1
|
Zhu G, Cao L, Wu J, Xu M, Zhang Y, Wu M, Li J. Co-morbid intersections of cancer and cardiovascular disease and targets for natural drug action: Reprogramming of lipid metabolism. Biomed Pharmacother 2024; 176:116875. [PMID: 38850662 DOI: 10.1016/j.biopha.2024.116875] [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: 04/01/2024] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024] Open
Abstract
Cancer and cardiovascular diseases are major contributors to global morbidity and mortality, and their seemingly separate pathologies are intricately intertwined. In the context of cancer, the cardiovascular disease encompasses not only the side effects arising from anti-tumor treatments but also the metabolic shifts induced by oncological conditions. A growing body of research indicates that lipid metabolic reprogramming serves as a distinctive hallmark of tumors. Furthermore, anomalies in lipid metabolism play a significant role in the development of cardiovascular disease. This study delves into the cardiac implications of lipid metabolic reprogramming within the cancer context, closely examining abnormalities in lipid metabolism present in tumors, cardiac tissue, and immune cells within the microenvironment. Additionally, we examined risk factors such as obesity and anti-tumor therapy. Despite progress, a gap remains in the availability of drugs targeting lipid metabolism modulation for treating tumors and mitigating cardiac risk, with limited advancement seen in prior studies. Here, we present a review of previous research on natural drugs that exhibit both shared and distinct therapeutic effects on tumors and cardiac health by modulating lipid metabolism. Our aim is to provide insights for potential drug development.
Collapse
Affiliation(s)
- Guanghui Zhu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Luchang Cao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Jingyuan Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China; Graduate School, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Manman Xu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China
| | - Ying Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Min Wu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Jie Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
2
|
Kreidieh F, McQuade J. Novel insights into cardiovascular toxicity of cancer targeted and immune therapies: Beyond ischemia with non-obstructive coronary arteries (INOCA). AMERICAN HEART JOURNAL PLUS : CARDIOLOGY RESEARCH AND PRACTICE 2024; 40:100374. [PMID: 38510501 PMCID: PMC10946000 DOI: 10.1016/j.ahjo.2024.100374] [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: 02/14/2024] [Accepted: 02/20/2024] [Indexed: 03/22/2024]
Abstract
Novel immune and targeted therapies approved over the past 2 decades have resulted in dramatic improvements in cancer-specific outcomes for many cancer patients. However, many of these agents can induce cardiovascular toxicity in a subset of patients. The field of cardio-oncology was established based on observations that anti-neoplastic chemotherapies and mantle radiation can lead to premature cardiomyopathy in cancer survivors. While conventional chemotherapy, targeted therapy, and immune therapies can all result in cardiovascular adverse events, the mechanisms, timing, and incidence of these events are inherently different. Many of these effects converge upon the coronary microvasculature to involve, through endocardial endothelial cells, a more direct effect through close proximity to cardiomyocyte with cellular communication and signaling pathways. In this review, we will provide an overview of emerging paradigms in the field of Cardio-Oncology, particularly the role of the coronary microvasculature in mediating cardiovascular toxicity of important cancer targeted and immune therapies. As the number of cancer patients treated with novel immune and targeted therapies grows exponentially and subsequently the number of long-term cancer survivors dramatically increases, it is critical that cardiologists and cardiology researchers recognize the unique potential cardiovascular toxicities of these agents.
Collapse
Affiliation(s)
- Firas Kreidieh
- Instructor of Clinical Medicine- Division of Hematology-Oncology; Associate Director- Internal Medicine Residency Program, American University of Beirut, Beirut, Lebanon
| | - Jennifer McQuade
- Associate Professor and Physician Scientist in Melanoma Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, United States of America
| |
Collapse
|
3
|
Di Marco A, Artioli G, Favaretto A, Cavasin N, Basso U. Multiorgan failure caused by pembrolizumab and axitinib in a woman affected by metastatic clear cell renal cell carcinoma: A case report and literature review. Medicine (Baltimore) 2024; 103:e37606. [PMID: 38552059 PMCID: PMC10977559 DOI: 10.1097/md.0000000000037606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/23/2024] [Indexed: 04/02/2024] Open
Abstract
RATIONALE Treatment with a combination of immune checkpoint inhibitors (ICIs) (pembrolizumab or nivolumab) and oral Tyrosine Kinase Inhibitors (TKI) targeting angiogenesis (axitinib, cabozantinib or lenvatinib) has shown benefits in terms of efficacy and survival in metastatic renal cell carcinoma (mRCC), with a favorable toxicity profile. However, some rare and serious treatment-related adverse events can be difficult to manage. PATIENT CONCERNS Here we report the first case of an mRCC patient who, after only 2 administrations of pembrolizumab-axitinib, experienced severe multiorgan failure (MOF) with heart failure, oliguria and acute hepatitis requiring aggressive supportive treatment in intensive care unit. DIAGNOSES A diagnosis of severe MOF induced by pembrolizumab plus axitinib was considered. INTERVENTIONS The patient was treated with dobutamine, levosimendan along with high-dose steroids under continuous cardiologic monitoring. OUTCOMES After treatment, the patient had a full recovery and was discharged from the hospital. LESSONS We reviewed all the other cases of MOF reported during treatment with combined ICI-TKI in cancer patients in order to summarize incidence, clinical manifestations and management with a specific focus on the need for prompt recognition and aggressive management under multidisciplinary care.
Collapse
Affiliation(s)
- Andrea Di Marco
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Grazia Artioli
- Department of Medical Oncology, AULSS 2 Marca Trevigiana, Ca’ Foncello Hospital, Treviso, Italy
| | - Adolfo Favaretto
- Department of Medical Oncology, AULSS 2 Marca Trevigiana, Ca’ Foncello Hospital, Treviso, Italy
| | - Nicolò Cavasin
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padua, Italy
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Umberto Basso
- Medical Oncology 1 Unit, Department of Oncology, Istituto Oncologico Veneto IOV IRCCS, Padua, Italy
| |
Collapse
|
4
|
Chen Y, Huang C, Lien L, Chen J, Hsieh F. Cardiovascular Toxicity of Angiogenesis Inhibitors Among Patients With Cancer in Taiwan: A Nested Case-Control Study. J Am Heart Assoc 2024; 13:e030263. [PMID: 38156594 PMCID: PMC10863808 DOI: 10.1161/jaha.123.030263] [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: 04/01/2023] [Accepted: 12/06/2023] [Indexed: 12/30/2023]
Abstract
BACKGROUND Research on the cardiovascular toxicity of angiogenesis inhibitors among patients with cancer in Taiwan is lacking. This observational study explored the risk of major adverse cardiovascular events (MACEs) associated with angiogenesis inhibitors in Taiwan. METHODS AND RESULTS We conducted a nested case-control study using the TCR (Taiwan Cancer Registry) linked with the Taiwan National Insurance Claim Database. We matched every case with 4 controls using risk-set sampling by index date, age, sex, cancer type, and cancer diagnosis date. Conditional logistic regression was used to evaluate the risks of MACEs and different cardiovascular events using propensity score adjustment or matching. Sensitivity analyses were used to evaluate the risks matched by cancer stages or exposure within 1 year. Among a cohort of 284 292 after the exclusion of prevalent cases, the incidences of MACEs among the overall cohort and those exposed to angiogenesis inhibitors were 22.5 and 32.5 events per 1000 person-years, respectively. We matched 17 817 cases with 70 740 controls, with a mean age of 74.9 years, and 56.8% of patients were men. After propensity score adjustment, angiogenesis inhibitors were associated with increased risks of MACEs (odds ratio, 4.56; 95% CI, 1.78-11.59). Significantly increased risks were noted for heart failure hospitalization, myocardial infarction, cerebrovascular accident, and venous thromboembolism, but not for new-onset atrial fibrillation. Similar results were observed after matching by cancer stage or restriction of 1-year exposure. CONCLUSIONS Angiogenesis inhibitors were associated with increased risks of MACEs among patients with various malignancies in Taiwan but were not associated with new-onset atrial fibrillation.
Collapse
Affiliation(s)
- Yen‐Chou Chen
- Division of Cardiology and Cardiovascular Research CentreTaipei Medical University HospitalTaipeiTaiwan
- Taipei Heart Institute, Taipei Medical UniversityTaipeiTaiwan
- School of Public Health, College of Public HealthTaipei Medical UniversityTaipeiTaiwan
| | - Chun‐Yao Huang
- Division of Cardiology and Cardiovascular Research CentreTaipei Medical University HospitalTaipeiTaiwan
- Taipei Heart Institute, Taipei Medical UniversityTaipeiTaiwan
- Department of Biomedical Sciences and EngineeringNational Central UniversityTao‐YuanTaiwan
| | - Li‐Ming Lien
- School of Medicine, College of MedicineTaipei Medical UniversityTaipeiTaiwan
- Department of NeurologyShin Kong Wu Ho‐Su Memorial HospitalTaipeiTaiwan
| | - Jin‐Hua Chen
- Graduate Institute of Data Science, College of ManagementTaipei Medical UniversityTaipeiTaiwan
- Health Data Analytics and Statistics Centre, Office of Data ScienceTaipei Medical UniversityTaipeiTaiwan
| | - Fang‐I Hsieh
- School of Public Health, College of Public HealthTaipei Medical UniversityTaipeiTaiwan
- Master Program in Clinical Genomics and Proteomics, College of PharmacyTaipei Medical UniversityTaipeiTaiwan
| |
Collapse
|
5
|
Liu S, Yue S, Guo Y, Han JY, Wang H. Sorafenib induces cardiotoxicity through RBM20-mediated alternative splicing of sarcomeric and mitochondrial genes. Pharmacol Res 2023; 198:107017. [PMID: 38006979 DOI: 10.1016/j.phrs.2023.107017] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 09/12/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023]
Abstract
Sorafenib, a multi-targeted tyrosine kinase inhibitor, is a first-line treatment for advanced solid tumors, but it induces many adverse cardiovascular events, including myocardial infarction and heart failure. These cardiac defects can be mediated by alternative splicing of genes critical for heart function. Whether alternative splicing plays a role in sorafenib-induced cardiotoxicity remains unclear. Transcriptome of rat hearts or human cardiomyocytes treated with sorafenib was analyzed and validated to define alternatively spliced genes and their impact on cardiotoxicity. In rats, sorafenib caused severe cardiotoxicity with decreased left ventricular systolic pressure, elongated sarcomere, enlarged mitochondria and decreased ATP. This was associated with alternative splicing of hundreds of genes in the hearts, many of which were targets of a cardiac specific splicing factor, RBM20. Sorafenib inhibited RBM20 expression in both rat hearts and human cardiomyocytes. The splicing of RBM20's targets, SLC25A3 and FHOD3, was altered into fetal isoforms with decreased function. Upregulation of RBM20 during sorafenib treatment reversed the pathogenic splicing of SLC25A3 and FHOD3, and enhanced the phosphate transport into mitochondria by SLC25A3, ATP synthesis and cell survival.We envision this regulation may happen in many drug-induced cardiotoxicity, and represent a potential druggable pathway for mitigating sorafenib-induced cardiotoxicity.
Collapse
Affiliation(s)
- Songming Liu
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China
| | - Shanshan Yue
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China
| | - Yuxuan Guo
- Peking University Institute of Cardiovascular Sciences, Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
| | - Jing-Yan Han
- Department of Integration of Chinese and Western Medicine, School of Basic Medical Sciences, Peking University, Beijing 100191, China; Tasly Microcirculation Research Center, Peking University Health Science Center, Beijing 100191, China.
| | - Huan Wang
- Department of Biomedical Informatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China; Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing 100191, China.
| |
Collapse
|
6
|
Alhazzani K, Almangour A, Alsalem A, Alqinyah M, Alhamed AS, Alhamami HN, Alanazi AZ. Examining the Effects of Dasatinib, Sorafenib, and Nilotinib on Vascular Smooth Muscle Cells: Insights into Proliferation, Migration, and Gene Expression Dynamics. Diseases 2023; 11:147. [PMID: 37873791 PMCID: PMC10594443 DOI: 10.3390/diseases11040147] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 10/17/2023] [Accepted: 10/21/2023] [Indexed: 10/25/2023] Open
Abstract
BACKGROUND Dasatinib, nilotinib, and sorafenib are clinically proven tyrosine kinase inhibitors (TKIs) used for the treatment of leukemia and hepatocellular carcinoma. However, there is a growing concern regarding cardiotoxicity associated with their use. The impact of these TKIs on vascular smooth muscle cells (VSMCs) remains unexplored. This study aims to investigate the effects of TKIs on VSMC proliferation and migration, as well as to elucidate the underlying mechanisms involving inflammatory and apoptotic pathways. METHODS VSMCs were extracted from albino rats and cultured in vitro. The cells were divided into four experimental groups: control, dasatinib, sorafenib, and nilotinib. The MTT assay was employed to assess the cytotoxic effects of TKIs on VSMCs. A scratch assay was conducted to evaluate the inhibitory potential of TKIs on VSMC migration. Flow cytometry analysis was used to detect apoptotic cells. Real-Time PCR expression was utilized to determine the differential gene expression of apoptotic and inflammatory markers. RESULTS Dasatinib, nilotinib, and sorafenib demonstrated significant inhibitory effects on VSMC viability and migration at low concentrations (<1 µmol/L, p < 0.05). Furthermore, gene expression analysis revealed up-regulation of inflammatory biomarkers (TNF-α, IL-6, and IL-1β) and apoptotic markers (P53, BAX), along with down-regulation of the anti-apoptotic biomarker BCL-2 in response to all TKIs. CONCLUSIONS This study demonstrates that dasatinib, nilotinib, and sorafenib inhibit VSMC proliferation and migration, suggesting their potential to induce vascular injury and remodeling by activating inflammation and apoptosis pathways. These findings highlight the need for further investigation into the cardiotoxic effects of these TKIs and the development of strategies to mitigate their adverse vascular effects.
Collapse
Affiliation(s)
- Khalid Alhazzani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | | | | | | | | | | | | |
Collapse
|
7
|
Airò G, Maffezzoli M, Lazzarin A, Bianconcini M, Greco A, Buti S, Leonetti A. Takotsubo syndrome in a patient with metastatic renal cell carcinoma treated with pembrolizumab plus axitinib. Immunotherapy 2022; 14:1297-1305. [DOI: 10.2217/imt-2022-0013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We report the case of a patient with metastatic renal cell carcinoma who developed Takotsubo syndrome (TTS) 6 days after starting pembrolizumab plus axitinib as first-line treatment. Coronary angiogram was negative for obstructive coronary artery disease and echocardiogram revealed a depressed left ventricular ejection fraction with apical akinesis. Axitinib was discontinued and myocardial contractile function fully recovered 23 days after the initial presentation. The treatment was safely resumed and granted a partial response of disease. A literature review regarding TTS in patients receiving VEGFR tyrosine kinase inhibitors and/or immune checkpoint inhibitors was performed. TTS is reported as a rare adverse event and the possible causal relationship between TTS and antineoplastic therapy is still unclear. Further research is warranted to better understand cardiotoxicity mechanisms and their management.
Collapse
Affiliation(s)
- Giulia Airò
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
| | - Michele Maffezzoli
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
| | | | | | - Alessandro Greco
- Cardiology Unit, University Hospital of Parma, Parma, 43126, Italy
| | - Sebastiano Buti
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine & Surgery, University of Parma, Parma, 43126, Italy
| | - Alessandro Leonetti
- Medical Oncology Unit, University Hospital of Parma, Parma, 43126, Italy
- Department of Medicine & Surgery, University of Parma, Parma, 43126, Italy
| |
Collapse
|
8
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J 2022; 43:4229-4361. [PMID: 36017568 DOI: 10.1093/eurheartj/ehac244] [Citation(s) in RCA: 897] [Impact Index Per Article: 448.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
|
9
|
Lyon AR, López-Fernández T, Couch LS, Asteggiano R, Aznar MC, Bergler-Klein J, Boriani G, Cardinale D, Cordoba R, Cosyns B, Cutter DJ, de Azambuja E, de Boer RA, Dent SF, Farmakis D, Gevaert SA, Gorog DA, Herrmann J, Lenihan D, Moslehi J, Moura B, Salinger SS, Stephens R, Suter TM, Szmit S, Tamargo J, Thavendiranathan P, Tocchetti CG, van der Meer P, van der Pal HJH. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J Cardiovasc Imaging 2022; 23:e333-e465. [PMID: 36017575 DOI: 10.1093/ehjci/jeac106] [Citation(s) in RCA: 108] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
|
10
|
Jiang H, Wang C, Zhang A, Li Y, Li J, Li Z, Yang X, Hou Y. ATF4 protects against sorafenib-induced cardiotoxicity by suppressing ferroptosis. Biomed Pharmacother 2022; 153:113280. [PMID: 35724508 DOI: 10.1016/j.biopha.2022.113280] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 06/05/2022] [Accepted: 06/08/2022] [Indexed: 11/02/2022] Open
Abstract
Sorafenib (SOR) is an effective chemotherapy drug for hepatocellular carcinoma, renal cell carcinoma, and differentiated thyroid carcinoma. However, a long-standing clinical issue associated with SOR use is an increased risk of cardiotoxicity, but the underlying mechanisms remain obscure. Here we report that ferroptosis of cardiomyocytes is responsible for SOR-induced cardiotoxicity. The specific ferroptosis inhibitor ferrostatin-1 and deferoxamine mesylate, an iron chelator, significantly alleviate SOR-induced cardiac damage. RNA-sequencing revealed that endoplasmic reticulum (ER) stress and the unfolded protein response were predominately activated, which might be attributed to the lipid reactive oxygen species-mediated perturbation of the ER. Activating transcription factor 4 (ATF4) is one of the most significantly up-regulated genes, knockdown of ATF4 exacerbates cardiomyocyte ferroptosis induced by SOR, while overexpression of ATF4 promotes cell survival. Mice with AAV-mediated ATF4 knockdown exhibit lipid peroxidation and more severe cardiomyopathy. Further experiments demonstrated that ATF4 exerts its protective role by elevating SLC7A11 expression, a transport subunit of system Xc-, which promotes cystine uptake and glutathione biosynthesis. The cardioprotective effect of ATF4 was diminished by SLC7A11 knockdown in cardiomyocytes subjected to SOR treatment. Taken together, these findings show that ferroptosis of cardiomyocytes is an important cause of SOR-related cardiotoxicity. ATF4 acts as a key regulator to promote cardiomyocytes survival by up-regulation of SLC7A11 and suppression of ferroptosis.
Collapse
Affiliation(s)
- Hui Jiang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China; Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Cong Wang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong 250014, China
| | - An Zhang
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China
| | - Yufeng Li
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Jianping Li
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Zhan Li
- Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong 250014, China
| | - Xin Yang
- Department of Pediatrics, The Second Hospital of Shandong University, Jinan, Shandong 250033, China
| | - Yinglong Hou
- Department of Cardiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong 250014, China; Department of Cardiology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Cardiac Electrophysiology and Arrhythmia, Jinan, Shandong 250014, China.
| |
Collapse
|
11
|
Cardio-onco-metabolism: metabolic remodelling in cardiovascular disease and cancer. Nat Rev Cardiol 2022; 19:414-425. [PMID: 35440740 PMCID: PMC10112835 DOI: 10.1038/s41569-022-00698-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/17/2022] [Indexed: 02/07/2023]
Abstract
Cardiovascular disease and cancer are the two leading causes of morbidity and mortality in the world. The emerging field of cardio-oncology has revealed that these seemingly disparate disease processes are intertwined, owing to the cardiovascular sequelae of anticancer therapies, shared risk factors that predispose individuals to both cardiovascular disease and cancer, as well the possible potentiation of cancer growth by cardiac dysfunction. As a result, interest has increased in understanding the fundamental biological mechanisms that are central to the relationship between cardiovascular disease and cancer. Metabolism, appropriate regulation of energy, energy substrate utilization, and macromolecular synthesis and breakdown are fundamental processes for cellular and organismal survival. In this Review, we explore the emerging data identifying metabolic dysregulation as an important theme in cardio-oncology. We discuss the growing recognition of metabolic reprogramming in cardiovascular disease and cancer and view the novel area of cardio-oncology through the lens of metabolism.
Collapse
|
12
|
Kwan JM, Oikonomou EK, Henry ML, Sinusas AJ. Multimodality Advanced Cardiovascular and Molecular Imaging for Early Detection and Monitoring of Cancer Therapy-Associated Cardiotoxicity and the Role of Artificial Intelligence and Big Data. Front Cardiovasc Med 2022; 9:829553. [PMID: 35369354 PMCID: PMC8964995 DOI: 10.3389/fcvm.2022.829553] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer mortality has improved due to earlier detection via screening, as well as due to novel cancer therapies such as tyrosine kinase inhibitors and immune checkpoint inhibitions. However, similarly to older cancer therapies such as anthracyclines, these therapies have also been documented to cause cardiotoxic events including cardiomyopathy, myocardial infarction, myocarditis, arrhythmia, hypertension, and thrombosis. Imaging modalities such as echocardiography and magnetic resonance imaging (MRI) are critical in monitoring and evaluating for cardiotoxicity from these treatments, as well as in providing information for the assessment of function and wall motion abnormalities. MRI also allows for additional tissue characterization using T1, T2, extracellular volume (ECV), and delayed gadolinium enhancement (DGE) assessment. Furthermore, emerging technologies may be able to assist with these efforts. Nuclear imaging using targeted radiotracers, some of which are already clinically used, may have more specificity and help provide information on the mechanisms of cardiotoxicity, including in anthracycline mediated cardiomyopathy and checkpoint inhibitor myocarditis. Hyperpolarized MRI may be used to evaluate the effects of oncologic therapy on cardiac metabolism. Lastly, artificial intelligence and big data of imaging modalities may help predict and detect early signs of cardiotoxicity and response to cardioprotective medications as well as provide insights on the added value of molecular imaging and correlations with cardiovascular outcomes. In this review, the current imaging modalities used to assess for cardiotoxicity from cancer treatments are discussed, in addition to ongoing research on targeted molecular radiotracers, hyperpolarized MRI, as well as the role of artificial intelligence (AI) and big data in imaging that would help improve the detection and prognostication of cancer-treatment cardiotoxicity.
Collapse
Affiliation(s)
- Jennifer M. Kwan
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Evangelos K. Oikonomou
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
| | - Mariana L. Henry
- Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Albert J. Sinusas
- Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, United States
- Department of Radiology and Biomedical Imaging, Yale University School of Medicine, New Haven, CT, United States
- Department of Biomedical Engineering, Yale University, New Haven, CT, United States
| |
Collapse
|
13
|
Hypoxia signaling and oxygen metabolism in cardio-oncology. J Mol Cell Cardiol 2022; 165:64-75. [PMID: 34979102 DOI: 10.1016/j.yjmcc.2021.12.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 12/05/2021] [Accepted: 12/27/2021] [Indexed: 12/26/2022]
Abstract
Cardio-oncology is a rapidly growing field in cardiology that focuses on the management of cardiovascular toxicities associated with cancer-directed therapies. Tumor hypoxia is a central driver of pathologic tumor growth, metastasis, and chemo-resistance. In addition, conditions that mimic hypoxia (pseudo-hypoxia) play a causal role in the pathogenesis of numerous types of cancer, including renal cell carcinoma. Therefore, therapies targeted at hypoxia signaling pathways have emerged over the past several years. Though efficacious, these therapies are associated with significant cardiovascular toxicities, ranging from hypertension to cardiomyopathy. This review focuses on oxygen metabolism in tumorigenesis, the role of targeting hypoxia signaling in cancer therapy, and the relevance of oxygen metabolism in cardio-oncology. This review will specifically focus on hypoxia signaling mediated by hypoxia-inducible factors and the prolyl hydroxylase oxygen-sensing enzymes, the cardiovascular effects of specific cancer targeted therapies mediated on VEGF and HIF signaling, hypoxic signaling in cardiovascular disease, and the role of oxygen in anthracycline cardiotoxicity. The implications of these therapies on myocardial biology and cardiac function are discussed, underlining the fine balance of hypoxia signaling in cardiac homeostasis. Understanding these cardiovascular toxicities will be important to optimize treatment for cancer patients while mitigating potentially severe cardiovascular side effects.
Collapse
|
14
|
Puliafito I, Esposito F, Prestifilippo A, Marchisotta S, Sciacca D, Vitale MP, Giuffrida D. Target Therapy in Thyroid Cancer: Current Challenge in Clinical Use of Tyrosine Kinase Inhibitors and Management of Side Effects. Front Endocrinol (Lausanne) 2022; 13:860671. [PMID: 35872981 PMCID: PMC9304687 DOI: 10.3389/fendo.2022.860671] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/12/2022] [Indexed: 01/18/2023] Open
Abstract
Thyroid cancer (TC) is the most common endocrine malignancy. TC is classified as differentiated TC (DTC), which includes papillary and follicular subtypes and Hürthle cell variants, medullary TC (MTC), anaplastic TC (ATC), and poorly differentiated TC (PDTC). The standard of care in DTC consists of surgery together with radioactive iodine (131I) therapy and thyroid hormone, but patients with MTC do not benefit from 131I therapy. Patients with advanced TC resistant to 131I treatment (RAI-R) have no chance of cure, as well as patients affected by ATC and progressive MTC, in which conventional therapy plays only a palliative role, representing, until a few years ago, an urgent unmet need. In the last decade, a better understanding of molecular pathways involved in the tumorigenesis of specific histopathological subtypes of TC has led to develop tyrosine kinase inhibitors (TKIs). TKIs represent a valid treatment in progressive advanced disease and were tested in all subtypes of TC, highlighting the need to improve progression-free survival. However, treatments using these novel therapeutics are often accompanied by side effects that required optimal management to minimize their toxicities and thereby enable patients who show benefit to continue treatment and obtain maximal clinical efficacy. The goal of this overview is to provide an update on the current use of the main drugs recently studied for advanced TC and the management of the adverse events.
Collapse
Affiliation(s)
- Ivana Puliafito
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | - Francesca Esposito
- IOM Ricerca Srl, Viagrande, Italy
- *Correspondence: Francesca Esposito, ; Dario Giuffrida,
| | - Angela Prestifilippo
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | | | - Dorotea Sciacca
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | - Maria Paola Vitale
- Hospital Pharmacy Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
| | - Dario Giuffrida
- Medical Oncology Unit, Istituto Oncologico del Mediterraneo SpA, Viagrande, Italy
- *Correspondence: Francesca Esposito, ; Dario Giuffrida,
| |
Collapse
|
15
|
Affiliation(s)
- Li-Ling Tan
- Cardiology, National University Heart Centre, Singapore
- Cardio-Oncology Service, Department of Cardiology, Royal Brompton and Harefield NHS Foundation Trust, London, UK
| | - Alexander Richard Lyon
- Cardio-Oncology Service, Department of Cardiology, Royal Brompton and Harefield NHS Foundation Trust, London, UK
- Cardiac Medicine, Imperial College London, London, UK
| |
Collapse
|
16
|
Asnani A, Moslehi JJ, Adhikari BB, Baik AH, Beyer AM, de Boer RA, Ghigo A, Grumbach IM, Jain S, Zhu H. Preclinical Models of Cancer Therapy-Associated Cardiovascular Toxicity: A Scientific Statement From the American Heart Association. Circ Res 2021; 129:e21-e34. [PMID: 33934611 DOI: 10.1161/res.0000000000000473] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Although cardiovascular toxicity from traditional chemotherapies has been well recognized for decades, the recent explosion of effective novel targeted cancer therapies with cardiovascular sequelae has driven the emergence of cardio-oncology as a new clinical and research field. Cardiovascular toxicity associated with cancer therapy can manifest as a broad range of potentially life-threatening complications, including heart failure, arrhythmia, myocarditis, and vascular events. Beyond toxicology, the intersection of cancer and heart disease has blossomed to include discovery of genetic and environmental risk factors that predispose to both. There is a pressing need to understand the underlying molecular mechanisms of cardiovascular toxicity to improve outcomes in patients with cancer. Preclinical cardiovascular models, ranging from cellular assays to large animals, serve as the foundation for mechanistic studies, with the ultimate goal of identifying biologically sound biomarkers and cardioprotective therapies that allow the optimal use of cancer treatments while minimizing toxicities. Given that novel cancer therapies target specific pathways integral to normal cardiovascular homeostasis, a better mechanistic understanding of toxicity may provide insights into fundamental pathways that lead to cardiovascular disease when dysregulated. The goal of this scientific statement is to summarize the strengths and weaknesses of preclinical models of cancer therapy-associated cardiovascular toxicity, to highlight overlapping mechanisms driving cancer and cardiovascular disease, and to discuss opportunities to leverage cardio-oncology models to address important mechanistic questions relevant to all patients with cardiovascular disease, including those with and without cancer.
Collapse
|
17
|
Moslehi J, Lichtman AH, Sharpe AH, Galluzzi L, Kitsis RN. Immune checkpoint inhibitor-associated myocarditis: manifestations and mechanisms. J Clin Invest 2021; 131:145186. [PMID: 33645548 DOI: 10.1172/jci145186] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment of various cancers, including malignancies once considered untreatable. These agents, however, are associated with inflammation and tissue damage in multiple organs. Myocarditis has emerged as a serious ICI-associated toxicity, because, while seemingly infrequent, it is often fulminant and lethal. The underlying basis of ICI-associated myocarditis is not completely understood. While the importance of T cells is clear, the inciting antigens, why they are recognized, and the mechanisms leading to cardiac cell injury remain poorly characterized. These issues underscore the need for basic and clinical studies to define pathogenesis, identify predictive biomarkers, improve diagnostic strategies, and develop effective treatments. An improved understanding of ICI-associated myocarditis will provide insights into the equilibrium between the immune and cardiovascular systems.
Collapse
Affiliation(s)
- Javid Moslehi
- Division of Cardiovascular Medicine and Division of Oncology, Cardio-Oncology Program, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Andrew H Lichtman
- Department of Pathology, Brigham and Women's Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Arlene H Sharpe
- Department of Immunology and Blavatnik Institute, Harvard Medical School, Boston, Massachusetts, USA.,Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Sandra and Edward Meyer Cancer Center, Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medical College, New York, New York, USA.,Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.,Université de Paris, Paris, France
| | - Richard N Kitsis
- Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, and Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
18
|
Protective Effect of RIVA Against Sunitinib-Induced Cardiotoxicity by Inhibiting Oxidative Stress-Mediated Inflammation: Probable Role of TGF-β and Smad Signaling. Cardiovasc Toxicol 2020; 20:281-290. [PMID: 31696377 DOI: 10.1007/s12012-019-09551-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Sunitinib (SUN) is an oral tyrosine kinase inhibitor approved in 2006 as a first-line treatment for metastatic renal cell cancer. However, weak selectivity to kinase receptors and cardiotoxicity have limited the use of sunitinib. Rivaroxaban (RIVA) is a Factor Xa inhibitor with cardioprotective action. It inhibits atherosclerosis and numerous inflammatory cascades. The present study was designed to investigate the cardioprotective effects of RIVA in sunitinib-induced cardiotoxicity. Thirty male Wistar rats were divided into five groups. Group 1 was the normal control (control). Group 2 was administered i.p. SUN 25 mg kg-1 thrice weekly for 3 weeks. Groups 3 and 4 received the same treatment as Group 2 followed by the administration of RIVA 5 mg kg-1 day-1 and 10 mg kg-1 day-1, respectively, for 3 weeks. Group 5 received only 10 mg kg-1 day-1 RIVA for 3 weeks. Serum levels of Ca2+, Mg2+, Fe3+/Fe2+, lipid profiles, and cardiac enzymes were measured. Cardiac tissues were isolated for the measurements of oxidant/antioxidant balance gene and protein expressions. Relative to the controls, the administration of SUN significantly altered serum levels of (Ca2+, Mg2+, Fe3+/Fe2+, lipid profiles, and cardiac enzymes), intracellular antioxidant enzymes, and the expression levels of the genes encoding certain proteins. RIVA treatment significantly restored these parameters to near-normal levels. RIVA treatment significantly mitigated SUN-induced cardiac injuries by restoring antioxidant enzyme levels and attenuating the proinflammatory cascades resulting from SUN-induced cardiac injuries.
Collapse
|
19
|
Miller JM, Meki MH, Ou Q, George SA, Gams A, Abouleisa RRE, Tang XL, Ahern BM, Giridharan GA, El-Baz A, Hill BG, Satin J, Conklin DJ, Moslehi J, Bolli R, Ribeiro AJS, Efimov IR, Mohamed TMA. Heart slice culture system reliably demonstrates clinical drug-related cardiotoxicity. Toxicol Appl Pharmacol 2020; 406:115213. [PMID: 32877659 DOI: 10.1016/j.taap.2020.115213] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 02/07/2023]
Abstract
The limited availability of human heart tissue and its complex cell composition are major limiting factors for the reliable testing of drug efficacy and toxicity. Recently, we developed functional human and pig heart slice biomimetic culture systems that preserve the viability and functionality of 300 μm heart slices for up to 6 days. Here, we tested the reliability of this culture system for testing the cardiotoxicity of anti-cancer drugs. We tested three anti-cancer drugs (doxorubicin, trastuzumab, and sunitinib) with known different mechanisms of cardiotoxicity at three concentrations and assessed the effect of these drugs on heart slice viability, structure, function and gene expression. Slices incubated with any of these drugs for 48 h showed diminished in viability as well as loss of cardiomyocyte structure and function. Mechanistically, RNA sequencing of doxorubicin-treated tissues demonstrated a significant downregulation of cardiac genes and upregulation of oxidative stress responses. Trastuzumab treatment downregulated cardiac muscle contraction-related genes consistent with its clinically known effect on cardiomyocytes. Interestingly, sunitinib treatment resulted in significant downregulation of angiogenesis-related genes, in line with its mechanism of action. Similar to hiPS-derived-cardiomyocytes, heart slices recapitulated the expected toxicity of doxorubicin and trastuzumab, however, slices were superior in detecting sunitinib cardiotoxicity and mechanism in the clinically relevant concentration range of 0.1-1 μM. These results indicate that heart slice culture models have the potential to become a reliable platform for testing and elucidating mechanisms of drug cardiotoxicity.
Collapse
Affiliation(s)
- Jessica M Miller
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Moustafa H Meki
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA
| | - Qinghui Ou
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Sharon A George
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Anna Gams
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA
| | - Riham R E Abouleisa
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Xian-Liang Tang
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Brooke M Ahern
- Department of Physiology, University of Kentucky, KY, USA
| | | | - Ayman El-Baz
- Department of Bioengineering, University of Louisville, KY, USA
| | - Bradford G Hill
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Jonathan Satin
- Department of Physiology, University of Kentucky, KY, USA
| | - Daniel J Conklin
- Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA
| | - Javid Moslehi
- Division of Cardiology, Cardio-Oncology Program, Vanderbilt University Medical Center, 2220 Pierce Avenue, Nashville, USA
| | - Roberto Bolli
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA
| | - Alexandre J S Ribeiro
- U.S. Food and Drug Administration, Center for Drug Evaluation and Research, Office of Translational Science, Office of Clinical Pharmacology, Division of Applied Regulatory Science, Silver Spring, MD, USA.
| | - Igor R Efimov
- Department of Biomedical Engineering, The George Washington University, Washington, DC, USA.
| | - Tamer M A Mohamed
- Institute of Molecular Cardiology, Department of Medicine, University of Louisville, KY, USA; Department of Bioengineering, University of Louisville, KY, USA; Envirome Institute, Diabetes and Obesity Center, Department of Medicine, University of Louisville, KY, USA; Department of Pharmacology and Toxicology, University of Louisville, KY, USA; Institute of Cardiovascular Sciences, University of Manchester, UK; Faculty of Pharmacy, Zagazig University, Egypt.
| |
Collapse
|
20
|
Tocchetti CG, Ameri P, de Boer RA, D’Alessandra Y, Russo M, Sorriento D, Ciccarelli M, Kiss B, Bertrand L, Dawson D, Falcao-Pires I, Giacca M, Hamdani N, Linke WA, Mayr M, van der Velden J, Zacchigna S, Ghigo A, Hirsch E, Lyon AR, Görbe A, Ferdinandy P, Madonna R, Heymans S, Thum T. Cardiac dysfunction in cancer patients: beyond direct cardiomyocyte damage of anticancer drugs: novel cardio-oncology insights from the joint 2019 meeting of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart. Cardiovasc Res 2020; 116:1820-1834. [DOI: 10.1093/cvr/cvaa222] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 06/17/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Abstract
In western countries, cardiovascular (CV) disease and cancer are the leading causes of death in the ageing population. Recent epidemiological data suggest that cancer is more frequent in patients with prevalent or incident CV disease, in particular, heart failure (HF). Indeed, there is a tight link in terms of shared risk factors and mechanisms between HF and cancer. HF induced by anticancer therapies has been extensively studied, primarily focusing on the toxic effects that anti-tumour treatments exert on cardiomyocytes. In this Cardio-Oncology update, members of the ESC Working Groups of Myocardial Function and Cellular Biology of the Heart discuss novel evidence interconnecting cardiac dysfunction and cancer via pathways in which cardiomyocytes may be involved but are not central. In particular, the multiple roles of cardiac stromal cells (endothelial cells and fibroblasts) and inflammatory cells are highlighted. Also, the gut microbiota is depicted as a new player at the crossroads between HF and cancer. Finally, the role of non-coding RNAs in Cardio-Oncology is also addressed. All these insights are expected to fuel additional research efforts in the field of Cardio-Oncology.
Collapse
Affiliation(s)
- Carlo Gabriele Tocchetti
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Interdepartmental Center of Clinical and Translational Sciences (CIRCET), Federico II University, Naples, Italy
| | - Pietro Ameri
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, Genova, Italy
- Department of Internal Medicine, University of Genova, Genova, Italy
| | - Rudolf A de Boer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, AB31, PO Box 30.001, 9700 RB Groningen, The Netherlands
| | - Yuri D’Alessandra
- Immunology and Functional Genomics Unit, Centro Cardiologico Monzino IRCCS, Milan, Italy
| | - Michele Russo
- Department of Translational Medical Sciences, Federico II University, via Pansini 5, 80131 Naples, Italy
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Daniela Sorriento
- Department of Advanced Biomedical Sciences, Federico II University, Naples, Italy
| | - Michele Ciccarelli
- Department of Medicine Surgery and Odontology, University of Salerno, Salerno, Italy
| | - Bernadett Kiss
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
| | - Luc Bertrand
- IREC Institute, Pole of Cardiovascular Research, Université Catholique de Louvain, Brussels, Belgium
| | - Dana Dawson
- School of Medicine and Dentistry, University of Aberdeen, Aberdeen, UK
| | - Ines Falcao-Pires
- Unidade de Investigação e Desenvolvimento Cardiovascular, Departamento de Cirurgia e Fisiologia, Faculdade de Medicina, Universidade do Porto, Portugal
| | - Mauro Giacca
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Nazha Hamdani
- Department of Molecular and Experimental Cardiology, Ruhr Universität Bochum, Bochum, Germany
- Department of Cardiology, St. Joseph Hospital, Ruhr University Bochum, Witten, Germany
| | | | - Manuel Mayr
- King’s British Heart Foundation Centre, King’s College London, London, UK
| | - Jolanda van der Velden
- Department of Physiology, Amsterdam UMC, Vrije Universiteit, Amsterdam Cardiovascular Sciences Institute, Amsterdam, The Netherlands
| | - Serena Zacchigna
- Department of Medicine, Surgery and Health Sciences and Cardiovascular Department, Centre for Translational Cardiology, Azienda Sanitaria Universitaria Integrata Trieste, Trieste, Italy
- International Center for Genetic Engineering and Biotechnology (ICGEB), Trieste, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Emilio Hirsch
- Department of Molecular Biotechnology and Health Sciences, Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Alexander R Lyon
- Cardio-Oncology Service, Royal Brompton Hospital, Imperial College London, London, UK
| | - Anikó Görbe
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Péter Ferdinandy
- Department of Pharmacology and Pharmacotherapy, Cardiometabolic Research Group and MTA-SE System Pharmacology Research Group, Semmelweis University, Budapest, Hungary
- Pharmahungary Group, Szeged, Hungary
| | - Rosalinda Madonna
- Institute of Cardiology, University of Pisa, Pisa, Italy
- Center for Cardiovascular Biology and Atherosclerosis Research, McGovern School of Medicine, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Stephane Heymans
- Department of Cardiology, Cardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Centre, Maastricht University, Maastricht, The Netherlands
- Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Thomas Thum
- Institute for Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Hannover, Germany
| |
Collapse
|
21
|
Sudasena D, Balanescu DV, Donisan T, Hassan S, Palaskas N, Kim P, Karimzad K, Lopez-Mattei J, Arain S, Gould KL, Iliescu C. Fulminant Vascular and Cardiac Toxicity Associated with Tyrosine Kinase Inhibitor Sorafenib. Cardiovasc Toxicol 2020; 19:382-387. [PMID: 30543051 DOI: 10.1007/s12012-018-9499-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The use of vascular endothelial growth factor inhibitors such as sorafenib is limited by a risk of severe cardiovascular toxicity. A 28-year-old man with acute myeloid leukemia treated with prednisone, tacrolimus, and sorafenib following stem cell transplantation presented with severe bilateral lower extremity claudication. The patient was discharged against medical advice prior to finalizing a cardiovascular evaluation, but returned 1 week later with signs suggestive of septic shock. Laboratory tests revealed troponin I of 12.63 ng/mL, BNP of 1690 pg/mL, and negative infectious workup. Electrocardiogram showed sinus tachycardia and new pathologic Q waves in the anterior leads. Coronary angiography revealed severe multivessel coronary artery disease. Peripheral angiography revealed severely diseased left anterior and posterior tibial arteries, tibioperoneal trunk, and peroneal artery, and subtotal occlusion of the right posterior tibial artery. Multiple coronary and peripheral drug-eluting stents were implanted. An intra-aortic balloon pump was placed. Cardiac magnetic resonance imaging revealed chronic left ventricular infarction with some viability, 17% ejection fraction, and left ventricular mural thrombi. The patient opted for medical management. Persistent symptoms 9 months later led to repeat angiography, showing total occlusion of the second obtuse marginal artery due to in-stent restenosis with proximal stent fracture, and chronic total occlusion of the right internal iliac artery extending to the pudendal branch. Cardiac positron emission tomography/computed tomography viability study demonstrated viable myocardium, deeming revascularization appropriate. Symptom resolution was obtained with no recurrences. Sorafenib-associated vasculopathy may follow a fulminant course. Multimodality cardiovascular imaging is essential for optimal management.
Collapse
Affiliation(s)
- Daryl Sudasena
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Dinu Valentin Balanescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA
| | - Teodora Donisan
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA
| | - Saamir Hassan
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA
| | - Nicolas Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA
| | - Peter Kim
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA
| | - Kaveh Karimzad
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA
| | - Juan Lopez-Mattei
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA
| | - Salman Arain
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - K Lance Gould
- Department of Internal Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Cezar Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, 1400 Pressler Street, 1451, 77030, Houston, TX, USA.
| |
Collapse
|
22
|
Avila MS, Siqueira SRR, Ferreira SMA, Bocchi EA. Prevention and Treatment of Chemotherapy-Induced Cardiotoxicity. Methodist Debakey Cardiovasc J 2020; 15:267-273. [PMID: 31988687 DOI: 10.14797/mdcj-15-4-267] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent decades have seen an increase in survival rates for cancer patients, partially explained by earlier diagnoses and new chemotherapeutic agents. However, chemotherapy may be associated with adverse cardiovascular events, including hypertension and pulmonary hypertension, supraventricular and ventricular arrhythmias, cardiomyopathy, and other forms of cardiovascular disease. For patients, the benefits of chemotherapy may be partially obfuscated by deleterious effects on the cardiovascular system, resulting in a significant increase in morbidity and mortality. In this article, we review strategies for prevention and treatment of chemotherapy-related cardiotoxicity.
Collapse
Affiliation(s)
- Monica Samuel Avila
- HOSPITAL DAS CLINICAS DA FACULDADE DE MEDICINA DA UNIVERSIDADE DE SAO PAULO, SAO PAULO, BRAZIL
| | | | | | - Edimar Alcides Bocchi
- HOSPITAL DAS CLINICAS DA FACULDADE DE MEDICINA DA UNIVERSIDADE DE SAO PAULO, SAO PAULO, BRAZIL
| |
Collapse
|
23
|
Aldemir MN, Simsek M, Kara AV, Ozcicek F, Mammadov R, Yazıcı GN, Sunar M, Coskun R, Gulaboglu M, Suleyman H. The effect of adenosine triphosphate on sunitinib-induced cardiac injury in rats. Hum Exp Toxicol 2020; 39:1046-1053. [PMID: 32131635 DOI: 10.1177/0960327120909874] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
In this study, we aimed to show the effect of adenosine 5'-triphosphate (ATP) on sunitinib-induced cardiac injury in rats. The rats (n = 30) were divided equally into three groups as sunitinib group (SG), sunitinib plus ATP group (SAG), and healthy group (HG); 2 mg/kg ATP was injected intraperitoneally (ip) to the SAG group. Same volume normal saline as solvent was administered ip to the other two groups. After 1 h, 25 mg/kg sunitinib was applied orally via catheter to stomach in the SAG and SG groups. This procedure was repeated once daily for 5 weeks. At the end of this period, all animals were sacrificed and their cardiac tissue was removed. Malondialdehyde (MDA), total glutathione (tGSH), tumor necrosis factor α (TNF-α), and nuclear factor κB (NF-κB) levels in rats' cardiac tissues and troponin I (Tp-I) levels in rats' blood samples were evaluated. Histopathological analysis was also performed in cardiac tissues of the animals. MDA, TNF-α, NF-κB, and Tp-I levels were higher in the SG group compared to the SAG and HG groups (p < 0.001). tGSH levels of the SG group were lower than the SAG and HG groups (p < 0.001). The structure and morphology of cardiac muscle fibers and blood vessels were normal in the control group. In the SG group, obvious cardiac muscle tissue damage with dilated myofibers, locally atrophic myofibers, and congested blood vessels were observed. In the SAG group, marked amelioration in these findings was observed. We showed this for the first time that ATP administration exerts a protective effect against cardiac effects of sunitinib.
Collapse
Affiliation(s)
- M N Aldemir
- Department of Medical Oncology, Faculty of Medicine, Yuzuncu Yil University, Van, Turkey
| | - M Simsek
- Department of Medical Oncology, Yozgat City Hospital, Yozgat, Turkey
| | - A V Kara
- Department of Nephrology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - F Ozcicek
- Department of Internal Medicine, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - R Mammadov
- Department Pharmacology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - G N Yazıcı
- Department of Histology and Embryology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - M Sunar
- Department of Anatomy, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - R Coskun
- Department of Cardiology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| | - M Gulaboglu
- Department of Biochemistry, Faculty of Pharmacy, Ataturk University, Erzurum, Turkey
| | - H Suleyman
- Department of Pharmacology, Faculty of Medicine, Erzincan University, Erzincan, Turkey
| |
Collapse
|
24
|
Campia U, Moslehi JJ, Amiri-Kordestani L, Barac A, Beckman JA, Chism DD, Cohen P, Groarke JD, Herrmann J, Reilly CM, Weintraub NL. Cardio-Oncology: Vascular and Metabolic Perspectives: A Scientific Statement From the American Heart Association. Circulation 2019; 139:e579-e602. [PMID: 30786722 DOI: 10.1161/cir.0000000000000641] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardio-oncology has organically developed as a new discipline within cardiovascular medicine as a result of the cardiac and vascular adverse sequelae of the major advances in cancer treatment. Patients with cancer and cancer survivors are at increased risk of vascular disease for a number of reasons. First, many new cancer therapies, including several targeted therapies, are associated with vascular and metabolic complications. Second, cancer itself serves as a risk factor for vascular disease, especially by increasing the risk for thromboembolic events. Finally, recent data suggest that common modifiable and genetic risk factors predispose to both malignancies and cardiovascular disease. Vascular complications in patients with cancer represent a new challenge for the clinician and a new frontier for research and investigation. Indeed, vascular sequelae of novel targeted therapies may provide insights into vascular signaling in humans. Clinically, emerging challenges are best addressed by a multidisciplinary approach in which cardiovascular medicine specialists and vascular biologists work closely with oncologists in the care of patients with cancer and cancer survivors. This novel approach realizes the goal of providing superior care through the creation of cardio-oncology consultative services and the training of a new generation of cardiovascular specialists with a broad understanding of cancer treatments.
Collapse
|
25
|
Ma W, Liu M, Liang F, Zhao L, Gao C, Jiang X, Zhang X, Zhan H, Hu H, Zhao Z. Cardiotoxicity of sorafenib is mediated through elevation of ROS level and CaMKII activity and dysregulation of calcium homoeostasis. Basic Clin Pharmacol Toxicol 2019; 126:166-180. [DOI: 10.1111/bcpt.13318] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Accepted: 08/28/2019] [Indexed: 12/13/2022]
Affiliation(s)
- Wenzhuo Ma
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Mei Liu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Fanfan Liang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Lili Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Chenying Gao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xixi Jiang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Xin Zhang
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Heqin Zhan
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
- Department of Pharmacology College of Pharmacy Xinxiang Medical University Xinxiang Henan 453003 China
| | - Hao Hu
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| | - Zhenghang Zhao
- Department of Pharmacology School of Basic Medical Sciences Xi'an Jiaotong University, Health Science Center Xi'an China
| |
Collapse
|
26
|
Adaptation of Human iPSC-Derived Cardiomyocytes to Tyrosine Kinase Inhibitors Reduces Acute Cardiotoxicity via Metabolic Reprogramming. Cell Syst 2019; 8:412-426.e7. [PMID: 31078528 DOI: 10.1016/j.cels.2019.03.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 01/25/2019] [Accepted: 03/15/2019] [Indexed: 12/31/2022]
Abstract
Tyrosine kinase inhibitors (TKIs) are widely used to treat solid tumors but can be cardiotoxic. The molecular basis for this toxicity and its relationship to therapeutic mechanisms remain unclear; we therefore undertook a systems-level analysis of human cardiomyocytes (CMs) exposed to four TKIs. CMs differentiated from human induced pluripotent stem cells (hiPSCs) were exposed to sunitinib, sorafenib, lapatinib, or erlotinib, and responses were assessed by functional assays, microscopy, RNA sequencing, and mass spectrometry (GEO: GSE114686; PRIDE: PXD012043). TKIs have diverse effects on hiPSC-CMs distinct from inhibition of tyrosine-kinase-mediated signal transduction; cardiac metabolism is particularly sensitive. Following sorafenib treatment, oxidative phosphorylation is downregulated, resulting in a profound defect in mitochondrial energetics. Cells adapt by upregulating aerobic glycolysis. Adaptation makes cells less acutely sensitive to sorafenib but may have long-term negative consequences. Thus, CMs exhibit adaptive responses to anti-cancer drugs conceptually similar to those previously shown in tumors to mediate drug resistance.
Collapse
|
27
|
Affiliation(s)
- Sarosh Rana
- From the Division of Maternal Fetal Medicine, Department of Obstetrics and Gynecology, University of Chicago, IL (S.R.)
| | - Elizabeth Lemoine
- Harvard Medical School, Boston, MA (E.L.)
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (E.L., S.A.K.)
| | - Joey P. Granger
- Department of Physiology, University of Mississippi Medical Center, Jackson (J.P.G.)
| | - S. Ananth Karumanchi
- Departments of Medicine, Obstetrics and Gynecology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA (S.A.K.)
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA (E.L., S.A.K.)
| |
Collapse
|
28
|
Yaranov D, Alexis JD. Heart Disease in Pregnancy: A Special Look at Peripartum Cardiomyopathy. CARDIOVASCULAR INNOVATIONS AND APPLICATIONS 2019. [DOI: 10.15212/cvia.2017.0066] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
|
29
|
Rhea IB, Oliveira GH. Cardiotoxicity of Novel Targeted Chemotherapeutic Agents. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2018; 20:53. [DOI: 10.1007/s11936-018-0649-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
30
|
Posterior reversible encephalopathy syndrome and takotsubo cardiomyopathy associated with lenvatinib therapy for thyroid cancer: a case report and review. Oncotarget 2018; 9:28281-28289. [PMID: 29963277 PMCID: PMC6021337 DOI: 10.18632/oncotarget.25606] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Accepted: 05/19/2018] [Indexed: 12/02/2022] Open
Abstract
As immunotherapies including tyrosine kinase inhibitors become more widely used for the treatment of a variety of malignancies, it is important for prescribers and patients to understand the potential adverse effects associated with these drugs. It is especially important to understand the potentially fatal side effects associated with these drugs to further determine risk factors for their development. The review presents a case of posterior reversible encephalopathy syndrome with concomitant Takotsubo cardiomyopathy, associated with use of lenvatinib therapy for thyroid cancer. It discusses the interventions performed and outcome. Potential mechanisms for development of these rare adverse effects, as well as cases in which these adverse effects are seen with use of other tyrosine-kinase inhibitors will be presented. It is important to continue to report these side effects, and further studies are needed to elucidate potential risk factors for their development, as well as to determine prognosis after development.
Collapse
|
31
|
Chang HM, Moudgil R, Scarabelli T, Okwuosa TM, Yeh ETH. Cardiovascular Complications of Cancer Therapy: Best Practices in Diagnosis, Prevention, and Management: Part 1. J Am Coll Cardiol 2017; 70:2536-2551. [PMID: 29145954 PMCID: PMC5825187 DOI: 10.1016/j.jacc.2017.09.1096] [Citation(s) in RCA: 250] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/24/2017] [Accepted: 09/26/2017] [Indexed: 12/27/2022]
Abstract
Modern cancer therapy has successfully cured many cancers and converted a terminal illness into a chronic disease. Because cancer patients often have coexisting heart diseases, expert advice from cardiologists will improve clinical outcome. In addition, cancer therapy can also cause myocardial damage, induce endothelial dysfunction, and alter cardiac conduction. Thus, it is important for practicing cardiologists to be knowledgeable about the diagnosis, prevention, and management of the cardiovascular complications of cancer therapy. In this first part of a 2-part review, we will review cancer therapy-induced cardiomyopathy and ischemia. This review is based on a MEDLINE search of published data, published clinical guidelines, and best practices in major cancer centers. With the number of cancer survivors expanding quickly, the time has come for cardiologists to work closely with cancer specialists to prevent and treat cancer therapy-induced cardiovascular complications.
Collapse
Affiliation(s)
- Hui-Ming Chang
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri
| | - Rohit Moudgil
- Department of Cardiology, University of Texas, MD Anderson Cancer Center, Houston, Texas
| | - Tiziano Scarabelli
- Division of Cardiology, Virginia Common Wealth University, Richmond, Virginia
| | - Tochukwu M Okwuosa
- Division of Cardiology, Rush University Medical Center, Chicago, Illinois
| | - Edward T H Yeh
- Center for Precision Medicine, Department of Medicine, University of Missouri, Columbia, Missouri.
| |
Collapse
|
32
|
Sandhu H, Cooper S, Hussain A, Mee C, Maddock H. Attenuation of Sunitinib-induced cardiotoxicity through the A3 adenosine receptor activation. Eur J Pharmacol 2017; 814:95-105. [DOI: 10.1016/j.ejphar.2017.08.011] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Revised: 08/08/2017] [Accepted: 08/09/2017] [Indexed: 12/11/2022]
|
33
|
Sheng CC, Amiri-Kordestani L, Palmby T, Force T, Hong CC, Wu JC, Croce K, Kim G, Moslehi J. 21st Century Cardio-Oncology: Identifying Cardiac Safety Signals in the Era of Personalized Medicine. JACC Basic Transl Sci 2016; 1:386-398. [PMID: 28713868 PMCID: PMC5508213 DOI: 10.1016/j.jacbts.2016.05.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 01/01/2023]
Abstract
Cardiotoxicity is a well-established complication of oncology therapies. Cardiomyopathy resulting from anthracyclines is a classic example. In the past decade, an explosion of novel cancer therapies, often targeted and more specific than traditional therapies, has revolutionized oncology therapy and dramatically changed cancer prognosis. However, some of these therapies have introduced an assortment of cardiovascular (CV) complications. At times, these devastating outcomes have only become apparent after drug approval and have limited the use of potent therapies. There is a growing need for better testing platforms, both for CV toxicity screening, as well as for elucidating mechanisms of cardiotoxicities of approved cancer therapies. This review discusses the utility of nonclinical models (in vitro, in vivo, & in silico) available and highlights recent advancements in modalities like human stem cell-derived cardiomyocytes for developing more comprehensive cardiotoxicity testing and new means of cardioprotection with targeted anticancer therapies.
Collapse
Affiliation(s)
- Calvin Chen Sheng
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Laleh Amiri-Kordestani
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Todd Palmby
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Thomas Force
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Charles C. Hong
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Research Medicine, Veterans Affairs Tennessee Valley Healthcare System, Nashville, Tennessee
- Accelerating Drug Repurposing Incubator, Vanderbilt Institute for Clinical and Translational Research, Nashville, Tennessee
| | - Joseph C. Wu
- Cardiovascular Division, Department of Medicine, Stanford University School of Medicine, Stanford, California
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, California
| | - Kevin Croce
- Division of Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts
| | - Geoffrey Kim
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, White Oak, Maryland
| | - Javid Moslehi
- Cardiovascular Division, Vanderbilt University School of Medicine, Nashville, Tennessee
- Cardio-Oncology Program, Vanderbilt University School of Medicine, Nashville, Tennessee
- Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| |
Collapse
|
34
|
Bellinger AM, Arteaga CL, Force T, Humphreys BD, Demetri GD, Druker BJ, Moslehi JJ. Cardio-Oncology: How New Targeted Cancer Therapies and Precision Medicine Can Inform Cardiovascular Discovery. Circulation 2016; 132:2248-58. [PMID: 26644247 DOI: 10.1161/circulationaha.115.010484] [Citation(s) in RCA: 91] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardio-oncology (the cardiovascular care of cancer patients) has developed as a new translational and clinical field based on the expanding repertoire of mechanism-based cancer therapies. Although these therapies have changed the natural course of many cancers, several may also lead to cardiovascular complications. Many new anticancer drugs approved over the past decade are "targeted" kinase inhibitors that interfere with intracellular signaling contributing to tumor progression. Unexpected cardiovascular and cardiometabolic effects of patient treatment with these inhibitors have provided unique insights into the role of kinases in human cardiovascular biology. Today, an ever-expanding number of cancer therapies targeting novel kinases and other specific cellular and metabolic pathways are being developed and tested in oncology clinical trials. Some of these drugs may affect the cardiovascular system in detrimental ways and others perhaps in beneficial ways. We propose that the numerous ongoing oncology clinical trials are an opportunity for closer collaboration between cardiologists and oncologists to study the cardiovascular and cardiometabolic changes caused by the modulation of these pathways in patients. In this regard, cardio-oncology represents an opportunity and a novel platform for basic and translational investigation and can serve as a potential avenue for optimization of anticancer therapies and for cardiovascular research and drug discovery.
Collapse
Affiliation(s)
- Andrew M Bellinger
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Carlos L Arteaga
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Thomas Force
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Benjamin D Humphreys
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - George D Demetri
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Brian J Druker
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.)
| | - Javid J Moslehi
- From Department of Medicine, Brigham and Women's Hospital, Boston, MA (A.M.B., G.D.D.); Vanderbilt-Ingram Cancer Center (C.L.A., J.M.), Cardiovascular Division (T.F., J.J.M.), and Cardio-Oncology Program (C.L.A., T.F., J.J.M.), Vanderbilt University School of Medicine, Nashville, TN; Department of Medicine, Washington University Medical Center, St. Louis, MO (B.D.H., J.M.); Dana-Farber Cancer Institute and Ludwig Center at Harvard, Harvard Medical School, Boston, MA (G.D.D.); and Knight Cancer Institute, Oregon Health & Science University and Howard Hughes Medical Institute, Portland, OR (B.J.D.).
| |
Collapse
|
35
|
Sundararajan S, Kumar A, Poongkunran M, Kannan A, Vogelzang NJ. Cardiovascular adverse effects of targeted antiangiogenic drugs: mechanisms and management. Future Oncol 2016; 12:1067-80. [DOI: 10.2217/fon.16.4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Anticancer treatment has evolved enormously over the last decade. Drugs targeting receptor tyrosine kinases, VEGFR and EGFR have changed the treatment landscape of certain cancers and have shifted the theme of anticancer therapy toward personalized care. However, these newer agents also come with unique side-effect profiles not seen with conventional chemotherapy including serious cardiovascular adverse effects. Hence, meticulous understanding of the adverse effects is crucial in maximizing clinical benefits and minimizing detrimental effects of these newer drugs. We have reviewed the cardiovascular adverse effects of anti-VEGF therapy in this article.
Collapse
Affiliation(s)
- Srinath Sundararajan
- Division of Hematology & Oncology, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Abhijeet Kumar
- Division of Hematology & Oncology, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Mugilan Poongkunran
- Department of Medicine, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Arun Kannan
- Division of Cardiology, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | - Nicholas J Vogelzang
- University of Nevada School of Medicine & US Oncology/Comprehensive Cancer Centers of Nevada, Las Vegas, NV, USA
| |
Collapse
|
36
|
Heart failure associated with small molecule tyrosine kinase inhibitors. Int J Cardiol 2016; 206:110-1. [DOI: 10.1016/j.ijcard.2016.01.059] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 01/02/2016] [Indexed: 01/07/2023]
|
37
|
|
38
|
Llurba E, Crispi F, Verlohren S. Update on the pathophysiological implications and clinical role of angiogenic factors in pregnancy. Fetal Diagn Ther 2015; 37:81-92. [PMID: 25659427 DOI: 10.1159/000368605] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/21/2014] [Indexed: 11/19/2022]
Abstract
Angiogenic markers are now being incorporated into clinical practice for the screening, diagnosing, and monitoring of preeclampsia. Pregnancy requires both vasculogenesis and angiogenesis in the fetal compartment and angiogenesis in the maternal compartment. Abnormal angiogenesis in the placenta determines impaired remodeling of the maternal spiral arteries and placental underperfusion that may ultimately lead to fetal growth restriction and maternal preeclampsia. The dysregulation of angiogenesis in the placenta and maternal-fetal circulation has emerged as one of the main pathophysiological features in the development of placental insufficiency and its clinical consequences. Abnormal angiogenesis has also been related to other obstetric and fetal conditions such as peripartum cardiomyopathy and fetal cardiac defects. This opens up new challenges for our understanding of angiogenic involvement in maternal cardiovascular function and fetal cardiac development, and it offers new clinical opportunities. This review summarizes the current knowledge of the pathophysiological implications and the clinical role of angiogenic factors in pregnancy.
Collapse
Affiliation(s)
- Elisa Llurba
- Department of Obstetrics, Maternal-Foetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain
| | | | | |
Collapse
|
39
|
Cross MJ, Berridge BR, Clements PJM, Cove-Smith L, Force TL, Hoffmann P, Holbrook M, Lyon AR, Mellor HR, Norris AA, Pirmohamed M, Tugwood JD, Sidaway JE, Park BK. Physiological, pharmacological and toxicological considerations of drug-induced structural cardiac injury. Br J Pharmacol 2015; 172:957-74. [PMID: 25302413 PMCID: PMC4314188 DOI: 10.1111/bph.12979] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/30/2014] [Accepted: 10/06/2014] [Indexed: 01/01/2023] Open
Abstract
The incidence of drug-induced structural cardiotoxicity, which may lead to heart failure, has been recognized in association with the use of anthracycline anti-cancer drugs for many years, but has also been shown to occur following treatment with the new generation of targeted anti-cancer agents that inhibit one or more receptor or non-receptor tyrosine kinases, serine/threonine kinases as well as several classes of non-oncology agents. A workshop organized by the Medical Research Council Centre for Drug Safety Science (University of Liverpool) on 5 September 2013 and attended by industry, academia and regulatory representatives, was designed to gain a better understanding of the gaps in the field of structural cardiotoxicity that can be addressed through collaborative efforts. Specific recommendations from the workshop for future collaborative activities included: greater efforts to identify predictive (i) preclinical; and (ii) clinical biomarkers of early cardiovascular injury; (iii) improved understanding of comparative physiology/pathophysiology and the clinical predictivity of current preclinical in vivo models; (iv) the identification and use of a set of cardiotoxic reference compounds for comparative profiling in improved animal and human cellular models; (v) more sharing of data (through publication/consortia arrangements) on target-related toxicities; (vi) strategies to develop cardio-protective agents; and (vii) closer interactions between preclinical scientists and clinicians to help ensure best translational efforts.
Collapse
Affiliation(s)
- M J Cross
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - B R Berridge
- Safety Assessment, GlaxoSmithKlineResearch Triangle Park, NC, USA
| | - P J M Clements
- David Jack Centre for Research & Development, GlaxoSmithKlineWare, Herts, UK
| | - L Cove-Smith
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - T L Force
- Center for Translational Medicine and Cardiology Division, Temple University School of MedicinePhiladelphia, PA, USA
| | - P Hoffmann
- Preclinical Safety, Novartis Pharm CorpEast Hanover, NJ, USA
| | - M Holbrook
- Safety Pharmacology, Covance Laboratories, Ltd.Harrogate, North Yorkshire, UK
| | - A R Lyon
- NIHR Cardiovascular Biomedical Research Unit, Royal Brompton Hospital and Imperial CollegeLondon, UK
| | - H R Mellor
- Drug Safety Evaluation, Vertex Pharmaceuticals (Europe), Ltd.Abingdon, Oxfordshire, UK
| | - A A Norris
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| | - M Pirmohamed
- The Wolfson Centre for Personalised Medicine, Institute of Translational Medicine, University of LiverpoolLiverpool, UK
| | - J D Tugwood
- Clinical & Experimental Pharmacology, Cancer Research UK Manchester Institute, University of ManchesterManchester, UK
| | - J E Sidaway
- Innovative Medicines, AstraZeneca R&DMacclesfield, UK
| | - B K Park
- MRC Centre for Drug Safety Science, Department of Molecular and Clinical Pharmacology, University of LiverpoolLiverpool, UK
| |
Collapse
|
40
|
Recognizing and managing left ventricular dysfunction associated with therapeutic inhibition of the vascular endothelial growth factor signaling pathway. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:335. [PMID: 25099086 DOI: 10.1007/s11936-014-0335-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OPINION STATEMENT Therapeutic inhibition of the vascular endothelial growth factor (VEGF) signaling pathway (VSP) is increasingly employed in the contemporary treatment of many cancer types. VSP inhibitors include the anti-VEGF monoclonal antibody (bevacizumab), soluble VEGF receptors (VEGF Trap), and small molecule tyrosine kinase inhibitors (TKIs) targeting the intracellular kinase domain of VEGF receptors. These agents are associated with cardiovascular toxicities such as hypertension, thrombosis, myocardial ischemia, and left ventricular (LV) dysfunction. Data on VSP inhibitor-associated LV dysfunction are largely limited to retrospective studies. Prospective studies are needed to establish the clinical significance of VSP inhibitor-associated LV dysfunction in the general population. Pre-clinical models of VSP inhibitor-associated LV dysfunction have identified mechanisms of cardiotoxicity and may improve our understanding of the pathophysiology underlying other cardiomyopathies. This review provides an overview of LV dysfunction that can occur in patients treated with VSP inhibitors. Potential strategies for clinical detection and management of this cardiotoxicity are explored, while acknowledging that currently available data specific to VSP-inhibitor LV dysfunction are limited. Avenues for future research are suggested.
Collapse
|
41
|
Duran JM, Makarewich CA, Trappanese D, Gross P, Husain S, Dunn J, Lal H, Sharp TE, Starosta T, Vagnozzi RJ, Berretta RM, Barbe M, Yu D, Gao E, Kubo H, Force T, Houser SR. Sorafenib cardiotoxicity increases mortality after myocardial infarction. Circ Res 2014; 114:1700-1712. [PMID: 24718482 DOI: 10.1161/circresaha.114.303200] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
RATIONALE Sorafenib is an effective treatment for renal cell carcinoma, but recent clinical reports have documented its cardiotoxicity through an unknown mechanism. OBJECTIVE Determining the mechanism of sorafenib-mediated cardiotoxicity. METHODS AND RESULTS Mice treated with sorafenib or vehicle for 3 weeks underwent induced myocardial infarction (MI) after 1 week of treatment. Sorafenib markedly decreased 2-week survival relative to vehicle-treated controls, but echocardiography at 1 and 2 weeks post MI detected no differences in cardiac function. Sorafenib-treated hearts had significantly smaller diastolic and systolic volumes and reduced heart weights. High doses of sorafenib induced necrotic death of isolated myocytes in vitro, but lower doses did not induce myocyte death or affect inotropy. Histological analysis documented increased myocyte cross-sectional area despite smaller heart sizes after sorafenib treatment, further suggesting myocyte loss. Sorafenib caused apoptotic cell death of cardiac- and bone-derived c-kit+ stem cells in vitro and decreased the number of BrdU+ (5-bromo-2'-deoxyuridine+) myocytes detected at the infarct border zone in fixed tissues. Sorafenib had no effect on infarct size, fibrosis, or post-MI neovascularization. When sorafenib-treated animals received metoprolol treatment post MI, the sorafenib-induced increase in post-MI mortality was eliminated, cardiac function was improved, and myocyte loss was ameliorated. CONCLUSIONS Sorafenib cardiotoxicity results from myocyte necrosis rather than from any direct effect on myocyte function. Surviving myocytes undergo pathological hypertrophy. Inhibition of c-kit+ stem cell proliferation by inducing apoptosis exacerbates damage by decreasing endogenous cardiac repair. In the setting of MI, which also causes large-scale cell loss, sorafenib cardiotoxicity dramatically increases mortality.
Collapse
Affiliation(s)
- Jason M Duran
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | | | - Danielle Trappanese
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Polina Gross
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Sharmeen Husain
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Jonathan Dunn
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Hind Lal
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Thomas E Sharp
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Timothy Starosta
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Ronald J Vagnozzi
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Remus M Berretta
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Mary Barbe
- Department of Anatomy and Cell Biology, Temple University School of Medicine, Philadelphia, PA
| | - Daohai Yu
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Erhe Gao
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Hajime Kubo
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| | - Thomas Force
- Center for Translational Medicine, Temple University School of Medicine, Philadelphia, PA
| | - Steven R Houser
- Cardiovascular Research Center, Temple University School of Medicine, Philadelphia, PA
| |
Collapse
|
42
|
Bracarda S, Sisani M, Marrocolo F, Hamzaj A, del Buono S, De Simone V. GOAL: An inverse toxicity-related algorithm for daily clinical practice decision making in advanced kidney cancer. Crit Rev Oncol Hematol 2014; 89:386-93. [DOI: 10.1016/j.critrevonc.2013.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 07/08/2013] [Accepted: 09/18/2013] [Indexed: 12/12/2022] Open
|
43
|
Zustovich F, Lombardi G, Farina P. Treating patients with metastatic renal carcinoma: an escape from Phase III. Expert Rev Anticancer Ther 2014; 12:919-27. [DOI: 10.1586/era.12.64] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
44
|
Chintalgattu V, Rees ML, Culver JC, Goel A, Jiffar T, Zhang J, Dunner K, Pati S, Bankson JA, Pasqualini R, Arap W, Bryan NS, Taegtmeyer H, Langley RR, Yao H, Kupferman ME, Entman ML, Dickinson ME, Khakoo AY. Coronary microvascular pericytes are the cellular target of sunitinib malate-induced cardiotoxicity. Sci Transl Med 2013; 5:187ra69. [PMID: 23720580 DOI: 10.1126/scitranslmed.3005066] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sunitinib malate is a multitargeted receptor tyrosine kinase inhibitor used in the treatment of human malignancies. A substantial number of sunitinib-treated patients develop cardiac dysfunction, but the mechanism of sunitinib-induced cardiotoxicity is poorly understood. We show that mice treated with sunitinib develop cardiac and coronary microvascular dysfunction and exhibit an impaired cardiac response to stress. The physiological changes caused by treatment with sunitinib are accompanied by a substantial depletion of coronary microvascular pericytes. Pericytes are a cell type that is dependent on intact platelet-derived growth factor receptor (PDGFR) signaling but whose role in the heart is poorly defined. Sunitinib-induced pericyte depletion and coronary microvascular dysfunction are recapitulated by CP-673451, a structurally distinct PDGFR inhibitor, confirming the role of PDGFR in pericyte survival. Thalidomide, an anticancer agent that is known to exert beneficial effects on pericyte survival and function, prevents sunitinib-induced pericyte cell death in vitro and prevents sunitinib-induced cardiotoxicity in vivo in a mouse model. Our findings suggest that pericytes are the primary cellular target of sunitinib-induced cardiotoxicity and reveal the pericyte as a cell type of concern in the regulation of coronary microvascular function. Furthermore, our data provide preliminary evidence that thalidomide may prevent cardiotoxicity in sunitinib-treated cancer patients.
Collapse
|
45
|
Verzoni E, de Braud F, Fabiani F, Grassi P, Testa I, Procopio G. Patient approach in advanced/metastatic renal cell carcinoma: focus on the elderly population and treatment-related toxicity. Future Oncol 2013; 9:1599-607. [DOI: 10.2217/fon.13.150] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Recent therapeutic advances have changed the treatment landscape of metastatic renal cell carcinoma. Unfortunately, the seven agents now available are not based on biomarkers that would indicate which one could provide the best benefit for every patient. We have reviewed the available information concerning the impact of each treatment on comorbidities or status that are frequently seen before commencing treatment for the advanced disease: elderly and patients with cardiovascular complications, metabolic and endocrinology disorders, and infections, as well as impaired organ function (kidney, liver and heart). Additional new drugs will be launched, but no predictive biomarkers are available. Head-to-head studies to evaluate the safety of the different drugs are rare. In this quite complex scenario, we believe that a decision-making approach focused on the patient may represent a suitable strategy.
Collapse
Affiliation(s)
- Elena Verzoni
- Department of Medical Oncology, Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Filippo de Braud
- Department of Medical Oncology, Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Francesca Fabiani
- Department of Emergency–Urgency, San Giuseppe General Hospital, Viale G. Boccaccio, Empoli (Florence), Italy
| | - Paolo Grassi
- Department of Medical Oncology, Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Isabella Testa
- Department of Medical Oncology, Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| | - Giuseppe Procopio
- Department of Medical Oncology, Unit 1, Fondazione IRCCS Istituto Nazionale dei Tumori, Via G. Venezian 1, 20133, Milan, Italy
| |
Collapse
|
46
|
Ky B, Vejpongsa P, Yeh ETH, Force T, Moslehi JJ. Emerging paradigms in cardiomyopathies associated with cancer therapies. Circ Res 2013; 113:754-64. [PMID: 23989717 DOI: 10.1161/circresaha.113.300218] [Citation(s) in RCA: 114] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The cardiovascular care of cancer patients (cardio-oncology) has emerged as a new discipline in clinical medicine, given recent advances in cancer therapy, and is driven by the cardiovascular complications that occur as a direct result of cancer therapy. Traditional therapies such as anthracyclines and radiation have been recognized for years to have cardiovascular complications. Less expected were the cardiovascular effects of targeted cancer therapies, which were initially thought to be specific to cancer cells and would spare any adverse effects on the heart. Cancers are typically driven by mutations, translocations, or overexpression of protein kinases. The majority of these mutated kinases are tyrosine kinases, though serine/threonine kinases also play key roles in some malignancies. Several agents were developed to target these kinases, but many more are in development. Major successes have been largely restricted to agents targeting human epidermal growth factor receptor-2 (mutated or overexpressed in breast cancer), BCR-ABL (chronic myelogenous leukemia and some cases of acute lymphoblastic leukemia), and c-Kit (gastrointestinal stromal tumor). Other agents targeting more complex malignancies, such as advanced solid tumors, have had successes, but have not extended life to the degree seen with chronic myelogenous leukemia. Years before the first targeted therapy, Judah Folkman correctly proposed that to address solid tumors one had to target the inherent neoangiogenesis. Unfortunately, emerging evidence confirms that angiogenesis inhibitors cause cardiac complications, including hypertension, thrombosis, and heart failure. And therein lies the catch-22. Nevertheless, cardio-oncology has the potential to be transformative as the human cardiomyopathies that arise from targeted therapies can provide insights into the normal function of the heart.
Collapse
Affiliation(s)
- Bonnie Ky
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | | | | | | | | |
Collapse
|
47
|
Llurba E, Sánchez O, Ferrer Q, Nicolaides KH, Ruíz A, Domínguez C, Sánchez-de-Toledo J, García-García B, Soro G, Arévalo S, Goya M, Suy A, Pérez-Hoyos S, Alijotas-Reig J, Carreras E, Cabero L. Maternal and foetal angiogenic imbalance in congenital heart defects. Eur Heart J 2013; 35:701-7. [PMID: 24159191 DOI: 10.1093/eurheartj/eht389] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
AIMS Animal models showed that angiogenesis is related to abnormal heart development. Our objectives were to ascertain whether a relationship exists between congenital heart defects (CHDs) and angiogenic/anti-angiogenic imbalance in maternal and foetal blood and study the expression of angiogenic factors in the foetal heart. METHODS AND RESULTS Maternal and cord blood placental growth factor (PlGF), soluble fms-like tyrosine kinase-1 (sFlt-1) and soluble endoglin (sEng) were compared in 65 cases of CHD and 204 normal controls. Angiogenic factor expression and markers of hypoxia were measured in heart tissue from 23 CHD foetuses and 8 controls. In the CHD group, compared with controls, plasma PlGF levels were significantly lower (367 ± 33 vs. 566 ± 26 pg/mL; P < 0.0001) and sFlt-1 significantly higher (2726 ± 450 vs. 1971 ± 130 pg/mL, P = 0.0438). Foetuses with CHD had higher cord plasma sFlt-1 (442 ± 76 vs. 274 ± 26 pg/mL; P = 0.0285) and sEng (6.76 ± 0.42 vs. 4.99 ± 0.49 ng/mL, P = 0.0041) levels. Expression of vascular endothelial growth factor (VEGF), sFlt-1, markers of chronic hypoxia, and antioxidant activity were significantly higher in heart tissue from CHD foetuses compared with normal hearts (VEGF, 1.59-fold; sFlt-1, 1.92-fold; hypoxia inducible factor (HIF)-2α, 1.45-fold; HO-1, 1.62-fold; SOD1, 1.31-fold). CONCLUSION An intrinsically angiogenic impairment exists in CHD that appears to be present in both the maternal and foetal circulation and foetal heart. Our data suggest that an imbalance of angiogenic-antiangiogenic factors is associated with developmental defects of the human heart.
Collapse
Affiliation(s)
- Elisa Llurba
- Department of Obstetrics, Maternal-Foetal Medicine Unit, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bair SM, Choueiri TK, Moslehi J. Cardiovascular complications associated with novel angiogenesis inhibitors: emerging evidence and evolving perspectives. Trends Cardiovasc Med 2013; 23:104-13. [PMID: 23290365 DOI: 10.1016/j.tcm.2012.09.008] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 09/11/2012] [Accepted: 09/12/2012] [Indexed: 12/21/2022]
Abstract
Novel cancer therapies targeting tumor angiogenesis have revolutionized treatment options in a variety of tumors. Specifically, VEGF signaling pathway (VSP) inhibitors have been introduced into clinical practice at a rapid pace over the last decade. It is becoming increasingly clear that VSP inhibitors can cause cardiovascular toxicities including hypertension, thrombosis, and heart failure. This review highlights these toxicities and proposes several strategies in their prevention and treatment. However, we recognize the dearth of data in this area and advocate a multi-disciplinary approach involving cardiologists and oncologists, as well as clinical and translational studies, in understanding and treating VSP-inhibitor associated toxicities.
Collapse
Affiliation(s)
- Steven M Bair
- Division of Cardiovascular Medicine, Department of Medicine, Brigham and Women's Hospital, MA 02115, USA
| | | | | |
Collapse
|
49
|
Gallucci G, Tartarone A, Tocchetti CG, Bochicchio AM, Coccaro M, Capobianco A, Maurea N, Improta G, Zupa A, Aieta M. Role of preeclampsia-related angiogenic factors in sunitinib cardiotoxicity: two cases and review of the literature. Future Oncol 2012; 9:127-33. [PMID: 23252570 DOI: 10.2217/fon.12.162] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Sunitinib is a multi-targeted tyrosine kinase inhibitor widely used in clear cell renal carcinoma and in imatinib-resistant gastrointestinal stromal tumors. Sunitinib-associated cardiotoxicity has been recognized and includes hypertension, left ventricular dysfunction and congestive heart failure; nevertheless, few data exist in the literature regarding the role of preeclampsia-related angiogenic factors in sunitinib cardiotoxicity. We report a case of sunitinib-induced severe left ventricular dysfunction that occurred in a hypertensive woman with metastatic renal carcinoma and a history of preeclampsia, and a case of sunitinib-induced preeclampsia-like syndrome in a normotensive patient with an imatinib-resistant gastrointestinal stromal tumor. Our experience confirms that inhibition of angiogenic factors to treat cancer is a novel challenge for the oncologist and requires the cardiologist's support.
Collapse
Affiliation(s)
- Giuseppina Gallucci
- Cardiology Unit, Centro di Riferimento Oncologico di Basilicata IRCCS, Via Padre Pio 1, 85028 Rionero in Vulture (PZ), Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Oliveira GH, Hardaway BW, Kucheryavaya AY, Stehlik J, Edwards LB, Taylor DO. Characteristics and survival of patients with chemotherapy-induced cardiomyopathy undergoing heart transplantation. J Heart Lung Transplant 2012; 31:805-10. [DOI: 10.1016/j.healun.2012.03.018] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2012] [Revised: 03/27/2012] [Accepted: 03/27/2012] [Indexed: 11/28/2022] Open
|