1
|
Almansouri NE, Bakkannavar S, Faheem Y, Jaiswal A, Shergill K, Boppana K, Nath TS. Efficacy of Angiotensin Receptor-Neprilysin Inhibitor and Its Renal Outcome in Heart Failure Patients: A Systematic Review of Randomized Clinical Trials. Cureus 2024; 16:e54501. [PMID: 38516430 PMCID: PMC10955452 DOI: 10.7759/cureus.54501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 02/19/2024] [Indexed: 03/23/2024] Open
Abstract
Heart failure (HF) is a major cause of morbidity and mortality and imposes a significant financial burden on healthcare systems globally. Angiotensin receptor-neprilysin inhibitor (ARNI), a novel neuroendocrine inhibitor, is frequently used in treating HF. However, there is still limited understanding regarding how it compares to other neuroendocrine inhibitors, such as angiotensin-converting enzyme inhibitors (ACEis) and angiotensin receptor blockers (ARBs). The purpose of this research is to present the most recent data regarding the efficacy and renal impact of ARNIs in the treatment of HF in comparison to ACE inhibitors and ARBs. Several large-scale randomized controlled trials (RCTs) have recently been conducted to evaluate the benefits of this drug in patients with different types of HF, regardless of their renal status. We searched multiple databases, including PubMed, PubMed Central (PMC), and Google Scholar, to find relevant RCTs. The efficacy outcome was a composite of the rate of death from cardiovascular causes, the frequency of HF hospitalizations (HFH), and alterations in N-terminal pro-brain natriuretic peptide (NT-proBNP) levels. The renal outcome was impairment of renal function. This systematic review analyzed large-scale RCTs involving 17,327 participants, with an average follow-up time of approximately 2.9 years. sacubitril/valsartan showed notable improvements compared to ACEis and ARBs in the following areas: reduction in NT-proBNP levels, prevention of further deterioration in renal function, and decreased hospitalizations for HF. Interestingly, there is no increased risk of mortality from cardiovascular causes with sacubitril or valsartan.
Collapse
Affiliation(s)
- Naiela E Almansouri
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
- Internal Medicine, University of Tripoli, Tripoli, LBY
| | - Saloni Bakkannavar
- Pediatrics, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Youmna Faheem
- Pediatrics, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Amisha Jaiswal
- Internal Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | - Kainaat Shergill
- Surgery, Maharishi Markandeshwar Institute of Medical Sciences and Research, Mullana, IND
| | - Kusalik Boppana
- Medicine, California Institute of Behavioral Neurosciences and Psychology, Fairfield, USA
| | | |
Collapse
|
2
|
Zhang J, Li W, Xue S, Gao P, Wang H, Chen H, Hong Y, Sun Q, Lu L, Wang Y, Wang Q. Qishen granule attenuates doxorubicin-induced cardiotoxicity by protecting mitochondrial function and reducing oxidative stress through regulation of Sirtuin3. JOURNAL OF ETHNOPHARMACOLOGY 2024; 319:117134. [PMID: 37714227 DOI: 10.1016/j.jep.2023.117134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 08/31/2023] [Accepted: 09/05/2023] [Indexed: 09/17/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Doxorubicin (DOX) is one of the most potent chemotherapy drugs available today. However, the adverse effect of cardiotoxicity limits its clinical application. New approaches are being investigated for the treatment of doxorubicin-induced cardiotoxicity (DIC). Doxorubicin is enriched in mitochondria and it could induce imbalance of protein modification, including acetylation of mitochondria proteins, thereby inducing DIC. Restoration of mitochondria function is an effective way to attenuate DIC. The formula for traditional Chinese medicine Granules of Qishen (QSG) was derived from the classic formula "Zhen-Wu-Tang" which has been extensively used in the treatment of myocardial infarction. It consists of six traditional Chinese medicines, including Astragalus membranaceus var. mongholicus (Bunge) P.K.Hsiao (Fabaceae), Salvia miltiorrhiza Bunge (Lamiaceae), Lonicera japonica Thunb. (Caprifoliaceae), Aconitum carmichaelii Debeaux (Ranunculaceae), Scrophularia ningpoensis Hemsl. (Scrophulariaceae), and Glycyrrhiza uralensis Fisch. (Fabaceae). QSG is a potential anti-DIC formula. A better understanding of the effectiveness and pharmacological mechanisms of QSG will aid in the prevention and treatment of DIC. AIM OF THE STUDY The purpose of this research was to explore the effectiveness of QSG in the treatment of DIC and to explore whether QSG could protect mitochondrial function and reduce oxidative damage by activating Sirtuin3(SIRT3)/Acetylated-superoxide dismutase 2(Ac-SOD2) signaling pathway. MATERIALS AND METHODS DOX was injected into mice through the tail vein to construct a mouse model of DOX-induced cardiotoxicity to explore the therapeutic effect of QSG in animals. Meanwhile, the H9C2 cell model was used to study the mechanism of QSG. The cardiac function was evaluated by echocardiography, hematoxylin-eosin (H&E) staining and measurement of serum levels of creatine kinase isoenzymes (CK-MB) and lactate dehydrogenase (LDH). Oxidative damage was evaluated by 2',7'-dichlorodihydro fluorescein diacetate (DCFH-DA) staining and Mito-SOX Red staining. Levels of total superoxide dismutase (SOD) activity and malondialdehyde (MDA) content were measured by following the instructions of commercially available kits. In order to detect the changes in mitochondrial membrane potential, cells were stained using the mitochondrial membrane potential detection kit (JC-1). Western blot analysis was applied to detect protein expressions of SIRT3, Ac-SOD2, Acetylation Lysine (Ac-Lys), Bax and Bcl-2. H9C2 cells were treated with SIRT3 inhibitor, in order to determine if QSG had effects via the SIRT3/Ac-SOD2 pathway. RESULTS In vivo studies showed that QSG ameliorated doxorubicin-induced damage of cardiac function in DIC mice model. The ejection fraction (EF) and fractional shortening (FS) were all up-regulated by QSG treatment. QSG decreased MDA levels and increased SOD activity. Meanwhile, doxorubicin induced high level of protein acetylation and QSG restored the acetylated protein back to normal levels. In particular, QSG upregulated expression of SIRT3 and downregulated Ac-SOD level. In vitro study demonstrated that QSG restored mitochondrial membrane potential, increased ATP level and reduced mitochondrial ROS production. When H9C2 cells were co-incubated with SIRT3 inhibitor, the efficacies of QSG on mitochondrial function were abrogated. Meanwhile, the regulative effects of QSG on SIRT3/Ac-SOD2 pathway were also abolished. CONCLUSION This study demonstrates that QSG is effective in treating DIC. QSG ameliorates oxidative damage and protects mitochondrial function partly by restoring protein acetylation level and by activating the SIRT3/Ac-SOD2 pathway.
Collapse
Affiliation(s)
- Jingmei Zhang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Weili Li
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Siming Xue
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Pengrong Gao
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Hui Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Huan Chen
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Yiqin Hong
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Qianbin Sun
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Linghui Lu
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, 100029, China.
| | - Yong Wang
- College of Chinese Medicine, Beijing University of Chinese Medicine, Beijing, 100029, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, 100029, China.
| | - Qiyan Wang
- School of Life Sciences, Beijing University of Chinese Medicine, Beijing, 100029, China; Beijing Key Laboratory of TCM Syndrome and Formula, Beijing University of Chinese Medicine, Beijing, 100029, China; Key Laboratory of TCM Syndrome and Formula (Beijing University of Chinese Medicine), Ministry of Education, Beijing, 100029, China.
| |
Collapse
|
3
|
Zhang S, Yahaya BH, Pan Y, Liu Y, Lin J. Menstrual blood-derived endometrial stem cell, a unique and promising alternative in the stem cell-based therapy for chemotherapy-induced premature ovarian insufficiency. Stem Cell Res Ther 2023; 14:327. [PMID: 37957675 PMCID: PMC10644549 DOI: 10.1186/s13287-023-03551-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Chemotherapy can cause ovarian dysfunction and infertility since the ovary is extremely sensitive to chemotherapeutic drugs. Apart from the indispensable role of the ovary in the overall hormonal milieu, ovarian dysfunction also affects many other organ systems and functions including sexuality, bones, the cardiovascular system, and neurocognitive function. Although conventional hormone replacement therapy can partly relieve the adverse symptoms of premature ovarian insufficiency (POI), the treatment cannot fundamentally prevent deterioration of POI. Therefore, effective treatments to improve chemotherapy-induced POI are urgently needed, especially for patients desiring fertility preservation. Recently, mesenchymal stem cell (MSC)-based therapies have resulted in promising improvements in chemotherapy-induced ovary dysfunction by enhancing the anti-apoptotic capacity of ovarian cells, preventing ovarian follicular atresia, promoting angiogenesis and improving injured ovarian structure and the pregnancy rate. These improvements are mainly attributed to MSC-derived biological factors, functional RNAs, and even mitochondria, which are directly secreted or indirectly translocated with extracellular vesicles (microvesicles and exosomes) to repair ovarian dysfunction. Additionally, as a novel source of MSCs, menstrual blood-derived endometrial stem cells (MenSCs) have exhibited promising therapeutic effects in various diseases due to their comprehensive advantages, such as periodic and non-invasive sample collection, abundant sources, regular donation and autologous transplantation. Therefore, this review summarizes the efficacy of MSCs transplantation in improving chemotherapy-induced POI and analyzes the underlying mechanism, and further discusses the benefit and existing challenges in promoting the clinical application of MenSCs in chemotherapy-induced POI.
Collapse
Affiliation(s)
- Shenghui Zhang
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute (IPPT), Universiti Sains Malaysia, Penang, Malaysia
| | - Ying Pan
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, , China
| | - Yanli Liu
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| | - Juntang Lin
- Stem Cell and Biotherapy Technology Research Center, Henan Joint International Research Laboratory of Stem Cell Medicine, Xinxiang Medical University, East of JinSui Road, Xinxiang, Henan, China.
| |
Collapse
|
4
|
Mikail N, Chequer R, Imperiale A, Meisel A, Bengs S, Portmann A, Gimelli A, Buechel RR, Gebhard C, Rossi A. Tales from the future-nuclear cardio-oncology, from prediction to diagnosis and monitoring. Eur Heart J Cardiovasc Imaging 2023; 24:1129-1145. [PMID: 37467476 PMCID: PMC10501471 DOI: 10.1093/ehjci/jead168] [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: 04/25/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
Cancer and cardiovascular diseases (CVD) often share common risk factors, and patients with CVD who develop cancer are at high risk of experiencing major adverse cardiovascular events. Additionally, cancer treatment can induce short- and long-term adverse cardiovascular events. Given the improvement in oncological patients' prognosis, the burden in this vulnerable population is slowly shifting towards increased cardiovascular mortality. Consequently, the field of cardio-oncology is steadily expanding, prompting the need for new markers to stratify and monitor the cardiovascular risk in oncological patients before, during, and after the completion of treatment. Advanced non-invasive cardiac imaging has raised great interest in the early detection of CVD and cardiotoxicity in oncological patients. Nuclear medicine has long been a pivotal exam to robustly assess and monitor the cardiac function of patients undergoing potentially cardiotoxic chemotherapies. In addition, recent radiotracers have shown great interest in the early detection of cancer-treatment-related cardiotoxicity. In this review, we summarize the current and emerging nuclear cardiology tools that can help identify cardiotoxicity and assess the cardiovascular risk in patients undergoing cancer treatments and discuss the specific role of nuclear cardiology alongside other non-invasive imaging techniques.
Collapse
Affiliation(s)
- Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Renata Chequer
- Department of Nuclear Medicine, Bichat University Hospital, AP-HP, University Diderot, 75018 Paris, France
| | - Alessio Imperiale
- Nuclear Medicine, Institut de Cancérologie de Strasbourg Europe (ICANS), University Hospitals of Strasbourg, 67093 Strasbourg, France
- Molecular Imaging-DRHIM, IPHC, UMR 7178, CNRS/Unistra, 67093 Strasbourg, France
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Kantonsspital Glarus, Burgstrasse 99, 8750 Glarus, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Alessia Gimelli
- Imaging Department, Fondazione CNR/Regione Toscana Gabriele Monasterio, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Cathérine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Cardiology, University Hospital Inselspital Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| |
Collapse
|
5
|
Tsigkas G, Vakka A, Apostolos A, Bousoula E, Vythoulkas-Biotis N, Koufou EE, Vasilagkos G, Tsiafoutis I, Hamilos M, Aminian A, Davlouros P. Dual Antiplatelet Therapy and Cancer; Balancing between Ischemic and Bleeding Risk: A Narrative Review. J Cardiovasc Dev Dis 2023; 10:135. [PMID: 37103014 PMCID: PMC10144375 DOI: 10.3390/jcdd10040135] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2023] [Revised: 03/15/2023] [Accepted: 03/19/2023] [Indexed: 04/28/2023] Open
Abstract
Cardiovascular (CV) events in patients with cancer can be caused by concomitant CV risk factors, cancer itself, and anticancer therapy. Since malignancy can dysregulate the hemostatic system, predisposing cancer patients to both thrombosis and hemorrhage, the administration of dual antiplatelet therapy (DAPT) to patients with cancer who suffer from acute coronary syndrome (ACS) or undergo percutaneous coronary intervention (PCI) is a clinical challenge to cardiologists. Apart from PCI and ACS, other structural interventions, such as TAVR, PFO-ASD closure, and LAA occlusion, and non-cardiac diseases, such as PAD and CVAs, may require DAPT. The aim of the present review is to review the current literature on the optimal antiplatelet therapy and duration of DAPT for oncologic patients, in order to reduce both the ischemic and bleeding risk in this high-risk population.
Collapse
Affiliation(s)
- Grigorios Tsigkas
- Department of Cardiology, University Hospital of Patras, 265 04 Patras, Greece; (A.V.); (A.A.); (N.V.-B.); (E.-E.K.); (G.V.); (P.D.)
| | - Angeliki Vakka
- Department of Cardiology, University Hospital of Patras, 265 04 Patras, Greece; (A.V.); (A.A.); (N.V.-B.); (E.-E.K.); (G.V.); (P.D.)
| | - Anastasios Apostolos
- Department of Cardiology, University Hospital of Patras, 265 04 Patras, Greece; (A.V.); (A.A.); (N.V.-B.); (E.-E.K.); (G.V.); (P.D.)
- First Department of Cardiology, Hippocration General Hospital, National and Kapodistrian University of Athens, 157 72 Athens, Greece
| | - Eleni Bousoula
- Department of Cardiology, Tzaneio General Hospital, 185 36 Piraeus, Greece;
| | - Nikolaos Vythoulkas-Biotis
- Department of Cardiology, University Hospital of Patras, 265 04 Patras, Greece; (A.V.); (A.A.); (N.V.-B.); (E.-E.K.); (G.V.); (P.D.)
| | - Eleni-Evangelia Koufou
- Department of Cardiology, University Hospital of Patras, 265 04 Patras, Greece; (A.V.); (A.A.); (N.V.-B.); (E.-E.K.); (G.V.); (P.D.)
| | - Georgios Vasilagkos
- Department of Cardiology, University Hospital of Patras, 265 04 Patras, Greece; (A.V.); (A.A.); (N.V.-B.); (E.-E.K.); (G.V.); (P.D.)
| | - Ioannis Tsiafoutis
- First Department of Cardiology, Red Cross Hospital, 115 26 Athens, Greece;
| | - Michalis Hamilos
- Department of Cardiology, Heraklion University Hospital, 715 00 Heraklion, Crete, Greece;
| | - Adel Aminian
- Department of Cardiology, Centre Hospitalier Universitaire de Charleroi, 6042 Charleroi, Belgium;
| | - Periklis Davlouros
- Department of Cardiology, University Hospital of Patras, 265 04 Patras, Greece; (A.V.); (A.A.); (N.V.-B.); (E.-E.K.); (G.V.); (P.D.)
| |
Collapse
|
6
|
Gao J, Zhao C, Zhang WZ, Liu S, Xin H, Lian ZX. Efficacy and safety profile of angiotensin receptor neprilysin inhibitors in the management of heart failure: a systematic review and meta-analysis of randomized controlled trials. Heart Fail Rev 2022:10.1007/s10741-022-10273-3. [PMID: 36184714 DOI: 10.1007/s10741-022-10273-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/19/2022] [Indexed: 11/29/2022]
Abstract
Several guidelines have recommended the use of angiotensin receptor neprilysin inhibitors (ARNIs) as replacement for angiotensin-converting enzyme inhibitors in the management of heart failure. Till date, there are no reviews done that comprehensively cover different aspects of efficacy and safety parameters. Hence, we have performed a comprehensive systematic review and meta-analysis on role of ARNIs for the management of heart failure patients. Searches were done in Embase, Scopus, China National Knowledge Infrastructure, Chinese Biomedical Literature Database, PubMed Central, Cochrane Library, MEDLINE, Google Scholar, ScienceDirect and Clinicaltrials.gov until June 2022. Risk of bias assessment was done with Cochrane's risk of bias tool. Meta-analysis was carried out using random-effects model. Pooled standardized mean difference (SMD)/mean difference (MD) and/or risk ratio (RR) with 95% confidence intervals (CIs) was reported. In total, we analysed 34 studies, with almost all of them had a high risk of bias. Pooled RR was 0.88 (95% CI: 0.82-0.95) for all-cause mortality, 0.84 (95% CI: 0.77-0.92) for cardiovascular mortality and 0.78 (95% CI: 0.70-0.87) for hospitalization. Pooled MD was 3.74 (95% CI: 1.93-5.55) for left ventricular ejection fraction, -2.16 (95% CI: -3.58 to -0.74) for left atrial volume index, -3.80 (95% CI: -6.60 to -1.00) for left ventricular end-diastolic dimension and -1.16 (95% CI: -1.98 to -0.35) for E/E' ratio. Regarding adverse events, pooled RR was 1.55 (95% CI: 1.31-1.85) for symptomatic hypotension, 0.93 (95% CI: 0.78-1.11) for worsening renal function, 1.09 (95% CI: 0.94-1.26) for hyperkalaemia and 1.29 (95% CI: 0.67-2.50) for angioedema. ARNIs had beneficial efficacy and safety profile on the management of heart failure especially patients with reduced ejection fraction.
Collapse
Affiliation(s)
- Juan Gao
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Cong Zhao
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Zhong Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Song Liu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Hui Xin
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhe-Xun Lian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China.
| |
Collapse
|
7
|
Yaseen IF, Farhan HA. Cardiovascular drug interventions in the cardio-oncology clinic by a cardiology pharmacist: ICOP-Pharm study. Front Cardiovasc Med 2022; 9:972455. [PMID: 36247485 PMCID: PMC9556995 DOI: 10.3389/fcvm.2022.972455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Accepted: 09/06/2022] [Indexed: 12/03/2022] Open
Abstract
Background Cardio-oncology is a rapidly growing field that requires a novel service design to deal with the increasing number of patients. It is reported that the volume of patients at the cardio-oncology clinic in the United Kingdom is 535 patients/5 years and in Canada is 779 patients/7 years. The pharmacist has a role in reducing the consultation time of physicians. Objective To identify the role of a qualified cardiology pharmacist at the cardio-oncology clinic using a new paradigm based on complementary interventions with the cardiologist for the management of patients with cancer and cardiovascular risk factors and/or cardiovascular diseases (CVRF/CVD). Methods A prospective observational study was conducted at the cardio-oncology clinic in the Medical City in Baghdad, Iraq between December 2020 and December 2021. Patients with CVRF/CVD were registered. The Iraqi Cardio-Oncology Program-Pharmacist (ICOP-Pharm) paradigm was designed to involve a qualified cardiology pharmacist for initial cardiovascular (CV) drug interventions. Results Among 333 patients who attended our clinic over the 1-year interval, 200 (60%) CVRF/CVD cases were enrolled in the study, and of them 79 (40%) patients had CV drug interventions. A total of 196 interventions were done, including 147 (75%) cases performed by the cardiology pharmacist, and 92 (63%) of the latter were CV drug initiations. Among the total CVRF/CVD treated initially by the cardiology pharmacist, hypertension 32 (26%) and cancer therapy-related cardiac dysfunction 29 (24%) were the main types. Conclusion The qualified cardiology pharmacist was responsible for three-quarters of the initial CV drug interventions at the cardio-oncology clinic in a complementary approach to the cardiologist. The role of the cardiology pharmacist in the ICOP-Pharm paradigm may be one of the reasons for the ability of the heart team to manage 3-fold of the patient volume when compared with those in the United Kingdom or Canada.
Collapse
Affiliation(s)
- Israa Fadhil Yaseen
- Baghdad Heart Center, Medical City, Baghdad, Iraq
- Scientific Council of Cardiology, Iraqi Board for Medical Specializations, Baghdad, Iraq
| | - Hasan Ali Farhan
- Baghdad Heart Center, Medical City, Baghdad, Iraq
- Scientific Council of Cardiology, Iraqi Board for Medical Specializations, Baghdad, Iraq
| |
Collapse
|
8
|
A Fibrosis Biomarker Early Predicts Cardiotoxicity Due to Anthracycline-Based Breast Cancer Chemotherapy. Cancers (Basel) 2022; 14:cancers14122941. [PMID: 35740602 PMCID: PMC9221256 DOI: 10.3390/cancers14122941] [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: 04/27/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/25/2022] Open
Abstract
Anthracycline-based cancer chemotherapy (ACC) causes myocardial fibrosis, a lesion contributing to left ventricular dysfunction (LVD). We investigated whether the procollagen-derived type-I C-terminal-propeptide (PICP): (1) associates with subclinical LVD (sLVD) at 3-months after ACC (3m-post-ACC); (2) predicts cardiotoxicity 1-year after ACC (12m-post-ACC) in breast cancer patients (BC-patients); and (3) associates with LVD in ACC-induced heart failure patients (ACC-HF-patients). Echocardiography, serum PICP and biomarkers of cardiomyocyte damage were assessed in two independent cohorts of BC-patients: CUN (n = 87) at baseline, post-ACC, and 3m and 12m (n = 65)-post-ACC; and HULAFE (n = 70) at baseline, 3m and 12m-post-ACC. Thirty-seven ACC-HF-patients were also studied. Global longitudinal strain (GLS)-based sLVD (3m-post-ACC) and LV ejection fraction (LVEF)-based cardiotoxicity (12m-post-ACC) were defined according to guidelines. BC-patients: all biomarkers increased at 3m-post-ACC versus baseline. PICP was particularly increased in patients with sLVD (interaction-p < 0.001) and was associated with GLS (p < 0.001). PICP increase at 3m-post-ACC predicted cardiotoxicity at 12m-post-ACC (odds-ratio ≥ 2.95 per doubling PICP, p ≤ 0.025) in both BC-cohorts, adding prognostic value to the early assessment of GLS and LVEF. ACC-HF-patients: PICP was inversely associated with LVEF (p = 0.004). In ACC-treated BC-patients, an early increase in PICP is associated with early sLVD and predicts cardiotoxicity 1 year after ACC. PICP is also associated with LVD in ACC-HF-patients.
Collapse
|
9
|
Vasileiou PVS, Siasos G, Gorgoulis VG. Molecular biomarkers in cardio-oncology: Where we stand and where we are heading. Bioessays 2022; 44:e2100234. [PMID: 35352831 DOI: 10.1002/bies.202100234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 03/01/2022] [Accepted: 03/15/2022] [Indexed: 11/08/2022]
Abstract
Until recently, cardiotoxicity in the setting of a malignant disease was attributed solely to the detrimental effects of chemo- and/or radio-therapy to the heart. On this account, the focus was on the evaluation of well-established cardiac biomarkers for the early detection of myocardial damage. Currently, this view has been revised. Cardiotoxicity is not restricted to a single organ but instead affects the endothelium as a whole. Indeed, it has come into light that not only cancer therapy but also malignant cells per se can impair the cardiovascular system, through a paracrine and endocrine mode of action. Even more intriguingly, a clear interplay between molecular pathways involved in cancer and cardiovascular disease has become prevalent, suggesting a common nominator that governs the pathophysiology of these two entities. Taken together, our strategy in the quest of novel biomarkers in the emerging field of cardio-oncology should be critically reshaped.
Collapse
Affiliation(s)
- Panagiotis V S Vasileiou
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Gerasimos Siasos
- Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Vassilis G Gorgoulis
- Molecular Carcinogenesis Group, Department of Histology and Embryology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Faculty Institute for Cancer Sciences, Manchester Academic Health Sciences Centre, University of Manchester, Manchester, UK.,Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Center for New Biotechnologies and Precision Medicine, Medical School, National and Kapodistrian University of Athens, Athens, Greece.,Faculty of Health and Medical Sciences, University of Surrey, Surrey, UK
| |
Collapse
|
10
|
Cardiotoxicity—current recommendations of prevention and treatment. MEMO - MAGAZINE OF EUROPEAN MEDICAL ONCOLOGY 2022. [DOI: 10.1007/s12254-021-00766-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SummaryTremendous advances in modern oncology therapies enable an increasing life expectancy of many cancer entities. Short or long-term cardiovascular side effects, however, gain importance. The current review focuses on recent recommendations for strategies of preventing and treating cardiotoxicity. A personalized assessment of the baseline risk of cardiotoxicity is recommended in all patients, without delaying the initiation of the cancer therapy. A baseline ECG, biomarkers (NT-proBNP, troponin), blood pressure and echocardiography should be obtained in all patients scheduled for potentially cardiotoxic treatments. Cardiac risk factors, e.g., coronary disease, hypertension, elevated lipids, should be promptly treated and optimized. Increased surveillance with more frequent cardiac imaging and sequential biomarker assessment during the cycles is recommended in high-risk cardiac patients. New imaging methods in echocardiography such as speckle tracking global longitudinal strain reflecting early myocardial ventricular deterioration are proposed in recent recommendations. Signs of cardiotoxicity should induce early treatment by, e.g., ACE-inhibitors, beta-blockers and/or other heart failure therapies. Immune therapies, e.g., checkpoint-inhibitors can induce cardiac events such as arrhythmias, acute coronary syndrome with plaque rupture, or myocarditis, even in negative magnetic resonance imaging or normal echocardiography findings. Troponin, BNP and ECG may help to identify these potentially deleterious side effects. Furthermore, there is a bidirectional influence of heart disease and cancer, e.g., by common inflammatory pathways. Pre-existent heart disease leads to worse prognosis in cancer, necessitating close follow-up and cardiac treatment during cancer therapy. On the other hand, cardiovascular mortality is increased after cancer survival and periodic cardiac follow-up is recommended long-term especially after chemotherapy and-or radiation.
Collapse
|
11
|
Lin J, Zhou J, Xie G, Liu J. Efficacy and safety of sacubitril-valsartan in patients with heart failure: a systematic review and meta-analysis of randomized clinical trials: A PRISMA-compliant article. Medicine (Baltimore) 2021; 100:e28231. [PMID: 34967357 PMCID: PMC8718238 DOI: 10.1097/md.0000000000028231] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 11/15/2021] [Accepted: 11/24/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND To investigate the efficacy and safety of sacubitril-valsartan in patients with heart failure, relevant randomized clinical trials (RCTs) were analyzed. METHODS We used Cochrane Library, PubMed web of science, CNKI, VIP, Medline, ISI Web of Science, CBMdisc, and Wanfang database to conduct a systematic literature research. A fixed-effects model was used to evaluate the standardized mean differences (SMDs) with 95% confidence intervals. We conducted sensitivity analysis and analyzed publication bias to comprehensively estimate the efficacy and safety of sacubitril-valsartan in patients with heart failure. RESULTS Among 132 retrieved studies, 5 relevant RCTs were included in the meta-analysis. The result showed that left ventricular ejection fraction (LVEF) was improved after sacubitril-valsartan in patients with heart failure, with an SMD (95% CI of 1.1 [1.01, 1.19] and P < .00001 fixed-effects model). Combined outcome indicators showed that, combined outcome indicators showed that, compared with control group, the left ventricular volume index (LAVI) (WMD = -2.18, 95% CI [-3.63, -0.74], P = .003), the E/e' (WMD = -1.01, 95% CI [-1.89, -0.12], P = .03), the cardiovascular death (RR = 0.89, 95% CI [0.83, 0.96], P = .003], and the rehospitalization rate of heart failure (RR = 0.83, 95% CI [0.78, 0.88], P < .01) decreased more significantly, but it had no effect on renal function (WMD = 0.74, 95% CI [0.54, 1.01], P = .06). CONCLUSIONS The present meta-analysis suggested that sacubitril-valsartan may improve the cardiac function of heart failure. Given the limited number of included studies, additional large sample-size RCTs are required to determine the long-term effect of cardiac function of sacubitril-valsartan in patients with heart failure.
Collapse
|
12
|
Stretti L, Zippo D, Coats AJ, Anker MS, von Haehling S, Metra M, Tomasoni D. A year in heart failure: an update of recent findings. ESC Heart Fail 2021; 8:4370-4393. [PMID: 34918477 PMCID: PMC9073717 DOI: 10.1002/ehf2.13760] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 11/28/2021] [Accepted: 12/01/2021] [Indexed: 12/22/2022] Open
Abstract
Major changes have occurred in these last years in heart failure (HF) management. Landmark trials and the 2021 European Society of Cardiology guidelines for the diagnosis and treatment of HF have established four classes of drugs for treatment of HF with reduced ejection fraction: angiotensin-converting enzyme inhibitors or an angiotensin receptor-neprilysin inhibitor, beta-blockers, mineralocorticoid receptor antagonists, and sodium-glucose co-transporter 2 inhibitors, namely, dapagliflozin or empagliflozin. These drugs consistently showed benefits on mortality, HF hospitalizations, and quality of life. Correction of iron deficiency is indicated to improve symptoms and reduce HF hospitalizations. AFFIRM-AHF showed 26% reduction in total HF hospitalizations with ferric carboxymaltose vs. placebo in patients hospitalized for acute HF (P = 0.013). The guanylate cyclase activator vericiguat and the myosin activator omecamtiv mecarbil improved outcomes in randomized placebo-controlled trials, and vericiguat is now approved for clinical practice. Treatment of HF with preserved ejection fraction (HFpEF) was a major unmet clinical need until this year when the results of EMPEROR-Preserved (EMPagliflozin outcomE tRial in Patients With chrOnic HFpEF) were issued. Compared with placebo, empagliflozin reduced by 21% (hazard ratio, 0.79; 95% confidence interval, 0.69 to 0.90; P < 0.001), the primary outcome of cardiovascular death or HF hospitalization. Advances in the treatment of specific phenotypes of HF, including atrial fibrillation, valvular heart disease, cardiomyopathies, cardiac amyloidosis, and cancer-related HF, also occurred. Coronavirus disease 2019 (COVID-19) pandemic still plays a major role in HF epidemiology and management. All these aspects are highlighted in this review.
Collapse
Affiliation(s)
- Lorenzo Stretti
- Cardiology, Cardio‐Thoracic Department, Civil Hospitals; Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Dauphine Zippo
- Cardiology, Cardio‐Thoracic Department, Civil Hospitals; Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | | | - Markus S. Anker
- Department of Cardiology (CBF)Charité ‐ Universitätsmedizin BerlinBerlinGermany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
- German Centre for Cardiovascular Research (DZHK), partner site BerlinBerlinGermany
| | - Stephan von Haehling
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CenterGöttingenGermany
- German Center for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Marco Metra
- Cardiology, Cardio‐Thoracic Department, Civil Hospitals; Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| | - Daniela Tomasoni
- Cardiology, Cardio‐Thoracic Department, Civil Hospitals; Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
| |
Collapse
|
13
|
Exercise to Reduce Anthracycline-Mediated Cardiovascular Complications in Breast Cancer Survivors. ACTA ACUST UNITED AC 2021; 28:4139-4156. [PMID: 34677269 PMCID: PMC8535000 DOI: 10.3390/curroncol28050351] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 01/12/2023]
Abstract
While developments in cancer therapeutics have greatly reduced morbidity and mortality in females with breast cancer, it comes at a cost of an increased risk of cardiovascular complications. In particular, anthracyclines, like doxorubicin, which are a mainstay of current chemotherapy regimens, are associated with dose-dependent cardiotoxicity. Exercise has been widely accepted as an effective intervention in reducing cardiovascular risk in a variety of different clinical conditions. However, the benefits of exercise in anthracycline-mediated cardiotoxicity are not clearly understood. First, this review discusses the pre-clinical studies which have elucidated the cardioprotective mechanisms of aerobic and resistance exercise in improving cardiovascular function in the setting of anthracycline treatment. Next, it aims to summarize the results of aerobic and resistance exercise clinical trials conducted in females with breast cancer who received anthracycline-based chemotherapy. The review further discusses the current exercise guidelines for women undergoing chemotherapy and contraindications for exercise. Finally, the review addresses gaps in research, specifically the need for further clinical trials to establish a recommended exercise prescription within this patient population.
Collapse
|
14
|
Boyle DA. Contemporary Insights into Cancer Cachexia for Oncology Nurses. Asia Pac J Oncol Nurs 2021; 8:462-470. [PMID: 34527776 PMCID: PMC8420918 DOI: 10.4103/apjon.apjon-2151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/21/2021] [Indexed: 12/02/2022] Open
Abstract
Cachexia is a complex, multiorgan phenomenon targeting skeletal muscle resulting from systemic metabolic imbalances. Multifocal in nature, It's ultimate outcome is significant muscle degradation and loss of adipose tissue exhibited as the "wasting syndrome" which is associated with significant functional decline. Currently, there are no approved biomarkers for screening nor therapeutic options to manage cancer cachexia. Furthermore, multiple psychosocial sequelae characterize the patient and family coping paradigm. Heightened education about the pathophysiology of cancer cachexia and awareness of intra-familial emotional distress can enhance oncology nurses' advocacy about, and attentiveness to, this common manifestation of advanced cancer.
Collapse
|
15
|
Frey MK, Bergler-Klein J. Echocardiographic evaluation of patients undergoing cancer therapy. Eur Heart J Cardiovasc Imaging 2021; 22:375-382. [PMID: 33393591 DOI: 10.1093/ehjci/jeaa341] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/02/2020] [Indexed: 12/28/2022] Open
Abstract
As advances in oncology therapies lead to significant improvement in life expectancy of many cancer entities, short-, and long-term cardiac side effects of oncology treatments gain increasing importance. In search of new screening modalities, echocardiography currently presents the best established and clinically easily feasible tool to detect cardiotoxicity in patients undergoing cancer therapy. This review focusses on the most commonly used oncology therapies and aims to give a practical approach to guide clinicians caring for this growing number of patients.
Collapse
Affiliation(s)
- Maria Klara Frey
- Department of Cardiology, Med. Univ. of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Jutta Bergler-Klein
- Department of Cardiology, Med. Univ. of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
16
|
Bergler-Klein J. Myocardial damage in anthracyclines and breast cancer: take a look at the bull's eye. Eur Heart J Cardiovasc Imaging 2021; 22:416-417. [PMID: 33569602 DOI: 10.1093/ehjci/jeab020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/22/2022] Open
Affiliation(s)
- Jutta Bergler-Klein
- Department of Cardiology, Univ. Clinic of Internal Med. II, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
17
|
Villa F, Bruno S, Costa A, Li M, Russo M, Cimino J, Altieri P, Ruggeri C, Gorgun C, De Biasio P, Paladini D, Coviello D, Quarto R, Ameri P, Ghigo A, Ravera S, Tasso R, Bollini S. The Human Fetal and Adult Stem Cell Secretome Can Exert Cardioprotective Paracrine Effects against Cardiotoxicity and Oxidative Stress from Cancer Treatment. Cancers (Basel) 2021; 13:cancers13153729. [PMID: 34359631 PMCID: PMC8345068 DOI: 10.3390/cancers13153729] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 12/28/2022] Open
Abstract
Simple Summary Anthracyclines, such as doxorubicin (Dox), are an important class of chemotherapeutic drugs. However, their use is hampered by the risk of developing heart failure. The aim of this study was to assess and compare the cardioprotective effects exerted by a set of factors, collectively named secretomes, secreted by either adult or fetal human stem cells. Both secretome formulations were effective in counteracting Dox-induced apoptosis and mitochondrial impairment in cardiomyocytes and cardiac fibroblasts. In vivo experiments in a mouse model of Dox-induced cardiomyopathy (DIC) indicated that early administration of both secretomes during Dox treatment exerted beneficial long-term effects, preserving cardiac function and body mass. These findings suggest that the stem cell secretome could represent a feasible option for future paracrine cardioprotective therapy against Dox-related cardiotoxicity during cancer treatment. Abstract Cardiovascular side effects are major shortcomings of cancer treatments causing cardiotoxicity and late-onset cardiomyopathy. While doxorubicin (Dox) has been reported as an effective chemotherapy agent, unspecific impairment in cardiomyocyte mitochondria activity has been documented. We demonstrated that the human fetal amniotic fluid-stem cell (hAFS) secretome, namely the secreted paracrine factors within the hAFS-conditioned medium (hAFS-CM), exerts pro-survival effects on Dox-exposed cardiomyocytes. Here, we provide a detailed comparison of the cardioprotective potential of hAFS-CM over the secretome of mesenchymal stromal cells from adipose tissue (hMSC-CM). hAFS and hMSC were preconditioned under hypoxia to enrich their secretome. The cardioprotective effects of hAFS/hMSC-CM were evaluated on murine neonatal ventricular cardiomyocytes (mNVCM) and on their fibroblast counterpart (mNVFib), and their long-term paracrine effects were investigated in a mouse model of Dox-induced cardiomyopathy. Both secretomes significantly contributed to preserving mitochondrial metabolism within Dox-injured cardiac cells. hAFS-CM and hMSC-CM inhibited body weight loss, improved myocardial function, reduced lipid peroxidation and counteracted the impairment of mitochondrial complex I activity, oxygen consumption, and ATP synthesis induced by Dox. The hAFS and hMSC secretomes can be exploited for inhibiting cardiotoxic detrimental side effects of Dox during cancer therapy, thus ensuring cardioprotection via combinatorial paracrine therapy in association with standard oncological treatments.
Collapse
Affiliation(s)
- Federico Villa
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.V.); (C.G.); (R.Q.)
| | - Silvia Bruno
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.B.); (A.C.); (S.R.)
| | - Ambra Costa
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.B.); (A.C.); (S.R.)
| | - Mingchuan Li
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (M.L.); (M.R.); (J.C.); (A.G.)
| | - Michele Russo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (M.L.); (M.R.); (J.C.); (A.G.)
| | - James Cimino
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (M.L.); (M.R.); (J.C.); (A.G.)
| | - Paola Altieri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine (DIMI), University of Genova, 16132 Genova, Italy; (P.A.); (C.R.); (P.A.)
| | - Clarissa Ruggeri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine (DIMI), University of Genova, 16132 Genova, Italy; (P.A.); (C.R.); (P.A.)
| | - Cansu Gorgun
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.V.); (C.G.); (R.Q.)
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.B.); (A.C.); (S.R.)
| | - Pierangela De Biasio
- Unit of Prenatal Diagnosis and Perinatal Medicine, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy;
| | - Dario Paladini
- Fetal Medicine and Surgery Unit, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Domenico Coviello
- Human Genetics Laboratory, IRCCS Istituto Giannina Gaslini, 16147 Genova, Italy;
| | - Rodolfo Quarto
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.V.); (C.G.); (R.Q.)
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.B.); (A.C.); (S.R.)
| | - Pietro Ameri
- Laboratory of Cardiovascular Biology, Department of Internal Medicine (DIMI), University of Genova, 16132 Genova, Italy; (P.A.); (C.R.); (P.A.)
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy
| | - Alessandra Ghigo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, 10126 Torino, Italy; (M.L.); (M.R.); (J.C.); (A.G.)
| | - Silvia Ravera
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.B.); (A.C.); (S.R.)
| | - Roberta Tasso
- Cellular Oncology Unit, IRCCS Ospedale Policlinico San Martino, 16132 Genova, Italy; (F.V.); (C.G.); (R.Q.)
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.B.); (A.C.); (S.R.)
- Correspondence: (R.T.); (S.B.); Tel.: +39-010-555-8394 (R.T.); +39-010-555-8257 (S.B.)
| | - Sveva Bollini
- Department of Experimental Medicine (DIMES), University of Genova, 16132 Genova, Italy; (S.B.); (A.C.); (S.R.)
- Correspondence: (R.T.); (S.B.); Tel.: +39-010-555-8394 (R.T.); +39-010-555-8257 (S.B.)
| |
Collapse
|
18
|
Cardiovascular toxicity of angiogenesis inhibitors and immune checkpoint inhibitors: synergistic anti-tumour effects at the cost of increased cardiovascular risk? Clin Sci (Lond) 2021; 135:1649-1668. [PMID: 34283204 DOI: 10.1042/cs20200300] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022]
Abstract
In the past two decades, treatment outcomes for a wide range of malignancies have improved remarkably due to the development of novel anti-cancer therapies, including vascular endothelial growth factor inhibitors (VEGFIs) and immune checkpoint inhibitors (ICIs). Despite their unprecedented anti-tumour effects, it is becoming increasingly clear that both types of agents are associated with specific cardiovascular toxicity, including hypertension, congestive heart failure, myocarditis and acceleration of atherosclerosis. Currently, VEGFI and ICI combination therapy is recommended for the treatment of advanced renal cell carcinoma (RCC) and has shown promising treatment efficacy in other tumour types as well. Consequently, VEGFI and ICI combination therapy will most likely become an important therapeutic strategy for various malignancies. However, this combinatory approach is expected to be accompanied by a substantial increase in cardiovascular risk, as both types of agents could act synergistically to induce cardiovascular sequelae. Therefore, a comprehensive baseline assessment and adequate monitoring by specialised cardio-oncology teams is essential in case these agents are used in combination, particularly in high-risk patients. This review summarises the mechanisms of action and treatment indications for currently registered VEGFIs and ICIs, and discusses their main vascular and cardiac toxicity. Subsequently, we provide the biological rationales for the observed promising synergistic anti-tumour effects of combined VEGFI/ICI administration. Lastly, we speculate on the increased risk for cardiovascular toxicity in case these agents are used in combination and its implications and future directions for the clinical situation.
Collapse
|
19
|
Anker MS, Sanz AP, Zamorano JL, Mehra MR, Butler J, Riess H, Coats AJS, Anker SD. Advanced cancer is also a heart failure syndrome: a hypothesis. Eur J Heart Fail 2021; 23:140-144. [PMID: 33247608 DOI: 10.1002/ejhf.2071] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 11/11/2020] [Accepted: 11/25/2020] [Indexed: 12/15/2022] Open
Abstract
We present the hypothesis that advanced stage cancer is also a heart failure syndrome. It can develop independently of or in addition to cardiotoxic effects of anti-cancer therapies. This includes an increased risk of ventricular arrhythmias. We suggest the pathophysiologic link for these developments includes generalized muscle wasting (i.e. sarcopenia) due to tissue homeostasis changes leading to cardiac wasting associated cardiomyopathy. Cardiac wasting with thinning of the ventricular wall increases ventricular wall stress, even in the absence of ventricular dilatation. In addition, arrhythmias may be facilitated by cellular wasting processes affecting structure and function of electrical cells and conduction pathways. We submit that in some patients with advanced cancer (but not terminal cancer), heart failure therapy or defibrillators may be relevant treatment options. The key points in selecting patients for such therapies may be the predicted life expectancy, quality of life at intervention time, symptomatic burden, and consequences for further anti-cancer therapies. The cause of death in advanced cancer is difficult to ascertain and consensus on event definitions in cancer is not established yet. Clinical investigations on this are called for. Broader ethical considerations must be taken into account when aiming to target cardiovascular problems in cancer patients. We suggest that focused attention to evaluating cardiac wasting and arrhythmias in cancer will herald a further evolution in the rapidly expanding field of cardio-oncology.
Collapse
Affiliation(s)
- Markus S Anker
- Department of Cardiology & Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Charité-Universitätsmedizin Berlin (Campus CVK), Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin (Campus CBF), Berlin, Germany
| | | | | | - Mandeep R Mehra
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Javed Butler
- Division of Cardiology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hanno Riess
- Department for Hematology, Oncology and Tumor Immunology (Campus CCM), Charite, University Medicine, Berlin, Germany
| | | | - Stefan D Anker
- Department of Cardiology & Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Charité-Universitätsmedizin Berlin (Campus CVK), Berlin, Germany
| |
Collapse
|
20
|
Anker MS, Sanz AP, Zamorano JL, Mehra MR, Butler J, Riess H, Coats AJS, Anker SD. Advanced cancer is also a heart failure syndrome: a hypothesis. J Cachexia Sarcopenia Muscle 2021; 12:533-537. [PMID: 33734609 PMCID: PMC8200419 DOI: 10.1002/jcsm.12694] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
We present the hypothesis that advanced stage cancer is also a heart failure syndrome. It can develop independently of or in addition to cardiotoxic effects of anti-cancer therapies. This includes an increased risk of ventricular arrhythmias. We suggest the pathophysiologic link for these developments includes generalized muscle wasting (i.e. sarcopenia) due to tissue homeostasis changes leading to cardiac wasting associated cardiomyopathy. Cardiac wasting with thinning of the ventricular wall increases ventricular wall stress, even in the absence of ventricular dilatation. In addition, arrhythmias may be facilitated by cellular wasting processes affecting structure and function of electrical cells and conduction pathways. We submit that in some patients with advanced cancer (but not terminal cancer), heart failure therapy or defibrillators may be relevant treatment options. The key points in selecting patients for such therapies may be the predicted life expectancy, quality of life at intervention time, symptomatic burden, and consequences for further anti-cancer therapies. The cause of death in advanced cancer is difficult to ascertain and consensus on event definitions in cancer is not established yet. Clinical investigations on this are called for. Broader ethical considerations must be taken into account when aiming to target cardiovascular problems in cancer patients. We suggest that focused attention to evaluating cardiac wasting and arrhythmias in cancer will herald a further evolution in the rapidly expanding field of cardio-oncology.
Collapse
Affiliation(s)
- Markus S Anker
- Department of Cardiology & Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Charité-Universitätsmedizin Berlin (Campus CVK), Berlin, Germany.,Department of Cardiology, Charité Universitätsmedizin Berlin (Campus CBF), Berlin, Germany
| | | | | | - Mandeep R Mehra
- Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Javed Butler
- Division of Cardiology, Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hanno Riess
- Department for Hematology, Oncology and Tumor Immunology (Campus CCM), Charite, University Medicine, Berlin, Germany
| | | | - Stefan D Anker
- Department of Cardiology & Berlin Institute of Health Center for Regenerative Therapies (BCRT), German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Charité-Universitätsmedizin Berlin (Campus CVK), Berlin, Germany
| |
Collapse
|
21
|
Galán-Arriola C, Villena-Gutiérrez R, Higuero-Verdejo MI, Díaz-Rengifo IA, Pizarro G, López GJ, de Molina-Iracheta A, Pérez-Martínez C, García RD, González-Calle D, Lobo M, Sánchez PL, Oliver E, Córdoba R, Fuster V, Sánchez-González J, Ibanez B. Remote ischaemic preconditioning ameliorates anthracycline-induced cardiotoxicity and preserves mitochondrial integrity. Cardiovasc Res 2021; 117:1132-1143. [PMID: 32597960 PMCID: PMC7983009 DOI: 10.1093/cvr/cvaa181] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 06/02/2020] [Accepted: 06/19/2020] [Indexed: 12/12/2022] Open
Abstract
AIMS Anthracycline-induced cardiotoxicity (AIC) is a serious adverse effect among cancer patients. A central mechanism of AIC is irreversible mitochondrial damage. Despite major efforts, there are currently no effective therapies able to prevent AIC. METHODS AND RESULTS Forty Large-White pigs were included. In Study 1, 20 pigs were randomized 1:1 to remote ischaemic preconditioning (RIPC, 3 cycles of 5 min leg ischaemia followed by 5 min reperfusion) or no pretreatment. RIPC was performed immediately before each intracoronary doxorubicin injections (0.45 mg/kg) given at Weeks 0, 2, 4, 6, and 8. A group of 10 pigs with no exposure to doxorubicin served as healthy controls. Pigs underwent serial cardiac magnetic resonance (CMR) exams at baseline and at Weeks 6, 8, 12, and 16, being sacrifice after that. In Study 2, 10 new pigs received 3 doxorubicin injections (with/out preceding RIPC) and were sacrificed at week 6. In Study 1, left ventricular ejection fraction (LVEF) depression was blunted animals receiving RIPC before doxorubicin (RIPC-Doxo), which had a significantly higher LVEF at Week 16 than doxorubicin treated pigs that received no pretreatment (Untreated-Doxo) (41.5 ± 9.1% vs. 32.5 ± 8.7%, P = 0.04). It was mainly due to conserved regional contractile function. In Study 2, transmission electron microscopy (TEM) at Week 6 showed fragmented mitochondria with severe morphological abnormalities in Untreated-Doxo pigs, together with upregulation of fission and autophagy proteins. At the end of the 16-week Study 1 protocol, TEM revealed overt mitochondrial fragmentation with structural fragmentation in Untreated-Doxo pigs, whereas interstitial fibrosis was less severe in RIPC+Doxo pigs. CONCLUSION In a translatable large-animal model of AIC, RIPC applied immediately before each doxorubicin injection resulted in preserved cardiac contractility with significantly higher long-term LVEF and less cardiac fibrosis. RIPC prevented mitochondrial fragmentation and dysregulated autophagy from AIC early stages. RIPC is a promising intervention for testing in clinical trials in AIC.
Collapse
Affiliation(s)
- Carlos Galán-Arriola
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Rocio Villena-Gutiérrez
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - María I Higuero-Verdejo
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
| | - Iván A Díaz-Rengifo
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
| | - Gonzalo Pizarro
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Complejo Hospitalario Ruber Juan Bravo, Madrid, Spain
| | - Gonzalo J López
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
| | - Antonio de Molina-Iracheta
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
| | | | - Rodrigo D García
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
| | - David González-Calle
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Department of Cardiology, Hospital Universitario Salamanca-IBSAL, Salamanca, Spain
| | - Manuel Lobo
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Department of Cardiology, IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Pedro L Sánchez
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Department of Cardiology, Hospital Universitario Salamanca-IBSAL, Salamanca, Spain
| | - Eduardo Oliver
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
| | - Raúl Córdoba
- Department of Cardiology, IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| | - Valentin Fuster
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- The Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | | | - Borja Ibanez
- Translational Laboratory for Cardiovascular Imaging and Therapy, Centro Nacional de Investigaciones Cardiovasculares (CNIC), c/Melchor Fernandez Almagro, 3. 28029 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Spain
- Department of Cardiology, IIS-Fundación Jiménez Díaz Hospital, Madrid, Spain
| |
Collapse
|
22
|
Mitochondrial Sirtuins and Doxorubicin-induced Cardiotoxicity. Cardiovasc Toxicol 2021; 21:179-191. [PMID: 33438065 DOI: 10.1007/s12012-020-09626-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Accepted: 12/11/2020] [Indexed: 12/20/2022]
Abstract
Doxorubicin (DOX) is the most effective and extensively used treatment for many tumors. However, its clinical use is hampered by its cardiotoxicity. DOX-induced mitochondrial dysfunction, which causes reactive oxygen species (ROS) generation, cardiomyocyte death, bioenergetic failure, and decreased cardiac function, is a very important mechanism of cardiotoxicity. These cellular processes are all linked by mitochondrial sirtuins (SIRT3-SIRT4). Mitochondrial sirtuins preserve mitochondrial function by increasing mitochondrial metabolism, inhibiting ROS generation by activating the antioxidant enzyme manganese-dependent superoxide dismutase (MnSOD), decreasing apoptosis by activating the forkhead homeobox type O (FOXO) and P53 pathways, and increasing autophagy through AMP-activated protein kinase (AMPK)/mTOR signaling. Thus, sirtuins function at the control point of many mechanisms involved in DOX-induced cardiotoxicity. In this review, we focus on the role of mitochondrial sirtuins in mitochondrial biology and DOX-induced cardiotoxicity. A further aim is to highlight other mitochondrial processes, such as autophagy (mitophagy) and mitochondrial quality control (MQC), for which the effect of mitochondrial sirtuins on cardiotoxicity is unknown.
Collapse
|
23
|
Jin K, Brennan PM, Poon MTC, Sudlow CLM, Figueroa JD. Raised cardiovascular disease mortality after central nervous system tumor diagnosis: analysis of 171,926 patients from UK and USA. Neurooncol Adv 2021; 3:vdab136. [PMID: 34647025 PMCID: PMC8500688 DOI: 10.1093/noajnl/vdab136] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Patients with central nervous system (CNS) tumors may be at risk of dying from cardiovascular disease (CVD). We examined CVD mortality risk in patients with different histological subtypes of CNS tumors. METHODS We analyzed UK(Wales)-based Secure Anonymized Information Linkage (SAIL) for 8743 CNS tumors patients diagnosed in 2000-2015, and US-based National Cancer Institute's Surveillance, Epidemiology, and End Results (SEER) for 163,183 patients in 2005-2015. We calculated age-, sex-, and calendar-year-adjusted standardized mortality ratios (SMRs) for CVD comparing CNS tumor patients to Wales and US residents. We used Cox regression models to examine factors associated with CVD mortality among CNS tumor patients. RESULTS CVD was the second leading cause of death for CNS tumor patients in SAIL (UK) and SEER (US). Patients with CNS tumors had higher CVD mortality than the general population (SAIL SMR = 2.64, 95% CI = 2.39-2.90, SEER SMR = 1.38, 95% CI = 1.35-1.42). Malignant CNS tumor patients had over 2-fold higher mortality risk in US and UK cohorts. SMRs for nonmalignant tumors were almost 2-fold higher in SAIL than in SEER. CVD mortality risk particularly cerebrovascular disease was substantially greater in patients diagnosed at age younger than 50 years, and within the first year after their cancer diagnosis (SAIL SMR = 2.98, 95% CI = 2.39-3.66, SEER SMR = 2.14, 95% CI = 2.03-2.25). Age, sex, race/ethnicity in USA, deprivation in UK and no surgery were associated with CVD mortality. CONCLUSIONS Patients with CNS tumors had higher risk for CVD mortality, particularly from cerebrovascular disease compared to the general population, supporting further research to improve mortality outcomes.
Collapse
Affiliation(s)
- Kai Jin
- Usher Institute, University of Edinburgh, Edinburgh, UK
- Brain Tumour Centre of Excellence, Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
| | - Paul M Brennan
- Brain Tumour Centre of Excellence, Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
- Translational Neurosurgery, Centre for Clinical Brain Sciences, University of Edinburgh, Edinburgh, UK
| | - Michael T C Poon
- Usher Institute, University of Edinburgh, Edinburgh, UK
- Brain Tumour Centre of Excellence, Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
| | - Cathie L M Sudlow
- Usher Institute, University of Edinburgh, Edinburgh, UK
- Brain Tumour Centre of Excellence, Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
| | - Jonine D Figueroa
- Usher Institute, University of Edinburgh, Edinburgh, UK
- Brain Tumour Centre of Excellence, Cancer Research UK Edinburgh Centre, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
24
|
Hajjar LA, Costa IBSDSD, Lopes MACQ, Hoff PMG, Diz MDPE, Fonseca SMR, Bittar CS, Rehder MHHDS, Rizk SI, Almeida DR, Fernandes GDS, Beck-da-Silva L, Campos CAHDM, Montera MW, Alves SMM, Fukushima JT, Santos MVCD, Negrão CE, Silva TLFD, Ferreira SMA, Malachias MVB, Moreira MDCV, Valente Neto MMR, Fonseca VCQ, Soeiro MCFDA, Alves JBS, Silva CMPDC, Sbano J, Pavanello R, Pinto IMF, Simão AF, Dracoulakis MDA, Hoff AO, Assunção BMBL, Novis Y, Testa L, Alencar Filho ACD, Cruz CBBV, Pereira J, Garcia DR, Nomura CH, Rochitte CE, Macedo AVS, Marcatti PTF, Mathias Junior W, Wiermann EG, Val RD, Freitas H, Coutinho A, Mathias CMDC, Vieira FMDAC, Sasse AD, Rocha V, Ramires JAF, Kalil Filho R. Brazilian Cardio-oncology Guideline - 2020. Arq Bras Cardiol 2020; 115:1006-1043. [PMID: 33295473 PMCID: PMC8452206 DOI: 10.36660/abc.20201006] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Ludhmila Abrahão Hajjar
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | | | - Paulo Marcelo Gehm Hoff
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Instituto D'or Pesquisa e Ensino, Rio de Janeiro, RJ - Brasil
| | - Maria Del Pilar Estevez Diz
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Silvia Moulin Ribeiro Fonseca
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Hospital Sírio Libanês, São Paulo, SP - Brasil
| | - Cristina Salvadori Bittar
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Hospital Sírio Libanês, São Paulo, SP - Brasil
| | - Marília Harumi Higuchi Dos Santos Rehder
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Hospital Sírio Libanês, São Paulo, SP - Brasil
| | - Stephanie Itala Rizk
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | | | - Luís Beck-da-Silva
- Hospital de Clínicas de Porto Alegre, Porto Alegre, RS - Brasil.,Universidade Federal do Rio Grande do Sul, Porto Alegre, RS - Brasil
| | | | | | | | - Júlia Tizue Fukushima
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Maria Verônica Câmara Dos Santos
- Sociedade Brasileira de Oncologia Pediátrica (SOBOPE), São Paulo, SP - Brasil.,Departamento de Cardiopatias Congênitas e Cardiologia Pediátrica (DCC/CP) da Sociedade Brasileira de Cardiologia (SBC), Rio de Janeiro, RJ - Brasil
| | - Carlos Eduardo Negrão
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | - Silvia Moreira Ayub Ferreira
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | | | | | | | | | - Juliana Barbosa Sobral Alves
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | - João Sbano
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Ricardo Pavanello
- Hospital do Coração da Associação do Sanatório Sírio - HCor, São Paulo, SP - Brasil
| | | | | | | | - Ana Oliveira Hoff
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | - Yana Novis
- Hospital Sírio Libanês, São Paulo, SP - Brasil
| | - Laura Testa
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | - Cecília Beatriz Bittencourt Viana Cruz
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Juliana Pereira
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Diego Ribeiro Garcia
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Cesar Higa Nomura
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Carlos Eduardo Rochitte
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Hospital do Coração (HCOR), São Paulo, SP - Brasil
| | | | | | - Wilson Mathias Junior
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | - Renata do Val
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | | | | | | | | | - André Deeke Sasse
- Grupo SOnhe, Campinas, SP - Brasil.,Universidade Estadual de Campinas (Unicamp), Campinas, SP - Brasil
| | - Vanderson Rocha
- Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - José Antônio Franchini Ramires
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| | - Roberto Kalil Filho
- Instituto do Coração do Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil.,Instituto do Câncer Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP - Brasil
| |
Collapse
|
25
|
Tomasoni D, Adamo M, Anker MS, von Haehling S, Coats AJS, Metra M. Heart failure in the last year: progress and perspective. ESC Heart Fail 2020; 7:3505-3530. [PMID: 33277825 PMCID: PMC7754751 DOI: 10.1002/ehf2.13124] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 11/11/2020] [Indexed: 12/11/2022] Open
Abstract
Research about heart failure (HF) has made major progress in the last years. We give here an update on the most recent findings. Landmark trials have established new treatments for HF with reduced ejection fraction. Sacubitril/valsartan was superior to enalapril in PARADIGM-HF trial, and its initiation during hospitalization for acute HF or early after discharge can now be considered. More recently, new therapeutic pathways have been developed. In the DAPA-HF and EMPEROR-Reduced trials, dapagliflozin and empagliflozin reduced the risk of the primary composite endpoint, compared with placebo [hazard ratio (HR) 0.74; 95% confidence interval (CI) 0.65-0.85; P < 0.001 and HR 0.75; 95% CI 0.65-0.86; P < 0.001, respectively]. Second, vericiguat, an oral soluble guanylate cyclase stimulator, reduced the composite endpoint of cardiovascular death or HF hospitalization vs. placebo (HR 0.90; 95% CI 0.82-0.98; P = 0.02). On the other hand, both the diagnosis and treatment of HF with preserved ejection fraction, as well as management of advanced HF and acute HF, remain challenging. A better phenotyping of patients with HF would be helpful for prognostic stratification and treatment selection. Further aspects, such as the use of devices, treatment of arrhythmias, and percutaneous treatment of valvular heart disease in patients with HF, are also discussed and reviewed in this article.
Collapse
Affiliation(s)
- Daniela Tomasoni
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
- Cardiology and Cardiac Catheterization Laboratory, Cardio‐thoracic DepartmentCivil HospitalsBresciaItaly
| | - Marianna Adamo
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
- Cardiology and Cardiac Catheterization Laboratory, Cardio‐thoracic DepartmentCivil HospitalsBresciaItaly
| | - Markus S. Anker
- Division of Cardiology and Metabolism, Department of Cardiology (CVK)Charité–University Medicine BerlinBerlinGermany
- Berlin Institute of Health Center for Regenerative Therapies (BCRT)BerlinGermany
- German Centre for Cardiovascular Research (DZHK), partner site BerlinBerlinGermany
- Department of Cardiology (CBF)Charité–University Medicine BerlinBerlinGermany
| | - Stephan von Haehling
- Department of Cardiology and PneumologyUniversity of Göttingen Medical CenterGöttingenGermany
- German Centre for Cardiovascular Research (DZHK), partner site GöttingenGöttingenGermany
| | - Andrew J. S. Coats
- Centre for Clinical and Basic Research, Department of Medical SciencesIRCCS San Raffaele PisanaRomeItaly
| | - Marco Metra
- Department of Medical and Surgical Specialties, Radiological Sciences, and Public HealthUniversity of BresciaBresciaItaly
- Cardiology and Cardiac Catheterization Laboratory, Cardio‐thoracic DepartmentCivil HospitalsBresciaItaly
| |
Collapse
|
26
|
[Tumor effects on the heart and circulation]. Internist (Berl) 2020; 61:1120-1124. [PMID: 33052455 DOI: 10.1007/s00108-020-00887-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
As a result of the continuous development of modern cancer treatment, more cancer patients can be cured every year. However, since many patients experience cardiovascular problems before, during and after their cancer treatment, cardio-oncology is becoming increasingly important. Numerous therapies can cause cardiotoxicity, such as chemotherapy, immunotherapy, antibody therapy and radiotherapy. If these remain undetected, the patient may develop, e.g. heart failure or severe heart valve damage. The broad spectrum of cardiovascular comorbidities has become an immense challenge for cardiologists and oncologists. Cardio-oncology also deals with the effects that cancer has on the cardiovascular system. New research indicates that the tumor itself also has direct negative effects on the heart, mediated by messenger substances. Therefore, it is important to understand which cancer patients are at increased cardiovascular risk, thereby enabling the development of new therapeutic approaches in the long term to maintain mobility and improve patient prognosis.
Collapse
|
27
|
Anthracycline-Related Heart Failure: Certain Knowledge and Open Questions : Where Do we Stand with Chemotherapyinduced Cardiotoxicity? Curr Heart Fail Rep 2020; 17:357-364. [PMID: 32964378 PMCID: PMC7683464 DOI: 10.1007/s11897-020-00489-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 12/24/2022]
Abstract
In the last decade, cardio-oncology has become a discipline on its own, with tremendous research going on to unravel the mechanisms underpinning different manifestations of cardiotoxicity caused by anticancer drugs. Although this domain is much broader than the effect of chemotherapy alone, a lot of questions about anthracycline-induced cardiotoxicity remain unknown. In this invited review, we provide insights in molecular mechanisms behind anthracycline-induced cardiotoxicity and put it in a clinical framework emphasizing the need for patients to understand, detect, and treat this detrimental condition.
Collapse
|
28
|
Beasley GS, Towbin JA. Acquired and modifiable cardiovascular risk factors in patients treated for cancer. J Thromb Thrombolysis 2020; 51:846-853. [PMID: 32918669 DOI: 10.1007/s11239-020-02273-7] [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] [Accepted: 09/02/2020] [Indexed: 11/28/2022]
Abstract
Cardiac mortality is the leading cause of death secondary to malignancy in survivors of cancer. The field of cardio-oncology is dedicated to identifying and, if possible, modifying risk factors that contribute to significant cardiac morbidity and mortality. Many risk factors for the development of cancer-related cardiotoxicity overlap with risk factors in cardiovascular disease such as hypertension, obesity, dyslipidemia, and diabetes among others. These risk factors are usually modifiable while others such as genetics, type of malignancy, and need for chemotherapy are less modifiable. This article summarizes acquired and modifiable risk factors in both pediatric and adult patients treated for cancer.
Collapse
Affiliation(s)
- Gary S Beasley
- Department of Pediatrics, Division of Cardiology Le Bonheur Children's Hospital and The Heart Institute, University of Tennessee Health Science Center, College of Medicine, 49 N. Dunlap Street, 3rd Floor, Faculty Office Building, Memphis, TN, 38105, USA. .,Cardio-Hema-Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| | - Jeffrey A Towbin
- Department of Pediatrics, Division of Cardiology Le Bonheur Children's Hospital and The Heart Institute, University of Tennessee Health Science Center, College of Medicine, 49 N. Dunlap Street, 3rd Floor, Faculty Office Building, Memphis, TN, 38105, USA.,Cardio-Hema-Oncology, St. Jude Children's Research Hospital, Memphis, TN, USA
| |
Collapse
|
29
|
Anker MS, Hadzibegovic S, Lena A, Belenkov Y, Bergler‐Klein J, de Boer RA, Farmakis D, von Haehling S, Iakobishvili Z, Maack C, Pudil R, Skouri H, Cohen‐Solal A, Tocchetti CG, Coats AJ, Seferović PM, Lyon AR. Recent advances in cardio-oncology: a report from the 'Heart Failure Association 2019 and World Congress on Acute Heart Failure 2019'. ESC Heart Fail 2019; 6:1140-1148. [PMID: 31884717 PMCID: PMC6989292 DOI: 10.1002/ehf2.12551] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Indexed: 02/06/2023] Open
Abstract
While anti-cancer therapies, including chemotherapy, immunotherapy, radiotherapy, and targeted therapy, are constantly advancing, cardiovascular toxicity has become a major challenge for cardiologists and oncologists. This has led to an increasing demand of cardio-oncology units in Europe and a growing interest of clinicians and researchers. The Heart Failure 2019 meeting of the Heart Failure Association of the European Society of Cardiology in Athens has therefore created a scientific programme that included four dedicated sessions on the topic along with several additional lectures. The major points that were discussed at the congress included the implementation and delivery of a cardio-oncology service, the collaboration among cardio-oncology experts, and the risk stratification, prevention, and early recognition of cardiotoxicity. Furthermore, sessions addressed the numerous different anti-cancer therapies associated with cardiotoxic effects and provided guidance on how to treat cancer patients who develop cardiovascular disease before, during, and after treatment.
Collapse
Affiliation(s)
- Markus S. Anker
- Division of Cardiology and Metabolism, Department of CardiologyCharité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research)partner site Berlin and Department of Cardiology, Charité Campus Benjamin FranklinBerlinGermany
| | - Sara Hadzibegovic
- Division of Cardiology and Metabolism, Department of CardiologyCharité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research)partner site Berlin and Department of Cardiology, Charité Campus Benjamin FranklinBerlinGermany
| | - Alessia Lena
- Division of Cardiology and Metabolism, Department of CardiologyCharité and Berlin Institute of Health Center for Regenerative Therapies (BCRT) and DZHK (German Centre for Cardiovascular Research)partner site Berlin and Department of Cardiology, Charité Campus Benjamin FranklinBerlinGermany
| | | | | | - Rudolf A. de Boer
- Department of CardiologyUniversity of GroningenUniversity Medical Center Groningen, GroningenThe Netherlands
| | - Dimitrios Farmakis
- University of Cyprus Medical SchoolNicosiaCyprus
- Department of Cardiology, Cardio‐Oncology Clinic, Heart Failure UnitAthens University Hospital ‘Attikon’, National and Kapodistrian University of AthensAthensGreece
| | - Stephan von Haehling
- Department of Cardiology and Pneumology, Heart Center Göttingen, German Center for Cardiovascular Medicine (DZHK)University of Göttingen Medical Center, Georg‐August‐UniversityGöttingenGermany
| | - Zaza Iakobishvili
- Department of Community Cardiology, Clalit Health Fund, and Sackler Faculty of MedicineTel Aviv UniversityTel AvivIsrael
| | - Christoph Maack
- Comprehensive Heart Failure Center (CHFC)University Clinic WürzburgWürzburgGermany
| | - Radek Pudil
- 1st Department of Medicine–Cardioangiology, Faculty of MedicineUniversity HospitalHradec KrálovéCzech Republic
| | - Hadi Skouri
- Department of Internal MedicineAmerican University of Beirut Medical CenterBeirutLebanon
| | - Alain Cohen‐Solal
- Department of Cardiology, Lariboisière Hospital and U942 INSERM, BIOCANVAS (Biomarqueurs Cardiovasculaires)Paris UniversityParisFrance
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences and Interdepartmental Center for Clinical and Translational Sciences (CIRCET)Federico II UniversityNaplesItaly
| | | | - Petar M. Seferović
- Faculty of Medicine and Heart Failure CenterBelgrade University Medical Center, University of BelgradeBelgradeSerbia
| | | | | |
Collapse
|