1
|
Mikail N, Chequer R, Imperiale A, Meisel A, Bengs S, Portmann A, Gimelli A, Buechel RR, Gebhard C, Rossi A. Tales from the future-nuclear cardio-oncology, from prediction to diagnosis and monitoring. Eur Heart J Cardiovasc Imaging 2023; 24:1129-1145. [PMID: 37467476 PMCID: PMC10501471 DOI: 10.1093/ehjci/jead168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 07/07/2023] [Indexed: 07/21/2023] Open
Abstract
Cancer and cardiovascular diseases (CVD) often share common risk factors, and patients with CVD who develop cancer are at high risk of experiencing major adverse cardiovascular events. Additionally, cancer treatment can induce short- and long-term adverse cardiovascular events. Given the improvement in oncological patients' prognosis, the burden in this vulnerable population is slowly shifting towards increased cardiovascular mortality. Consequently, the field of cardio-oncology is steadily expanding, prompting the need for new markers to stratify and monitor the cardiovascular risk in oncological patients before, during, and after the completion of treatment. Advanced non-invasive cardiac imaging has raised great interest in the early detection of CVD and cardiotoxicity in oncological patients. Nuclear medicine has long been a pivotal exam to robustly assess and monitor the cardiac function of patients undergoing potentially cardiotoxic chemotherapies. In addition, recent radiotracers have shown great interest in the early detection of cancer-treatment-related cardiotoxicity. In this review, we summarize the current and emerging nuclear cardiology tools that can help identify cardiotoxicity and assess the cardiovascular risk in patients undergoing cancer treatments and discuss the specific role of nuclear cardiology alongside other non-invasive imaging techniques.
Collapse
Affiliation(s)
- Nidaa Mikail
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Renata Chequer
- Department of Nuclear Medicine, Bichat University Hospital, AP-HP, University Diderot, 75018 Paris, France
| | - Alessio Imperiale
- Nuclear Medicine, Institut de Cancérologie de Strasbourg Europe (ICANS), University Hospitals of Strasbourg, 67093 Strasbourg, France
- Molecular Imaging-DRHIM, IPHC, UMR 7178, CNRS/Unistra, 67093 Strasbourg, France
| | - Alexander Meisel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Kantonsspital Glarus, Burgstrasse 99, 8750 Glarus, Switzerland
| | - Susan Bengs
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Angela Portmann
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| | - Alessia Gimelli
- Imaging Department, Fondazione CNR/Regione Toscana Gabriele Monasterio, Via G. Moruzzi 1, 56124 Pisa, Italy
| | - Ronny R Buechel
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
| | - Cathérine Gebhard
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
- Department of Cardiology, University Hospital Inselspital Bern, Freiburgstrasse 18, 3010 Bern, Switzerland
| | - Alexia Rossi
- Department of Nuclear Medicine, University Hospital Zurich, Rämistrasse 100, 8091 Zurich, Switzerland
- Center for Molecular Cardiology, University of Zurich, Wagistrasse 12, 8952 Schlieren, Switzerland
| |
Collapse
|
2
|
Leo I, Vidula M, Bisaccia G, Procopio MC, Licordari R, Perotto M, La Vecchia G, Miaris N, Bravo PE, Bucciarelli-Ducci C. The Role of Advanced Cardiovascular Imaging Modalities in Cardio-Oncology: From Early Detection to Unravelling Mechanisms of Cardiotoxicity. J Clin Med 2023; 12:4945. [PMID: 37568347 PMCID: PMC10419705 DOI: 10.3390/jcm12154945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Advances in cancer therapies have led to a global improvement in patient survival rates. Nevertheless, the price to pay is a concomitant increase in cardiovascular (CV) morbidity and mortality in this population. Increased inflammation and disturbances of the immune system are shared by both cancer and CV diseases. Immunological effects of anti-cancer treatments occur with both conventional chemotherapy and, to a greater extent, with novel biological therapies such as immunotherapy. For these reasons, there is growing interest in the immune system and its potential role at the molecular level in determining cardiotoxicity. Early recognition of these detrimental effects could help in identifying patients at risk and improve their oncological management. Non-invasive imaging already plays a key role in evaluating baseline CV risk and in detecting even subclinical cardiac dysfunction during surveillance. The aim of this review is to highlight the role of advanced cardiovascular imaging techniques in the detection and management of cardiovascular complications related to cancer treatment.
Collapse
Affiliation(s)
- Isabella Leo
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Mahesh Vidula
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA (P.E.B.)
- Divisions of Nuclear Medicine and Cardiothoracic Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giandomenico Bisaccia
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Neuroscience, Imaging and Clinical Sciences, “G. d’Annunzio” University of Chieti-Pescara, 66100 Chieti, Italy
| | - Maria Cristina Procopio
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Clinical and Experimental Medicine, University of Messina, 98122 Messina, Italy
| | - Roberto Licordari
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Biomedical and Dental Sciences and of Morphological and Functional Images, University of Messina, 98122 Messina, Italy
| | - Maria Perotto
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
| | - Giulia La Vecchia
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- Department of Cardiovascular and Pulmonary Science, Catholic University of Sacred Heart, 00168 Rome, Italy
| | - Nikolaos Miaris
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
| | - Paco E. Bravo
- Division of Cardiovascular Medicine, Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA (P.E.B.)
- Divisions of Nuclear Medicine and Cardiothoracic Imaging, Department of Radiology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Chiara Bucciarelli-Ducci
- Royal Brompton and Harefield Hospitals, Guy’s and St Thomas’ NHS Foundation Trust, London SE1 7EH, UK; (I.L.)
- School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London WC2R 2LS, UK
| |
Collapse
|
3
|
Bauckneht M, Sambuceti G. Functional innervation imaging in the evaluation of cardiotoxicity: Just the beginning of the journey. J Nucl Cardiol 2022; 29:2292-2294. [PMID: 34228327 DOI: 10.1007/s12350-021-02668-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Accepted: 05/04/2021] [Indexed: 10/20/2022]
Affiliation(s)
- Matteo Bauckneht
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy.
| | - Gianmario Sambuceti
- Nuclear Medicine, IRCCS Ospedale Policlinico San Martino, Largo Rosanna Benzi 10, 16132, Genoa, Italy
- Department of Health Sciences, University of Genoa, Genoa, Italy
| |
Collapse
|
4
|
Arrais TR, Cavalli GD, Dos Santos BT, Pereira GB, Migliavaca CB, Grossman GB, Biolo A. MIBG cardiac imaging compared to ejection fraction in evaluation of cardiotoxicity: a systematic review. J Nucl Cardiol 2022; 29:2274-2291. [PMID: 34228328 DOI: 10.1007/s12350-021-02610-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Accepted: 03/15/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND Advances in diagnosis and treatment of cancer has improved survival but resulted in increased cardiotoxic effects. The decrease in left ventricular ejection fraction (EF), one of the pillars of diagnosis of cardiotoxicity, seems to be a late process in the evolution of the disease, so 123I-metaiodobenzylguanidine (MIBG) cardiac imaging has been proposed to detect early cardiac impairment. The aim of this systematic review was to evaluate the performance of MIBG cardiac scan in this scenario. METHODS AND RESULTS A systematic search was conducted in five international databases comparing MIBG parameters with EF for evaluation of cardiotoxicity. Twelve studies were included and separated in three groups. First, studies evaluating patients with established cardiotoxicity, in which EF was reduced and MIBG parameters were abnormal. Second, studies analyzing patients during or after treatment compared to controls, with MIBG parameters significantly different between groups in most studies, even when EF remained normal. Finally, studies analyzing anthracycline (ATC) dose-related changes, with alteration in MIBG parameters occurring even when EF was preserved. CONCLUSION Although studies had high methodological variability, cardiac sympathetic innervation imaging seems to be a promising tool for assessing early cardiotoxicity. Further studies are needed to analyze its diagnostic value in this scenario.
Collapse
Affiliation(s)
- Thaís Rossato Arrais
- Nuclear Medicine Department, Hospital Moinhos de Vento, Rua Ramiro Barcelos, 910-201, Porto Alegre, RS, 90035-001, Brazil.
- Post-graduate Program in Cardiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.
| | | | | | | | - Celina Borges Migliavaca
- Post-graduate Program in Epidemiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Gabriel Blacher Grossman
- Nuclear Medicine Department, Hospital Moinhos de Vento, Rua Ramiro Barcelos, 910-201, Porto Alegre, RS, 90035-001, Brazil
| | - Andréia Biolo
- Post-graduate Program in Cardiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Cardiology Department, Hospital de Clínicas de Porto Alegre, Porto Alegre, Brazil
| |
Collapse
|
5
|
Abstract
PURPOSE OF REVIEW Successful treatment of cancer can be hampered by the attendant risk of cardiotoxicity, manifesting as cardiomyopathy, left ventricle systolic dysfunction and, in some cases, heart failure. This risk can be mitigated if the injury to the heart is detected before the onset to irreversible cardiac impairment. The gold standard for cardiac imaging in cardio-oncology is echocardiography. Despite improvements in the application of this modality, it is not typically sensitive to sub-clinical or early-stage dysfunction. We identify in this review some emerging tracers for detecting incipient cardiotoxicity by positron emission tomography (PET). RECENT FINDINGS Vectors labeled with positron-emitting radionuclides (e.g., carbon-11, fluorine-18, gallium-68) are now available to study cardiac function, metabolism, and tissue repair in preclinical models. Many of these probes are highly sensitive to early damage, thereby potentially addressing the limitations of current imaging approaches, and show promise in preliminary clinical evaluations. The overlapping pathophysiology between cardiotoxicity and heart failure significantly expands the number of imaging tools available to cardio-oncology. This is highlighted by the emergence of radiolabeled probes targeting fibroblast activation protein (FAP) for sensitive detection of dysregulated healing process that underpins adverse cardiac remodeling. The growth of PET scanner technology also creates an opportunity for a renaissance in metabolic imaging in cardio-oncology research.
Collapse
Affiliation(s)
- James M. Kelly
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
| | - John W. Babich
- Division of Radiopharmaceutical Sciences and Molecular Imaging Innovations Institute (MI3), Weill Cornell Medicine, Belfer Research Building, Room BB-1604, 413 East 69th St, New York, NY 10021 USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY 10021 USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021 USA
| |
Collapse
|
6
|
Soufer A, Liu C, Henry ML, Baldassarre LA. Nuclear cardiology in the context of multimodality imaging to detect cardiac toxicity from cancer therapeutics: Established and emerging methods. J Nucl Cardiol 2020; 27:1210-1224. [PMID: 30868378 DOI: 10.1007/s12350-019-01671-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 02/12/2019] [Indexed: 12/21/2022]
Abstract
The complexity of cancer therapies has vastly expanded in the last decade, along with type and severity of cardiac toxicities associated with these treatments. Prevention of pre-clinical cardiotoxicity may improve cardiovascular outcomes and circumvent the decision to place life-sustaining chemotherapeutic agents on hold, making the early detection of cancer therapeutic related cardiac toxicity with non-invasive imaging essential to the care of these patients. There are several established methods of cardiac imaging in the areas of nuclear cardiology, echocardiography, computed tomography, and cardiac magnetic resonance imaging that are used to assess for cardiovascular toxicity of cancer treatments, with several methods under development. The following review will provide an overview of current and emerging imaging techniques in these areas.
Collapse
Affiliation(s)
- Aaron Soufer
- Department of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA.
| | - Chi Liu
- Department of Radiology and Biomedical Engineering, Yale University School of Medicine, New Haven, CT, USA
| | - Mariana L Henry
- Department of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| | - Lauren A Baldassarre
- Department of Cardiovascular Medicine, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
7
|
Laursen AH, Ripa RS, Hasbak P, Kjær A, Elming MB, Køber L, Hutchings M, Thune JJ. 123I-MIBG for detection of subacute doxorubicin-induced cardiotoxicity in patients with malignant lymphoma. J Nucl Cardiol 2020; 27:931-939. [PMID: 30569409 DOI: 10.1007/s12350-018-01566-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2018] [Accepted: 11/21/2018] [Indexed: 12/22/2022]
Abstract
BACKGROUND Doxorubicin is the mainstay of curative lymphoma treatment but is associated with a dose-dependent cardiotoxicity that is often recognized too late to avoid substantial irreversible cardiac injury. Iodine-123 metaiodobenzylguanidine (123I-MIBG) is a gamma-emitting tracer that mimics noradrenaline uptake, storage, and release mechanisms in adrenergic presynaptic neurons. 123I-MIBG scintigraphy can be used for assessment of doxorubicin-induced injury to myocardial adrenergic neurons during treatment and could be the tool for early detection of doxorubicin cardiotoxicity, which is currently lacking. METHODS AND RESULTS A total of 37 lymphoma patients scheduled for doxorubicin treatment were included in our study. 123I-MIBG imaging was performed prior to chemotherapy and after a median of 4 cycles of doxorubicin. Early and late heart-to-mediastinum ratios (H/Mearly and H/Mlate) and washout rate (WOR) were used for evaluation of cardiotoxicity. The prognostic value of 123I-MIBG results was assessed using left ventricular ejection fraction (LVEF) as measured by cardiac magnetic resonance at 1-year follow-up. We found a post-therapy increase in WOR (including nine patients with > 10% increase), which was not statistically significant (18.6 vs 23.4%, P = 0.09). The difference appeared to be driven by an increase in H/Mearly. LVEF decreased from baseline to 1-year follow-up (64 vs 58%, P = 0.03). LVEF change was not associated with changes in WOR (P = 0.5). CONCLUSION The present study does not provide evidence for 123I-MIBG imaging as a clinically applicable tool for early detection of doxorubicin-induced cardiotoxicity.
Collapse
Affiliation(s)
- Adam Høgsbro Laursen
- Department of Hematology, Rigshospitalet, University of Copenhagen, section 9322, Blegdamsvej 9, 2100, Copenhagen, Denmark.
| | - Rasmus Sejersten Ripa
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Philip Hasbak
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Andreas Kjær
- Department of Clinical Physiology, Nuclear Medicine & PET and Cluster for Molecular Imaging, Department of Biomedical Sciences, Rigshospitalet, University of Copenhagen, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Marie Bayer Elming
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Lars Køber
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Martin Hutchings
- Department of Hematology, Rigshospitalet, University of Copenhagen, section 9322, Blegdamsvej 9, 2100, Copenhagen, Denmark
| | - Jens Jakob Thune
- Department of Cardiology, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
- Department of Cardiology, Bispebjerg and Frederiksberg Hospital, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
8
|
Wan N, Travin MI. Cardiac Imaging With 123I-meta-iodobenzylguanidine and Analogous PET Tracers: Current Status and Future Perspectives. Semin Nucl Med 2020; 50:331-348. [PMID: 32540030 DOI: 10.1053/j.semnuclmed.2020.03.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Autonomic innervation plays an important role in proper functioning of the cardiovascular system. Altered cardiac sympathetic function is present in a variety of diseases, and can be assessed with radionuclide imaging using sympathetic neurotransmitter analogues. The most studied adrenergic radiotracer is cardiac 123I-meta-iodobenzylguanidine (123I-mIBG). Cardiac 123I-mIBG uptake can be evaluated using both planar and tomographic imaging, thereby providing insight into global and regional sympathetic innervation. Standardly assessed imaging parameters are the heart-to-mediastinum ratio and washout rate, customarily derived from planar images. Focal tracer deficits on tomographic imaging also show prognostic utility, with some data suggesting that the best approach to tomographic image interpretation may differ from conventional methods. Cardiac 123I-mIBG image findings strongly correlate with the severity and prognosis of many cardiovascular diseases, especially heart failure and ventricular arrhythmias. Cardiac 123I-mIBG imaging in heart failure is FDA approved for prognostic purposes. With the robustly demonstrated ability to predict occurrence of potentially fatal arrhythmias, cardiac 123I-mIBG imaging shows promise for better selecting patients who will benefit from an implantable cardioverter defibrillator, but clinical use has been hampered by lack of the randomized trial needed for incorporation into societal guidelines. In patients with ischemic heart disease, cardiac 123I-mIBG imaging aids in assessing the extent of damage and in identifying arrhythmogenic regions. There have also been studies using cardiac 123I-mIBG for other conditions, including patients following heart transplantation, diabetic related cardiac abnormalities and chemotherapy induced cardiotoxicity. Positron emission tomographic adrenergic radiotracers, that improve image quality, have been investigated, especially 11C-meta-hydroxyephedrine, and most recently 18F-fluorbenguan. Cadmium-zinc-telluride cameras also improve image quality. With better spatial resolution and quantification, PET tracers and advanced camera technologies promise to expand the clinical utility of cardiac sympathetic imaging.
Collapse
Affiliation(s)
- Ningxin Wan
- Division of Nuclear Medicine, Department of Radiology, and Division of Cardiology, Department of Medicine, Montefiore Medical Center and The Albert Einstein College of Medicine, Bronx, NY
| | - Mark I Travin
- Division of Nuclear Medicine, Department of Radiology, and Division of Cardiology, Department of Medicine, Montefiore Medical Center and The Albert Einstein College of Medicine, Bronx, NY.
| |
Collapse
|
9
|
McCluskey SP, Haslop A, Coello C, Gunn RN, Tate EW, Southworth R, Plisson C, Long NJ, Wells LA. Imaging of Chemotherapy-Induced Acute Cardiotoxicity with 18F-Labeled Lipophilic Cations. J Nucl Med 2019; 60:1750-1756. [PMID: 31147403 DOI: 10.2967/jnumed.119.226787] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Accepted: 05/29/2019] [Indexed: 12/25/2022] Open
Abstract
Many chemotherapy agents are toxic to the heart, such that increasing numbers of cancer survivors are now living with the potentially lethal cardiovascular consequences of their treatment. Earlier and more sensitive detection of chemotherapy-induced cardiotoxicity may allow improved treatment strategies and increase long-term survival. Lipophilic cation PET tracers may be suitable for early detection of cardiotoxicity. This study aimed to evaluate an 18F-labeled lipophilic phosphonium cation, [1-(2-18F-fluoroethyl),1H[1,2,3]triazole-4-ethylene]triphenylphosphonium bromide (18F-MitoPhos), as a cardiac imaging agent, comparing it with leading PET and SPECT lipophilic cationic tracers before further assessing its potential for imaging cardiotoxicity in an acute doxorubicin model. Methods: Cardiac uptake and response to decreased mitochondrial membrane potential of 18F-MitoPhos and 99mTc-sestamibi were tested in isolated perfused rat hearts. Baseline pharmacokinetic profiles of 18F-MitoPhos and 18F-fluorobenzyltriphenylphosphonium and their response to acute doxorubicin-induced cardiotoxicity were assessed in rats in vivo (10, 15, or 20 mg of doxorubicin per kilogram, intravenously, 48 h beforehand). Results: Cardiac retention of 18F-MitoPhos was more than double that of 99mTc-sestamibi in isolated perfused rat hearts. A favorable biodistribution of 18F-MitoPhos in vivo was observed, with heart-to-tissue ratios of 304 ± 186, 11.2 ± 1.2, and 3.8 ± 0.6 for plasma, liver, and lung, respectively (60 min). A significant dose-dependent loss of cardiac retention of 18F-MitoPhos was observed on doxorubicin treatment, with average cardiac SUV from 30 to 60 min (mean ± SD) decreasing from 3.5 ± 0.5 (control) to 1.8 ± 0.1 (doxorubicin, 20 mg/kg). Other assessed biomarkers showed no alterations. Conclusion: 18F-MitoPhos showed pharmacokinetic parameters suitable for cardiac imaging. A significant dose response of cardiac uptake to doxorubicin treatment was observed before detectable biomarker alterations. 18F-MitoPhos is therefore a promising tracer for imaging chemotherapy-induced cardiotoxicity. To our knowledge, this is the first demonstration of radiolabeled lipophilic cations being used for the PET imaging of chemotherapy-induced cardiotoxicity and indicates the potential application of these compounds in this area.
Collapse
Affiliation(s)
- Stuart P McCluskey
- Department of Chemistry, Imperial College London, London, United Kingdom.,Invicro LLC, London, United Kingdom
| | - Anna Haslop
- Department of Chemistry, Imperial College London, London, United Kingdom
| | | | - Roger N Gunn
- Invicro LLC, London, United Kingdom.,Division of Brain Sciences, Imperial College London, Imperial College Centre for Drug Discovery Science, London, United Kingdom; and
| | - Edward W Tate
- Department of Chemistry, Imperial College London, London, United Kingdom
| | - Richard Southworth
- Biomedical Engineering and Imaging Sciences, King's College London, London, United Kingdom
| | | | - Nicholas J Long
- Department of Chemistry, Imperial College London, London, United Kingdom
| | | |
Collapse
|
10
|
Proctor P, Hage FG. Serial Cardiac Nuclear Imaging: Opportunities and Challenges. CURRENT CARDIOVASCULAR IMAGING REPORTS 2018. [DOI: 10.1007/s12410-018-9442-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
11
|
Abstract
PURPOSE OF REVIEW Cardio-oncology focuses increased effort to decrease cancer treatment-related cardiotoxicity while continuing to improve outcomes. We sought to synthesize the latest in nuclear cardiology as it pertains to the assessment of left ventricular function in preventative guidelines and comparison to other modalities, novel molecular markers of pre-clinical cardiotoxicity, and its role in cardiac amyloid diagnosis. RECENT FINDINGS Planar ERNA (equilibrium radionuclide angiocardiography) provides a reliable and proven means of monitoring and preventing anthracycline cardiotoxicity, and SPECT ERNA using solid-state gamma cameras may provide reproducible assessments of left ventricular function with reduced radiation exposure. While certain chemotherapeutics have vascular side effects, the use of stress perfusion imaging has still not been adequately studied for routine use. Similarly, markers of apoptosis, inflammation, and sympathetic nerve dysfunction are promising, but are still not ready for uniform usage. SPECT tracers can assist in nonbiopsy diagnosis of cardiac amyloid. Nuclear cardiology is a significant contributor to the multimodality approach to cardio-oncology.
Collapse
Affiliation(s)
- Jorge A Alvarez
- Warren Alpert Medical School, Brown University, Providence, RI, USA
| | - Raymond R Russell
- Warren Alpert Medical School, Brown University, Providence, RI, USA.
| |
Collapse
|
12
|
Abstract
PURPOSE OF REVIEW Autonomic innervation is crucial for regulating cardiac function. Sympathetic innervation imaging with 123I-mIBG and analogous PET tracers assesses disease in ways that differ from customary methods. This review describes practical use in various clinical scenarios, discusses recent guidelines, presents new data confirming risk stratification power, describes an ongoing prospective study, and looks forward to wider use in patient management. RECENT FINDINGS ASNC 123I-mIBG guidelines are available, expanding on European guidelines. ADMIRE-HF patient follow-up increased to 2 years in ADMIRE HFX, demonstrating independent mortality risk reclassification. ADMIRE-HF findings were substantiated in a Japanese consortium study and in the PAREPET 11C-HED PET study. Exciting potential uses of adrenergic imaging are management of LVADs and VT ablation. CZT cameras provide advantages, but derived parameters differ from Anger camera values. Independent risk stratification utility of adrenergic imaging with 123I-mIBG and PET tracers is continuously being confirmed. An ongoing prospective randomized study promises to establish patient management utility. There is potential for wider use and improved images with newer cameras and PET.
Collapse
Affiliation(s)
- Mark I Travin
- Department of Radiology/Division of Nuclear Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East-210th Street, Bronx, NY, 10467-2490, USA.
| |
Collapse
|
13
|
Chen-Scarabelli C, McRee C, Leesar MA, Hage FG, Scarabelli TM. Comprehensive review on cardio-oncology: Role of multimodality imaging. J Nucl Cardiol 2017; 24:906-935. [PMID: 27225513 DOI: 10.1007/s12350-016-0535-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 04/19/2016] [Indexed: 10/21/2022]
Abstract
Cancer and cardiovascular disease are the two leading causes of mortality worldwide. Evolving oncologic therapy, including the use of newer targeted agents, has led to an improvement in survival from childhood- and adult-onset cancers. Consequently, there has been a growing realization of cardiotoxic complications related to cancer therapy, with some complications manifesting over months to decades after completion of cancer treatment. This paper reviews cancer therapeutics-related cardiovascular toxicity and its manifestations, multimodality imaging techniques for surveillance and detection of this complication, and the current state of knowledge in this emerging field.
Collapse
Affiliation(s)
- Carol Chen-Scarabelli
- Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Chad McRee
- Division of Cardiovascular Disease, University of Alabama at Birmingham, 1530 3rd Avenue, South Tinsley Harrison Tower, Birmingham, Alabama, 35294-0006, USA
| | - Massoud A Leesar
- Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Cardiovascular Disease, University of Alabama at Birmingham, 1530 3rd Avenue, South Tinsley Harrison Tower, Birmingham, Alabama, 35294-0006, USA
| | - Fadi G Hage
- Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, Alabama, USA
- Division of Cardiovascular Disease, University of Alabama at Birmingham, 1530 3rd Avenue, South Tinsley Harrison Tower, Birmingham, Alabama, 35294-0006, USA
| | - Tiziano M Scarabelli
- Birmingham Veterans Affairs Medical Center, University of Alabama at Birmingham, Birmingham, Alabama, USA.
- Division of Cardiovascular Disease, University of Alabama at Birmingham, 1530 3rd Avenue, South Tinsley Harrison Tower, Birmingham, Alabama, 35294-0006, USA.
| |
Collapse
|
14
|
Pepe A, Pizzino F, Gargiulo P, Perrone-Filardi P, Cadeddu C, Mele D, Monte I, Novo G, Zito C, Di Bella G. Cardiovascular imaging in the diagnosis and monitoring of cardiotoxicity: cardiovascular magnetic resonance and nuclear cardiology. J Cardiovasc Med (Hagerstown) 2017; 17 Suppl 1 Special issue on Cardiotoxicity from Antiblastic Drugs and Cardioprotection:e45-e54. [PMID: 27755242 DOI: 10.2459/jcm.0000000000000380] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Chemotherapy-induced cardiotoxicity (CTX) is a determining factor for the quality of life and mortality of patients administered potentially cardiotoxic drugs and in long-term cancer survivors. Therefore, prevention and early detection of CTX are highly desirable, as is the exploration of alternative therapeutic strategies and/or the proposal of potentially cardioprotective treatments. In recent years, cardiovascular imaging has acquired a pivotal role in this setting. Although echocardiography remains the diagnostic method most used to monitor cancer patients, the need for more reliable, reproducible and accurate detection of early chemotherapy-induced CTX has encouraged the introduction of second-line advanced imaging modalities, such as cardiac magnetic resonance (CMR) and nuclear techniques, into the clinical setting. This review of the Working Group on Drug Cardiotoxicity and Cardioprotection of the Italian Society of Cardiology aims to afford an overview of the most important findings from the literature about the role of CMR and nuclear techniques in the management of chemotherapy-treated patients, describe conventional and new parameters for detecting CTX from both diagnostic and prognostic perspectives and provide integrated insight into the role of CMR and nuclear techniques compared with other imaging tools and versus the positions of the most important international societies.
Collapse
Affiliation(s)
- Alessia Pepe
- aU.O.C. Magnetic Resonance Imaging, Fondazione Toscana G. Monasterio C.N.R., Pisa bDepartment of Clinical and Experimental Medicine. Section of Cardiology A.O.U. Policlinico 'G. Martino' University of Messina, Messina cSDN, Institute of Diagnostic and Nuclear Development dDepartment of Advanced Biomedical Sciences, 'Federico II' University, Naples eDepartment of Medical Sciences 'Mario Aresu', University of Cagliari, Cagliari fCardiology Unit, University Hospital of Ferrara, Ferrara gDepartment of General Surgery and Medical-Surgery Specialities, University of Catania, Catania hChair and Division of Cardiology, University of Palermo, Palermo, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Laursen AH, Thune JJ, Hutchings M, Hasbak P, Kjaer A, Elming MB, Ripa RS. 123
I-MIBG imaging for detection of anthracycline-induced cardiomyopathy. Clin Physiol Funct Imaging 2017; 38:176-185. [DOI: 10.1111/cpf.12419] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 01/19/2017] [Indexed: 01/08/2023]
Affiliation(s)
- Adam H. Laursen
- Department of Haematology; Rigshospitalet; Copenhagen Denmark
| | | | | | - Philip Hasbak
- Department of Clinical Physiology; Nuclear Medicine & PET and Cluster for Molecular Imaging; Rigshospitalet and University of Copenhagen; Copenhagen Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology; Nuclear Medicine & PET and Cluster for Molecular Imaging; Rigshospitalet and University of Copenhagen; Copenhagen Denmark
| | - Marie B. Elming
- Department of Cardiology; Rigshospitalet; Copenhagen Denmark
| | - Rasmus S. Ripa
- Department of Clinical Physiology; Nuclear Medicine & PET and Cluster for Molecular Imaging; Rigshospitalet and University of Copenhagen; Copenhagen Denmark
| |
Collapse
|
16
|
Dos Santos MJ, da Rocha ET, Verberne HJ, da Silva ET, Aragon DC, Junior JS. Assessment of late anthracycline-induced cardiotoxicity by 123I-mIBG cardiac scintigraphy in patients treated during childhood and adolescence. J Nucl Cardiol 2017; 24:256-264. [PMID: 26601670 DOI: 10.1007/s12350-015-0309-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 10/01/2015] [Indexed: 12/26/2022]
Abstract
PURPOSE The goal of this study was to evaluate late cardiotoxic effects of anthracyclines (ATC) by evaluating cardiac sympathetic activity in a cohort of asymptomatic patients previously treated with ATC for childhood cancers. METHODS We studied 89 asymptomatic patients previously treated with ATC with a normal echocardiogram (49 men and 40 women) and a control group of 40 healthy individuals (26 men and 14 women). Both groups underwent planar myocardial 123I-meta-iodobenzylguanidine scintigraphy (123I-mIBG). From these images, the early and late heart-to-mediastinum (H/M) ratio and washout rate (WR) were assessed. RESULTS The mean survival at the time of the 123I-mIBG scintigraphy was 5.3 ± 3.4 years. Patients treated with ATC had a lower but clinical normal left ventricular ejection fraction (LVEF) compared to controls (60.44 ± 6.5 vs 64.1 ± 6.0%, P < 0.01). Both the late H/M ratio and WR were not able to discriminate ATC treated patients from controls. The cumulative ATC dose was the only independent predictor of the LVEF, explaining approximately 12% of the variation in LVEF (P = 0.01). CONCLUSIONS Although the pathophysiology behind ATC cardiotoxicity is most likely multifactorial, myocardial sympathetic activity is not associated with a reduction in LVEF 5-years after completion of chemotherapy.
Collapse
Affiliation(s)
- Marcelo José Dos Santos
- Department of Nuclear Medicine, Pio XII Foundation, Barretos Cancer Hospital, Barretos, São Paulo, Zip code: 14.784-700, Brazil.
| | - Euclides Timóteo da Rocha
- Department of Nuclear Medicine, Pio XII Foundation, Barretos Cancer Hospital, Barretos, São Paulo, Zip code: 14.784-700, Brazil
| | - Hein J Verberne
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | | | - Davi Casale Aragon
- Department of Pediatrics, Medical School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - José Soares Junior
- The Heart Institute (Incor), Medical School of São Paulo, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
17
|
Russell RR, Alexander J, Jain D, Poornima IG, Srivastava AV, Storozynsky E, Schwartz RG. The role and clinical effectiveness of multimodality imaging in the management of cardiac complications of cancer and cancer therapy. J Nucl Cardiol 2016; 23:856-84. [PMID: 27251147 DOI: 10.1007/s12350-016-0538-8] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 12/24/2022]
Abstract
With the increasing number of individuals living with a current or prior diagnosis of cancer, it is important for the cardiovascular specialist to recognize the various complications of cancer and its therapy on the cardiovascular system. This is true not only for established cancer therapies, such as anthracyclines, that have well established cardiovascular toxicities, but also for the new targeted therapies that can have "off target" effects in the heart and vessels. The purpose of this informational statement is to provide cardiologists, cardiac imaging specialists, cardio-oncologists, and oncologists an understanding of how multimodality imaging may be used in the diagnosis and management of the cardiovascular complications of cancer therapy. In addition, this document is meant to provide useful general information concerning the cardiovascular complications of cancer and cancer therapy as well as established recommendations for the monitoring of specific cardiotoxic therapies.
Collapse
Affiliation(s)
- Raymond R Russell
- Rhode Island Cardiovascular Institute, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, 593 Eddy Street, APC 737, Providence, RI, 02903, USA.
| | - Jonathan Alexander
- Cardiology Division, Western Connecticut Medical Center at Danbury Hospital, Danbury, CT, USA
| | - Diwakar Jain
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Indu G Poornima
- Division of Cardiology, Allegheny Health Network, Pittsburgh, PA, USA
| | - Ajay V Srivastava
- Division of Cardiovascular Medicine, Scripps Clinic, La Jolla, CA, USA
| | - Eugene Storozynsky
- Cardiology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Ronald G Schwartz
- Cardiology Division, Department of Medicine, University of Rochester Medical Center, Rochester, NY, USA
- Nuclear Medicine Division, Department of Imaging Sciences, University of Rochester Medical Center, Rochester, NY, USA
| |
Collapse
|
18
|
Khouri MG, Klein MR, Velazquez EJ, Jones LW. Current and emerging modalities for detection of cardiotoxicity in cardio-oncology. Future Cardiol 2015; 11:471-84. [PMID: 26235924 DOI: 10.2217/fca.15.16] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Advancements in diagnostic tools and curative-intent therapies have improved cancer-specific survival. With prolonged survival, patients are now subject to increased aging and development of cardiovascular risk factors such that further improvements in cancer-specific mortality are at risk of being offset by increased cardiovascular mortality. Moreover, established and novel adjuvant therapies used in cancer treatment are associated with unique and varying degrees of direct as well as indirect myocardial and cardiovascular injury (i.e., cardiotoxicity). Current approaches for evaluating anticancer therapy-induced injury have limitations, particularly lack of sensitivity for early detection of subclinical cardiac and cardiovascular dysfunction. With emerging evidence suggesting early prevention and treatment can mitigate the degree of cardiotoxicity and limit interruption of life-saving cancer therapy, the importance of early detection is increasingly paramount. Newer imaging modalities, functional capacity testing and blood biomarkers have the potential to improve early detection of cardiotoxicity and reduce cardiovascular morbidity and mortality.
Collapse
Affiliation(s)
- Michel G Khouri
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Michael R Klein
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Eric J Velazquez
- Department of Medicine, Division of Cardiology, Duke University Medical Center, Durham, NC, USA
| | - Lee W Jones
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
19
|
Kongbundansuk S, Hundley WG. Noninvasive imaging of cardiovascular injury related to the treatment of cancer. JACC Cardiovasc Imaging 2014; 7:824-38. [PMID: 25124015 PMCID: PMC4183055 DOI: 10.1016/j.jcmg.2014.06.007] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Revised: 06/20/2014] [Accepted: 06/26/2014] [Indexed: 01/03/2023]
Abstract
The introduction of multiple treatments for cancer, including chemotherapeutic agents and radiation therapy, has significantly reduced cancer-related morbidity and mortality. However, these therapies can promote a variety of toxicities, among the most severe being the ones involving the cardiovascular system. Currently, for many surviving cancer patients, cardiovascular (CV) events represent the primary cause of morbidity and mortality. Recent data suggest that CV injury occurs early during cancer treatment, creating a substrate for subsequent cardiovascular events. Researchers have investigated the utility of noninvasive imaging strategies to detect the presence of CV injury during and after completion of cancer treatment because it starts early during cancer therapy, often preceding the development of chemotherapy or cancer therapeutics related cardiac dysfunction. In this State-of-the-Art Paper, we review the utility of current clinical and investigative CV noninvasive modalities for the identification and characterization of cancer treatment-related CV toxicity.
Collapse
Affiliation(s)
- Suwat Kongbundansuk
- Department of Internal Medicine (Section on Cardiology), Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - W Gregory Hundley
- Department of Internal Medicine (Section on Cardiology), Wake Forest School of Medicine, Winston-Salem, North Carolina; Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina.
| |
Collapse
|
20
|
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: 1.7] [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
|
21
|
Ong DS, Scherrer-Crosbie M, Coelho-Filho O, Francis SA, Neilan TG. Imaging methods for detection of chemotherapy-associated cardiotoxicity and dysfunction. Expert Rev Cardiovasc Ther 2014; 12:487-97. [DOI: 10.1586/14779072.2014.893824] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
22
|
Schwartz RG, Jain D, Storozynsky E. Traditional and novel methods to assess and prevent chemotherapy-related cardiac dysfunction noninvasively. J Nucl Cardiol 2013; 20:443-64. [PMID: 23572315 DOI: 10.1007/s12350-013-9707-1] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The field of cardio-oncology is challenged to address an ever greater spectrum of cardiotoxicity associated with combination chemotherapy, greater dose intensity, extremes of age, and enhanced patient survival which exposes more protracted risk of developing congestive heart failure (CHF). Recent reports of chemotherapy-induced hypertension as a common adverse effect of angiogenesis inhibitors and immunosuppressants clarify the need for routine blood pressure (BP) monitoring and guideline-based management of hypertension as an integral strategy to preserve LV function. Serial monitoring of radionuclide left ventricular ejection fraction (LVEF) in adults and echocardiography in children continues to provide outcome based, cost-effective prevention of CHF in high risk patients receiving chemotherapy. To optimize treatment and monitoring strategies to eliminate late-onset LV dysfunction and CHF, traditional and novel candidate methods for assessment of chemotherapy-induced LV dysfunction are reviewed. These include serial assessment of LV volume indices by gated SPECT ERNA and gated SPECT MPI, 3D echocardiography and contrast 2D echocardiography; longitudinal strain imaging, diastolic functional parameters, (123)I-MIBG, (111)In-Antimyosin antibody imaging, and (99m)Tc-Annexin V apoptosis imaging, biomarkers including troponins and BNP; genetic markers, and both functional and tissue characterization techniques with T1 weighted and T2 weighted images with cardiac magnetic resonance imaging (CMR). In our quest to optimize strategies for long-term cancer survival and prevention of CHF for patients receiving chemotherapy, rigorous modality and guideline-specific clinical outcome trials are required. A new multi-modality monitoring approach is proposed, which integrates evidence-based strengths of CMR, echocardiography, ERNA, biomarkers, and BP management for surveillance and validation of cardiotoxicity and prevention of clinical heart failure in patients receiving a broad spectrum of cancer therapies.
Collapse
|
23
|
Abstract
Radionuclide cardiac imaging has potential to assess underlying molecular, electrophysiologic, and pathophysiologic processes of cardiac disease. An area of current interest is cardiac autonomic innervation imaging with a radiotracer such as (123)I-meta-iodobenzylguanidine ((123)I-mIBG), a norepinephrine analogue. Cardiac (123)I-mIBG uptake can be assessed by planar and SPECT techniques, involving determination of global uptake by a heart-to-mediastinal ratio, tracer washout between early and delayed images, and focal defects on tomographic images. Cardiac (123)I-mIBG findings have consistently been shown to correlate strongly with heart failure severity, pre-disposition to cardiac arrhythmias, and poor prognosis independent of conventional clinical, laboratory, and image parameters. (123)I-mIBG imaging promises to help monitor a patient's clinical course and response to therapy, showing potential to help select patients for an ICD and other advanced therapies better than current methods. Autonomic imaging also appears to help diagnose ischemic heart disease and identify higher risk, as well as risk-stratify patients with diabetes. Although more investigations in larger populations are needed to strengthen prior findings and influence modifications of clinical guidelines, cardiac (123)I-mIBG imaging shows promise as an emerging technique for recognizing and following potentially life-threatening conditions, as well as improving our understanding of the pathophysiology of various diseases.
Collapse
Affiliation(s)
- Mark I Travin
- Division of Nuclear Medicine, Department of Radiology, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East-210th Street, Bronx, NY 10467-2490, USA.
| |
Collapse
|
24
|
Jiji RS, Kramer CM, Salerno M. Non-invasive imaging and monitoring cardiotoxicity of cancer therapeutic drugs. J Nucl Cardiol 2012; 19:377-88. [PMID: 22351492 PMCID: PMC3314105 DOI: 10.1007/s12350-012-9512-2] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Cardiotoxicity due to administration of cancer therapeutic agents such as anthracyclines and herceptin are well described. Established guidelines to screen for chemotherapy-related cardiotoxicity (CRC) are primarily based on serial assessment of left ventricular (LV) ejection fraction (EF). However, other parameters such as LV volume, diastolic function, and strain may also be useful in screening for cardiotoxicity. More recent advances in molecular imaging of apoptosis and tissue characterization by cardiac MRI are techniques which might allow early detection of patients at high risk for developing cardiotoxicity prior to a drop in EF. This comprehensive multi-modality review will discuss both the current established imaging techniques as well as the emerging technologies which may revolutionize the future of screening and evaluation for CRC.
Collapse
Affiliation(s)
- Ronny S Jiji
- Cardiovascular Division, Departments of Medicine and Radiology and the Cardiovascular Imaging Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | |
Collapse
|
25
|
Abstract
Cardiac autonomic innervation plays a key role in maintaining hemodynamic and electrophysiologic harmony. Cardiac sympathetic function is adversely altered in many disease states, such as congestive heart failure, myocardial ischemia, and diabetes. (123)I-mIBG, a sympathetic neurotransmitter radionuclide analog, aids in the detection of sympathetic innervation abnormalities and can be imaged with planar and single-photon emission computed tomographic techniques. Cardiac (123)I-mIBG uptake can be assessed by the heart mediastinal ratio (H/M), tracer washout rate, and focal uptake defects. These parameters have been widely studied and shown to correlate strongly and independently with congestive heart failure progression, cardiac arrhythmias, cardiac death, and all-cause mortality. There is accumulating evidence that (123)I-mIBG imaging can help to monitor a patient's clinical course and response to therapy. The ability to predict potentially lethal ventricular arrhythmias promises to help more accurately select patients for implantable cardioverter defibrillators, limiting unnecessary devices and identifying additional patients at risk who do not meet current guidelines. (123)I-mIBG shows potential to help determine whether greater risk and usually more expensive ventricular assist device therapies or cardiac transplantation might be needed. Although more investigation in larger populations is needed to strengthen previous findings, cardiac (123)I-mIBG imaging shows promise as a new technique for recognizing and following potentially life-threatening cardiac conditions.
Collapse
Affiliation(s)
- Amala Chirumamilla
- Department of Nuclear Medicine and Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | | |
Collapse
|
26
|
de Geus-Oei LF, Mavinkurve-Groothuis AMC, Bellersen L, Gotthardt M, Oyen WJG, Kapusta L, van Laarhoven HWM. Scintigraphic techniques for early detection of cancer treatment-induced cardiotoxicity. J Nucl Med 2011; 41:170-81. [PMID: 21421717 DOI: 10.2967/jnumed.110.082784] [Citation(s) in RCA: 79] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
New antitumor agents have resulted in significant survival benefits for cancer patients. However, several agents may have serious cardiovascular side effects. Left ventricular ejection fraction measurement by (99m)Tc multigated radionuclide angiography is regarded as the gold standard to measure cardiotoxicity in adult patients. It identifies left ventricular dysfunction with high reproducibility and low interobserver variability. A decrease in left ventricular ejection fraction, however, is a relatively late manifestation of myocardial damage. Nuclear cardiologic techniques that visualize pathophysiologic processes at the tissue level could detect myocardial injury at an earlier stage. These techniques may give the opportunity for timely intervention to prevent further damage and could provide insights into the mechanisms and pathophysiology of cardiotoxicity caused by anticancer agents. This review provides an overview of past, current, and promising newly developed radiopharmaceuticals and describes the role and recent advances of scintigraphic techniques to measure cardiotoxicity. Both first-order functional imaging techniques (visualizing mechanical [pump] function), such as (99m)Tc multigated radionuclide angiography and (99m)Tc gated blood-pool SPECT, and third-order functional imaging techniques (visualizing pathophysiologic and neurophysiologic processes at the tissue level) are discussed. Third-order functional imaging techniques comprise (123)I-metaiodobenzylguanidine scintigraphy, which images the efferent sympathetic nervous innervations; sympathetic neuronal PET, with its wide range of tracers; (111)In-antimyosin, which is a specific marker for myocardial cell injury and necrosis; (99m)Tc-annexin V scintigraphy, which visualizes apoptosis and cell death; fatty-acid-use scintigraphy, which visualizes the storage of free fatty acids in the lipid pool of the cytosol (which can be impaired by cardiotoxic agents); and (111)In-trastuzumab imaging, to study trastuzumab targeting to the myocardium. To define the prognostic importance and clinical value of each of these functional imaging techniques, prospective clinical trials are warranted.
Collapse
Affiliation(s)
- Lioe-Fee de Geus-Oei
- Department of Nuclear Medicine, Radboud University Nijmegen Medical Centre, Nijmegen, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
27
|
Das J, Ghosh J, Manna P, Sil PC. Taurine suppresses doxorubicin-triggered oxidative stress and cardiac apoptosis in rat via up-regulation of PI3-K/Akt and inhibition of p53, p38-JNK. Biochem Pharmacol 2011; 81:891-909. [PMID: 21295553 DOI: 10.1016/j.bcp.2011.01.008] [Citation(s) in RCA: 159] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Revised: 12/21/2010] [Accepted: 01/18/2011] [Indexed: 12/16/2022]
Abstract
The objective of the present study was to investigate the signaling mechanisms involved in the beneficial role of taurine against doxorubicin-induced cardiac oxidative stress. Male rats were administered doxorubicin. Hearts were collected 3 weeks after the last dose of doxorubicin and were analyzed. Doxorubicin administration retarded the growth of the body and the heart and caused injury in the cardiac tissue because of increased oxidative stress. Similar experiments with doxorubicin showed reduced cell viability, increased ROS generation, intracellular Ca(2+) and DNA fragmentation, disrupted mitochondrial membrane potential and apoptotic cell death in primary cultured neonatal rat cardiomyocytes. Signal transduction studies showed that doxorubicin increased p53, JNK, p38 and NFκB phosphorylation; decreased the levels of phospho ERK and Akt; disturbed the Bcl-2 family protein balance; activated caspase 12, caspase 9 and caspase 3; and induced cleavage of the PARP protein. However, taurine treatment or cardiomyocyte incubation with taurine suppressed all of the adverse effects of doxorubicin. Studies with several inhibitors, including PS-1145 (an IKK inhibitor), SP600125 (a JNK inhibitor), SB203580 (a p38 inhibitor) and LY294002 (a PI3-K/Akt inhibitor), demonstrated that the mechanism of taurine-induced cardio protection involves activation of specific survival signals and PI3-K/Akt as well as the inhibition of p53, JNK, p38 and NFκB. These novel findings suggest that taurine might have clinical implications for the prevention of doxorubicin-induced cardiac oxidative stress.
Collapse
Affiliation(s)
- Joydeep Das
- Division of Molecular Medicine, Bose Institute, Kolkata, West Bengal, India
| | | | | | | |
Collapse
|
28
|
Abstract
Cardiac autonomic function plays a crucial role in health and disease, with abnormalities both reflecting the severity of the disease and contributing specifically to clinical deterioration and poor prognosis. Radiotracer analogs of the sympathetic mediator norepinephrine have been investigated extensively, and are at the brink of potential widespread clinical use. The most widely studied SPECT tracer, I-123 metaiodobenzylguanidine ((123)I-mIBG) has consistently shown a strong, independent ability to risk stratify patients with advanced congestive heart failure. Increased global cardiac uptake appears to have a high negative predictive value in terms of cardiac events, especially death and arrhythmias, and therefore and may have a role in guiding therapy, particularly by helping to better select patients unresponsive to conventional medical therapies who would benefit from device therapies such as an ICD (implantable cardioverter defibrillator), CRT (cardiac resynchronization therapy), LVAD (left ventricular assist device), or cardiac transplantation. Cardiac autonomic imaging with SPECT and PET tracers also shows potential to assess patients following cardiac transplant, those with primary arrhythmic condition, coronary artery disease, diabetes mellitus, and during cardiotoxic chemotherapy. Radiotracer imaging of cardiac autonomic function allows visualization and quantitative measurements of underlying molecular aspects of cardiac disease, and should therefore provide a perspective that other cardiac tests cannot.
Collapse
Affiliation(s)
- Sang Yong Ji
- Department of Nuclear Medicine, Montefiore Medical Center, Albert Einstein College of Medicine, 111 East-210th Street, Bronx, NY 10467-2490, USA
| | | |
Collapse
|
29
|
|
30
|
Altena R, Perik PJ, van Veldhuisen DJ, de Vries EG, Gietema JA. Cardiovascular toxicity caused by cancer treatment: strategies for early detection. Lancet Oncol 2009; 10:391-9. [PMID: 19341970 DOI: 10.1016/s1470-2045(09)70042-7] [Citation(s) in RCA: 178] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cardiovascular toxicity is one of the most devastating complications of cancer treatment and can arise during or shortly after treatment, or even several years later. Identification of the left ventricular ejection fraction (LVEF) is the most common method to screen for toxic effects on the heart; however, this approach underestimates cardiac damage and additional strategies for the monitoring of treatment-induced cardiotoxicity are being explored. Guidelines for monitoring have been formulated for several cancer treatments; however, appropriate underlying evidence is still largely absent. In this Review, we summarise conventional and contemporary methods for early detection of cardiotoxicity and designate a level of evidence for the basis of each method.
Collapse
Affiliation(s)
- Renske Altena
- Department of Medical Oncology, University of Groningen and University Medical Centre Groningen, Groningen, Netherlands
| | | | | | | | | |
Collapse
|
31
|
Choi HJ, Seon MR, Lim SS, Kim JS, Chun HS, Park JHY. Hexane/ethanol extract of Glycyrrhiza uralensis licorice suppresses doxorubicin-induced apoptosis in H9c2 rat cardiac myoblasts. Exp Biol Med (Maywood) 2008; 233:1554-60. [PMID: 18849542 DOI: 10.3181/0807-rm-221] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Doxorubicin (DOX) is an anthracycline antibiotic, and has been recognized as one of the most effective anti-neoplastic agents in cancer chemotherapy. However, its usefulness is limited by its profound cardiotoxicity. Licorice is one of the most frequently prescribed agents in traditional herbal medicine, and is also employed as a natural sweetening additive. In traditional Chinese medicine, licorice root is added to a variety of herbal preparations to detoxify the effects of the other herbs in the preparation. In the present study, we explored the possibility that Glycyrrhiza uralensis licorice may alleviate DOX-induced cardiotoxicity. The hexane/ethanol extract of Glycyrrhiza uralensis (HEGU), which lacks glycyrrhizin, was prepared because glycyrrhizin intake has previously been reported to induce hypertension. In an effort to determine whether HEGU ameliorates DOX-induced cytotoxicity in H9c2 rat cardiac myoblasts, the cells were pretreated with 0-15 mg/L HEGU, then treated with doxorubicin. The pretreatment of cells with HEGU resulted in a significant mitigation of DOX-induced reductions in cell numbers (34 +/- 7%) and increases in apoptosis (53 +/- 1%). The Western blot analysis of cell lysates showed that HEGU suppressed DOX-induced increases in the levels of p53, phospho-p53 (Ser 15), and Bax. In addition, HEGU induced an increase in the levels of Bcl-xL, regardless of DOX-treatment. HEGU inhibited the DOX-induced cleavage of caspases 9, 3, and 7, as well as DOX-induced poly(ADP-ribose) polymerase cleavage. Furthermore, HEGU caused reductions in the viable cell numbers of HT-29 human colon cancer cells (IC50 = 10.7 +/- 0.3 mg/L), MDA-MB-231 human breast cancer cells (IC50 = 7.5 +/- 0.1 mg/L), and DU145 human prostate cancer cells (IC50 = 4.7 +/- 0.5 mg/L). HEGU augmented DOX-induced reductions in the viability of DU145 cells (15 +/- 1%). These results indicate that HEGU may potentially be an effective agent for the alleviation of DOX-induced cardiotoxicity.
Collapse
Affiliation(s)
- Hyun Ju Choi
- Department of Food Science and Nutrition, Hallym University, Chuncheon 200-702, South Korea
| | | | | | | | | | | |
Collapse
|
32
|
Deng S, Kulle B, Hosseini M, Schlüter G, Hasenfuss G, Wojnowski L, Schmidt A. Dystrophin-deficiency increases the susceptibility to doxorubicin-induced cardiotoxicity. Eur J Heart Fail 2007; 9:986-94. [PMID: 17888722 DOI: 10.1016/j.ejheart.2007.07.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/22/2006] [Revised: 06/15/2007] [Accepted: 07/17/2007] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND AIM The clinical use of doxorubicin (DOX) and other anthracyclines is limited by a dosage-dependent cardiotoxicity, which can lead to cardiomyopathy. The role of the individual genetic makeup in this disorder is poorly understood. Alterations in genes encoding cardiac cytoskeleton or sarcolemma proteins may increase the susceptibility to doxorubicin-related cardiotoxicity. METHODS Female dystrophin-deficient mice (MDX) and age-matched wild-type mice underwent chronic treatment with doxorubicin. Cardiac function and tissue damage were assessed by echocardiography and histopathology, respectively. Gene expression changes were investigated using microarrays. RESULTS DOX treatment resulted in mortality, cardiac insufficiency, and cardiac interstitial fibrosis. These alterations were more pronounced in DOX-treated MDX mice than in DOX-treated wild-type mice. Changes in gene expression were more numerous in MDX mice, including genes involved in cell adhesion, oxidative stress, cytoskeleton organization, inflammatory and immune response and cell death. CONCLUSIONS Dystrophin deficiency facilitates the development and progression of doxorubicin-induced cardiac injury. The underlying mechanisms may involve changes in cell adhesion, in cytoskeleton, as well as in inflammatory and immune responses. Genetic variants of cytoskeletal proteins in humans may affect the individual susceptibility to doxorubicin. Cardiotoxic drugs may accelerate the manifestation of pre-clinical cardiomyopathies caused by deficiencies in cytoskeletal or sarcolemma proteins.
Collapse
Affiliation(s)
- Shiwei Deng
- Department of Pharmacology, Johannes Gutenberg University, 55101 Mainz, Germany
| | | | | | | | | | | | | |
Collapse
|
33
|
Rudzinski T, Ciesielczyk M, Religa W, Bednarkiewicz Z, Krzeminska-Pakula M. Doxorubicin-induced ventricular arrhythmia treated by implantation of an automatic cardioverter-defibrillator. ACTA ACUST UNITED AC 2007; 9:278-80. [PMID: 17383986 DOI: 10.1093/europace/eum033] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Anthracyclines are a group of potent antitumour agents and cardiotoxicity is an important factor limiting their therapeutic effectiveness. Although cardiomyopathy is the most widely recognized type of cardiotoxic reaction, early arrhythmia following anthracycline administration may be of clinical significance as well. We report a case of ventricular tachycardia causing cardiac arrest in a female treated with doxorubicin as adjuvant therapy of breast cancer. Due to recurrence of the arrhythmia and a desire to continue chemotherapy, an automatic cardioverter-defibrillator was implanted with excellent effect.
Collapse
Affiliation(s)
- Tomasz Rudzinski
- 2nd Department of Cardiology, Medical University of Lodz, Kniaziewicza 1/5, 91-347 Lodz, Poland.
| | | | | | | | | |
Collapse
|
34
|
Panjrath GS, Jain D. Monitoring chemotherapy-induced cardiotoxicity: role of cardiac nuclear imaging. J Nucl Cardiol 2007; 13:415-26. [PMID: 16750786 DOI: 10.1016/j.nuclcard.2006.03.002] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Cardiotoxicity may result from a range of chemotherapeutic agents. The prevalence of cardiotoxicity from certain cytotoxic agents is reported to be significantly high. In addition to serious side effects and increased long-lasting morbidity and mortality, dose limitation and suboptimal usage is an important adverse effect. Nuclear cardiac imaging has played a quintessential and important role in identifying patients at risk and in the prevention and reduction of cardiac injury resulting from cytotoxic agents. Despite exploring a number of other diagnostic imaging or biochemical tools for identification of cardiac injury, nuclear cardiac imaging in the form of radionuclide angiocardiography continues to be the most suitable and cost-effective tool for reducing the prevalence of cases of cardiac dysfunction resulting from chemotherapy. This article reviews the prevalence, mechanisms, and prevention strategies for cardiotoxicity associated with some of the commonly known cytotoxic agents and the role of nuclear cardiac imaging in its monitoring and prevention, along with recent advances in this area.
Collapse
Affiliation(s)
- Gurusher Singh Panjrath
- Department of Internal Medicine, St. Luke's-Roosevelt Hospital Center, New York, NY 10025, USA.
| | | |
Collapse
|
35
|
Abstract
The use of anthracyclines is limited by dose-dependent cardiotoxicity. Three forms of anthracycline cardiotoxicity are described; an immediate pericarditis-myocarditis syndrome, an early onset chronic progressive CHF developing during or shortly after therapy and late-onset cardiotoxicity presenting years following treatment. A number of risk factors have been reported, including; cumulative dose, administration schedule, mediastinal radiotherapy, old and young age, concurrent cardiovascular disease, combination therapy, gender, ethnicity and chromosomal abnormalities. Evaluation of left ventricular ejection fraction has been widely adopted as a means of monitoring and assessing anthracycline-induced cardiotoxicity. Biochemical markers and other techniques, such as endomyocardial biopsy, metaiodobenzylguanidine and indium-111-antimyosin scintigraphy are not routinely used. Methods employed to prevent cardiotoxicity include cumulative dose limitation, alteration of administration schedule, anthracycline analogues, liposomal formulations and the cardioprotective agent, dexrazoxane. With the growing number of paediatric malignancy survivors and the increasing use of anthracyclines in the adjuvant treatment of breast cancer, the cardiotoxicity associated with these agents will remain a formidable issue for physicians. Further work is required to identify patients at increased risk of cardiotoxicity and to develop novel methods of protecting and treating this adverse effect.
Collapse
Affiliation(s)
- Robin L Jones
- Royal Marsden Hospital, Department of Medicine, Fulham Road, London, SW3 6JJ, UK.
| | | | | |
Collapse
|
36
|
Abstract
Nuclear cardiology has made significant advances since the first reports of planar scintigraphy for the evaluation of left ventricular perfusion and function. While the current "state of the art" of gated myocardial perfusion single-photon emission computed tomographic (SPECT) imaging offers invaluable diagnostic and prognostic information for the evaluation of patients with suspected or known coronary artery disease (CAD), advances in the cellular and molecular biology of the cardiovascular system have helped to usher in a new modality in nuclear cardiology, namely, molecular imaging. In this review, we will discuss the current state of the art in nuclear cardiology, which includes SPECT and positron emission tomographic evaluation of myocardial perfusion, evaluation of left ventricular function by gated myocardial perfusion SPECT and gated blood pool SPECT, and the evaluation of myocardial viability with PET and SPECT methods. In addition, we will discuss the future of nuclear cardiology and the role that molecular imaging will play in the early detection of CAD at the level of the vulnerable plaque, the evaluation of cardiac remodeling, and monitoring of important new therapies including gene therapy and stem cell therapy.
Collapse
|
37
|
Ueno M, Kakinuma Y, Yuhki KI, Murakoshi N, Iemitsu M, Miyauchi T, Yamaguchi I. Doxorubicin induces apoptosis by activation of caspase-3 in cultured cardiomyocytes in vitro and rat cardiac ventricles in vivo. J Pharmacol Sci 2006; 101:151-8. [PMID: 16766856 DOI: 10.1254/jphs.fp0050980] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Doxorubicin (DOX) is widely used to treat patients suffering from cancer, but the usage for patients is limited because of the dose-dependent cardiotoxicity. We hypothesized that DOX induces apoptosis through caspase activation in cardiomyocytes, and we examined this hypothesis using both rat primary cultured cardiomyocytes and rat hearts from an animal model. Cardiomyocytes were treated with DOX for 24 h. The activity of caspase-3 was significantly increased by DOX treatment. In rats with DOX injected intravenously once a week for 5 weeks, left ventricular fractional shortening evaluated by echocardiography was significantly decreased at age 14 weeks, 2 weeks after the end of DOX-administration. At 16 weeks of age, endothelin-1 mRNA and atrial natriuretic peptide mRNA were also significantly increased, likewise, and TUNEL positive cells were significantly increased in the ventricles of DOX-treated rats. The activity of caspase-3 in the ventricles was also significantly increased compared to that of untreated rats at 16 weeks. However, the activity of caspase-8 and the expression level of Fas-ligand mRNA were comparable with those of the untreated rats. In conclusion, DOX induces apoptosis through the activation of caspase-3, suggesting that apoptosis has an important role in the progression of cardiomyopathy due to DOX.
Collapse
Affiliation(s)
- Michihiko Ueno
- Cardiovascular Division, Department of Internal Medicine, Institute of Clinical Medicine, Tsukaba, Ibaraki, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
The sympathetic nervous system has great influence on cardiovascular physiology. Cardiac neurotransmission single photon emission computed tomography (SPECT) imaging allows in vivo noninvasive assessment of presynaptic reuptake and storage of neurotransmitters, which offers characterization of the cardiac neuronal function in different diseases of the heart and other altered metabolic or functional conditions. Therefore assessment of the integrity of cardiac sympathetic innervation may help in the diagnosis of these disorders, as well as in prognostication, and will result in better therapy and outcome. At present, the most widely available SPECT tracer by which to assess cardiac neurotransmission is metaiodobenzylguanidine labeled with iodine 123. This article focuses on reviewing the characteristics of cardiac SPECT imaging with I-123 metaiodobenzylguanidine and its role in the assessment of pathophysiologic changes during relevant clinical conditions.
Collapse
Affiliation(s)
- Albert Flotats
- Nuclear Medicine Department, Hospital de la Santa Creu i Sant Pau, 08025 Barcelona, Spain.
| | | |
Collapse
|
39
|
Taniyama Y, Walsh K. Elevated myocardial Akt signaling ameliorates doxorubicin-induced congestive heart failure and promotes heart growth. J Mol Cell Cardiol 2002; 34:1241-7. [PMID: 12392981 DOI: 10.1006/jmcc.2002.2068] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Doxorubicin is a chemotherapeutic agent that can induce cardiotoxicity and congestive heart failure (CHF). In this study we tested whether intracoronary Akt1 gene delivery could inhibit doxorubicin-induced CHF. Saline or a replication defective adenoviral vector expressing constitutively-active Akt1 (myrAkt) or beta-galactosidase (betagal) was delivered to the myocardium of 8 week old rats one day prior to initiating doxorubicin administration. In animals receiving saline or betagal, doxorubicin resulted in significant decreases in cardiac function and retarded post-natal heart growth at the 5 weeks time point. In contrast, transduction of myrAkt protected hearts against doxorubicin-induced decreases in fractional shortening and cardiac index, and improved left ventricular function at 5 weeks time point. Delivery of myrAkt also reversed the doxorubicin-induced reduction in post-natal heart growth and diminished lung edema. These data show that myocardial Akt can inhibit doxorubicin-induced reductions in cardiac function and growth, suggesting that manipulation of this signaling pathway may have utility for the treatment of congestive heart failure.
Collapse
Affiliation(s)
- Yoshiaki Taniyama
- Molecular Cardiology, Whitaker Cardiovascular Institute, Boston University School of Medicine, 715 Albany Street, Boston, MA 02118, USA
| | | |
Collapse
|
40
|
Goethals I, De Winter O, De Bondt P, De Sutter J, Dierckx R, Van De Wiele C. The clinical value of nuclear medicine in the assessment of irradiation-induced and anthracycline-associated cardiac damage. Ann Oncol 2002; 13:1331-9. [PMID: 12196357 DOI: 10.1093/annonc/mdf318] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Two groups of patients, those treated for Hodgkin's disease and breast cancer, are particularly at risk of developing late myocardial damage, since radiotherapy (RT) techniques for both patient groups may include (large) parts of the heart, and adjuvant systemic therapy is frequently administered to these patients, in particular anthracycline-containing chemotherapy. Available literature on the monitoring and prediction of RT-induced and anthracycline-associated cardiac damage using nuclear medicine techniques is presented. Based on relevant studies, the risk of overall cardiac disease post-RT and overt congestive heart failure during anthracycline-containing chemotherapy is probably low. Conventional nuclear medicine imaging, i.e. myocardial perfusion scintigraphy, may be of complementary use to echocardiographical evaluation for routine follow-up after RT with modern techniques, in a subgroup of patients with known cardiovascular risk factors. Left ventricle ejection fraction (LVEF) measurements, as assessed by radionuclide angiography for the monitoring of anthracycline-associated cardiac injury, are not very sensitive and early detection will probably be enhanced by combining LVEF measurements with other cardiac function parameters. Also, it may be expected that nuclear medicine techniques using molecular radioligands will constitute an essential future step in the evaluation of subclinical cardiac injury afforded by the combined effect of RT and cardiotoxic chemotherapy.
Collapse
Affiliation(s)
- I Goethals
- Division of Nuclear Medicine, Department of Cardiology, Ghent University Hospital, Ghent, Belgium.
| | | | | | | | | | | |
Collapse
|
41
|
Postma A, Elzenga NJ, Haaksma J, Schasfoort-Van Leeuwen MJM, Kamps WA, Bink-Boelkens MTE. Cardiac status in bone tumor survivors up to nearly 19 years after treatment with doxorubicin: a longitudinal study. MEDICAL AND PEDIATRIC ONCOLOGY 2002; 39:86-92. [PMID: 12116055 DOI: 10.1002/mpo.10074] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
BACKGROUND Longitudinal assessment of cardiac toxicity in anthracycline-treated long-term bone tumor survivors. PROCEDURES Cardiac status was assessed in 29 patients 14.1 (range 7-18.7) years after treatment with doxorubicin (DOXO) 360 mg/m(2) (median 225-550). The median age of the patients at the time of the study was 32.5 years (range 19.7-52). The evaluation consisted of an electrocardiogram (ECG), 24-hr ambulatory ECG with analysis of heart rate variability (HRV) and echocardiography. The results were compared to those of a study of the same patients that was performed 5 years earlier 8.9 years (range 2.3-14.1) after treatment. [Postma et al.: Med Pediatr Oncol 26:230-237, 1996] RESULTS We found no progression of ECG abnormalities, arrhythmias, or echocardiographic abnormalities. Females were at risk for reduced contractility (P = 0.006). HRV was significantly reduced compared to age- and sex-matched controls and compared to the previous results. CONCLUSIONS Anthracycline-related late echocardiographic abnormalities and arrhythmias detected 8.9 years after treatment, showed no further deterioration with ongoing follow-up. However, there was a significant reduction of HRV. This suggests that HRV might be a sensitive test for detection of anthracycline-induced cardiac toxicity.
Collapse
Affiliation(s)
- A Postma
- Department of Pediatrics, Division of Pediatric Oncology/Hematology, University of Groningen/University Hospital, Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
42
|
Brown HR, Ni H, Benavides G, Yoon L, Hyder K, Giridhar J, Gardner G, Tyler RD, Morgan KT. Correlation of simultaneous differential gene expression in the blood and heart with known mechanisms of adriamycin-induced cardiomyopathy in the rat. Toxicol Pathol 2002; 30:452-69. [PMID: 12187937 DOI: 10.1080/01926230290105604] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
As the genomes of mammalian species become sequenced and gene functions are ascribed, the use of differential gene expression (DGE) to evaluate organ function will become common in the experimental evaluation of new drug therapies. The ability to translate this technology into useful information for human exposures depends on tissue sampling that is impractical or generally not possible in man. The possibility that the DGE of nucleated cells, reticulocytes, or platelets in blood may present the necessary link with target organ toxicity provides an opportunity to correlate preclinical with clinical outcomes. Adriamycin is highly effective alone and more frequently in combination with other chemotherapeutic agents in the treatment of a variety of susceptible malignancies. Adriamycin-induced cardiomyopathy was examined as an endpoint to measure the utility of DOE on whole blood as a predictor of cardiac toxicity. Statistically significant gene changes were observed between relevant blood and cardiac gene profiles that corroborated the accepted mechanisms of toxicity (oxidative stress, effects on carnitine transport, DNA intercalation). There were, however, clear indications that other target organs (bone marrow and intestinal tract) were affected. The divergent expression of some genes between the blood and the heart on day 7 may also indicate the timing and mechanism of development of the cardiomyopathy and confirm current therapeutic approaches for its prevention. The data demonstrate that whole blood gene expression particularly in relation to oxidative stress, in conjunction with standard hematology and clinical chemistry, may be useful in monitoring and predicting cardiac damage secondary to adriamycin administration. Appendices A & B, referenced in this paper, are not printed in this issue of Toxicologic Pathology. They are available as downloadable text files at http://taylorandfrancis.metapress.com/openurl.asp?genre=journal&issn=0192-6233. To access them, click on the issue link for 30(4), then select this article. A download option appears at the bottom of this abstract. In order to access the full article online, you must either have an individual subscription or a member subscription accessed through www.toxpath.org.
Collapse
Affiliation(s)
- H Roger Brown
- Drug Safety, GlaxoSmithKline, Inc, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Nousiainen T, Jantunen E, Vanninen E, Hartikainen J. Early decline in left ventricular ejection fraction predicts doxorubicin cardiotoxicity in lymphoma patients. Br J Cancer 2002; 86:1697-700. [PMID: 12087452 PMCID: PMC2375393 DOI: 10.1038/sj.bjc.6600346] [Citation(s) in RCA: 119] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2001] [Revised: 03/05/2002] [Accepted: 04/09/2002] [Indexed: 11/30/2022] Open
Abstract
Thirty adult patients with non-Hodgkin's lymphoma were studied to evaluate prospectively the significance of early decline in left ventricular ejection fraction after low cumulative doxorubicin dose (200 mg x m(-2)) in predicting the later impairment of left ventricular function. Cardiac function was monitored with radionuclide ventriculography at baseline and after cumulative doxorubicin doses of 200, 400 and 500 mg x m(-2). Cardiotoxicity was defined as a decrease in left ventricular ejection fraction of more than 10% units to a final left ventricular ejection fraction < or =50%. Twenty-eight patients received doxorubicin > or =400 mg x m(-2) and were evaluable for cardiotoxicity. Clinical heart failure developed in two patients (7%) after a cumulative doxorubicin dose of 500 mg m(-2). Left ventricular ejection fraction decreased more than 10% absolute ejection fraction units to a final left ventricular ejection fraction < or =50% in 10 patients (36%). Left ventricular ejection fraction decreased from 56+/-1.5% to 53.6+/-1.5% (P=0.016) in patients with no cardiotoxicity, and from 60.8+/-2.4% to 41.8+/-2.0% (P<0.001) in patients with cardiotoxicity. For patients who developed cardiotoxicity, the fall in left ventricular ejection fraction after a cumulative doxorubicin dose of only 200 mg x m(-2) was highly significant (left ventricular ejection fraction 49.7+/-1.8%, P=0.001 vs baseline). In receiver operator characteristic analysis, the area under the curve for the decrease in left ventricular ejection fraction at a cumulative doxorubicin dose of 200 mg x m(-2) for predicting cardiotoxicity in all patients was 0.858. The decrease in left ventricular ejection fraction of more than 4% units after a cumulative doxorubicin dose of 200 mg x m(-2) had a 90% sensitivity and 72% specificity for predicting later cardiotoxicity. Our results show that the significant impairment of left ventricular function during doxorubicin therapy can be predicted early, already at low cumulative doxorubicin doses. This finding may be of value in identifying patients at high or low risk for the development of anthracycline cardiotoxicity.
Collapse
Affiliation(s)
- T Nousiainen
- Department of Medicine, Kuopio University Hospital and University of Kuopio, FIN 70211 Kuopio, Finland.
| | | | | | | |
Collapse
|
44
|
Affiliation(s)
- Amar D Patel
- Division of Cardiovascular Disease, Department of Medicine, University of Alabama at Birmingham, Birmingham 35294-0006, USA
| | | |
Collapse
|
45
|
Nousiainen T, Vanninen E, Jantunen E, Remes J, Kuikka J, Hartikainen J. Anthracycline-induced cardiomyopathy: long-term effects on myocardial cell integrity, cardiac adrenergic innervation and fatty acid uptake. CLINICAL PHYSIOLOGY (OXFORD, ENGLAND) 2001; 21:123-8. [PMID: 11168306 DOI: 10.1046/j.1365-2281.2001.00292.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Cardiotoxicity of anthracyclines is a clinical challenge in cancer chemotherapy. Limited data is available on the physiological mechanisms responsible for anthracycline-induced heart failure or its recovery. We studied four patients with a history of severe anthracycline-induced heart failure manifested 2-116 months earlier by using radionuclide ventriculography for the measurement of left ventricular function, indium-111-antimyosin scintigraphy for the detection of myocardial cell injury and iodine-123-metaiodobenzylguanidine (MIBG) scintigraphy for the assessment of cardiac adrenergic innervation. Myocardial perfusion and fatty acid utilization were assessed with iodine-123-paraphenyl pentadecanoid acid (pPPA) and single photon emission computed tomography (SPECT). Symptoms of congestive heart failure (CHF) were still present in two patients whereas the others were asymptomatic at the time of the study. The patients who showed complete clinical recovery had normal or near normal left ventricular ejection fraction (LVEF) (47 and 52%), whereas the patients with symptoms of heart failure had low ejection fractions (21 and 31%). All patients presented with abnormal antimyosin uptake and decreased myocardial MIBG uptake. Patients with low ejection fraction tended to have higher antimyosin uptake suggesting more severe, persistent myocyte injury. All but one patient showed normal fatty acid utilization. These data suggest that patients with a history of severe anthracycline-induced cardiomyopathy have persistent myocardial cell injury and adrenergic dysfunction up to 10 years after the development of heart failure. These findings seem to be present regardless of recovery of left ventricular function.
Collapse
Affiliation(s)
- T Nousiainen
- Department of Medicine, Kuopio University Hospital, 70211 Kuopio, Finland
| | | | | | | | | | | |
Collapse
|
46
|
Nakamura T, Ueda Y, Juan Y, Katsuda S, Takahashi H, Koh E. Fas-mediated apoptosis in adriamycin-induced cardiomyopathy in rats: In vivo study. Circulation 2000; 102:572-8. [PMID: 10920071 DOI: 10.1161/01.cir.102.5.572] [Citation(s) in RCA: 162] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND The precise molecular mechanism of Adriamycin-induced cardiomyopathy (ADR-CM) is still unknown. We address the demonstration of apoptotic myocardial cell death and the apoptosis-inducing molecules in ADR-CM induced in rats. METHODS AND RESULTS Until 8 weeks after the first administration of ADR, there was no increase in the number of labeled cells by terminal deoxynucleotidyl transferase assay (TUNEL method). Apoptotic indices increased significantly at weeks 9 and 10 in hearts of the ADR-treated group but not in those of the control group (0.42+/-0.12% versus 0.10+/-0.02% and 0.86+/-0.11% versus 0.09+/-0.04% at weeks 9 and 10, respectively). DNA ladder formation was also observed in the myocardial tissues during the late stages of the ADR-CM of rats. There was no significant difference in expression of p53 gene between the ADR group and the control group at either the message or the protein level. An overexpression of Fas antigen was shown in myocardial cells of ADR-treated hearts at weeks 9 and 10 by both Western blotting and immunofluorescent staining. Furthermore, we confirmed that neutralization of anti-Fas ligand antibody inhibited ADR-induced apoptosis. CONCLUSIONS Apoptotic cell death was observed in the hearts of ADR-CM rats, and the number of apoptotic myocardial cells increased with the deterioration of morphological findings and cardiac function, indicating that apoptosis may be an important mechanism of loss of myocardial cells and cardiac dysfunction in ADR-CM. Apoptosis in ADR-CM rats is not p53-dependent but rather is executed through a Fas-mediated pathway.
Collapse
Affiliation(s)
- T Nakamura
- Department of Pediatrics, Kanazawa Medical University, Ishikawa, Japan
| | | | | | | | | | | |
Collapse
|
47
|
Meinardi MT, van der Graaf WT, van Veldhuisen DJ, Gietema JA, de Vries EG, Sleijfer DT. Detection of anthracycline-induced cardiotoxicity. Cancer Treat Rev 1999; 25:237-47. [PMID: 10448132 DOI: 10.1053/ctrv.1999.0128] [Citation(s) in RCA: 103] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
The use of anthracyclines, a group of potent anti-cancer agents incorporated into the treatment of a wide variety of solid and haematological tumours, is limited by its cardiotoxicity that can result in congestive heart failure (CHF). The best method to detect cardiotoxicity at an early stage in order to prevent severe deterioration, is still an unsolved problem. Although endomyocardial biopsy is considered to be the most sensitive and specific test for this purpose, its use is limited by its invasiveness. In daily practice, oncologists make use of parameters of systolic function (left ventricular ejection fraction, or fractional shortening) to detect cardiotoxicity, but these methods are not able to identify cardiotoxicity at an early stage. Based on increasing knowledge into the pathophysiology of anthracycline-induced cardiotoxicity and heart failure in general, new methods including the determination of diastolic function parameters, anti-myosin scintigraphy, assessment of heart rate variability, and the determination of biochemical markers have been proposed to identify patients at risk of the development of CHF in an early stage. However, most of these newer methods have not yet been adequately evaluated to allow them to be recommended for use in routine clinical practice.
Collapse
Affiliation(s)
- M T Meinardi
- Division of Medical Oncology, Department of Internal Medicine, University Hospital Groningen, The Netherlands.
| | | | | | | | | | | |
Collapse
|
48
|
Calzas J, Lianes P, Cortés-Funes H. [Heart pathology of extracardiac origin. VII. Heart and neoplasms]. Rev Esp Cardiol 1998; 51:314-31. [PMID: 9608805 DOI: 10.1016/s0300-8932(98)74751-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Cardiac alterations of neoplastic diseases can be due to direct invasion produced by primary cardiac tumors or more frequently secondary to local compression of vascular structures by extracardiac neoplasms, such as superior vena cava syndrome. One of the most important alterations is the cardiotoxicity of anticancer treatments, either chemotherapy drugs or radiotherapy techniques. These treatments cause acute and/or chronic cardiotoxicity that the oncologist and the cardiologist must be aware of. For instance, 4.5% to 7% of patients that have been treated with anthracyclines may suffer cardiac failure in their lifetime. The pathogenesis is still not clear. There is currently a lot of research on cardioprotectors, but nowadays the only one approved by the FDA is dexrazoxane, which is used on breast cancer patients treated with adriamycin.
Collapse
Affiliation(s)
- J Calzas
- Servicio de Oncología Médica, Hospital Universitario 12 de Octubre, Madrid
| | | | | |
Collapse
|
49
|
Giantris A, Abdurrahman L, Hinkle A, Asselin B, Lipshultz SE. Anthracycline-induced cardiotoxicity in children and young adults. Crit Rev Oncol Hematol 1998; 27:53-68. [PMID: 9548017 DOI: 10.1016/s1040-8428(97)10007-5] [Citation(s) in RCA: 159] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- A Giantris
- Department of Pediatrics, Children's Hospital at Strong, University of Rochester Medical Center, NY 14642, USA
| | | | | | | | | |
Collapse
|
50
|
Valdés Olmos RA, ten Bokkel Huinink WW, Dewit LG, Hoefnagel CA, Liem IH, van Tinteren H. Iodine-123 metaiodobenzylguanidine in the assessment of late cardiac effects from cancer therapy. EUROPEAN JOURNAL OF NUCLEAR MEDICINE 1996; 23:453-8. [PMID: 8612668 DOI: 10.1007/bf01247376] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Recognition of adverse late cardiac effects from cancer therapy may enable identification of patients with risk of cardiotoxicity upon cancer retreatment. In this study the feasibility of using iodine-123 metaiodobenzylguanidine (123I-MIBG) heart scintigraphy to detect abnormalities of the myocardial adrenergic neurone function in the late period after cancer therapy was evaluated in relation to the left ventricle ejection fraction (LVEF) in 18 cancer patients: 11 had undergone thoracic irradiation involving the heart, in five cases in combination with anthracycline therapy, 11-228 months (median 60 months) before radionuclide tests, while seven had not received previous anthracycline and/or radiotherapy (controls). The 123I-MIBG cardiac uptake, expressed as a heart-to-mediastinum ratio on planar images after 4h, ranged from 1.21 to 1.76 (median 1.56) in cancer therapy patients, which was significantly decreased (P=0.0006) in comparison with controls (range 1.81- 2.06, median 1.9). The myocardial 123I-MIBG washout, calculated from planar images after 15 min and 4 h, and LVEF also showed significant differences, but with some overlap in individual cases. In cancer therapy patients, cardiac abnormalities seen on planar images and additional single-photon emission tomographic images varied from focal defects to diffusely reduced myocardial uptake. It is concluded that 123I-MIBG heart scintigraphy, which is able to identify cardiac adrenergic neurone abnormalities in the follow-up period after cancer therapy, may help to identify relapsed patients who are at increased risk of developing cardiotoxicity during retreatment with cardiotoxic therapy modalities.
Collapse
Affiliation(s)
- R A Valdés Olmos
- Department of Nuclear Medicine, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|