51
|
Kim EK, Cho J, Kim JY, Chang SA, Park SJ, Choi JO, Lee SC, Ahn JS, Park SW, Im YH, Jeon ES, Park YH. Early Decline in Left Ventricular Ejection Fraction Can Predict Trastuzumab-Related Cardiotoxicity in Patients with Breast Cancer: A Study Using 13 Years of Registry Data. Cancer Res Treat 2018; 51:727-736. [PMID: 30177584 PMCID: PMC6473274 DOI: 10.4143/crt.2018.262] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2018] [Accepted: 09/03/2018] [Indexed: 12/16/2022] Open
Abstract
Purpose While concerns regarding trastuzumab-related cardiac dysfunction (TRCD) in patients with breast cancer are increasing, there is a lack of evidence supporting the current recommendations for TRCD monitoring. We aimed to investigate the clinical predictors of TRCD in the adjuvant setting of human epidermal growth factor receptor 2–positive breast cancer patients. Materials and Methods From August 2003 to April 2016, consecutive 998 patients who were treated with adjuvant trastuzumab for breast cancer were retrospectively evaluated. TRCD was defined as a decrease ≥10% in left ventricular ejection fraction (LVEF), with a decline below the normal limit or symptomatic heart failure. Results Among 787 eligible patients who had complete data sets consisting of both baseline and follow-up assessment of left ventricular systolic function by echocardiography (mean age, 49.9±9.5 years), 58 (7.4%) developed TRCD. TRCD patients had lower baseline LVEF (63% [59–66] vs. 65% [61–68], p=0.016) and more frequently administered Adriamycin (98% vs. 89%, p=0.022) than those without TRCD. On follow-up echocardiography, a drop in LVEF ≥5% within the first 3 months was more frequent in TRCD patients (78.3% vs. 38.4%, p<0.001). Regardless of baseline LVEF and Adriamycin treatment, a drop in LVEF ≥5% within the first 3 months of trastuzumab administration was strongly associated with the development of TRCD (adjusted hazard ratio, 45.1[17.0–127.6], p<0.001). Conclusion The overall incidence of TRCD was 7.4% in Asian breast cancer patients treated with adjuvant trastuzumab. A decline in LVEF ≥5% within the first 3 months of trastuzumab initiation was strongly associated with TRCD development in patients with breast cancer.
Collapse
Affiliation(s)
- Eun Kyoung Kim
- Division of Cardiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jinhyun Cho
- Division of Hematology-Oncology, Department of Medicine, Inha University Hospital, Inha University School of Medicine, Incheon, Korea
| | - Ji-Yeon Kim
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung-A Chang
- Division of Cardiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sung-Ji Park
- Division of Cardiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Oh Choi
- Division of Cardiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Sang Chol Lee
- Division of Cardiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Jin Seok Ahn
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Seung Woo Park
- Division of Cardiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Young-Hyuck Im
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Eun Seok Jeon
- Division of Cardiology, Heart Vascular Stroke Institute, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Yeon Hee Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
52
|
Jordan JH, Todd RM, Vasu S, Hundley WG. Cardiovascular Magnetic Resonance in the Oncology Patient. JACC Cardiovasc Imaging 2018; 11:1150-1172. [PMID: 30092971 PMCID: PMC6242266 DOI: 10.1016/j.jcmg.2018.06.004] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 06/05/2018] [Accepted: 06/14/2018] [Indexed: 01/20/2023]
Abstract
Patients with or receiving potentially cardiotoxic treatment for cancer are susceptible to developing decrements in left ventricular mass, diastolic function, or systolic function. They may also experience valvular heart disease, pericardial disease, or intracardiac masses. Cardiovascular magnetic resonance may be used to assess cardiac anatomy, structure, and function and to characterize myocardial tissue. This combination of features facilitates the diagnosis and management of disease processes in patients with or those who have survived cancer. This report outlines and describes prior research involving cardiovascular magnetic resonance for assessing cardiovascular disease in patients with or previously having received treatment for cancer.
Collapse
Affiliation(s)
- Jennifer H Jordan
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina.
| | - Ryan M Todd
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Sujethra Vasu
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - W Gregory Hundley
- Department of Internal Medicine, Section on Cardiovascular Medicine at the Wake Forest School of Medicine, Winston-Salem, North Carolina
| |
Collapse
|
53
|
Is There Added Value of the Third Imaging Dimension in Cardio-Oncology? JACC Cardiovasc Imaging 2018; 11:1069-1071. [DOI: 10.1016/j.jcmg.2018.01.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2018] [Accepted: 01/25/2018] [Indexed: 11/17/2022]
|
54
|
Zhu Q, Kirova YM, Cao L, Arsene-Henry A, Chen J. Cardiotoxicity associated with radiotherapy in breast cancer: A question-based review with current literatures. Cancer Treat Rev 2018; 68:9-15. [DOI: 10.1016/j.ctrv.2018.03.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/25/2022]
|
55
|
Venneri L, Zoppellaro G, Khattar RS. Cardio-oncology: the role of advanced echocardiography in cancer patients. Expert Rev Cardiovasc Ther 2018; 16:249-258. [PMID: 29457984 DOI: 10.1080/14779072.2018.1443394] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
INTRODUCTION Cardio-oncology is a rapidly growing field aimed at improving the quality of care of cancer patients by preventing and monitoring cardiovascular complications resulting from cancer treatment. Cardiac imaging, and in particular, transthoracic echocardiography, plays an essential
role in the baseline assessment and serial follow-up of cardio-oncology patients. Areas covered: This review article discusses the role of cardiac imaging with a focus on advanced echocardiography for the detection and management of cancer therapy related cardiovascular complications, in particular, left ventricular dysfunction and heart failure. Expert commentary: While traditional imaging based assessment of left ventricular ejection fraction still has its place in cardiac monitoring, more advanced echocardiographic modalities, in particular, myocardial deformation imaging with speckle tracking strain analysis, show great potential for detecting early signs of cardiotoxicity. Larger studies are needed to determine both the clinical role of strain measurement in influencing initiation of cardioprotective agents and its prognostic value in long term outcome.
Collapse
Affiliation(s)
- Lucia Venneri
- a Department of Echocardiography , Royal Brompton Hospital , London , UK
| | - Giacomo Zoppellaro
- a Department of Echocardiography , Royal Brompton Hospital , London , UK
| | - Rajdeep S Khattar
- a Department of Echocardiography , Royal Brompton Hospital , London , UK
| |
Collapse
|
56
|
Litvak A, Batukbhai B, Russell SD, Tsai HL, Rosner GL, Jeter SC, Armstrong D, Emens LA, Fetting J, Wolff AC, Silhy R, Stearns V, Connolly RM. Racial disparities in the rate of cardiotoxicity of HER2-targeted therapies among women with early breast cancer. Cancer 2018; 124:1904-1911. [PMID: 29381193 DOI: 10.1002/cncr.31260] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2017] [Revised: 12/04/2017] [Accepted: 12/29/2017] [Indexed: 01/03/2023]
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2)-targeted therapies are highly effective at preventing breast cancer recurrence but are associated with cardiotoxicity in some patients, and minimal data are available regarding racial disparities in the incidence of this toxicity. The authors conducted a retrospective study to analyze the association of black or white race with treatment-induced cardiotoxicity and incomplete therapy among patients with HER2-positive early breast cancer. METHODS Women with HER2-positive, stage I through III breast cancer who initiated (neo)adjuvant HER2-targeted therapy (trastuzumab with or without pertuzumab) from January 2005 to March 2015 at the authors' institution were eligible. We analyzed differences in the incidence of cardiotoxicity (a decline in the left ventricular ejection fraction to <50% AND an absolute drop in the left ventricular ejection fraction of ≥10% from baseline) and incomplete therapy (<52 weeks of HER2-targeted therapy) between black and white women in univariate and multivariable analyses. RESULTS The authors identified 59 black patients and 157 white patients who had a median follow-up 5.2 years. The median patient age was 53 years and was similar for black and white patients. The 1-year cardiotoxicity incidence was 12% overall (95% confidence interval [CI], 7%-16%), 24% in black women (95% CI, 12%-34%), and 7% in white women (95% CI, 3%-11%). Black patients had a significantly greater probability of incomplete therapy compared with white patients (odds ratio, 4.61; 95% CI, 1.70-13.07; P = .002). High correlation was observed between a cardiotoxicity event and incomplete therapy (96% concordance). CONCLUSIONS Black patients have a higher rate of cardiotoxicity and resultant incomplete adjuvant HER2-targeted therapy than white patients. This patient population may benefit from enhanced cardiac surveillance, cardioprotective strategies, and early referral to cardiology when appropriate. Cancer 2018;124:1904-11. © 2018 American Cancer Society.
Collapse
Affiliation(s)
- Anya Litvak
- Department of Medicine, The Johns Hopkins Hospital, Baltimore, Maryland
| | | | - Stuart D Russell
- Department of Medicine, The Johns Hopkins Hospital, Baltimore, Maryland
| | - Hua-Ling Tsai
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Gary L Rosner
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Stacie C Jeter
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Deborah Armstrong
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Leisha A Emens
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - John Fetting
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Antonio C Wolff
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Raquel Silhy
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Vered Stearns
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| | - Roisin M Connolly
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Baltimore, Maryland
| |
Collapse
|
57
|
Jain D, Russell RR, Schwartz RG, Panjrath GS, Aronow W. Cardiac Complications of Cancer Therapy: Pathophysiology, Identification, Prevention, Treatment, and Future Directions. Curr Cardiol Rep 2018; 19:36. [PMID: 28374177 DOI: 10.1007/s11886-017-0846-x] [Citation(s) in RCA: 60] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
PURPOSE OF REVIEW Cardiotoxicity is an important complication of cancer therapy. With a significant improvement in the overall survival and prognosis of patients undergoing cancer therapy, cardiovascular toxicity of cancer therapy has become an important public health issue. Several well-established as well as newer anticancer therapies such as anthracyclines, trastuzumab, and other HER2 receptor blockers, antimetabolites, alkylating agents, tyrosine kinase inhibitors, angiogenesis inhibitors, checkpoint inhibitors, and thoracic irradiation are associated with significant cardiotoxicity. RECENT FINDINGS Cardiovascular imaging employing radionuclide imaging, echocardiography, and magnetic resonance imaging is helpful in early detection of the cardiotoxicity and prevention of overt heart failure. These techniques also provide important tools for understanding the mechanism of cardiotoxicity of these modalities, which would help develop strategies for the prevention of cardiac morbidity and mortality related to the use of these agents. An understanding of the mechanism of the cardiotoxicity of cancer therapies can help prevent and treat their adverse cardiovascular consequences. Clinical implementation of algorithms based upon cardiac imaging and several non-imaging biomarkers can prevent cardiac morbidity and mortality associated with the use of cardiotoxic cancer therapies.
Collapse
Affiliation(s)
- Diwakar Jain
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, 100 Woods Road, Valhalla, NY, USA.
| | - Raymond R Russell
- Rhode Island Cardiovascular Institute, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI, 02903, 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
| | - Gurusher S Panjrath
- Heart and Vascular Institute, George Washington University School of Medicine and Health Sciences, Washington, DC, USA
| | - Wilbert Aronow
- Section of Cardiovascular Medicine, New York Medical College and Westchester Medical Center, 100 Woods Road, Valhalla, NY, USA
| |
Collapse
|
58
|
Jain D, Ahmad T, Cairo M, Aronow W. Cardiotoxicity of cancer chemotherapy: identification, prevention and treatment. ANNALS OF TRANSLATIONAL MEDICINE 2017; 5:348. [PMID: 28936442 DOI: 10.21037/atm.2017.06.35] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cardiotoxicity is an important complication of several cancer therapeutic agents. Several well established and newer anticancer therapies such as anthracyclines, trastuzumab and other HER2 receptor blockers, antimetabolites, alkylating agents, tyrosine kinase inhibitors (TKIs), angiogenesis inhibitors, and checkpoint inhibitors are associated with significant cardiotoxicity. Cardiovascular imaging employing radionuclide imaging, echocardiography and magnetic resonance imaging are helpful in early detection and prevention of overt heart failure secondary to cardiotoxicity of cancer therapy. An understanding of the mechanism of the cardiotoxicity of cancer therapies can help prevent and treat their adverse cardiovascular consequences. Clinical implementation of algorithms based upon cardiac imaging and several non-imaging biomarkers can prevent cardiac morbidity and mortality associated with the use of cardiotoxic cancer therapies.
Collapse
Affiliation(s)
- Diwakar Jain
- Section of Cardiovascular Medicine, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Tauseef Ahmad
- Section of Oncology and Hematology, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Mitchel Cairo
- Section of Pediatric Hematology and Oncology, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| | - Wilbert Aronow
- Section of Cardiovascular Medicine, Department of Pediatrics, New York Medical College and Westchester Medical Center, Valhalla, NY, USA
| |
Collapse
|
59
|
Cardiac Imaging: Multimodality Advances and Surveillance Strategies in Detection of Cardiotoxicity. Curr Oncol Rep 2017; 19:63. [PMID: 28791609 DOI: 10.1007/s11912-017-0622-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Contemporary cancer management has increased the overall number of cancer survivors, but cardiotoxicity remains a subject of concern, which is a major cause of noncancer mortality among survivors. Among the potential cardiovascular complications, left ventricular (LV) systolic dysfunction is a poor prognostic factor. The importance of its early detection is based on the principle that the likelihood of response to heart failure (HF) treatment is temporally related to the initiation of HF treatment. For these reasons, cardiac monitoring is commonly applied in general practice, based on serial measurements of LV ejection fraction (LVEF); transthoracic echocardiography (TTE) is generally used. However, the LVEF, as a diagnostic and predictive parameter, has significant limitations, which calls for more effective multimodality imaging strategies. This approach requires further study, but there is increasing available data in the literature, encouraging the combination of multimodality imaging parameters and techniques for early cancer therapeutic-related cardiac dysfunction (CTRCD) detection.
Collapse
|
60
|
Kang Y, Wang W, Zhao H, Qiao Z, Shen X, He B. Assessment of Subclinical Doxorubicin-induced Cardiotoxicity in a Rat Model by Speckle-Tracking Imaging. Arq Bras Cardiol 2017; 109:0. [PMID: 28700019 PMCID: PMC5576117 DOI: 10.5935/abc.20170097] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/20/2017] [Accepted: 02/20/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUNDS: Despite their clear therapeutic benefits, anthracycline-induced cardiotoxicity is a major concern limiting the ability to reduce morbidity and mortality associated with cancers. The early identification of anthracycline-induced cardiotoxicity is of vital importance to assess the cardiac risk against the potential cancer treatment. OBJECTIVE: To investigate whether speckle-tracking analysis can provide a sensitive and accurate measurement when detecting doxorubicin-induced left ventricular injury. METHODS: Wistar rats were divided into 4 groups with 8 rats each, given doxorubicin intraperitoneally at weekly intervals for up to 4 weeks. Group 1: 2.5 mg/kg/week; group 2: 3 mg/kg/week; group 3: 3.5mg/kg/week; group 4: 4mg/kg/week. An additional 5 rats were used as controls. Echocardiographic images were obtained at baseline and 1 week after the last dose of treatment. Radial (Srad) and circumferential (Scirc) strains, radial (SRrad) and circumferential (SRcirc) strain rates were analyzed. After the experiment, cardiac troponin I (cTnI) was analyzed and the heart samples were histologically evaluated. RESULTS: After doxorubicin exposure, LVEF was significantly reduced in group 4 (p = 0.006), but remained stable in the other groups. However, after treatment, Srads were reduced in groups 2, 3 and 4 (p all < 0.05). The decrease in Srads was correlated with cTnI (rho = -0.736, p = 0.000) and cardiomyopathy scores (rho = -0.797, p = 0.000). CONCLUSION: Radial strain could provide a sensitive and noninvasive index in early detection of doxorubicin-induced myocardial injury. The changes in radial strain had a significant correlation with myocardial lesions and serum cardiac troponin I levels, indicating that this parameter could accurately evaluate cardiotoxicity severity. FUNDAMENTO: Apesar dos seus claros benefícios terapêuticos, a cardiotoxicidade induzida pela antraciclina é uma grande preocupação que limita a capacidade de reduzir a morbidade e mortalidade associadas com cânceres. A identificação precoce da cardiotoxicidade induzida por antraciclina é de vital importância para o equilíbrio entre o risco cardíaco e o potencial tratamento do câncer. OBJETIVO: Investigar se a análise por speckle-tracking pode fornecer uma medida sensível e precisa na detecção de lesão ventricular esquerda induzida por doxorrubicina. MÉTODOS: Ratos Wistar foram divididos em 4 grupos de 8 ratos cada, e doxorrubicina foi administrada intraperitonealmente em intervalos semanais de até 4 semanas. Grupo 1: 2,5 mg/kg/semana; Grupo 2: 3 mg/kg/semana; Grupo 3: 3,5 mg/kg/semana; Grupo 4: 4 mg/kg/semana. Foram utilizados 5 ratos adicionais como controles. As imagens ecocardiográficas foram obtidas na linha basal e 1 semana após a última dose do tratamento. Foram analisados o strain radial (Srad) e circunferencial (Scirc) e as taxas de strain radial (TSrad) e circunferencial (TScirc). Após o experimento, a troponina cardíaca I (cTnI) foi analisada e as amostras cardíacas foram avaliadas histologicamente. RESULTADOS: Após a exposição à doxorrubicina, a FEVE foi significativamente reduzida no grupo 4 (p = 0,006), mas permaneceu estável nos outros grupos. Entretanto, após o tratamento, os Srads foram reduzidos nos grupos 2, 3 e 4 (p < 0,05). A diminuição dos Srads foi correlacionada com cTnI (rho = -0,736, p = 0,000) e os escores de cardiomiopatia (rho = -0,797, p = 0,000). CONCLUSÃO: O strain radial pode fornecer um índice sensível e não-invasivo na detecção precoce da lesão miocárdica induzida pela doxorrubicina. As alterações do strain radial apresentaram correlação significativa com lesões miocárdicas e níveis séricos de troponina I cardíaca, indicando que esse parâmetro pode avaliar com precisão a gravidade da cardiotoxicidade.
Collapse
Affiliation(s)
- Yu Kang
- Renji Hospital, School of medicine, Shanghai Jiaotong University,
Shanghai, China
| | - Wei Wang
- Renji Hospital, School of medicine, Shanghai Jiaotong University,
Shanghai, China
| | - Hang Zhao
- Renji Hospital, School of medicine, Shanghai Jiaotong University,
Shanghai, China
| | - Zhiqing Qiao
- Renji Hospital, School of medicine, Shanghai Jiaotong University,
Shanghai, China
| | - Xuedong Shen
- Renji Hospital, School of medicine, Shanghai Jiaotong University,
Shanghai, China
| | - Ben He
- Renji Hospital, School of medicine, Shanghai Jiaotong University,
Shanghai, China
| |
Collapse
|
61
|
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: 1.0] [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
|
62
|
López-Fernández T, Thavendiranathan P. Nuevas técnicas de imagen cardiaca en la detección precoz de cardiotoxicidad secundaria a tratamientos oncológicos. Rev Esp Cardiol 2017. [PMID: 28189542 DOI: 10.1016/j.recesp.2016.12.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
63
|
Manrique CR, Park M, Tiwari N, Plana JC, Garcia MJ. Diagnostic Strategies for Early Recognition of Cancer Therapeutics-Related Cardiac Dysfunction. CLINICAL MEDICINE INSIGHTS-CARDIOLOGY 2017; 11:1179546817697983. [PMID: 28469492 PMCID: PMC5392033 DOI: 10.1177/1179546817697983] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 02/01/2017] [Indexed: 12/31/2022]
Abstract
Cardiovascular toxicity in the form of cardiac dysfunction continues to be an obstacle for patients with cancer. Survival and quality of life of cancer survivors are frequently affected by increased incidence of cardiovascular disease. The involvement of the cardiovascular system by primary or secondary malignancies, as well as its dysfunction secondary to the administration of antineoplastics, has led to the development of a new discipline called Cardio-Oncology, an exciting cardiology subspecialty with more questions than answers and as a result an enormous opportunity for research in the field. Multidisciplinary efforts have been focused on the prevention, diagnosis, and treatment of cancer therapeutics-related cardiovascular dysfunction (CTRCD). This review article will focus on the early diagnosis of left ventricular dysfunction associated with chemotherapy. Currently, the identification of cardiac toxicity associated with cancer treatment is the cornerstone for critical decisions regarding anticancer therapy and cardioprotective strategies. Its early detection, especially in subclinical phases, allows immediate intervention to prevent further impairment of the myocardium and other cardiovascular structures. The most significant published studies were selected for this revision, providing an updated document for the health professionals involved in the care of patients with cancer. We examined the current evidence and recommendations for biochemical and noninvasive diagnostic techniques, including their specific role for identification of CTRCD. Traditional and advanced imaging modalities, used alone or in combination with cardiovascular biomarkers, are essential for the recognition of cardiotoxicity during cancer therapy. Evolving basic and clinical research are focused on the development of more sensitive and specific diagnostic tools and for the recognition of cardiac toxicity.
Collapse
Affiliation(s)
- Carlos R Manrique
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Michael Park
- Division of Cardiology, Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Nidhish Tiwari
- Division of Cardiology, Department of Medicine, Jacobi Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Juan Carlos Plana
- Division of Cardiology, Baylor College of Medicine, Houston, TX, USA
| | - Mario J Garcia
- Division of Cardiology, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
64
|
Huang H, Nijjar PS, Misialek JR, Blaes A, Derrico NP, Kazmirczak F, Klem I, Farzaneh-Far A, Shenoy C. Accuracy of left ventricular ejection fraction by contemporary multiple gated acquisition scanning in patients with cancer: comparison with cardiovascular magnetic resonance. J Cardiovasc Magn Reson 2017; 19:34. [PMID: 28335788 PMCID: PMC5364623 DOI: 10.1186/s12968-017-0348-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/24/2017] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Multiple gated acquisition scanning (MUGA) is a common imaging modality for baseline and serial assessment of left ventricular ejection fraction (LVEF) for cardiotoxicity risk assessment prior to, surveillance during, and surveillance after administration of potentially cardiotoxic cancer treatment. The objective of this study was to compare the accuracy of left ventricular ejection fractions (LVEF) obtained by contemporary clinical multiple gated acquisition scans (MUGA) with reference LVEFs from cardiovascular magnetic resonance (CMR) in consecutive patients with cancer. METHODS In a cross-sectional study, we compared MUGA clinical and CMR reference LVEFs in 75 patients with cancer who had both studies within 30 days. Misclassification was assessed using the two most common thresholds of LVEF used in cardiotoxicity clinical studies and practice: 50 and 55%. RESULTS Compared to CMR reference LVEFs, MUGA clinical LVEFs were only lower by a mean of 1.5% (48.5% vs. 50.0%, p = 0.17). However, the limits of agreement between MUGA clinical and CMR reference LVEFs were wide at -19.4 to 16.5%. At LVEF thresholds of 50 and 55%, there was misclassification of 35 and 20% of cancer patients, respectively. CONCLUSIONS MUGA clinical LVEFs are only modestly accurate when compared with CMR reference LVEFs. These data have significant implications on clinical research and patient care of a population with, or at risk for, cardiotoxicity.
Collapse
Affiliation(s)
- Hans Huang
- Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN USA
| | - Prabhjot S. Nijjar
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical Center, 420 Delaware Street SE, MMC 508, Minneapolis, MN 55455 USA
| | - Jeffrey R. Misialek
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical Center, 420 Delaware Street SE, MMC 508, Minneapolis, MN 55455 USA
| | - Anne Blaes
- Division of Hematology, Oncology and Transplantation, Department of Medicine, University of Minnesota Medical Center, Minneapolis, MN USA
| | | | - Felipe Kazmirczak
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical Center, 420 Delaware Street SE, MMC 508, Minneapolis, MN 55455 USA
| | - Igor Klem
- Duke Cardiovascular Magnetic Resonance Center, Duke University Medical Center, Durham, NC USA
- Division of Cardiology, Duke University Medical Center, Durham, NC USA
| | - Afshin Farzaneh-Far
- Division of Cardiology, Duke University Medical Center, Durham, NC USA
- Division of Cardiology, Department of Medicine, University of Illinois at Chicago, Chicago, IL USA
| | - Chetan Shenoy
- Cardiovascular Division, Department of Medicine, University of Minnesota Medical Center, 420 Delaware Street SE, MMC 508, Minneapolis, MN 55455 USA
| |
Collapse
|
65
|
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: 0.8] [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
|
66
|
López-Fernández T, Thavendiranathan P. Emerging Cardiac Imaging Modalities for the Early Detection of Cardiotoxicity Due to Anticancer Therapies. ACTA ACUST UNITED AC 2017; 70:487-495. [PMID: 28189542 DOI: 10.1016/j.rec.2017.01.004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 12/16/2016] [Indexed: 01/14/2023]
Abstract
The undeniable advances in the field of oncology have finally led to a decrease in overall cancer-related mortality. However, this population of long-term cancer survivors is now facing a shift toward a substantial increase in cardiovascular morbidity and mortality. Because the development of overt cardiotoxicity can be associated with poor outcomes, preclinical identification of cardiac toxicity is important. This will promote early instauration of treatments to prevent overt heart dysfunction and allow oncologists to continue cancer therapy in an uninterrupted manner. Surveillance strategies for the early detection of cardiac injury include cardiac imaging and biomarkers during treatment. In this review, we outline existing cardiac imaging modalities to detect myocardial changes in patients undergoing cancer treatment and in survivors, and their strengths and limitations.
Collapse
Affiliation(s)
- Teresa López-Fernández
- Servicio de Cardiología, Unidad de Imagen Cardiaca, Unidad de Cardio-Oncología, Hospital Universitario La Paz, IdiPaz, Madrid, Spain.
| | - Paaladinesh Thavendiranathan
- Peter Munk Cardiac Center, Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
67
|
|
68
|
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: 4.4] [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
|
69
|
Reuvekamp EJ, Bulten BF, Nieuwenhuis AA, Meekes MRA, de Haan AFJ, Tol J, Maas AHEM, Elias-Smale SE, de Geus-Oei LF. Does diastolic dysfunction precede systolic dysfunction in trastuzumab-induced cardiotoxicity? Assessment with multigated radionuclide angiography (MUGA). J Nucl Cardiol 2016; 23:824-32. [PMID: 26048264 PMCID: PMC4956716 DOI: 10.1007/s12350-015-0164-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Accepted: 04/10/2015] [Indexed: 01/09/2023]
Abstract
BACKGROUND Trastuzumab is successfully used for the treatment of HER2-positive breast cancer. Because of its association with cardiotoxicity, LVEF is monitored by MUGA, though this is a relatively late measure of cardiac function. Diastolic dysfunction (DD) is believed to be an early predictor of cardiac impairment. We evaluate the merit of MUGA-derived diastolic function parameters in the early detection of trastuzumab-induced cardiotoxicity (TIC). METHODS AND RESULTS 77 trastuzumab-treated patients with normal baseline systolic and diastolic function were retrospectively selected (n = 77). All serial MUGA examinations were re-analyzed for systolic and diastolic function parameters. 36 patients (47%) developed SD and 45 patients (58%) DD during treatment. Both systolic and diastolic parameters significantly decreased. Of the patients with SD, 24 (67%) also developed DD. DD developed prior to systolic impairment in 54% of cases, in 42% vice versa, while time to occurrence did not differ significantly (P = .52). This also applied to the subgroup of advanced stage breast cancer patients (P = .1). CONCLUSIONS Trastzumab-induced SD and DD can be detected by MUGA. An impairment of MUGA-derived diastolic parameters does not occur prior to SD and therefore cannot be used as earlier predictors of TIC.
Collapse
Affiliation(s)
- E J Reuvekamp
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | - B F Bulten
- Biomedical Photonic Imaging Group, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, PO Box 217, 7500 AE, Enschede, The Netherlands.
| | - A A Nieuwenhuis
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | - M R A Meekes
- Department of Radiology and Nuclear Medicine, Radboudumc, Nijmegen, The Netherlands
| | - A F J de Haan
- Department for Health Evidence, Radboudumc, Nijmegen, The Netherlands
| | - J Tol
- Department of Medical Oncology, Radboudumc, Nijmegen, The Netherlands
| | - A H E M Maas
- Department of Cardiology, Radboudumc, Nijmegen, The Netherlands
| | - S E Elias-Smale
- Department of Cardiology, Radboudumc, Nijmegen, The Netherlands
| | - L F de Geus-Oei
- Biomedical Photonic Imaging Group, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, PO Box 217, 7500 AE, Enschede, The Netherlands
- Department of Radiology, Leiden University Medical Centre, Leiden, The Netherlands
| |
Collapse
|
70
|
Dias A, Claudino W, Sinha R, Perez C, Jain D. Human epidermal growth factor antagonists and cardiotoxicity—A short review of the problem and preventative measures. Crit Rev Oncol Hematol 2016; 104:42-51. [DOI: 10.1016/j.critrevonc.2016.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Revised: 03/09/2016] [Accepted: 04/27/2016] [Indexed: 01/21/2023] Open
|
71
|
Echocardiography and Alternative Cardiac Imaging Strategies for Long-Term Cardiotoxicity Surveillance of Cancer Survivors Treated with Chemotherapy and/or Radiation Exposure. Curr Oncol Rep 2016; 18:52. [PMID: 27461436 DOI: 10.1007/s11912-016-0532-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
72
|
Cardiac toxicity of trastuzumab in elderly patients with breast cancer. JOURNAL OF GERIATRIC CARDIOLOGY : JGC 2016; 13:355-63. [PMID: 27403145 PMCID: PMC4921548 DOI: 10.11909/j.issn.1671-5411.2016.04.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Breast cancer (BC) is diagnosed in ≥ 65 year old women in about half of cases. Experts currently recommend that systemic therapy is offered to elderly patients with BC, if, based on their overall conditions and life expectancy, it can be reasonably anticipated that the benefits will outweigh the risks of treatment. Like for young subjects, the monoclonal antibody against human epidermal growth factor receptor-2 (HER-2), trastuzumab, represents a valid therapeutic option when BC over-expresses this receptor. Unfortunately, administration of trastuzumab is associated with the occurrence of left ventricular dysfunction and chronic heart failure (CHF), possibly because of interference with the homeostatic functions of HER-2 in the heart. Registry-based, retrospective analyses have reported an incidence of CHF around 25% in elderly women receiving trastuzumab compared with 10%-15% in those not given any therapy for BC, and the risk of CHF has been estimated to be two-fold higher in > 60-65 year old trastuzumab users vs. non-users. Extremely advanced age and preexisting cardiac disease have been shown to predispose to trastuzumab cardiotoxicity. Therefore, selection of older patients for treatment with trastuzumab should be primarily based on their general status and the presence of comorbidities; previous chemotherapy, especially with anthracyclines, should be also taken into account. Once therapy has started, efforts should be made to ensure regular cardiac surveillance. The role of selected biomarkers, such as cardiac troponin, or new imaging techniques (three-dimension, tissue Doppler echocardiography, magnetic resonance imaging) is promising, but must be further investigated especially in the elderly. Moreover, additional studies are needed in order to better understand the mechanisms by which trastuzumab affects the old heart.
Collapse
|
73
|
Virani SA, Dent S, Brezden-Masley C, Clarke B, Davis MK, Jassal DS, Johnson C, Lemieux J, Paterson I, Sebag IA, Simmons C, Sulpher J, Thain K, Thavendiranathan P, Wentzell JR, Wurtele N, Côté MA, Fine NM, Haddad H, Hayley BD, Hopkins S, Joy AA, Rayson D, Stadnick E, Straatman L. Canadian Cardiovascular Society Guidelines for Evaluation and Management of Cardiovascular Complications of Cancer Therapy. Can J Cardiol 2016; 32:831-41. [DOI: 10.1016/j.cjca.2016.02.078] [Citation(s) in RCA: 117] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Revised: 02/17/2016] [Accepted: 02/17/2016] [Indexed: 12/30/2022] Open
|
74
|
Nair N, Gongora E. Heart failure in chemotherapy-related cardiomyopathy: Can exercise make a difference? BBA CLINICAL 2016; 6:69-75. [PMID: 27413695 PMCID: PMC4925806 DOI: 10.1016/j.bbacli.2016.06.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 06/09/2016] [Accepted: 06/09/2016] [Indexed: 12/20/2022]
Abstract
Medical therapies in oncology have resulted in better survival resulting in a large population who are at risk of early and late cardiac complications of chemotherapy. Cardiotoxicity related to chemotherapy can manifest decades after treatment with a threefold higher mortality rate as compared to idiopathic dilated cardiomyopathy. The leading cause of death in cancer survivors seems to be cardiac. Early detection and intervention could prevent progression of heart failure to end stage disease requiring advanced therapies such as implantation of ventricular assist devices or cardiac transplantation. This review focuses on the role of exercise in cardioprotection in this population. The current practice of depending on ejection fraction for diagnosis of heart failure is suboptimal to detect subclinical disease. It is also important to diagnose and treat early diastolic dysfunction as this tends to lead to heart failure with preserved ejection fraction. Hence we suggest an algorithm here that is based on using strain rate and tissue Doppler imaging modalities to detect subclinical systolic and diastolic dysfunction. Further research is warranted in terms of defining exercise prescriptions in this population. Human studies with multicenter participation in randomized controlled trials should be done to elucidate the intricacies of aerobic exercise intervention in cardiotoxicity dependent heart failure. It is also necessary to assess the utility of exercise interventions in the different chemotherapeutic regimens as they impact the outcomes. The need for exercise prescription to prevent cardiotoxicity in chemotherapy patients The molecular basis of exercise as an intervention Summary of existing evidence Need for further studies on the role of exercise in different chemotherapeutic regimens
Collapse
Affiliation(s)
- Nandini Nair
- Division of Cardiology, Texas Tech Health Sciences Center, Lubbock, TX 79382, United States
| | - Enrique Gongora
- Memorial Cardiac and Vascular Institute, Hollywood, FL 33021, United States
| |
Collapse
|
75
|
Matos E, Jug B, Blagus R, Zakotnik B. A Prospective Cohort Study on Cardiotoxicity of Adjuvant Trastuzumab Therapy in Breast Cancer Patients. Arq Bras Cardiol 2016; 107:40-7. [PMID: 27305108 PMCID: PMC4976955 DOI: 10.5935/abc.20160084] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 02/23/2016] [Indexed: 01/03/2023] Open
Abstract
Background Cardiotoxicity is an important side effect of trastuzumab therapy and cardiac
surveillance is recommended. Objectives The aim of our study was to prospectively assess baseline patients'
characteristics, level of N-terminal pro-brain natriuretic peptide
(NT-proBNP) and echocardiographic parameters as possible predictors of
trastuzumab-related cardiac dysfunction. Methods In a prospective cohort study, clinical, echocardiographic and neurohumoral
assessment was performed at baseline, after 4, 8 and 12 months in breast
cancer patients undergoing post-anthracycline (3-4 cycles) adjuvant therapy
with trastuzumab. Trastuzumab-related cardiac dysfunction was defined as a
decline of ≥ 10% in left ventricular ejection fraction (LVEF). Results 92 patients (mean age, 53.6 ± 9.0 years) were included. Patients who
developed trastuzumab-related LVEF decline ≥ 10% (20.6%) during
treatment had significantly higher baseline LVEF (70.7 ± 4.4%) than
those without (64.8 ± 5.5%) (p = 0.0035). All other measured baseline
parameters (age, body mass index, arterial hypertension, level of NT-proBNP
and other echocardiographic parameters) were not identified as
significant. Conclusions Our findings suggest that baseline patient' characteristics, level of
NT-proBNP and echocardiographic parameters, as long as they are within
normal range, are not a reliable tool to predict early trastuzumab-related
cardiac dysfunction in patients undergoing post-low dose anthracycline
adjuvant trastuzumab therapy. A LVEF decline in patients with high-normal
baseline level although statistically significant is not clinically
relevant.
Collapse
Affiliation(s)
- Erika Matos
- Division of Medical Oncology, Institute of Oncology Ljubljana, Slovenia
| | - Borut Jug
- Department of Vascular Diseases, University Medical Centre Ljubljana, Slovenia
| | - Rok Blagus
- Institute for Biostatistics and Medical Informatics, University of Ljubljana, Slovenia
| | - Branko Zakotnik
- Division of Medical Oncology, Institute of Oncology Ljubljana, Slovenia
| |
Collapse
|
76
|
Murtagh G, Yu Z, Harrold E, Cooke J, Keegan N, Fukuda S, Addetia K, Kim JH, Spencer KT, Takeuchi M, Kennedy J, Ward RP, Patel AR, Lang RM, DeCara JM. Monitoring Ionizing Radiation Exposure for Cardiotoxic Effects of Breast Cancer Treatment. Am J Cardiol 2016; 117:1678-1682. [PMID: 27040573 DOI: 10.1016/j.amjcard.2016.02.047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 02/18/2016] [Accepted: 02/18/2016] [Indexed: 01/03/2023]
Abstract
Serial assessments of left ventricular ejection fraction (LVEF) are customary in patients with breast cancer receiving trastuzumab. Radionuclide angiography (RNA) is often used; however, a typical monitoring schedule could include 5 scans in a year. We evaluated the proportion of imaging-related ionizing radiation attributable to RNA in 115 patients with breast cancer, from 3 medical centers in the United States, Ireland, and Japan, who completed 12 months of trastuzumab treatment. Estimated radiation dose (ERD) was used to calculate exposure associated with imaging procedures spanning the 18 months before and after trastuzumab therapy. In addition, 20 cardiologists and oncologists from participating centers were surveyed for their opinions regarding the contribution of RNA to overall radiation exposure during trastuzumab treatment. When RNA was used to monitor LVEF, the mean ERD from imaging was substantial (34 ± 24.3 mSv), with the majority attributable solely to RNA (24.7 ± 14.8 mSv, 72.6%). Actual ERD associated with RNA in this population differed significantly from the perception in surveyed cardiologists and oncologists; 70% of respondents believed that RNA typically accounted for 0% to 20% of overall radiation exposure from imaging; RNA actually accounted for more than 70% of ERD. In conclusion, RNA was used to monitor LVEF in most patients in this cohort during and after trastuzumab therapy. This significantly increased ERD and accounted for a greater proportion of radiation than that perceived by surveyed physicians. ERD should be taken into account when choosing a method of LVEF surveillance. Alternative techniques that do not use radiation should be strongly considered.
Collapse
|
77
|
Maurea N, Spallarossa P, Cadeddu C, Madonna R, Mele D, Monte I, Novo G, Pagliaro P, Pepe A, Tocchetti CG, Zito C, Mercuro G. A recommended practical approach to the management of target therapy and angiogenesis inhibitors cardiotoxicity: an opinion paper of the working group on drug cardiotoxicity and cardioprotection, Italian Society of Cardiology. J Cardiovasc Med (Hagerstown) 2016; 17 Suppl 1:e93-e104. [PMID: 27183530 PMCID: PMC4927319 DOI: 10.2459/jcm.0000000000000383] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Indexed: 12/21/2022]
Abstract
The US National Cancer Institute estimates that cardiotoxicity (CTX) from target therapy refers mostly to four groups of drugs: epidermal growth factor receptor 2 inhibitors, angiogenic inhibitors, directed Abelson murine leukemia viral oncogene homolog inhibitors, and proteasome inhibitors. The main cardiotoxic side-effects related to antiepidermal growth factor receptor 2 therapy are left ventricular systolic dysfunction and heart failure. Angiogenesis inhibitors are associated with hypertension, left ventricular dysfunction/heart failure, myocardial ischemia, QT prolongation, and thrombosis. Moreover, other agents may be related to CTX induced by treatment. In this study, we review the guidelines for a practical approach for the management of CTX in patients under anticancer target therapy.
Collapse
Affiliation(s)
- Nicola Maurea
- Division of Cardiology, Istituto Nazionale per lo Studio e la Cura dei Tumori ‘Fondazione Giovanni Pascale’ – IRCCS, Naples
| | | | - Christian Cadeddu
- Department of Medical Sciences ‘Mario Aresu’, University of Cagliari, Cagliari
| | - Rosalinda Madonna
- Institute of Cardiology, Center of Excellence on Aging, ‘G. d’Annunzio’ University, Chieti
| | - Donato Mele
- Cardiology Unit, University Hospital of Ferrara, Ferrara
| | - Ines Monte
- Department of General Surgery and Medical-Surgery Specialties, University of Catania, Catania
| | - Giuseppina Novo
- Chair and Division of Cardiology, University of Palermo, Palermo
| | - Pasquale Pagliaro
- Department of Clinical and Biological Sciences, University of Turin, Turin
| | - Alessia Pepe
- U.O.C. Magnetic Resonance Imaging, Fondazione G. Monasterio C.N.R., Pisa
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, Federico II University, Naples
| | - Concetta Zito
- U.O.C. Cardiology Intensive Unit, A.O.U. Policlinico ‘G. Martino’, University of Messina, Messina, Italy
| | - Giuseppe Mercuro
- Department of Medical Sciences ‘Mario Aresu’, University of Cagliari, Cagliari
| |
Collapse
|
78
|
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) 2016; 17 Suppl 1:e45-e54. [PMID: 27183525 DOI: 10.2459/jcm.0000000000000380] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [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
|
79
|
Cardiovascular surveillance in breast cancer treatment: A more individualized approach is needed. Maturitas 2016; 89:58-62. [PMID: 27180161 DOI: 10.1016/j.maturitas.2016.04.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 04/11/2016] [Accepted: 04/18/2016] [Indexed: 12/18/2022]
Abstract
Newly developed treatment strategies for breast cancer have reduced mortality rates over the past decades. Patients with breast cancer represent a heterogeneous population. Differences in the severity of the disease require diverse treatment options. Women have distinct individual risk patterns for cardiovascular disease that may affect their susceptibility to cardiotoxicity during therapy. While breast cancer treatment is targeted more on tumor and patient characteristics, a tailored individual approach with early and late cardiosurveillance is not yet implemented in routine care. Newly available cardiac imaging techniques are better suited to the early detection of cardiotoxicity and should be used more often in those patients at highest risk, as the early intervention afforded will improve their quality of life and prognosis.
Collapse
|
80
|
Chaudry M, Banchs J, Chavez-MacGregor M. Anthracycline or trastuzumab-related cardiotoxicity: do we have a predictive biomarker? Biomark Med 2016; 10:315-28. [DOI: 10.2217/bmm.15.124] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Chemotherapy-induced cardiotoxicity, is a well-known and potentially serious complication strongly impacting the quality of life and overall survival of breast cancer patients. The current diagnostic approach to detect cardiac damage is the estimation of left ventricular ejection fraction by echocardiography. However, this approach exhibits less sensitivity toward early prediction of cardiomyopathy, not allowing for preventive strategies. Measurement of serum cardiac-specific biomarkers can be a valid diagnostic tool for identifying patients prone to developing cardiotoxocity and in whom closer cardiac monitoring and preventive strategies are pivotal. In this article, we review work done on biomarkers in recent years, with an emphasis on troponin and B-type natriuretic peptide, which are currently the most studied in this field. We also briefly discuss current and emerging imaging techniques for early detection of cardiomyopathy.
Collapse
Affiliation(s)
- Misbat Chaudry
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-3721, USA
| | - Jose Banchs
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Mariana Chavez-MacGregor
- Department of Breast Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030-3721, USA
- Department of Health Services Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| |
Collapse
|
81
|
Yu AF, Ky B. Roadmap for biomarkers of cancer therapy cardiotoxicity. Heart 2015; 102:425-30. [PMID: 26674989 DOI: 10.1136/heartjnl-2015-307894] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 11/17/2015] [Indexed: 01/07/2023] Open
Abstract
Contemporary cancer treatment uses multiple modalities such as chemotherapy, targeted therapy and radiotherapy. These therapies, often used in combination, are associated with an increased risk of cardiotoxicity, specifically cardiomyopathy and heart failure. Cardiologists and oncologists are faced with the challenge of maximising the clinical benefit from cancer therapy while minimising the risk of early and late-onset cardiotoxicity. The current paradigm for cardiotoxicity detection and management relies primarily upon the assessment of left ventricular ejection fraction (LVEF). However, LVEF alone is limited in both diagnostic and prognostic ability. There is growing enthusiasm over the identification of newer biomarkers of cardiotoxicity that can detect cardiac injury at earlier stages of disease and could be used as an adjunctive prognostic measure to routine LVEF assessment. Thus, imaging and circulating biomarkers are currently under active investigation for use throughout the continuum of cancer care-for risk stratification of cardiotoxicity prior to treatment, detection of early cardiotoxicity during treatment and diagnosis of late cardiotoxicity in survivorship. Myocardial strain, cardiac troponin and brain natriuretic peptide are the most prominent biomarkers currently being studied, although data on novel circulating biomarkers are emerging.
Collapse
Affiliation(s)
- Anthony F Yu
- Department of Medicine, Cardiology Service, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Bonnie Ky
- Division of Cardiovascular Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
82
|
Molinaro M, Ameri P, Marone G, Petretta M, Abete P, Di Lisa F, De Placido S, Bonaduce D, Tocchetti CG. Recent Advances on Pathophysiology, Diagnostic and Therapeutic Insights in Cardiac Dysfunction Induced by Antineoplastic Drugs. BIOMED RESEARCH INTERNATIONAL 2015; 2015:138148. [PMID: 26583088 PMCID: PMC4637019 DOI: 10.1155/2015/138148] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Accepted: 07/01/2015] [Indexed: 12/28/2022]
Abstract
Along with the improvement of survival after cancer, cardiotoxicity due to antineoplastic treatments has emerged as a clinically relevant problem. Potential cardiovascular toxicities due to anticancer agents include QT prolongation and arrhythmias, myocardial ischemia and infarction, hypertension and/or thromboembolism, left ventricular (LV) dysfunction, and heart failure (HF). The latter is variable in severity, may be reversible or irreversible, and can occur soon after or as a delayed consequence of anticancer treatments. In the last decade recent advances have emerged in clinical and pathophysiological aspects of LV dysfunction induced by the most widely used anticancer drugs. In particular, early, sensitive markers of cardiac dysfunction that can predict this form of cardiomyopathy before ejection fraction (EF) is reduced are becoming increasingly important, along with novel therapeutic and cardioprotective strategies, in the attempt of protecting cardiooncologic patients from the development of congestive heart failure.
Collapse
Affiliation(s)
- Marilisa Molinaro
- Department of Medicine and Health Sciences, University of Molise, 86100 Campobasso, Italy
| | - Pietro Ameri
- Department of Internal Medicine, University of Genova, 16132 Genova, Italy
| | - Giancarlo Marone
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Mario Petretta
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, 35121 Padova, Italy
- National Researches Council, Neuroscience Institute, University of Padova, 35121 Padova, Italy
| | - Sabino De Placido
- Department of Clinical Medicine and Surgery, Federico II University, 80131 Naples, Italy
| | - Domenico Bonaduce
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| | - Carlo G. Tocchetti
- Department of Translational Medical Sciences, Division of Internal Medicine, Federico II University, 80131 Naples, Italy
| |
Collapse
|
83
|
Abstract
The identification of patients at risk of cardiac toxicity (cardiotoxicity) from cancer therapy is challenging. There is an increasing focus on early detection of cardiotoxicity such that interventions can be instituted to prevent advanced heart failure. Clinical risk prediction tools are limited and clinical symptoms are not specific. Direct assessment of myocardial function before and during cancer treatment using cardiac imaging appears to be an objective method to identify patients at risk. Although, multiple imaging modalities and measures of cardiac function are available, the best modality or the optimal measure of function is unknown. Measurement of left ventricular ejection fraction is most commonly used; however, growing literature suggests that it is inadequate for the detection of early cardiac injury. Other measures include left ventricular diastolic function, myocardial deformation, and myocardial tissue characterization. This review will provide an overview of the clinically available measures for the assessment of cardiotoxicity.
Collapse
|
84
|
|
85
|
Bordun KA, Premecz S, daSilva M, Mandal S, Goyal V, Glavinovic T, Cheung M, Cheung D, White CW, Chaudhary R, Freed DH, Villarraga HR, Herrmann J, Kohli M, Ravandi A, Thliveris J, Pitz M, Singal PK, Mulvagh S, Jassal DS. The utility of cardiac biomarkers and echocardiography for the early detection of bevacizumab- and sunitinib-mediated cardiotoxicity. Am J Physiol Heart Circ Physiol 2015; 309:H692-701. [DOI: 10.1152/ajpheart.00172.2015] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Accepted: 06/17/2015] [Indexed: 01/09/2023]
Abstract
The recent introduction of novel anticancer therapies, including bevacizumab (BVZ) and sunitinib (SNT), is associated with an increased risk of cardiotoxicity. However, early identification of left ventricular (LV) systolic dysfunction may facilitate dose modification and avoid the development of advanced heart failure. Using a murine model of BVZ- and SNT-mediated cardiotoxicity, we investigated whether cardiac biomarkers and/or tissue velocity imaging (TVI) using echocardiography can detect early changes in cardiac function, before a decrease in LV ejection fraction is identified. A total of 75 wild-type C57Bl/6 male mice were treated with either 0.9% saline, BVZ, or SNT. Serial monitoring of blood pressure, high-sensitivity troponin I, and echocardiographic indexes were performed over a 14-day study period, after which the mice were euthanized for histological and biochemical analyses. Mice treated with either BVZ or SNT developed systemic hypertension as early as day 7, which increased by day 14. Cardiac biomarkers, specifically high-sensitivity troponin I, were not predictive of early LV systolic dysfunction. Although conventional LV ejection fraction values decreased at day 13 in mice treated with either BVZ or SNT, TVI confirmed early LV systolic dysfunction at day 8. Histological and biochemical analysis demonstrated loss of cellular integrity, increased oxidative stress, and increased cardiac apoptosis in mice treated with BVZ or SNT therapy at day 14. In a murine model of BVZ- or SNT-mediated cardiomyopathy, noninvasive assessment by TVI detected early LV systolic dysfunction before alterations in conventional echocardiographic indexes.
Collapse
Affiliation(s)
- Kimberly-Ann Bordun
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sheena Premecz
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Megan daSilva
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Soma Mandal
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Vineet Goyal
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Tamara Glavinovic
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Matthew Cheung
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - David Cheung
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Christopher W. White
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Rakesh Chaudhary
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Darren H. Freed
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Hector R. Villarraga
- Department of Internal Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Joerg Herrmann
- Department of Internal Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Manish Kohli
- Division of Medical Oncology, Department of Oncology, Mayo Clinic, Rochester, Minnesota
| | - Amir Ravandi
- Section of Cardiology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
| | - James Thliveris
- Department of Human Anatomy and Cell Science, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Marshall Pitz
- Section of Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; and
| | - Pawan K. Singal
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Sharon Mulvagh
- Department of Internal Medicine, Division of Cardiovascular Diseases, Mayo Clinic, Rochester, Minnesota
| | - Davinder S. Jassal
- Institute of Cardiovascular Sciences, St. Boniface Research Centre, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Cardiology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada
- Section of Oncology, Department of Internal Medicine, University of Manitoba, Winnipeg, Manitoba, Canada; and
- Department of Radiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
86
|
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: 1.3] [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
|
87
|
Multimodality Imaging in Cardiooncology. JOURNAL OF ONCOLOGY 2015; 2015:263950. [PMID: 26300915 PMCID: PMC4537747 DOI: 10.1155/2015/263950] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2014] [Revised: 10/13/2014] [Accepted: 10/17/2014] [Indexed: 01/06/2023]
Abstract
Cardiotoxicity represents a rising problem influencing prognosis and quality of life of chemotherapy-treated patients. Anthracyclines and trastuzumab are the drugs most commonly associated with development of a cardiotoxic effect. Heart failure, myocardial ischemia, hypertension, myocarditis, and thrombosis are typical manifestation of cardiotoxicity by chemotherapeutic agents. Diagnosis and monitoring of cardiac side-effects of cancer treatment is of paramount importance. Echocardiography and nuclear medicine methods are widely used in clinical practice and left ventricular ejection fraction is the most important parameter to asses myocardial damage secondary to chemotherapy. However, left ventricular ejection decrease is a delayed phenomenon, occurring after a long stage of silent myocardial damage that classic imaging methods are not able to detect. New imaging techniques including three-dimensional echocardiography, speckle tracking echocardiography, and cardiac magnetic resonance have demonstrated high sensitivity in detecting the earliest alteration of left ventricular function associated with future development of chemotherapy-induced cardiomyopathy. Early diagnosis of cardiac involvement in cancer patients can allow for timely and adequate treatment management and the introduction of cardioprotective strategies.
Collapse
|
88
|
Luskin MR, Banerjee R, Del Percio S, Loren AW. A Pound of Cure Requires An Ounce (or More) of Prevention: Survivorship and Complications of Therapy for Hematologic Malignancies. Curr Hematol Malig Rep 2015; 10:225-36. [PMID: 26162948 DOI: 10.1007/s11899-015-0274-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Patients treated for a hematologic malignancy are at risk for treatment-related complications. As the goal of therapy is frequently curative, treatments are especially intensive and long-term toxicity is common. Chemotherapy and radiation are associated with increased risk for cardiac and pulmonary disease, endocrine disorders, infertility, sexual dysfunction, second cancers, and psychosocial distress. The risk for each complication is dictated by patient characteristics including age, co-morbidities, and genetic predispositions, as well as the specifics of therapy. Survivors of pediatric cancers and allogeneic hematopoietic stem cell transplantation have unique risks due to vulnerable age at time of toxic exposure and ongoing immune dysfunction, respectively.
Collapse
Affiliation(s)
- Marlise R Luskin
- Division of Hematology-Oncology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA,
| | | | | | | |
Collapse
|
89
|
Abdel-Rahman O, Alorabi M. Use of angiotensin-converting enzyme inhibitors in the prophylaxis of anthracycline or trastuzumab-related cardiac dysfunction: preclinical and clinical considerations. Expert Rev Anticancer Ther 2015; 15:829-837. [DOI: 10.1586/14737140.2015.1047766] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|
90
|
Abdel-Qadir H, Amir E, Thavendiranathan P. The use of myocardial strain and newer echocardiography imaging techniques in cancer patients. Future Oncol 2015. [DOI: 10.2217/fon.15.122] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Affiliation(s)
- Husam Abdel-Qadir
- Division of Cardiology, Women's College Hospital, Toronto, Canada
- Division of Cardiology, St Michael's Hospital, Toronto, Canada
| | - Eitan Amir
- Division of Medical Oncology, Princess Margaret Cancer Centre, University of Toronto, Toronto, Canada
| | - Paaladinesh Thavendiranathan
- Division of Cardiology, Peter Munk Cardiac Centre, Ted Rogers Program in Cardiotoxicity Prevention, University Health Network, University of Toronto, Toronto, Canada
| |
Collapse
|
91
|
Abstract
Improvements in therapies have significantly changed survival of cancer patients. However, the clinical history and oncologic treatment put cancer patients at higher risk for developing cardiovascular problems. Anthracyclines, but also the targeted therapy and angiogenesis inhibitors, are all treatments associated with cardiotoxicity. The most common adverse event is a reduction in left ventricular ejection fraction that may progress to overt heart failure. Recognition of a cardiac impairment during or after a potential cardiotoxic treatment requires a stringent assessment of clinical symptoms and signs of heart failure associated with an evaluation of the left ventricular ejection fraction, which, however, detects the damage already installed. Circulating cardiac biomarkers are promising in detecting cardiotoxicity and will likely change the approach for identifying patients at risk.
Collapse
Affiliation(s)
- Michela Salvatici
- Division of Laboratory Medicine, European Institute of Oncology, via Ripamonti, 435, 20141 Milan, Italy
| | - Maria T Sandri
- Division of Laboratory Medicine, European Institute of Oncology, via Ripamonti, 435, 20141 Milan, Italy
| |
Collapse
|
92
|
O'Hare M, Murphy K, Mookadam F, Sharma A, Lee H. Cardio-oncology Part II: the monitoring, prevention, detection and treatment of chemotherapeutic cardiac toxicity. Expert Rev Cardiovasc Ther 2015; 13:519-27. [PMID: 25864865 DOI: 10.1586/14779072.2015.1027686] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cancer treatment has advanced in recent years with new drugs, complex regimes and multiple modalities of treatment; which has improved survival of cancer patients. Cardiotoxicity from chemotherapy is an emerging, important issue. Currently, echocardiographic evaluation of ejection fraction is the most commonly employed diagnostic tool for detecting chemotherapy-induced cardiotoxicity. However, novel echocardiographic techniques assessing myocardial mechanics using strain imaging can detect earlier changes. New imaging techniques and biomarkers can risk stratify patients to identify those requiring closer monitoring. Cardiologists collaborating with oncologists can detect and treat cardiovascular chemotherapeutic complications earlier, reducing morbidity and mortality. While cardiac MRI and multigated acquisition nuclear scanning are alternatives, echocardiography has become the mainstream for assessing cardiac function due to its portability, efficiency and low cost. Current recommendations regarding cardiac monitoring of cancer patients are based on expert consensus opinion. There is a need for prospective controlled trials to support specific guidelines.
Collapse
Affiliation(s)
- Meabh O'Hare
- From the Division of Cardiovascular Diseases, Mayo Clinic, 13400 East Shea Boulevard, Scottsdale, AZ 85259, USA
| | | | | | | | | |
Collapse
|
93
|
Evaluation, prevention and management of cancer therapy-induced cardiotoxicity: a contemporary approach for clinicians. Curr Opin Cardiol 2015; 30:197-204. [PMID: 25574894 DOI: 10.1097/hco.0000000000000145] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW While targeted therapies have improved cancer outcomes, unique cardiovascular toxicities are increasingly recognized, particularly when administered sequentially after anthracyclines or radiation. Patients with cancer therapy-induced cardiotoxicity benefit from collaborative care involving cardiology and oncology, leading to a new interdisciplinary field called cardio-oncology. The present review will highlight contemporary clinical issues in cardio-oncology. RECENT FINDINGS Recently, risk factors for cancer therapy-induced cardiotoxicity have been evaluated in real-world rather than in clinical trial patients. Biomarkers and advanced echocardiography are emerging as sensitive tools for preclinical identification of cancer therapy-induced cardiotoxicity. Single-center studies suggest that cancer therapy-induced cardiotoxicity responds to prompt heart failure medical treatment, and such therapy may even prevent cardiotoxicity. SUMMARY Modern cancer therapy has short-term cardiac risk that may require collaborative management by clinicians with expertise in cardiology and oncology. The increased effectiveness of modern cancer therapy is resulting in a growing population of cancer survivors who are at long-term risk for cardiovascular disease. The present review of contemporary clinical issues in cardio-oncology will be of interest to healthcare providers who manage cardiotoxicity during cancer therapy, and who follow patients who survive cancer but face increased long-term cardiovascular risk.
Collapse
|
94
|
Yu AF, Steingart RM, Fuster V. Cardiomyopathy associated with cancer therapy. J Card Fail 2014; 20:841-52. [PMID: 25151211 PMCID: PMC5972392 DOI: 10.1016/j.cardfail.2014.08.004] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Revised: 08/08/2014] [Accepted: 08/14/2014] [Indexed: 01/03/2023]
Abstract
Chemotherapy-associated cardiomyopathy is a well known cardiotoxicity of contemporary cancer treatment and a cause of increasing concern for both cardiologists and oncologists. As cancer outcomes improve, cardiovascular disease has become a leading cause of morbidity and mortality among cancer survivors. Asymptomatic or symptomatic left ventricular systolic dysfunction in the setting of cardiotoxic chemotherapy is an important entity to recognize. Early diagnosis of cardiac injury through the use of novel blood-based biomarkers or noninvasive imaging modalities may allow for the initiation of cardioprotective medications or modification of chemotherapy regimen to minimize or prevent further damage. Several clinical trials are currently underway to determine the efficacy of cardioprotective medications for the prevention of chemotherapy-associated cardiomyopathy. Implementing a strategy that includes both early detection and prevention of cardiotoxicity will likely have a significant impact on the overall prognosis of cancer survivors. Continued coordination of care between cardiologists and oncologists remains critical to maximizing the oncologic benefit of cancer therapy while minimizing any early or late cardiovascular effects.
Collapse
Affiliation(s)
- Anthony F Yu
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York.
| | - Richard M Steingart
- Cardiology Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Valentin Fuster
- Zena and Michael A. Wiener Cardiovascular Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
95
|
Tian S, Hirshfield KM, Jabbour SK, Toppmeyer D, Haffty BG, Khan AJ, Goyal S. Serum biomarkers for the detection of cardiac toxicity after chemotherapy and radiation therapy in breast cancer patients. Front Oncol 2014; 4:277. [PMID: 25346912 PMCID: PMC4191171 DOI: 10.3389/fonc.2014.00277] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/23/2014] [Indexed: 12/31/2022] Open
Abstract
Multi-modality cancer treatments that include chemotherapy, radiation therapy, and targeted agents are highly effective therapies. Their use, especially in combination, is limited by the risk of significant cardiac toxicity. The current paradigm for minimizing cardiac morbidity, based on serial cardiac function monitoring, is suboptimal. An alternative approach based on biomarker testing, has emerged as a promising adjunct and a potential substitute to routine echocardiography. Biomarkers, most prominently cardiac troponins and natriuretic peptides, have been evaluated for their ability to describe the risk of potential cardiac dysfunction in clinically asymptomatic patients. Early rises in cardiac troponin concentrations have consistently predicted the risk and severity of significant cardiac events in patients treated with anthracycline-based chemotherapy. Biomarkers represent a novel, efficient, and robust clinical decision tool for the management of cancer therapy-induced cardiotoxicity. This article aims to review the clinical evidence that supports the use of established biomarkers such as cardiac troponins and natriuretic peptides, as well as emerging data on proposed biomarkers.
Collapse
Affiliation(s)
- Sibo Tian
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Kim M Hirshfield
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Salma K Jabbour
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Deborah Toppmeyer
- Division of Medical Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Bruce G Haffty
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Atif J Khan
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| | - Sharad Goyal
- Department of Radiation Oncology, Rutgers Cancer Institute of New Jersey and Rutgers Robert Wood Johnson Medical School , New Brunswick, NJ , USA
| |
Collapse
|
96
|
Nolan MT, Lowenthal RM, Venn A, Marwick TH. Chemotherapy-related cardiomyopathy: a neglected aspect of cancer survivorship. Intern Med J 2014; 44:939-50. [DOI: 10.1111/imj.12532] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2014] [Accepted: 07/09/2014] [Indexed: 12/12/2022]
Affiliation(s)
- M. T. Nolan
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| | - R. M. Lowenthal
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| | - A. Venn
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| | - T. H. Marwick
- Cardiovascular Research Department; Menzies Research Institute Tasmania; Hobart Tasmania Australia
| |
Collapse
|
97
|
Preemptive Cardioprotective Strategies in Patients Receiving Chemotherapy. CURRENT CARDIOVASCULAR RISK REPORTS 2014. [DOI: 10.1007/s12170-014-0406-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
98
|
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: 4.6] [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
|
99
|
D’Amore C, Gargiulo P, Paolillo S, Pellegrino AM, Formisano T, Mariniello A, Della Ratta G, Iardino E, D’Amato M, La Mura L, Fabiani I, Fusco F, Perrone Filardi P. Nuclear imaging in detection and monitoring of cardiotoxicity. World J Radiol 2014; 6:486-492. [PMID: 25071889 PMCID: PMC4109100 DOI: 10.4329/wjr.v6.i7.486] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Revised: 04/21/2014] [Accepted: 05/29/2014] [Indexed: 02/06/2023] Open
Abstract
Cardiotoxicity as a result of cancer treatment is a novel and serious public health issue that has a significant impact on a cancer patient’s management and outcome. The coexistence of cancer and cardiac disease in the same patient is more common because of aging population and improvements in the efficacy of antitumor agents. Left ventricular dysfunction is the most typical manifestation and can lead to heart failure. Left ventricular ejection fraction measurement by echocardiography and multigated radionuclide angiography is the most common diagnostic approach to detect cardiac damage, but it identifies a late manifestation of myocardial injury. Early non-invasive imaging techniques are needed for the diagnosis and monitoring of cardiotoxic effects. Although echocardiography and cardiac magnetic resonance are the most commonly used imaging techniques for cardiotoxicity assessment, greater attention is focused on new nuclear cardiologic techniques, which can identify high-risk patients in the early stage and visualize the pathophysiologic process at the tissue level before clinical manifestation. The aim of this review is to summarize the role of nuclear imaging techniques in the non-invasive detection of myocardial damage related to antineoplastic therapy at the reversible stage, focusing on the current role and future perspectives of nuclear imaging techniques and molecular radiotracers in detection and monitoring of cardiotoxicity.
Collapse
|
100
|
Jensen MM, Schmidt U, Huang C, Zerahn B. Gated tomographic radionuclide angiography using cadmium-zinc-telluride detector gamma camera; comparison to traditional gamma cameras. J Nucl Cardiol 2014; 21:384-96. [PMID: 24366823 DOI: 10.1007/s12350-013-9844-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 12/02/2013] [Indexed: 10/25/2022]
Abstract
PURPOSE Estimation of left ventricular ejection fraction (LVEF) with equilibrium 99MTc-HSA equilibrium radionuclide angiography (MUGA) is frequently used for assessing cardiac function. The purpose of this study was to compare intra- and interobserver variation between three different gamma cameras. MATERIALS AND METHODS Eighty-two patients, scanned in the same sequential order on the three cameras. Each acquisition was analyzed twice by two technologists. Inter- and intraobserver variations were calculated as the coefficient of variation and the 95% confidence interval for limits of agreement between each sequence of analyses for each of the three cameras. RESULTS The lowest intraobserver variations in LVEF for the two NaI-detector cameras were 3.1% (-4.0% to 3.5%) for the planar and 3.4% (-4.2% to 4.5%) for SPECT (P ≤ 0.001-0.019), the highest result for the CZT SPECT camera was 2.6% (-2.9% to 3.1%). Similarly, interobserver variation was 4.8% (-4.8% to 6.4%) and 4.9% (-5.4% to 7.5%), respectively, for each of the NaI-detector cameras and 3.3% (-3.4% to 4.3%) for the CZT SPECT camera (P ≤ 0.001-0.008). DISCUSSION The CZT detector camera was superior to both NaI detector cameras regarding intra- and interobserver variation. The CZT SPECT camera may identify changes in LVEF with greater certainty than its NaI detector-equipped counterparts.
Collapse
Affiliation(s)
- Maria Maj Jensen
- Department of Clinical Physiology and Nuclear Medicine, Copenhagen University Hospital Herlev, Herstedvesterstræde 54, 2620, Albertslund, Denmark,
| | | | | | | |
Collapse
|