201
|
Experimental determination of diagnostic window of cardiac troponins in the development of chronic anthracycline cardiotoxicity and estimation of its predictive value. Int J Cardiol 2015; 201:358-67. [PMID: 26310978 DOI: 10.1016/j.ijcard.2015.07.103] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Revised: 06/17/2015] [Accepted: 07/30/2015] [Indexed: 01/23/2023]
Abstract
BACKGROUND Cardiac troponins (cTns) seem to be more sensitive for the detection of anthracycline cardiotoxicity than the currently recommended method of monitoring LV systolic function. However, the optimal timing of blood sampling remains unknown. Hence, the aims of the present study were to determine the precise diagnostic window for cTns during the development of chronic anthracycline cardiotoxicity and to evaluate their predictive value. METHODS Cardiotoxicity was induced in rabbits with daunorubicin (3mg/kg, weekly, for 8 weeks). Blood samples were collected 2-168 h after the 1st, 5th and 8th drug administrations, and concentrations of cTns were determined using highly sensitive assays: hs cTnT (Roche) and hs cTnI (Abbott). RESULTS The plasma levels of cTns progressively increased with the rising number of chemotherapy cycles. While only a mild non-significant increase in both cTn levels occurred after the first daunorubicin dose, a significant rise was observed after the 5th and 8th administrations. Two hours after these administrations, a significant increase occurred with a peak between 4-6h and a decline until 24h. Discrete cTn release continued even after cessation of the therapy. While greater variability of cTn levels was observed around the peak concentrations, the values did not correspond well with the severity of LV systolic dysfunction. Unlike AMI in cardiotoxicity, cTn elevations may be better associated with cumulative dose and concentrations at steady state than cmax. CONCLUSIONS To the best of our knowledge, this is the first study to precisely describe the diagnostic window and predictive value of cTns in anthracycline cardiotoxicity.
Collapse
|
202
|
Putt M, Hahn VS, Januzzi JL, Sawaya H, Sebag IA, Plana JC, Picard MH, Carver JR, Halpern EF, Kuter I, Passeri J, Cohen V, Banchs J, Martin RP, Gerszten RE, Scherrer-Crosbie M, Ky B. Longitudinal Changes in Multiple Biomarkers Are Associated with Cardiotoxicity in Breast Cancer Patients Treated with Doxorubicin, Taxanes, and Trastuzumab. Clin Chem 2015. [PMID: 26220066 DOI: 10.1373/clinchem.2015.241232] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
BACKGROUND Biomarkers may play an important role in identifying patients at risk for cancer therapy cardiotoxicity. Our objectives were to define the patterns of change in biomarkers with cancer therapy and their associations with cardiotoxicity. METHODS In a multicenter cohort of 78 breast cancer patients undergoing doxorubicin and trastuzumab therapy, 8 biomarkers were evaluated at baseline and every 3 months over a maximum follow-up of 15 months. These biomarkers, hypothesized to be mechanistically relevant to cardiotoxicity, included high-sensitivity cardiac troponin I (hs-cTnI), high-sensitivity C-reactive protein (hsCRP), N-terminal pro-B-type natriuretic peptide (NT-proBNP), growth differentiation factor 15 (GDF-15), myeloperoxidase (MPO), placental growth factor (PlGF), soluble fms-like tyrosine kinase receptor-1 (sFlt-1), and galectin 3 (gal-3). We determined if biomarker increases were associated with cardiotoxicity at the same visit and the subsequent visit over the entire course of therapy. Cardiotoxicity was defined by the Cardiac Review and Evaluation Criteria; alternative definitions were also considered. RESULTS Across the entire cohort, all biomarkers except NT-proBNP and gal-3 demonstrated increases by 3 months; these increases persisted for GDF-15, PlGF, and hs-cTnI at 15 months. Increases in MPO, PlGF, and GDF-15 were associated with cardiotoxicity at the same visit [MPO hazard ratio 1.38 (95% CI 1.10-1.71), P = 0.02; PlGF 3.78 (1.30-11.0), P = 0.047; GDF-15 1.71 (1.15-2.55), P = 0.01] and the subsequent visit. MPO was robust to alternative outcome definitions. CONCLUSIONS Increases in MPO are associated with cardiotoxicity over the entire course of doxorubicin and trastuzumab therapy. Assessment with PlGF and GDF-15 may also be of value. These findings motivate validation studies in additional cohorts.
Collapse
Affiliation(s)
- Mary Putt
- University of Pennsylvania, Philadelphia, PA
| | | | - James L Januzzi
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Heloisa Sawaya
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Igal A Sebag
- Sir Mortimer B. Davis-Jewish General Hospital and McGill University, Montreal, CA
| | | | - Michael H Picard
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | - Elkan F Halpern
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Irene Kuter
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Jonathan Passeri
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | - Victor Cohen
- Sir Mortimer B. Davis-Jewish General Hospital and McGill University, Montreal, CA
| | | | | | - Robert E Gerszten
- Massachusetts General Hospital and Harvard Medical School, Boston, MA
| | | | - Bonnie Ky
- University of Pennsylvania, Philadelphia, PA;
| |
Collapse
|
203
|
Hess CN, Roe MT, Clare RM, Chiswell K, Kelly J, Tcheng JE, Hagstrom E, James SK, Khouri MG, Hirsch BR, Kong DF, Abernethy AP, Krucoff MW. Relationship Between Cancer and Cardiovascular Outcomes Following Percutaneous Coronary Intervention. J Am Heart Assoc 2015; 4:JAHA.115.001779. [PMID: 26150477 PMCID: PMC4608066 DOI: 10.1161/jaha.115.001779] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Background Cardiovascular disease and cancer increasingly coexist, yet relationships between cancer and long-term cardiovascular outcomes post–percutaneous coronary intervention (PCI) are not well studied. Methods and Results We examined stented PCI patients at Duke (1996–2010) using linked data from the Duke Information Systems for Cardiovascular Care and the Duke Tumor Registry (a cancer treatment registry). Our primary outcome was cardiovascular mortality. Secondary outcomes included composite cardiovascular mortality, myocardial infarction, or repeat revascularization and all-cause mortality. We used adjusted cause-specific hazard models to examine outcomes among cancer patients (cancer treatment pre-PCI) versus controls (no cancer treatment pre-PCI). Cardiovascular mortality was explored in a cancer subgroup with recent (within 1 year pre-PCI) cancer and in post-PCI cancer patients using post-PCI cancer as a time-dependent variable. Among 15 008 patients, 3.3% (n=496) were cancer patients. Observed rates of 14-year cardiovascular mortality (31.4% versus 27.7%, P=0.31) and composite cardiovascular death, myocardial infarction, or revascularization (51.1% versus 55.8%, P=0.37) were similar for cancer versus control groups; all-cause mortality rates were higher (79.7% versus 49.3%, P<0.01). Adjusted risk of cardiovascular mortality was similar for cancer patients versus controls (hazard ratio 0.95; 95% CI 0.76 to 1.20) and for patients with versus without recent cancer (hazard ratio 1.46; 95% CI 0.92 to 2.33). Post-PCI cancer, present in 4.3% (n=647) of patients, was associated with cardiovascular mortality (adjusted hazard ratio 1.51; 95% CI 1.11 to 2.03). Conclusions Cancer history was present in a minority of PCI patients but was not associated with worse long-term cardiovascular outcomes. Further investigation into PCI outcomes in this population is warranted.
Collapse
Affiliation(s)
- Connie N Hess
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC (C.N.H., M.T.R., J.E.T., M.G.K., D.F.K., M.W.K.) Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.)
| | - Matthew T Roe
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC (C.N.H., M.T.R., J.E.T., M.G.K., D.F.K., M.W.K.) Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.)
| | - Robert M Clare
- Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.)
| | - Karen Chiswell
- Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.)
| | - Joseph Kelly
- Center for Learning Healthcare, Duke Clinical Research Institute, Durham, NC (J.K., A.P.A.)
| | - James E Tcheng
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC (C.N.H., M.T.R., J.E.T., M.G.K., D.F.K., M.W.K.) Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.)
| | - Emil Hagstrom
- Department of Medical Sciences, Cardiology, and Uppsala Clinical Research Center, Uppsala University, Sweden (E.H., S.K.J.)
| | - Stefan K James
- Department of Medical Sciences, Cardiology, and Uppsala Clinical Research Center, Uppsala University, Sweden (E.H., S.K.J.)
| | - Michel G Khouri
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC (C.N.H., M.T.R., J.E.T., M.G.K., D.F.K., M.W.K.)
| | - Bradford R Hirsch
- Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.) Duke Cancer Institute, Durham, NC (B.R.H., A.P.A.)
| | - David F Kong
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC (C.N.H., M.T.R., J.E.T., M.G.K., D.F.K., M.W.K.) Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.)
| | - Amy P Abernethy
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, NC (A.P.A.) Center for Learning Healthcare, Duke Clinical Research Institute, Durham, NC (J.K., A.P.A.) Duke Cancer Institute, Durham, NC (B.R.H., A.P.A.)
| | - Mitchell W Krucoff
- Division of Cardiology, Department of Medicine, Duke University Medical Center, Durham, NC (C.N.H., M.T.R., J.E.T., M.G.K., D.F.K., M.W.K.) Duke Clinical Research Institute, Durham, NC (C.N.H., M.T.R., R.M.C., K.C., J.E.T., B.R.H., D.F.K., M.W.K.)
| |
Collapse
|
204
|
Montoro N, López-Sendón JL. Heart failure years after cancer treatment. Future Cardiol 2015; 11:433-40. [DOI: 10.2217/fca.15.36] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Progress in cancer treatment has significantly improved survival of patients with cancer. However, the incidence of cardiovascular diseases such as left ventricular dysfunction (LVD) and chronic heart failure (HF) is increasing due to the long-term toxic effects of chemotherapy and radiotherapy. Cardio-oncology teams are necessary to ensure the implementation of primary prevention strategies and screening protocols for early recognition of LVD. Moreover, early administration of advanced treatment for HF is crucial to achieve left ventricular recovery. In this article we will focus on the prevalence of chronic HF among cancer survivors, the main risk factors of LVD and chronic HF, the prevention strategies and management based on the current evidence and, finally, the future perspectives in this field.
Collapse
Affiliation(s)
- Nieves Montoro
- Cardiology Department, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain
| | - Jose Luis López-Sendón
- Cardiology Department, La Paz University Hospital, Paseo de la Castellana 261, 28046 Madrid, Spain
| |
Collapse
|
205
|
Piper SE, McDonagh TA. Chemotherapy-related Cardiomyopathy. Eur Cardiol 2015; 10:19-24. [PMID: 30310418 PMCID: PMC6159418 DOI: 10.15420/ecr.2015.10.01.19] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Accepted: 07/23/2015] [Indexed: 11/04/2022] Open
Abstract
Advances in chemotherapeutic agents have resulted in significantly improved cancer survival rates. Cardiac toxicity, however, has emerged as a leading cause of morbidity, both during and years after treatment. One of the most common manifestations of cardiotoxicity is that of heart failure and left ventricular systolic dysfunction. In this review, current opinions and guidelines in this field are discussed, with particular focus on the most common culprits, the anthracyclines, and the monoclonal antibody, trastuzumab.
Collapse
Affiliation(s)
- Susan E Piper
- King's College London, The James Black Centre, London, UK; Kings College Hospital NHS Foundation Trust, London, UK
| | - Theresa A McDonagh
- King's College London, The James Black Centre, London, UK; Kings College Hospital NHS Foundation Trust, London, UK
| |
Collapse
|
206
|
Clarke E, Lenihan D. Cardio-oncology: a new discipline in medicine to lead us into truly integrative care. Future Cardiol 2015; 11:359-61. [DOI: 10.2217/fca.15.55] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Affiliation(s)
- Ellen Clarke
- Future Science Group, Unitec House, 2 Albert Place, London, N3 1QB, UK
| | - Daniel Lenihan
- Vanderbilt University, 1215 21st Ave South, Suite 5209, Nashville, TN 37232, USA
| |
Collapse
|
207
|
Abstract
Improvements in therapies have significantly changed survival of cancer patients. However, the clinical history and oncologic treatment put cancer patients at higher risk for developing cardiovascular problems. Anthracyclines, but also the targeted therapy and angiogenesis inhibitors, are all treatments associated with cardiotoxicity. The most common adverse event is a reduction in left ventricular ejection fraction that may progress to overt heart failure. Recognition of a cardiac impairment during or after a potential cardiotoxic treatment requires a stringent assessment of clinical symptoms and signs of heart failure associated with an evaluation of the left ventricular ejection fraction, which, however, detects the damage already installed. Circulating cardiac biomarkers are promising in detecting cardiotoxicity and will likely change the approach for identifying patients at risk.
Collapse
Affiliation(s)
- Michela Salvatici
- Division of Laboratory Medicine, European Institute of Oncology, via Ripamonti, 435, 20141 Milan, Italy
| | - Maria T Sandri
- Division of Laboratory Medicine, European Institute of Oncology, via Ripamonti, 435, 20141 Milan, Italy
| |
Collapse
|
208
|
Caro Codón J, Rosillo Rodríguez SO, López Fernández T. Cardiotoxicity from the cardiologist's perspective. Future Cardiol 2015; 11:425-32. [DOI: 10.2217/fca.15.47] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Screening programs and contemporary multidisciplinary management of oncologic patients have resulted in lower mortality and improved outcomes. Nevertheless, treatment-related cardiac toxicity has been recognized as a major side effect, negatively affecting quality of life and prognosis in cancer survivors. Physicians involved in the care of these patients should be familiar with the effects of chemotherapy and radiotherapy on the heart, as well as the tests that may facilitate early diagnosis and prompt referral to cardiology units with expertise in the management of oncologic patients. This special report focuses on the mechanisms of cardiotoxicity and the techniques (including basic and advanced imaging and biomarkers) used in the cardiac evaluation of cancer patients.
Collapse
Affiliation(s)
- Juan Caro Codón
- Cardio Oncology Unit, La Paz University Hospital, IdiPaz Research Institute, Madrid, Spain
| | | | - Teresa López Fernández
- Cardio Oncology Unit, La Paz University Hospital, IdiPaz Research Institute, Madrid, Spain
| |
Collapse
|
209
|
Abstract
Advances in chemotherapeutic agents over the past two decades have resulted in significantly improved cancer survival rates. Cardiac toxicity, however, has emerged as a leading cause of morbidity, both during and years after treatment. One of the most common manifestations of cardiotoxicity is that of heart failure and left ventricular systolic dysfunction. Consequently, the field of cardio-oncology is a rapidly emerging field of sub-specialty, with growing research interests in all aspects of management. In this review, current opinions and guidelines in this field are discussed, with particular focus on the most common culprits, the anthracyclines and the monoclonal antibody, trastuzumab.
Collapse
Affiliation(s)
- Susan Piper
- Department of Cardiovascular Research, King's College London, The James Black Center, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| | - Theresa McDonagh
- Department of Cardiovascular Research, King's College London, The James Black Center, 125 Coldharbour Lane, London SE5 9NU, UK
- Department of Cardiology, King's College Hospital NHS Foundation Trust, Denmark Hill, London SE5 9RS, UK
| |
Collapse
|
210
|
A comparative study of folate receptor-targeted doxorubicin delivery systems: Dosing regimens and therapeutic index. J Control Release 2015; 208:106-20. [DOI: 10.1016/j.jconrel.2015.04.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 04/03/2015] [Accepted: 04/09/2015] [Indexed: 01/07/2023]
|
211
|
Cardinale D, Colombo A, Bacchiani G, Tedeschi I, Meroni CA, Veglia F, Civelli M, Lamantia G, Colombo N, Curigliano G, Fiorentini C, Cipolla CM. Early detection of anthracycline cardiotoxicity and improvement with heart failure therapy. Circulation 2015; 131:1981-8. [PMID: 25948538 DOI: 10.1161/circulationaha.114.013777] [Citation(s) in RCA: 1078] [Impact Index Per Article: 107.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 03/23/2015] [Indexed: 12/16/2022]
Abstract
BACKGROUND Three types of anthracycline-induced cardiotoxicities are currently recognized: acute, early-onset chronic, and late-onset chronic. However, data supporting this classification are lacking. We prospectively evaluated incidence, time of occurrence, clinical correlates, and response to heart failure therapy of cardiotoxicity. METHODS AND RESULTS We assessed left ventricular ejection fraction (LVEF), at baseline, every 3 months during chemotherapy and for the following year, every 6 months over the following 4 years, and yearly afterward in a heterogeneous cohort of 2625 patients receiving anthracycline-containing therapy. In case of cardiotoxicity (LVEF decrease >10 absolute points, and <50%), heart failure therapy was initiated. Recovery from cardiotoxicity was defined as partial (LVEF increase >5 absolute points and >50%) or full (LVEF increase to the baseline value). The median follow-up was 5.2 (quartile 1 to quartile 3, 2.6-8.0) years. The overall incidence of cardiotoxicity was 9% (n=226). The median time elapsed between the end of chemotherapy and cardiotoxicity development was 3.5 (quartile 1 to quartile 3, 3-6) months. In 98% of cases (n=221), cardiotoxicity occurred within the first year. Twenty-five (11%) patients had full recovery, and 160 (71%) patients had partial recovery. At multivariable analysis, end-chemotherapy LVEF (hazard ratio, 1.37; 95% confidence interval, 1.33-1.42 for each percent unit decrement) and cumulative doxorubicin dose (hazard ratio, 1.09; 95% confidence interval, 1.04-1.15 for each 50 mg/m(2) increment) were independent correlates of cardiotoxicity. CONCLUSIONS Most cardiotoxicity after anthracycline-containing therapy occurs within the first year and is associated with anthracycline dose and LVEF at the end of treatment. Early detection and prompt therapy of cardiotoxicity appear crucial for substantial recovery of cardiac function.
Collapse
Affiliation(s)
- Daniela Cardinale
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.).
| | - Alessandro Colombo
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Giulia Bacchiani
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Ines Tedeschi
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Carlo A Meroni
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Fabrizio Veglia
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Maurizio Civelli
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Giuseppina Lamantia
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Nicola Colombo
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Giuseppe Curigliano
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Cesare Fiorentini
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| | - Carlo M Cipolla
- From European Institute of Oncology, Milan, Italy (D.C., A.C., G.B., I.T., C.A.M., M.C., G.L., N.C., G.C., C.M.C.); and Centro Cardiologico Monzino (I.R.C.C.S.), Milan, Italy (F.V., C.F.)
| |
Collapse
|
212
|
Ghatalia P, Morgan CJ, Je Y, Nguyen PL, Trinh QD, Choueiri TK, Sonpavde G. Congestive heart failure with vascular endothelial growth factor receptor tyrosine kinase inhibitors. Crit Rev Oncol Hematol 2015; 94:228-37. [DOI: 10.1016/j.critrevonc.2014.12.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2014] [Revised: 10/11/2014] [Accepted: 12/11/2014] [Indexed: 11/16/2022] Open
|
213
|
Metra M. The EJHF last Editor's legacy: how can a high impact factor be built? ESC Heart Fail 2015; 2:50-57. [PMID: 28834654 PMCID: PMC6410532 DOI: 10.1002/ehf2.12032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 03/23/2015] [Indexed: 11/09/2022] Open
Abstract
The European Journal of Heart Failure (EJHF) has reached a high impact factor making it one of the most important cardiology journals. I discuss herein what could be the main causes of such high ranking. Publication of the European Society of Cardiology guidelines for the diagnosis and treatment of acute and chronic heart failure has had the most important role with a number of citations, which has been approximately 10 times that of the other most cited articles of the same year. Other position statements, reviews, design papers, and research articles about landmark topics have given major contributions. With respect to the different clinical presentations, articles about heart failure with preserved ejection fraction and about advanced heart failure have gained many citations. Epidemiology, biomarkers, medical treatment, and devices have attracted most of the interest. In conclusion, being able to look ahead and to publish what is going to become important remains a major challenge. That of EJHF has been a success story, to date, and learning from the past may help to build upon this achievement.
Collapse
Affiliation(s)
- Marco Metra
- Cardiology, Department of Medical and Surgical Specialties, Radiological Sciences and Public Health, University of Brescia, Brescia, Italy
| |
Collapse
|
214
|
De Angelis A, Piegari E, Cappetta D, Russo R, Esposito G, Ciuffreda LP, Ferraiolo FAV, Frati C, Fagnoni F, Berrino L, Quaini F, Rossi F, Urbanek K. SIRT1 activation rescues doxorubicin-induced loss of functional competence of human cardiac progenitor cells. Int J Cardiol 2015; 189:30-44. [PMID: 25889431 DOI: 10.1016/j.ijcard.2015.03.438] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 03/12/2015] [Accepted: 03/31/2015] [Indexed: 12/17/2022]
Abstract
BACKGROUND The search for compounds able to counteract chemotherapy-induced heart failure is extremely important at the age of global cancer epidemic. The role of SIRT1 in the maintenance of progenitor cell homeostasis may contribute to its cardioprotective effects. SIRT1 activators, by preserving progenitor cells, could have a clinical relevance for the prevention of doxorubicin (DOXO)-cardiotoxicity. METHODS To determine whether SIRT1 activator, resveratrol (RES), interferes with adverse effects of DOXO on cardiac progenitor cells (CPCs): 1) human CPCs (hCPCs) were exposed in vitro to DOXO or DOXO+RES and their regenerative potential was tested in vivo in an animal model of DOXO-induced heart failure; 2) the in vivo effects of DOXO+RES co-treatment on CPCs were studied in a rat model. RESULTS In contrast to healthy cells, DOXO-exposed hCPCs were ineffective in a model of anthracycline cardiomyopathy. The in vitro activation of SIRT1 decreased p53 acetylation, overcame suppression of the IGF-1/Akt pro-survival and anti-apoptotic signaling, enhanced oxidative stress defense and prevented senescence and growth arrest of hCPCs. Priming with RES counterbalanced the onset of dysfunctional phenotype in DOXO-exposed hCPCs, partly restoring their ability to repair the damage with improvement in cardiac function and animal survival. The in vivo co-treatment DOXO+RES prevented the anthracycline-induced alterations in CPCs, partly preserving cardiac function. CONCLUSION SIRT1 activation protects DOXO-exposed CPCs and re-establishes their proper function. Pharmacological intervention at the level of tissue-specific progenitor cells may provide cardiac benefits for the growing population of long-term cancer survivors that are at risk of chemotherapy-induced cardiovascular toxicity.
Collapse
Affiliation(s)
- Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy.
| | - Elena Piegari
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Donato Cappetta
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Rosa Russo
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Grazia Esposito
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Loreta Pia Ciuffreda
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | | | - Caterina Frati
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, Parma, Italy
| | - Francesco Fagnoni
- Immunohematology and Transfusional Medicine Unit, University-Hospital of Parma, Via Gramsci 14, Parma, Italy
| | - Liberato Berrino
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Federico Quaini
- Department of Clinical and Experimental Medicine, University of Parma, Via Gramsci 14, Parma, Italy
| | - Francesco Rossi
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| | - Konrad Urbanek
- Department of Experimental Medicine, Section of Pharmacology, Second University of Naples, Via Costantinopoli 16, 80138 Naples, Italy
| |
Collapse
|
215
|
Xu R, Wang Q. Large-scale automatic extraction of side effects associated with targeted anticancer drugs from full-text oncological articles. J Biomed Inform 2015; 55:64-72. [PMID: 25817969 DOI: 10.1016/j.jbi.2015.03.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2014] [Revised: 02/12/2015] [Accepted: 03/20/2015] [Indexed: 10/23/2022]
Abstract
Targeted anticancer drugs such as imatinib, trastuzumab and erlotinib dramatically improved treatment outcomes in cancer patients, however, these innovative agents are often associated with unexpected side effects. The pathophysiological mechanisms underlying these side effects are not well understood. The availability of a comprehensive knowledge base of side effects associated with targeted anticancer drugs has the potential to illuminate complex pathways underlying toxicities induced by these innovative drugs. While side effect association knowledge for targeted drugs exists in multiple heterogeneous data sources, published full-text oncological articles represent an important source of pivotal, investigational, and even failed trials in a variety of patient populations. In this study, we present an automatic process to extract targeted anticancer drug-associated side effects (drug-SE pairs) from a large number of high profile full-text oncological articles. We downloaded 13,855 full-text articles from the Journal of Oncology (JCO) published between 1983 and 2013. We developed text classification, relationship extraction, signaling filtering, and signal prioritization algorithms to extract drug-SE pairs from downloaded articles. We extracted a total of 26,264 drug-SE pairs with an average precision of 0.405, a recall of 0.899, and an F1 score of 0.465. We show that side effect knowledge from JCO articles is largely complementary to that from the US Food and Drug Administration (FDA) drug labels. Through integrative correlation analysis, we show that targeted drug-associated side effects positively correlate with their gene targets and disease indications. In conclusion, this unique database that we built from a large number of high-profile oncological articles could facilitate the development of computational models to understand toxic effects associated with targeted anticancer drugs.
Collapse
Affiliation(s)
- Rong Xu
- Medical Informatics Program, Center for Clinical Investigation, Case Western Reserve University, Cleveland, OH 44106, United States.
| | - QuanQiu Wang
- ThinTek, LLC, Palo Alto, CA 94306, United States.
| |
Collapse
|
216
|
Srikanthan A, Ethier JL, Ocana A, Seruga B, Krzyzanowska MK, Amir E. Cardiovascular toxicity of multi-tyrosine kinase inhibitors in advanced solid tumors: a population-based observational study. PLoS One 2015; 10:e0122735. [PMID: 25815472 PMCID: PMC4376902 DOI: 10.1371/journal.pone.0122735] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2014] [Accepted: 02/12/2015] [Indexed: 01/08/2023] Open
Abstract
Background Treatment with small molecule tyrosine kinase inhibitors (TKIs) has improved survival in many cancers, yet has been associated with an increased risk of adverse events. Warnings of cardiovascular events are common in drug labels of many TKIs. Despite these warnings, cardiovascular toxicity of patients treated with TKIs remains unclear. Here, we evaluate the cardiovascular outcomes of advanced cancer patients treated with small molecule tyrosine kinase inhibitors. Methods A population based cohort study was undertaken involving adults aged >18 years in Ontario, Canada, diagnosed with any advanced malignancy between 2006 and 2012. Data were extracted from linked administrative governmental databases. Adults with advanced cancer receiving TKIs were identified and followed throughout the time period. The main outcomes of interest were rates of hospitalization for ischemic heart disease (acute myocardial infarction and angina) or cerebrovascular accidents and death. Results 1642 patients with a mean age of 62.5 years were studied; 1046 were treated with erlotinib, 166 with sorafenib and 430 with sunitinib. Over the 380 day median follow-up period (range 6-1970 days), 1.1% of all patients had ischemic heart events, 0.7% had cerebrovascular accidents and 72.1% died. Rates of cardiovascular events were similar to age and gender-matched individuals without cancer. In a subgroup analysis of treatment patients with a prior history of ischemic heart disease, 3.3% had ischemic heart events while 1.2% had cerebrovascular accidents. Conclusions TKIs do not appear to increase the cause-specific hazard of ischemic heart disease and cerebrovascular accidents compared to age and gender-matched individuals without advanced cancer.
Collapse
Affiliation(s)
- Amirrtha Srikanthan
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
| | - Josee-Lyne Ethier
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Alberto Ocana
- Medical Oncology Department and Translational Research Unit, Albacete University Hospital, Edificio de Investigación, Calle Francisco Javier de Moya, Albacete, Spain
| | - Bostjan Seruga
- Department of Medical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Monika K. Krzyzanowska
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Eitan Amir
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre and Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Institute of Health Policy, Management and Evaluation, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
217
|
Morrison VA, Hamlin P, Soubeyran P, Stauder R, Wadhwa P, Aapro M, Lichtman S. Diffuse large B-cell lymphoma in the elderly: Impact of prognosis, comorbidities, geriatric assessment, and supportive care on clinical practice. An International Society of Geriatric Oncology (SIOG) Expert Position Paper. J Geriatr Oncol 2015; 6:141-52. [DOI: 10.1016/j.jgo.2014.11.004] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/02/2014] [Accepted: 11/20/2014] [Indexed: 12/19/2022]
|
218
|
Evaluation, prevention and management of cancer therapy-induced cardiotoxicity: a contemporary approach for clinicians. Curr Opin Cardiol 2015; 30:197-204. [PMID: 25574894 DOI: 10.1097/hco.0000000000000145] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW While targeted therapies have improved cancer outcomes, unique cardiovascular toxicities are increasingly recognized, particularly when administered sequentially after anthracyclines or radiation. Patients with cancer therapy-induced cardiotoxicity benefit from collaborative care involving cardiology and oncology, leading to a new interdisciplinary field called cardio-oncology. The present review will highlight contemporary clinical issues in cardio-oncology. RECENT FINDINGS Recently, risk factors for cancer therapy-induced cardiotoxicity have been evaluated in real-world rather than in clinical trial patients. Biomarkers and advanced echocardiography are emerging as sensitive tools for preclinical identification of cancer therapy-induced cardiotoxicity. Single-center studies suggest that cancer therapy-induced cardiotoxicity responds to prompt heart failure medical treatment, and such therapy may even prevent cardiotoxicity. SUMMARY Modern cancer therapy has short-term cardiac risk that may require collaborative management by clinicians with expertise in cardiology and oncology. The increased effectiveness of modern cancer therapy is resulting in a growing population of cancer survivors who are at long-term risk for cardiovascular disease. The present review of contemporary clinical issues in cardio-oncology will be of interest to healthcare providers who manage cardiotoxicity during cancer therapy, and who follow patients who survive cancer but face increased long-term cardiovascular risk.
Collapse
|
219
|
Tamargo J, Caballero R, Delpón E. Cancer Chemotherapy and Cardiac Arrhythmias: A Review. Drug Saf 2015; 38:129-52. [DOI: 10.1007/s40264-014-0258-4] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
220
|
Hinojar R, Botnar R, Kaski JC, Prasad S, Nagel E, Puntmann VO. Individualized cardiovascular risk assessment by cardiovascular magnetic resonance. Future Cardiol 2015; 10:273-89. [PMID: 24762254 DOI: 10.2217/fca.13.102] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular magnetic resonance (CMR) is gaining clinical importance in preventive medicine. Evidence on diagnostic accuracy and prognostic value, in addition to the development of faster imaging, increased availability of equipment and imaging expertise have led to a wide-spread use of CMR in a growing number of clinical indications. The first part of this review summarizes the role of CMR biomarkers for risk assessment focusing on the patients groups that benefit from the use of CMR. In the second part, the future directions for CMR are discussed and their role in prevention of cardiovascular disease.
Collapse
Affiliation(s)
- Rocio Hinojar
- Cardiovascular Imaging Department, Division of Imaging Sciences & Biomedical Engineering, King's College London, London, UK
| | | | | | | | | | | |
Collapse
|
221
|
Gimelli A, Lancellotti P, Badano LP, Lombardi M, Gerber B, Plein S, Neglia D, Edvardsen T, Kitsiou A, Scholte AJHA, Schroder S, Cosyns B, Gargiulo P, Zamorano JL, Perrone-Filardi P. Non-invasive cardiac imaging evaluation of patients with chronic systolic heart failure: a report from the European Association of Cardiovascular Imaging (EACVI). Eur Heart J 2014; 35:3417-25. [DOI: 10.1093/eurheartj/ehu433] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
|
222
|
Christenson ES, James T, Agrawal V, Park BH. Use of biomarkers for the assessment of chemotherapy-induced cardiac toxicity. Clin Biochem 2014; 48:223-35. [PMID: 25445234 DOI: 10.1016/j.clinbiochem.2014.10.013] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2014] [Revised: 10/25/2014] [Accepted: 10/28/2014] [Indexed: 12/17/2022]
Abstract
OBJECTIVES To review the evidence for the use of various biomarkers in the detection of chemotherapy associated cardiac damage. DESIGN AND METHODS Pubmed.gov was queried using the search words chemotherapy and cardiac biomarkers with the filters of past 10years, humans, and English language. An emphasis was placed on obtaining primary research articles looking at the utility of biomarkers for the detection of chemotherapy-mediated cardiac injury. RESULTS Biomarkers may help identify patients undergoing treatment who are at high risk for cardiotoxicity and may assist in identification of a low risk cohort that does not necessitate continued intensive screening. cTn assays are the best studied biomarkers in this context and may represent a promising and potentially valuable modality for detecting cardiac toxicity in patients undergoing chemotherapy. Monitoring cTnI levels may provide information regarding the development of cardiac toxicity before left ventricular dysfunction becomes apparent on echocardiography or via clinical symptoms. A host of other biomarkers have been evaluated for their utility in the field of chemotherapy related cardiac toxicity with intermittent success; further trials are necessary to determine what role they may end up playing for prediction and prognostication in this setting. CONCLUSIONS Biomarkers represent an exciting potential complement or replacement for echocardiographic monitoring of chemotherapy related cardiac toxicity which may allow for earlier realization of the degree of cardiac damage occurring during treatment, creating the opportunity for more timely modulation of therapy.
Collapse
Affiliation(s)
- Eric S Christenson
- Johns Hopkins University, Department of Medicine, Baltimore, MD 21287, USA.
| | - Theodore James
- Johns Hopkins University, Department of Medicine, Baltimore, MD 21287, USA
| | - Vineet Agrawal
- Johns Hopkins University, Department of Medicine, Baltimore, MD 21287, USA
| | - Ben H Park
- Johns Hopkins University, Department of Oncology, Baltimore, MD 21287, USA
| |
Collapse
|
223
|
Abdalla AME, Xiao L, Ouyang C, Yang G. Engineered nanoparticles: thrombotic events in cancer. NANOSCALE 2014; 6:14141-14152. [PMID: 25347245 DOI: 10.1039/c4nr04825c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Engineered nanoparticles are being increasingly produced for specific applications in medicine. Broad selections of nano-sized constructs have been developed for applications in diagnosis, imaging, and drug delivery. Nanoparticles as contrast agents enable conjugation with molecular markers which are essential for designing effective diagnostic and therapeutic strategies. Such investigations can also lead to a better understanding of disease mechanisms such as cancer-associated thrombosis which remains unpredictable with serious bleeding complications and high risk of death. Here we review the recent and current applications of engineered nanoparticles in diagnosis and therapeutic strategies, noting their toxicity in relation to specific markers as a target.
Collapse
Affiliation(s)
- Ahmed M E Abdalla
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | | | | | | |
Collapse
|
224
|
Sharp TE, George JC. Stem cell therapy and breast cancer treatment: review of stem cell research and potential therapeutic impact against cardiotoxicities due to breast cancer treatment. Front Oncol 2014; 4:299. [PMID: 25405100 PMCID: PMC4217360 DOI: 10.3389/fonc.2014.00299] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2014] [Accepted: 10/14/2014] [Indexed: 12/16/2022] Open
Abstract
A new problem has emerged with the ever-increasing number of breast cancer survivors. While early screening and advances in treatment have allowed these patients to overcome their cancer, these treatments often have adverse cardiovascular side effects that can produce abnormal cardiovascular function. Chemotherapeutic and radiation therapy have both been linked to cardiotoxicity; these therapeutics can cause a loss of cardiac muscle and deterioration of vascular structure that can eventually lead to heart failure (HF). This cardiomyocyte toxicity can leave the breast cancer survivor with a probable diagnosis of dilated or restrictive cardiomyopathy (DCM or RCM). While current HF standard of care can alleviate symptoms, other than heart transplantation, there is no therapy that replaces cardiac myocytes that are killed during cancer therapies. There is a need to develop novel therapeutics that can either prevent or reverse the cardiac injury caused by cancer therapeutics. These new therapeutics should promote the regeneration of lost or deteriorating myocardium. Over the last several decades, the therapeutic potential of cell-based therapy has been investigated for HF patients. In this review, we discuss the progress of pre-clinical and clinical stem cell research for the diseased heart and discuss the possibility of utilizing these novel therapies to combat cardiotoxicity observed in breast cancer survivors.
Collapse
Affiliation(s)
- Thomas E Sharp
- Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, PA , USA
| | - Jon C George
- Cardiovascular Research Center, Temple University School of Medicine , Philadelphia, PA , USA ; Division of Cardiovascular Medicine, Temple University Hospital , Philadelphia, PA , USA
| |
Collapse
|
225
|
Messinger-Rapport BJ, Gammack JK, Little MO, Morley JE. Clinical Update on Nursing Home Medicine: 2014. J Am Med Dir Assoc 2014; 15:786-801. [DOI: 10.1016/j.jamda.2014.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 09/02/2014] [Indexed: 12/18/2022]
|
226
|
Argilés JM, Busquets S, Stemmler B, López-Soriano FJ. Cancer cachexia: understanding the molecular basis. Nat Rev Cancer 2014; 14:754-62. [PMID: 25291291 DOI: 10.1038/nrc3829] [Citation(s) in RCA: 909] [Impact Index Per Article: 82.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cancer cachexia is a devastating, multifactorial and often irreversible syndrome that affects around 50-80% of cancer patients, depending on the tumour type, and that leads to substantial weight loss, primarily from loss of skeletal muscle and body fat. Since cachexia may account for up to 20% of cancer deaths, understanding the underlying molecular mechanisms is essential. The occurrence of cachexia in cancer patients is dependent on the patient response to tumour progression, including the activation of the inflammatory response and energetic inefficiency involving the mitochondria. Interestingly, crosstalk between different cell types ultimately seems to result in muscle wasting. Some of the recent progress in understanding the molecular mechanisms of cachexia may lead to new therapeutic approaches.
Collapse
Affiliation(s)
- Josep M Argilés
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; and Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | - Sílvia Busquets
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; and Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| | | | - Francisco J López-Soriano
- Cancer Research Group, Departament de Bioquímica i Biologia Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain; and Institut de Biomedicina de la Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
227
|
Zhabyeyev P, McLean B, Patel VB, Wang W, Ramprasath T, Oudit GY. Dual loss of PI3Kα and PI3Kγ signaling leads to an age-dependent cardiomyopathy. J Mol Cell Cardiol 2014; 77:155-9. [PMID: 25451171 DOI: 10.1016/j.yjmcc.2014.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 10/16/2014] [Accepted: 10/17/2014] [Indexed: 10/24/2022]
Abstract
Phosphatidylinositide 3-kinase (PI3K) signaling plays a critical role in maintaining normal cardiac structure and function. PI3Kα and PI3Kγ are the dominant cardiac isoforms and have both adaptive and maladaptive roles in heart disease. Broad spectrum PI3K inhibitors are emerging as potential new chemotherapeutic agents which may have deleterious long-term effects on the heart. We created a double mutant (PI3KDM) model by crossing p110γ(-/-) (PI3KγKO) with cardiac-specific PI3KαDN mice and studied cardiac structure and function at 1-year of age. Pressure-volume loop analysis and echocardiographic assessment showed PI3KDM mice developed marked impairment in systolic function while the wildtype, PI3KαDN, and PI3KγKO mice maintained normal systolic and diastolic function at 1-year of age. The PI3KDM hearts displayed increased expression of disease markers, increased myocardial fibrosis and matrix metalloproteinase (MMP) activity, depolymerization of intracellular F-actin, loss of phospho(threonine-308)-Akt, and normalization of phospho-Erk1/2 signaling. Dual loss of PI3Kα and PI3Kγ isoforms results in an age-dependent cardiomyopathy implying that long-term exposure to pan-PI3K inhibitors may lead to severe cardiotoxicity.
Collapse
Affiliation(s)
- Pavel Zhabyeyev
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Brent McLean
- Department of Physiology, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Vaibhav B Patel
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Wang Wang
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Department of Physiology, University of Alberta, Edmonton, Canada
| | - Tharmarajan Ramprasath
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada
| | - Gavin Y Oudit
- Division of Cardiology, Department of Medicine, University of Alberta, Edmonton, Canada; Department of Physiology, University of Alberta, Edmonton, Canada; Mazankowski Alberta Heart Institute, University of Alberta, Edmonton, Canada.
| |
Collapse
|
228
|
HGF/Met Axis in Heart Function and Cardioprotection. Biomedicines 2014; 2:247-262. [PMID: 28548070 PMCID: PMC5344277 DOI: 10.3390/biomedicines2040247] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2014] [Revised: 09/18/2014] [Accepted: 10/13/2014] [Indexed: 12/27/2022] Open
Abstract
Hepatocyte growth factor (HGF) and its tyrosine kinase receptor (Met) play important roles in myocardial function both in physiological and pathological situations. In the developing heart, HGF influences cardiomyocyte proliferation and differentiation. In the adult, HGF/Met signaling controls heart homeostasis and prevents oxidative stress in normal cardiomyocytes. Thus, the possible cardiotoxicity of current Met-targeted anti-cancer therapies has to be taken in consideration. In the injured heart, HGF plays important roles in cardioprotection by promoting: (1) prosurvival (anti-apoptotic and anti-autophagic) effects in cardiomyocytes, (2) angiogenesis, (3) inhibition of fibrosis, (4) anti-inflammatory and immunomodulatory signals, and (5) regeneration through activation of cardiac stem cells. Furthermore, we discuss the putative role of elevated HGF as prognostic marker of severity in patients with cardiac diseases. Finally, we examine the potential of HGF-based molecules as new therapeutic tools for the treatment of cardiac diseases.
Collapse
|
229
|
Nolan MT, Lowenthal RM, Venn A, Marwick TH. Chemotherapy-related cardiomyopathy: a neglected aspect of cancer survivorship. Intern Med J 2014; 44:939-50. [DOI: 10.1111/imj.12532] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/09/2014] [Indexed: 12/12/2022]
Affiliation(s)
- M. T. Nolan
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| | - R. M. Lowenthal
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| | - A. Venn
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| | - T. H. Marwick
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| |
Collapse
|
230
|
Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M, Ganame J, Sebag IA, Agler DA, Badano LP, Banchs J, Cardinale D, Carver J, Cerqueira M, DeCara JM, Edvardsen T, Flamm SD, Force T, Griffin BP, Jerusalem G, Liu JE, Magalhães A, Marwick T, Sanchez LY, Sicari R, Villarraga HR, Lancellotti P. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. Eur Heart J Cardiovasc Imaging 2014; 15:1063-93. [PMID: 25239940 PMCID: PMC4402366 DOI: 10.1093/ehjci/jeu192] [Citation(s) in RCA: 653] [Impact Index Per Article: 59.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
| | | | - Ana Barac
- Medstar Washington Hospital Center, Washington, District of Columbia
| | - Michael S Ewer
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Bonnie Ky
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Igal A Sebag
- Jewish General Hospital and McGill University, Montreal, Quebec, Canada
| | | | | | - Jose Banchs
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | | | - Joseph Carver
- Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Thor Edvardsen
- Oslo University Hospital and University of Oslo, Oslo, Norway
| | | | | | | | | | - Jennifer E Liu
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | | | - Liza Y Sanchez
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Rosa Sicari
- CNR Institute of Clinical Physiology, Pisa, Italy
| | | | | |
Collapse
|
231
|
Sturgeon K, Schadler K, Muthukumaran G, Ding D, Bajulaiye A, Thomas NJ, Ferrari V, Ryeom S, Libonati JR. Concomitant low-dose doxorubicin treatment and exercise. Am J Physiol Regul Integr Comp Physiol 2014; 307:R685-92. [PMID: 25009215 PMCID: PMC4166763 DOI: 10.1152/ajpregu.00082.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 07/04/2014] [Indexed: 01/08/2023]
Abstract
Cardiotoxicity is a side effect for cancer patients treated with doxorubicin (DOX). We tested the hypothesis that low-intensity aerobic exercise concomitant with DOX treatment would offset DOX-induced cardiotoxicity while also improving the therapeutic efficacy of DOX on tumor progression. B16F10 melanoma cells (3 × 10(5)) were injected subcutaneously into the scruff of 6- to 8-wk-old male C57BL/6 mice (n = 48). A 4 mg/kg cumulative dose of DOX was administered over 2 wk, and exercise (EX) consisted of treadmill walking (10 m/min, 45 min/day, 5 days/wk, 2 wk). Four experimental groups were tested: 1) sedentary (SED) + vehicle, 2) SED + DOX, 3) EX + vehicle, and 4) EX + DOX. Tumor volume was attenuated in DOX and lowest in EX + DOX. DOX-treated animals had less gain in body weight, reduced heart weights (HW), smaller HW-to-body weight ratios, and shorter tibial lengths by the end of the protocol; and exercise did not reverse the cardiotoxic effects of DOX. Despite decreased left ventricular (LV) mass with DOX, cardiomyocyte cross-sectional area, β-myosin heavy chain gene expression, and whole heart systolic (fractional shortening) and diastolic (E/A ratio) function were similar among groups. DOX also resulted in increased LV fibrosis with lower LV end diastolic volume and stroke volume. Myocardial protein kinase B activity was increased with both DOX and EX treatments, and tuberous sclerosis 2 (TSC2) abundance was reduced with EX. Downstream phosphorylation of TSC2 and mammalian target of rapamycin were similar across groups. We conclude that exercise increases the efficacy of DOX in inhibiting tumor growth without mitigating subclinical DOX-induced cardiotoxicity in a murine model of melanoma.
Collapse
Affiliation(s)
- Kathleen Sturgeon
- University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania; and
| | - Keri Schadler
- University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania; and
| | | | - Dennis Ding
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Akinyemi Bajulaiye
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Nicholas J Thomas
- University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania; and
| | - Victor Ferrari
- University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania; and
| | - Sandra Ryeom
- University of Pennsylvania, School of Medicine, Philadelphia, Pennsylvania; and
| | - Joseph R Libonati
- School of Nursing, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
232
|
Abstract
The Patient-Reported Outcomes Safety Event Reporting (PROSPER) Consortium was convened to improve safety reporting by better incorporating the perspective of the patient. PROSPER comprises industry, regulatory authority, academic, private sector and patient representatives who are interested in the area of patient-reported outcomes of adverse events (PRO-AEs). It has developed guidance on PRO-AE data, including the benefits of wider use and approaches for data capture and analysis. Patient-reported outcomes (PROs) encompass the full range of self-reporting, rather than only patient reports collected by clinicians using validated instruments. In recent years, PROs have become increasingly important across the spectrum of healthcare and life sciences. Patient-centred models of care are integrating shared decision making and PROs at the point of care; comparative effectiveness research seeks to include patients as participatory stakeholders; and industry is expanding its involvement with patients and patient groups as part of the drug development process and safety monitoring. Additionally, recent pharmacovigilance legislation from regulatory authorities in the EU and the USA calls for the inclusion of patient-reported information in benefit-risk assessment of pharmaceutical products. For patients, technological advancements have made it easier to be an active participant in one's healthcare. Simplified internet search capabilities, electronic and personal health records, digital mobile devices, and PRO-enabled patient online communities are just a few examples of tools that allow patients to gain increased knowledge about conditions, symptoms, treatment options and side effects. Despite these changes and increased attention on the perceived value of PROs, their full potential has yet to be realised in pharmacovigilance. Current safety reporting and risk assessment processes remain heavily dependent on healthcare professionals, though there are known limitations such as under-reporting and discordant perspectives between patient reports and clinician perceptions of adverse outcomes. PROSPER seeks to support the wider use of PRO-AEs. The scope of this guidance document, which was completed between July 2011 and March 2013, considered a host of domains related to PRO-AEs, including definitions and suitable taxonomies, the range of datasets that could be used, data collection mechanisms, and suitable analytical methodologies. PROSPER offers an innovative framework to differentiate patient populations. This framework considers populations that are prespecified (such as those in clinical trials, prospective observational studies and some registries) and non-prespecified populations (such as those in claims databases, PRO-enabled online patient networks, and social websites in general). While the main focus of this guidance is on post-approval PRO-AEs from both prespecified and non-prespecified population groups, PROSPER has also considered pre-approval, prespecified populations. The ultimate aim of this guidance is to ensure that the patient 'voice' and perspective feed appropriately into collection of safety data. The guidance also covers a minimum core dataset for use by industry or regulators to structure PRO-AEs (accessible in the online appendix) and how data, once collected, might be evaluated to better inform on the safe and effective use of medicinal products. Structured collection of such patient data can be considered both a means to an end (improving patient safety) as well as an end in itself (expressing the patient viewpoint). The members of the PROSPER Consortium therefore direct this PRO-AE guidance to multiple stakeholders in drug safety, including industry, regulators, prescribers and patients. The use of this document across the entirety of the drug development life cycle will help to better define the benefit-risk profile of new and existing medicines. Because of the clinical relevance of 'real-world' data, PROs have the potential to contribute important new knowledge about the benefits and risks of medicinal products, communicated through the voice of the patient.
Collapse
|
233
|
Plana JC, Galderisi M, Barac A, Ewer MS, Ky B, Scherrer-Crosbie M, Ganame J, Sebag IA, Agler DA, Badano LP, Banchs J, Cardinale D, Carver J, Cerqueira M, DeCara JM, Edvardsen T, Flamm SD, Force T, Griffin BP, Jerusalem G, Liu JE, Magalhães A, Marwick T, Sanchez LY, Sicari R, Villarraga HR, Lancellotti P. Expert consensus for multimodality imaging evaluation of adult patients during and after cancer therapy: a report from the American Society of Echocardiography and the European Association of Cardiovascular Imaging. J Am Soc Echocardiogr 2014; 27:911-39. [PMID: 25172399 DOI: 10.1016/j.echo.2014.07.012] [Citation(s) in RCA: 888] [Impact Index Per Article: 80.7] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
| | | | - Ana Barac
- Medstar Washington Hospital Center, Washington, District of Columbia
| | - Michael S Ewer
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Bonnie Ky
- University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Igal A Sebag
- Jewish General Hospital and McGill University, Montreal, Quebec, Canada
| | | | | | - Jose Banchs
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | | | - Joseph Carver
- Abramson Cancer Center at the University of Pennsylvania, Philadelphia, Pennsylvania
| | | | | | - Thor Edvardsen
- Oslo University Hospital and University of Oslo, Oslo, Norway
| | | | | | | | | | - Jennifer E Liu
- Memorial Sloan-Kettering Cancer Center, New York, New York
| | | | | | - Liza Y Sanchez
- MD Anderson Cancer Center, University of Texas, Houston, Texas
| | - Rosa Sicari
- CNR Institute of Clinical Physiology, Pisa, Italy
| | | | | |
Collapse
|
234
|
Shelburne N, Adhikari B, Brell J, Davis M, Desvigne-Nickens P, Freedman A, Minasian L, Force T, Remick SC. Cancer treatment-related cardiotoxicity: current state of knowledge and future research priorities. J Natl Cancer Inst 2014; 106:dju232. [PMID: 25210198 PMCID: PMC4176042 DOI: 10.1093/jnci/dju232] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2014] [Revised: 06/27/2014] [Accepted: 07/01/2014] [Indexed: 12/31/2022] Open
Abstract
Cardiotoxicity resulting from direct myocyte damage has been a known complication of cancer treatment for decades. More recently, the emergence of hypertension as a clinically significant side effect of several new agents has been recognized as adversely affecting cancer treatment outcomes. With cancer patients living longer, in part because of treatment advances, these adverse events have become increasingly important to address. However, little is known about the cardiovascular pathogenic mechanisms associated with cancer treatment and even less about how to optimally prevent and manage short- and long-term cardiovascular complications, leading to improved patient safety and clinical outcomes. To identify research priorities, allocate resources, and establish infrastructure required to address cardiotoxicity associated with cancer treatment, the National Cancer Institute (NCI) and National Heart, Lung and Blood Institute (NHLBI) sponsored a two-day workshop, "Cancer treatment-related cardiotoxicity: Understanding the current state of knowledge and future research priorities," in March 2013 in Bethesda, MD. Participants included leading oncology and cardiology researchers and health professionals, patient advocates and industry representatives, with expertise ranging from basic to clinical science. Attendees were charged with identifying research opportunities to advance the understanding of cancer treatment-related cardiotoxicity across basic and clinical science. This commentary highlights the key discussion points and overarching recommendations from that workshop.
Collapse
Affiliation(s)
- Nonniekaye Shelburne
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV.
| | - Bishow Adhikari
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| | - Joanna Brell
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| | - Myrtle Davis
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| | - Patrice Desvigne-Nickens
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| | - Andrew Freedman
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| | - Lori Minasian
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| | - Thomas Force
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| | - Scot C Remick
- Division of Cancer Control and Population Sciences (NS, AF) and Division of Cancer Prevention (JB, LM) and Division of Cancer Treatment and Diagnosis (MD), National Cancer Institute, Rockville, MD; Division of Cardiovascular Sciences, National Heart, Lung and Blood Institute, Bethesda, MD (BA, PDN); Cancer Center, MetroHealth Medical Center and Casewestern Reserve University, Cleveland, OH (JB); Vanderbilt Heart and Vascular Institute, Vanderbilt University School of Medicine, Nashville, TN (TF); Mary Babb Randolph Cancer Center, West Virginia University (SCR), Morgantown, WV
| |
Collapse
|
235
|
Left ventricular function in long-term survivors of childhood lymphoma. Am J Cardiol 2014; 114:483-90. [PMID: 24948492 DOI: 10.1016/j.amjcard.2014.04.055] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/29/2014] [Accepted: 04/29/2014] [Indexed: 11/23/2022]
Abstract
Survivors of childhood lymphoma (CL) have markedly increased risk of developing heart failure. Echocardiographic studies after cardiotoxic treatment have primarily demonstrated left ventricular (LV) systolic dysfunction. In the present study, we hypothesized that longer follow-up and a more comprehensive echocardiographic examination would reveal more cardiac abnormalities. We conducted a cross-sectional study with echocardiography 20.4 ± 8.6 years after diagnosis in 125 survivors of CL, grouped according to treatment methods, and compared with matched controls. Treatment included mediastinal radiotherapy (median 40.0 Gy) in 66 and anthracyclines (median dose 160 mg/m(2)) in 92 survivors of CL. Abnormal LV function, left-sided valve dysfunction, or both occurred in 62 patients (50%). Diastolic dysfunction occurred in 29%. Compared with control subjects, mitral annular early diastolic velocities (e') were reduced in patients (septal e' 0.09 ± 0.03 vs 0.12 ± 0.03 m/s, p <0.001), and the E/e' ratio was increased, particularly after mediastinal radiotherapy (10.6 ± 6.4 vs 5.6 ± 1.3, p <0.001). Survivors of CL had lower fractional shortening than control subjects (32 ± 6 vs 36 ± 7, p <0.001), but mean ejection fraction was equal and overt systolic dysfunction was infrequent. After mediastinal radiotherapy alone, global longitudinal myocardial strain was lower (p <0.05) compared with other treatment groups. Left-sided valvular dysfunction occurred in 55% of patients after mediastinal radiotherapy. In conclusion, survivors of CL had reduced LV diastolic function assessed by tissue Doppler imaging. This was more pronounced after mediastinal radiotherapy, which also frequently led to valvular disease. Systolic function was normal in most survivors of CL.
Collapse
|
236
|
D’Amore C, Gargiulo P, Paolillo S, Pellegrino AM, Formisano T, Mariniello A, Della Ratta G, Iardino E, D’Amato M, La Mura L, Fabiani I, Fusco F, Perrone Filardi P. Nuclear imaging in detection and monitoring of cardiotoxicity. World J Radiol 2014; 6:486-492. [PMID: 25071889 PMCID: PMC4109100 DOI: 10.4329/wjr.v6.i7.486] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/21/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Cardiotoxicity as a result of cancer treatment is a novel and serious public health issue that has a significant impact on a cancer patient’s management and outcome. The coexistence of cancer and cardiac disease in the same patient is more common because of aging population and improvements in the efficacy of antitumor agents. Left ventricular dysfunction is the most typical manifestation and can lead to heart failure. Left ventricular ejection fraction measurement by echocardiography and multigated radionuclide angiography is the most common diagnostic approach to detect cardiac damage, but it identifies a late manifestation of myocardial injury. Early non-invasive imaging techniques are needed for the diagnosis and monitoring of cardiotoxic effects. Although echocardiography and cardiac magnetic resonance are the most commonly used imaging techniques for cardiotoxicity assessment, greater attention is focused on new nuclear cardiologic techniques, which can identify high-risk patients in the early stage and visualize the pathophysiologic process at the tissue level before clinical manifestation. The aim of this review is to summarize the role of nuclear imaging techniques in the non-invasive detection of myocardial damage related to antineoplastic therapy at the reversible stage, focusing on the current role and future perspectives of nuclear imaging techniques and molecular radiotracers in detection and monitoring of cardiotoxicity.
Collapse
|
237
|
Vasti C, Hertig CM. Neuregulin-1/erbB activities with focus on the susceptibility of the heart to anthracyclines. World J Cardiol 2014; 6:653-662. [PMID: 25068025 PMCID: PMC4110613 DOI: 10.4330/wjc.v6.i7.653] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2013] [Revised: 02/11/2014] [Accepted: 05/19/2014] [Indexed: 02/06/2023] Open
Abstract
Neuregulin-1 (NRG1) signaling through the tyrosine kinase receptors erbB2 and erbB4 is required for cardiac morphogenesis, and it plays an essential role in maintaining the myocardial architecture during adulthood. The tyrosine kinase receptor erbB2 was first linked to the amplification and overexpression of erbb2 gene in a subtype of breast tumor cells, which is indicative of highly proliferative cells and likely a poor prognosis following conventional chemotherapy. The development of targeted therapies to block the survival of erbB2-positive cancer cells revealed that impaired NRG1 signaling through erbB2/erbB4 heterodimers combined with anthracycline chemotherapy may lead to dilated cardiomyopathy in a subpopulation of treated patients. The ventricular-specific deletion of either erbb2 or erbb4 manifested dilated cardiomyopathy, which is aggravated by the administration of doxorubicin. Based on the exacerbated toxicity displayed by the combined treatment, it is expected that the relevant pathways would be affected in a synergistic manner. This review examines the NRG1 activities that were monitored in different model systems, focusing on the emerging pathways and molecular targets, which may aid in understanding the acquired dilated cardiomyopathy that occurs under the conditions of NRG1-deficient signaling.
Collapse
|
238
|
Salazar-Mendiguchía J, González-Costello J, Roca J, Ariza-Solé A, Manito N, Cequier A. Anthracycline-mediated cardiomyopathy: basic molecular knowledge for the cardiologist. ARCHIVOS DE CARDIOLOGIA DE MEXICO 2014; 84:218-23. [PMID: 25001055 DOI: 10.1016/j.acmx.2013.08.006] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 08/12/2013] [Accepted: 08/20/2013] [Indexed: 11/28/2022] Open
Abstract
Anthracyclines are cytostatic antibiotics discovered almost half a century ago exerting their action through inhibition of topoisomerase II. The two most representative drugs are doxorubicin and daunorubicin and they have been proven as useful antineoplastics and are widely prescribed in daily oncology practice; unfortunately, cardiotoxicity has been a limiting factor when it comes to their use. Diverse mechanisms have been involved in anthracycline cardiotoxicity, none of which are capable of causing the whole clinical picture by itself. Traditionally, reactive oxygen species (ROS) have received more attention, although recently basic research has proven other factors to be as important as ROS. These factors mainly involve sarcomeric structure disruption, toxic accumulation of metabolites, iron metabolism, energetic alterations and inflammation. The role of genetics has been studied by some groups, although a clear genotype-response relationship is yet to be elucidated. With the improved survival from different oncologic diseases we are witnessing more cases of chemotherapy-induced cardiotoxicity and the advent of new anticancer drugs poses several challenges for the cardiologist, highlighting the importance of a deep knowledge of the main mechanisms inducing this toxicity.
Collapse
Affiliation(s)
- Joel Salazar-Mendiguchía
- Unidad de Miocardiopatías, Insuficiencia Cardíaca y Trasplante, Área de Enfermedades del Corazón, Hospital Universitari de Bellvitge, Barcelona, Spain.
| | - José González-Costello
- Unidad de Miocardiopatías, Insuficiencia Cardíaca y Trasplante, Área de Enfermedades del Corazón, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Josep Roca
- Unidad de Miocardiopatías, Insuficiencia Cardíaca y Trasplante, Área de Enfermedades del Corazón, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Albert Ariza-Solé
- Unidad de Miocardiopatías, Insuficiencia Cardíaca y Trasplante, Área de Enfermedades del Corazón, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Nicolás Manito
- Unidad de Miocardiopatías, Insuficiencia Cardíaca y Trasplante, Área de Enfermedades del Corazón, Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Angel Cequier
- Unidad de Miocardiopatías, Insuficiencia Cardíaca y Trasplante, Área de Enfermedades del Corazón, Hospital Universitari de Bellvitge, Barcelona, Spain
| |
Collapse
|
239
|
Aleman BM, Moser EC, Nuver J, Suter TM, Maraldo MV, Specht L, Vrieling C, Darby SC. Cardiovascular disease after cancer therapy. EJC Suppl 2014; 12:18-28. [PMID: 26217163 PMCID: PMC4250533 DOI: 10.1016/j.ejcsup.2014.03.002] [Citation(s) in RCA: 89] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 03/26/2014] [Indexed: 12/15/2022] Open
Abstract
Improvements in treatment and earlier diagnosis have both contributed to increased survival for many cancer patients. Unfortunately, many treatments carry a risk of late effects including cardiovascular diseases (CVDs), possibly leading to significant morbidity and mortality. In this paper we describe current knowledge of the cardiotoxicity arising from cancer treatments, outline gaps in knowledge, and indicate directions for future research and guideline development, as discussed during the 2014 Cancer Survivorship Summit organised by the European Organisation for Research and Treatment of Cancer (EORTC). Better knowledge is needed of the late effects of modern systemic treatments and of radiotherapy to critical structures of the heart, including the effect of both radiation dose and volume of the heart exposed. Research elucidating the extent to which treatments interact in causing CVD, and the mechanisms involved, as well as the extent to which treatments may increase CVD indirectly by increasing cardiovascular risk factors is also important. Systematic collection of data relating treatment details to late effects is needed, and great care is needed to obtain valid and generalisable results. Better knowledge of these cardiac effects will contribute to both primary and secondary prevention of late complications where exposure to cardiotoxic treatment is unavoidable. Also surrogate markers would help to identify patients at increased risk of cardiotoxicity. Evidence-based screening guidelines for CVD following cancer are also needed. Finally, risk prediction models should be developed to guide primary treatment choice and appropriate follow up after cancer treatment.
Collapse
Affiliation(s)
- Berthe M.P. Aleman
- Department of Radiation Oncology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Elizabeth C. Moser
- Department of Radiotherapy and Breast Unit, Champalimaud Foundation, Lisbon, Portugal
| | - Janine Nuver
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Thomas M. Suter
- Department of Cardiology, Bern University Hospital, Bern, Switzerland
| | - Maja V. Maraldo
- Department of Oncology and Haematology, Rigshospitalet, University of Copenhagen, Denmark
| | - Lena Specht
- Department of Oncology and Haematology, Rigshospitalet, University of Copenhagen, Denmark
| | - Conny Vrieling
- Department of Radiotherapy, Clinique des Grangettes, Geneva, Switzerland
| | - Sarah C. Darby
- Clinical Trial Service Unit, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
240
|
Magnano LC, Martínez Cibrian N, Andrade González X, Bosch X. Cardiac Complications of Chemotherapy: Role of Prevention. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:312. [DOI: 10.1007/s11936-014-0312-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
241
|
Cardiotoxicity of systemic agents used in breast cancer. Breast 2014; 23:317-28. [PMID: 24794210 DOI: 10.1016/j.breast.2014.04.002] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 03/21/2014] [Accepted: 04/03/2014] [Indexed: 11/21/2022] Open
Abstract
Several breast cancer therapies can lead to cardiovascular toxicity: drugs such anthracyclines can cause permanent damage, anti-HER2 agents may cause transitory and reversible cardiac dysfunction and others, such as those used in endocrine therapy, primarily disturb lipid metabolism. Considering the seriousness of these complications, trials are now being conducted to address cardiotoxicity associated with new drugs; however, to fully understand their toxicity profiles, longer follow-up is needed. In this review, we compile the information available about cardiac toxicity related to well-established systemic breast cancer treatments, as well as newer drugs, including antiangiogenics, mTOR inhibitors and novel anti-HER2 agents. We also describe current and next generation cardiac biomarkers and functional tests that can optimize treatment and reduce and prevent the incidence of treatment-related cardiotoxicity.
Collapse
|
242
|
Colombo A, Sandri MT, Salvatici M, Cipolla CM, Cardinale D. Cardiac complications of chemotherapy: role of biomarkers. CURRENT TREATMENT OPTIONS IN CARDIOVASCULAR MEDICINE 2014; 16:313. [PMID: 24771223 DOI: 10.1007/s11936-014-0313-6] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OPINION STATEMENT Both conventional and novel antineoplastic drugs may cause damage to the heart, ultimately affecting patients' survival and quality of life. In fact, the most frequent and typical clinical manifestation of cardiotoxicity, asymptomatic or symptomatic left ventricular dysfunction, may be induced not only by conventional cancer therapy, like anthracyclines, but also by new antitumoral targeted therapy such as trastuzumab. At present, left ventricular ejection fraction assessment represents the main standard practice for cardiac monitoring during cancer therapy, but it detects myocardial damage only when a functional impairment has already occurred, not allowing for early preventive strategies. In the last decade, a newer approach based on the measurement of cardiospecific biomarkers has been proposed, proving to have higher prognostic value than imaging modalities. In particular, cardiac troponin elevation during chemotherapy allows us to identify patients who are more prone to develop myocardial dysfunction and cardiac events during follow up. In these patients, the use of an angiotensin-converting enzyme inhibitor, such as enalapril, has shown to be effective in improving clinical outcome, giving the chance for a cardioprotective strategy in a selected population.
Collapse
Affiliation(s)
- Alessandro Colombo
- Cardiology Division, European Institute of Oncology, I.R.C.C.S., Via Ripamonti 435, 20141, Milan, Italy,
| | | | | | | | | |
Collapse
|
243
|
Hahn VS, Lenihan DJ, Ky B. Cancer therapy-induced cardiotoxicity: basic mechanisms and potential cardioprotective therapies. J Am Heart Assoc 2014; 3:e000665. [PMID: 24755151 PMCID: PMC4187516 DOI: 10.1161/jaha.113.000665] [Citation(s) in RCA: 186] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2013] [Accepted: 02/26/2014] [Indexed: 01/03/2023]
Affiliation(s)
- Virginia Shalkey Hahn
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (V.S.H., B.K.)
| | - Daniel J. Lenihan
- Cardiovascular Medicine, Vanderbilt University School of Medicine, Nashville, TN (D.J.L.)
| | - Bonnie Ky
- Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (V.S.H., B.K.)
- Penn Cardiovascular Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (B.K.)
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA (B.K.)
| |
Collapse
|
244
|
Thavendiranathan P, Poulin F, Lim KD, Plana JC, Woo A, Marwick TH. Use of myocardial strain imaging by echocardiography for the early detection of cardiotoxicity in patients during and after cancer chemotherapy: a systematic review. J Am Coll Cardiol 2014; 63:2751-68. [PMID: 24703918 DOI: 10.1016/j.jacc.2014.01.073] [Citation(s) in RCA: 767] [Impact Index Per Article: 69.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 01/24/2014] [Accepted: 01/28/2014] [Indexed: 01/29/2023]
Abstract
The literature exploring the utility of advanced echocardiographic techniques (such as deformation imaging) in the diagnosis and prognostication of patients receiving potentially cardiotoxic cancer therapy has involved relatively small trials in the research setting. In this systematic review of the current literature, we describe echocardiographic myocardial deformation parameters in 1,504 patients during or after cancer chemotherapy for 3 clinically-relevant scenarios. The systematic review was performed following the PRISMA (Preferred Reporting Items for Systematic Reviews and Meta-Analyses) guidelines using the EMBASE (1974 to November 2013) and MEDLINE (1946 to November 2013) databases. All studies of early myocardial changes with chemotherapy demonstrate that alterations of myocardial deformation precede significant change in left ventricular ejection fraction (LVEF). Using tissue Doppler-based strain imaging, peak systolic longitudinal strain rate has most consistently detected early myocardial changes during therapy, whereas with speckle tracking echocardiography (STE), peak systolic global longitudinal strain (GLS) appears to be the best measure. A 10% to 15% early reduction in GLS by STE during therapy appears to be the most useful parameter for the prediction of cardiotoxicity, defined as a drop in LVEF or heart failure. In late survivors of cancer, measures of global radial and circumferential strain are consistently abnormal, even in the context of normal LVEF, but their clinical value in predicting subsequent ventricular dysfunction or heart failure has not been explored. Thus, this systematic review confirms the value of echocardiographic myocardial deformation parameters for the early detection of myocardial changes and prediction of cardiotoxicity in patients receiving cancer therapy.
Collapse
Affiliation(s)
- Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada; Cardiac Conditions in Oncology Program, Mount Sinai Hospital, University of Toronto, Toronto, Ontario, Canada.
| | - Frédéric Poulin
- Division of Cardiology, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Ki-Dong Lim
- Division of Cardiology, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | - Juan Carlos Plana
- Cardio-Oncology Center, Section of Cardiovascular Imaging, Department of Cardiovascular Medicine, Cleveland Clinic, Cleveland, Ohio
| | - Anna Woo
- Division of Cardiology, Peter Munk Cardiac Center, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Ontario, Canada
| | | |
Collapse
|
245
|
Tarone G, Balligand JL, Bauersachs J, Clerk A, De Windt L, Heymans S, Hilfiker-Kleiner D, Hirsch E, Iaccarino G, Knöll R, Leite-Moreira AF, Lourenço AP, Mayr M, Thum T, Tocchetti CG. Targeting myocardial remodelling to develop novel therapies for heart failure. Eur J Heart Fail 2014; 16:494-508. [DOI: 10.1002/ejhf.62] [Citation(s) in RCA: 81] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 01/02/2014] [Accepted: 01/18/2014] [Indexed: 02/04/2023] Open
Affiliation(s)
- Guido Tarone
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute; Università di Torino; Torino Italy
| | - Jean-Luc Balligand
- Institut de Recherche Expérimentale et Clinique (IREC), Pole de Pharmacologie et Thérapeutique (UCL-FATH) and Department of Medicine, Cliniques Saint-Luc; Université catholique de Louvain; Bruxelles Belgium
| | - Johann Bauersachs
- Department of Cardiology and Angiology; Medizinische Hochschule-Hannover; Hannover Germany
| | - Angela Clerk
- School of Biological Sciences; University of Reading; Reading UK
| | - Leon De Windt
- Department of Cardiology, CARIM School for Cardiovascular Diseases; Maastricht University; Maastricht The Netherlands
| | - Stephane Heymans
- Center for Heart Failure Research, Cardiovascular Research Institute Maastricht (CARIM); Maastricht University; The Netherlands
| | - Denise Hilfiker-Kleiner
- Molecular Cardiology, Department of Cardiology and Angiology; Medizinische Hochschule-Hannover; Hannover Germany
| | - Emilio Hirsch
- Dipartimento di Biotecnologie Molecolari e Scienze per la Salute; Università di Torino; Torino Italy
| | - Guido Iaccarino
- Facoltà di Medicina; Università di Salerno; Salerno Italy
- IRCCS ‘Multimedica’; Milano Italy
| | - Ralph Knöll
- National Heart & Lung Institute; Imperial College London; London UK
| | - Adelino F. Leite-Moreira
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine; University of Porto; Porto Portugal
| | - André P. Lourenço
- Department of Physiology and Cardiothoracic Surgery, Faculty of Medicine; University of Porto; Porto Portugal
| | - Manuel Mayr
- King's British Heart Foundation Centre; King's College London; London UK
| | - Thomas Thum
- National Heart & Lung Institute; Imperial College London; London UK
- Institute of Molecular and Translational Therapeutic Strategies; Hannover Medical School; Hannover Germany
| | | |
Collapse
|
246
|
Vasiliadis I, Kolovou V, Kolovou G. Acute Cardiotoxic Effects of Adjuvant Trastuzumab Treatment. Angiology 2014; 65:950. [DOI: 10.1177/0003319714524048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Ioannis Vasiliadis
- Department of Clinical Biochemistry (Vascular Prevention Clinic), Royal Free Campus, University College London Medical School, University College London (UCL), London, United Kingdom
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| | - Vana Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| | - Genovefa Kolovou
- Cardiology Department, Onassis Cardiac Surgery Center, Athens, Greece
| |
Collapse
|
247
|
Abstract
Modeling integrated human physiology in vitro is a formidable task not yet achieved with any of the existing cell/tissue systems. However, tissue engineering is becoming increasingly successful at authentic representation of the actual environmental milieu of tissue development, regeneration and disease progression, and in providing real-time insights into morphogenic events. Functional human tissue units engineered to combine biological fidelity with the high-throughput screening and real-time measurement of physiological responses are poised to transform drug screening and predictive modeling of disease. In this review, we focus on the in vitro engineering of functional human myocardium that mimics heart tissue for analysis of myocardial function, in the context of physiological studies, drug screening for therapeutics, and safety pharmacology.
Collapse
|
248
|
McMurray JJ, Adamopoulos S, Anker SD, Auricchio A, Böhm M, Dickstein K, Falk V, Filippatos G, Fonseca C, Gomez-Sanchez MA, Jaarsma T, Køber L, Lip GY, Maggioni AP, Parkhomenko A, Pieske BM, Popescu BA, Rønnevik PK, Rutten FH, Schwitter J, Seferovic P, Stepinska J, Trindade PT, Voors AA, Zannad F, Zeiher A, Bax JJ, Baumgartner H, Ceconi C, Dean V, Deaton C, Fagard R, Funck-Brentano C, Hasdai D, Hoes A, Kirchhof P, Knuuti J, Kolh P, McDonagh T, Moulin C, Popescu BA, Reiner Ž, Sechtem U, Sirnes PA, Tendera M, Torbicki A, Vahanian A, Windecker S, McDonagh T, Sechtem U, Bonet LA, Avraamides P, Ben Lamin HA, Brignole M, Coca A, Cowburn P, Dargie H, Elliott P, Flachskampf FA, Guida GF, Hardman S, Iung B, Merkely B, Mueller C, Nanas JN, Nielsen OW, Ørn S, Parissis JT, Ponikowski P. ESC Guidelines for the diagnosis and treatment of acute and chronic heart failure 2012. Eur J Heart Fail 2014; 14:803-69. [PMID: 22828712 DOI: 10.1093/eurjhf/hfs105] [Citation(s) in RCA: 1829] [Impact Index Per Article: 166.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
249
|
Lüscher TF, Ruschitzka F, Landmesser U, Voors AA, van Gilst WH, van Veldhuisen DJ. TheEuropean Heart Journaland theEuropean Journal of Heart Failure: partners in scientific publishing. Eur J Heart Fail 2014; 14:1075-82. [DOI: 10.1093/eurjhf/hfs137] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Affiliation(s)
- Thomas F. Lüscher
- Editorial Office, European Heart Journal; Zurich Heart House; Moussonstreet 4 8091 Zürich Switzerland
| | - Frank Ruschitzka
- Editorial Office, European Heart Journal; Zurich Heart House; Moussonstreet 4 8091 Zürich Switzerland
| | - Ulf Landmesser
- Editorial Office, European Heart Journal; Zurich Heart House; Moussonstreet 4 8091 Zürich Switzerland
| | - Adriaan A. Voors
- Editorial Office, European Journal of Heart Failure; University of Groningen; Groningen The Netherlands
- Kingston-upon-Hull UK
| | - Wiek H. van Gilst
- Editorial Office, European Journal of Heart Failure; University of Groningen; Groningen The Netherlands
- Kingston-upon-Hull UK
| | - Dirk J. van Veldhuisen
- Editorial Office, European Journal of Heart Failure; University of Groningen; Groningen The Netherlands
- Kingston-upon-Hull UK
| |
Collapse
|
250
|
Horacek JM, Vasatova M, Pudil R, Tichy M, Zak P, Jakl M, Jebavy L, Maly J. Biomarkers for the early detection of anthracycline-induced cardiotoxicity: current status. Biomed Pap Med Fac Univ Palacky Olomouc Czech Repub 2014; 158:511-7. [PMID: 24457832 DOI: 10.5507/bp.2014.004] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2013] [Accepted: 01/15/2014] [Indexed: 11/23/2022] Open
Abstract
BACKGROUND Cardiotoxicity is a well-known and potentially serious complication of anticancer therapy. Anthracycline-based chemotherapy represents the greatest risk. Early detection of cardiotoxicity is crucial for applying preventive and supportive therapeutic strategies. METHODS AND RESULTS Various methods have been recommended for monitoring of cardiotoxicity. In our conditions, echocardiography and electrocardiography are routinely used. However, this approach shows low sensitivity for the early prediction of cardiomyopathy when the possibilities of appropriate management could still improve the patient's outcome. Recently, biomarkers of cardiac injury have been investigated in the assessment of chemotherapy-induced cardiotoxicity. Cardiospecific biomarkers, such as cardiac troponins, show high diagnostic efficacy in the early subclinical phase of the disease before the clinical onset of cardiomyopathy. Increase in their concentrations correlates with disease severity. As for natriuretic peptides, some studies, including ours, have shown promising results. Definitive evidence of their diagnostic and prognostic role in this context is still lacking and natriuretic peptides have not been routinely used for monitoring of cardiotoxicity in clinical practice. Other perspective biomarkers of cardiotoxicity in oncology are under study, especially heart-type fatty acid-binding protein (H-FABP) and glycogen phosphorylase BB (GPBB). Our studies using GPBB have provided encouraging results. However, the available data are limited and their practical use in this context cannot be recommended until their clinical efficacy is clearly defined. CONCLUSIONS This review covers the current status of biomarkers for the early detection of anthracycline-induced cardiotoxicity. The authors present in brief, their own experience with multiple biomarkers in the detection of cardiotoxicity.
Collapse
Affiliation(s)
- Jan M Horacek
- Department of Internal Medicine, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|