study was to identify the impact of HFE C282Y and H63D SNP on the development of subclinical heart damage during and/or after doxorubicin-based chemotherapy in breast cancer patients. Data of 81 women with breast cancer treated with doxorubicin-based chemotherapy in the outpatient clinic were analyzed and SNP RT-PCR tests were performed. Statistically significant association between H63D and ABCC after completion of chemotherapy was observed (p < 0.005). Consequently, our study demonstrated that H63D SNP has an important role in the development of ABCC. HFE SNP mutation status could be used as one of important tools to identify high-risk patients for ABCC.
Collapse
|
25
|
Mercurio V, Cuomo A, Cadeddu Dessalvi C, Deidda M, Di Lisi D, Novo G, Manganaro R, Zito C, Santoro C, Ameri P, Spallarossa P, Arboscello E, Tocchetti CG, Penna C. Redox Imbalances in Ageing and Metabolic Alterations: Implications in Cancer and Cardiac Diseases. An Overview from the Working Group of Cardiotoxicity and Cardioprotection of the Italian Society of Cardiology (SIC). Antioxidants (Basel) 2020; 9:E641. [PMID: 32708201 PMCID: PMC7402085 DOI: 10.3390/antiox9070641] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/14/2020] [Accepted: 07/16/2020] [Indexed: 12/14/2022] Open
Abstract
Metabolic syndrome (MetS) is a well established risk factor for cardiovascular (CV) diseases. In addition, several studies indicate that MetS correlates with the increased risk of cancer in adults. The mechanisms linking MetS and cancer are not fully understood. Several risk factors involved in MetS are also cancer risk factors, such as the consumption of high calorie-food or high fat intake, low fibre intake, and sedentary lifestyle. Other common aspects of both cancer and MetS are oxidative stress and inflammation. In addition, some anticancer treatments can induce cardiotoxicity, including, for instance, left ventricular (LV) dysfunction and heart failure (HF), endothelial dysfunction and hypertension. In this review, we analyse several aspects of MetS, cancer and cardiotoxicity from anticancer drugs. In particular, we focus on oxidative stress in ageing, cancer and CV diseases, and we analyse the connections among CV risk factors, cancer and cardiotoxicity from anticancer drugs.
Collapse
Affiliation(s)
- Valentina Mercurio
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy; (V.M.); (A.C.)
| | - Alessandra Cuomo
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy; (V.M.); (A.C.)
| | - Christian Cadeddu Dessalvi
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy; (C.C.D.); (M.D.)
| | - Martino Deidda
- Department of Medical Sciences and Public Health, University of Cagliari, 09042 Cagliari, Italy; (C.C.D.); (M.D.)
| | - Daniela Di Lisi
- Cardiology Unit AUOP Policlinico, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.N.)
| | - Giuseppina Novo
- Cardiology Unit AUOP Policlinico, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (D.D.L.); (G.N.)
| | - Roberta Manganaro
- Cardiology with Coronary Intensive Care Unit, Department of Clinical and Experimental Medicine, University Hospital Policlinico “G. Martino”, University of Messina, 98124 Messina, Italy; (R.M.); (C.Z.)
| | - Concetta Zito
- Cardiology with Coronary Intensive Care Unit, Department of Clinical and Experimental Medicine, University Hospital Policlinico “G. Martino”, University of Messina, 98124 Messina, Italy; (R.M.); (C.Z.)
| | - Ciro Santoro
- Department of Advanced Biomedical Sciences, Federico II University, 80131 Naples, Italy;
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy; (P.A.); (P.S.); (E.A.)
| | - Paolo Spallarossa
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy; (P.A.); (P.S.); (E.A.)
| | - Eleonora Arboscello
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy—IRCCS Italian Cardiovascular Network & Department of Internal Medicine, University of Genova, 16121 Genova, Italy; (P.A.); (P.S.); (E.A.)
| | - Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, 80131 Naples, Italy; (V.M.); (A.C.)
- Interdepartmental Center of Clinical and Translational Sciences, Federico II University, 80131 Naples, Italy
| | - Claudia Penna
- Department of Clinical and Biological Sciences, University of Turin, 10043 Torino, Italy
| |
Collapse
|