1
|
Uslu H, Atila Uslu G, Çiçek B, Bolat İ, Yıldırım S. Trigonelline alkaloid is effective in preventing doxorubicin-induced lung damage. Arch Physiol Biochem 2024:1-8. [PMID: 39287053 DOI: 10.1080/13813455.2024.2404097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/21/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND One of the most popular chemotherapy medications is doxorubicin (DOX), however it can have non-negligible damage. When the underlying mechanisms of damage are investigated, the most prominent pathways are oxidative stress, inflammation and apoptosis. AIM We investigated the NF-κB/MAPK inflammatory pathway and cellular apoptosis to determine the efficacy of trigonelline alkaloid (TRIG) in preventing DOX-induced lung injury. METHODOLOGY The study consisted of C, TRIG, DOX and TRIG+DOX groups. TRIG and TRIG+DOX groups received 50 mg/kg TRIG for 7 days. On day 8, DOX and TRIG+DOX groups received a single dose of 15 mg/kg DOX. RESULTS Our results showed that apoptosis markers and inflammation were higher in the DOX group. In contrast, TRIG pretreatment partially suppressed apoptosis and decreased inflammation by blocking the activation of the MAPK/NF-κB pathway, lowering IL-6 levels, and protecting the lung from apoptotic cell death. CONCLUSION Assessing TRIG's effectiveness in lung tissue injury, this study may be a crucial first step.
Collapse
Affiliation(s)
- Hamit Uslu
- Department of Physiology, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Gözde Atila Uslu
- Department of Physiology, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - Betül Çiçek
- Department of Physiology, Erzincan Binali Yıldırım University, Erzincan, Türkiye
| | - İsmail Bolat
- Department of Pathology, Atatürk University, Erzurum, Türkiye
| | - Serkan Yıldırım
- Department of Pathology, Atatürk University, Erzurum, Türkiye
| |
Collapse
|
2
|
Bertolini D, Pizzi C, Donal E, Galli E. Cancer and Heart Failure: Dangerous Liaisons. J Cardiovasc Dev Dis 2024; 11:263. [PMID: 39330321 PMCID: PMC11432566 DOI: 10.3390/jcdd11090263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/28/2024] Open
Abstract
Cancer and heart failure (HF) are increasingly relevant worldwide, both from an epidemiologic and clinical point of view. This review aims to explore the relationship between cancer and HF by underscoring risk factors and disclosing the cardiotoxic effects of the current chemotherapy agents. We also deal with the current evidence on the diagnosis and management of HF related to cancer therapy. Finally, we will address the main gaps in knowledge and future perspectives in this field.
Collapse
Affiliation(s)
- Davide Bertolini
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Carmine Pizzi
- Cardiology Unit, Cardiac Thoracic and Vascular Department, IRCCS Azienda Ospedaliera-Universitaria di Bologna, 40138 Bologna, Italy
- Department of Medical and Surgical Sciences-DIMEC, Alma Mater Studiorum, University of Bologna, 40138 Bologna, Italy
| | - Erwan Donal
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| | - Elena Galli
- Univ Rennes, CHU Rennes, Inserm, LTSI-UMR 1099, F-35000 Rennes, France
| |
Collapse
|
3
|
Bostany G, Chen Y, Francisco L, Dai C, Meng Q, Sparks J, Sessions M, Nabell L, Stringer-Reasor E, Khoury K, Lenneman C, Keene K, Armenian S, Landier W, Bhatia S. Cardiac Dysfunction Among Breast Cancer Survivors: Role of Cardiotoxic Therapy and Cardiovascular Risk Factors. J Clin Oncol 2024:JCO2301779. [PMID: 38833638 DOI: 10.1200/jco.23.01779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Revised: 03/15/2024] [Accepted: 04/04/2024] [Indexed: 06/06/2024] Open
Abstract
PURPOSE Cardiac dysfunction is the leading cause of mortality among 10-year breast cancer survivors. Limited information regarding long-term risks of cardiac dysfunction after cardiotoxic therapy (anthracyclines, trastuzumab/pertuzumab, radiation) has precluded development of surveillance guidelines for the survivors. METHODS Patients with breast cancer who completed cardiotoxic therapy underwent echocardiographic screening every 2 years. New-onset cardiac dysfunction was defined as left ventricular ejection fraction (LVEF) <50% after cardiotoxic therapy initiation and included early- and late-onset cardiac dysfunction. RESULTS We evaluated 2,808 echocardiograms in 829 breast cancer survivors; the median age at breast cancer diagnosis was 54.2 years (range, 20.3-86.3); the median follow-up was 8.6 years (1.8-39.8); 39.7% received anthracyclines, 16% received trastuzumab/pertuzumab, 6.2% received both anthracyclines and trastuzumab/pertuzumab, and 38.1% received radiation alone. The cumulative incidence of cardiac dysfunction increased from 1.8% at 2 years to 15.3% at 15 years from cardiotoxic therapy initiation. Multivariable Cox regression analysis identified the following risk factors: non-Hispanic Black race (hazard ratio [HR], 2.15 [95% CI], 1.37 to 3.38), cardiotoxic therapies (anthracyclines: HR, 2.35 [95% CI, 1.25 to 4.4]; anthracyclines and trastuzumab/pertuzumab: HR, 3.92 [95% CI, 1.74 to 8.85]; reference: left breast radiation alone), selective estrogen receptor modulators (HR, 2.0 [95% CI, 1.2 to 3.33]), and precancer hypertension (HR, 3.16 [95% CI, 1.63 to 6.1]). Late-onset cardiac dysfunction was most prevalent among anthracycline- and radiation-exposed patients; early-onset cardiac dysfunction was most prevalent among patients exposed to anthracyclines and trastuzumab/pertuzumab; equal prevalence of both early- and late-onset cardiac dysfunction was observed in trastuzumab-/pertuzumab-exposed patients. Adjusted longitudinal analyses revealed an annual decline in LVEF by 0.29% (P = .009) over 20 years from breast cancer diagnosis. CONCLUSION These findings provide evidence to support echocardiographic surveillance for several years after cardiotoxic therapy and also suggest a need to examine the efficacy of management of cardiovascular risk factors to mitigate risk.
Collapse
Affiliation(s)
- Geoffrey Bostany
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Yanjun Chen
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Liton Francisco
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Chen Dai
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Qingrui Meng
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Jessica Sparks
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Min Sessions
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
| | - Lisle Nabell
- Division of Hematology/Oncology, UAB, Birmingham, AL
| | | | - Katia Khoury
- Division of Hematology/Oncology, UAB, Birmingham, AL
| | | | | | - Saro Armenian
- Department of Pediatric Oncology, City of Hope, Duarte, CA
| | - Wendy Landier
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
- Division of Pediatric Hematology/Oncology, UAB, Birmingham, AL
| | - Smita Bhatia
- Institute for Cancer Outcomes and Survivorship, University of Alabama at Birmingham (UAB) Heersink School of Medicine (SOM), Birmingham, AL
- Division of Pediatric Hematology/Oncology, UAB, Birmingham, AL
| |
Collapse
|
4
|
Li B, Zhang J, Ma N, Li W, You G, Chen G, Zhao L, Wang Q, Zhou H. PEG-conjugated bovine haemoglobin enhances efficiency of chemotherapeutic agent doxorubicin with alleviating DOX-induced splenocardiac toxicity in the breast cancer. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:120-130. [PMID: 36905212 DOI: 10.1080/21691401.2023.2176865] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
Doxorubicin (DOX) is an effective chemotherapeutic agent widely used for cancer treatment. However, hypoxia in tumour tissue and obvious adverse effects particularly cardiotoxicity restricts the clinical usage of DOX. Our study is based on the co-administration of haemoglobin-based oxygen carriers (HBOCs) and DOX in a breast cancer model to investigate HBOCs' ability to enhance chemotherapeutic effectiveness and its capabilities to alleviate the side effects induced by DOX. In an in-vitro study, the results suggested the cytotoxicity of DOX was significantly improved when combined with HBOCs in a hypoxic environment, and produced more γ-H2AX indicating higher DNA damage than free DOX did. Compared with administration of free DOX, combined therapy exhibited a stronger tumour suppressive effect in an in-vivo study. Further mechanism studies showed that the expression of various proteins such as hypoxia-inducible factor-1α (HIF-1α), CD31, CD34, and vascular endothelial growth factor (VEGF) in tumour tissues was also significantly reduced in the combined treatment group. In addition, HBOCs can significantly reduce the splenocardiac toxicity induced by DOX, according to the results of the haematoxylin and eosin (H&E) staining and histological investigation. This study suggested that PEG-conjugated bovine haemoglobin may not only reduce the hypoxia in tumours and increase the efficiency of chemotherapeutic agent DOX, but also alleviate the irreversible heart toxicity caused by DOX-inducted splenocardiac dysregulation.
Collapse
Affiliation(s)
- Bingting Li
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China
| | - Jun Zhang
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China.,The Western Theater General Hospital, Chengdu, P. R. China
| | - Ning Ma
- Clinical Laboratory of Beijing Huairou Hospital, Beijing, P. R. China
| | - Weidan Li
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China
| | - Guoxing You
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China
| | - Gan Chen
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China
| | - Lian Zhao
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China
| | - Quan Wang
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China
| | - Hong Zhou
- Institute of Health Service and Transfusion Medicine, Beijing, P. R. China
| |
Collapse
|
5
|
Xu C, Ou E, Li Z, Chen Z, Jia Q, Xu X, Luo L, Xu G, Liu J, Yuan Z, Zhao Y. Synthesis and in vivo evaluation of new steviol derivatives that protect against cardiomyopathy by inhibiting ferroptosis. Bioorg Chem 2022; 129:106142. [PMID: 36150232 DOI: 10.1016/j.bioorg.2022.106142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/30/2022] [Accepted: 09/06/2022] [Indexed: 11/02/2022]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of death globally. Inhibiting ferroptosis and thus preventing cardiac cell death is a promising and effective strategy for cardiomyopathy prevention and therapy. Steviol, an ent-kaurene diterpenoid, possesses broad-spectrum bioactivity. In the present study, with the aim to discover new agents for CVDs treatment, 30 derivatives of steviol, including 22 new ones, were synthesized, and evaluated their protective activity in vivo using the doxorubicin (DOX) induced zebrafish cardiomyopathy model. Our results firstly demonstrated that steviol has promising cardioprotective activity and further modification of steviol can greatly improve the activity. Among the new derivatives, 16d and 16e show the most potent activity. Both 16d (1 μM) and 16e (0.1 μM) effectively maintain the normal heart shape and prevent the cardiac dysfunction impaired by DOX in zebrafish. Their therapeutic efficacy is much superior to the parent natural product, steviol, and positive drug, levosimendan. Further study demonstrated that 16d and 16e inhibit DOX-induced ferroptosis and thus protect cardiomyopathy, by suppressing the glutathione depletion, iron accumulation, and lipid peroxidation, decreasing reactive oxygen species overaccumulation, and restoring the mitochondrial membrane potential. Consequently, due to their unique structure and significant cardioprotective activity with ferroptosis inhibition, new steviol derivatives 16d and 16e merit further research for the development of new cardioprotective drug candidates.
Collapse
Affiliation(s)
- Chao Xu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - E Ou
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhiyin Li
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhenyu Chen
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Qi Jia
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Xiaojia Xu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Liping Luo
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Geng Xu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Jiansong Liu
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China
| | - Zhengqiang Yuan
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| | - Yu Zhao
- School of Biomedical and Pharmaceutical Sciences, Guangdong University of Technology, Guangzhou 510006, China.
| |
Collapse
|
6
|
Ganz PA, Hays RD, Spritzer KL, Rogatko A, Ko CY, Colangelo LH, Arora A, Hopkins JO, Evans TL, Yothers G. Health-related quality of life outcomes after neoadjuvant chemoradiotherapy for rectal cancer in NRG Oncology/NSABP R-04. Cancer 2022; 128:3233-3242. [PMID: 35749631 PMCID: PMC9540677 DOI: 10.1002/cncr.34341] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/10/2022] [Accepted: 05/12/2022] [Indexed: 11/17/2022]
Abstract
Background There has been limited evaluation of health‐related quality of life (HRQOL) in rectal cancer patients receiving neoadjuvant chemoradiotherapy. HRQOL outcomes in the National Surgical Adjuvant Breast and Bowel Project R‐04 trial are examined in this article. Methods Between 2004 and 2010, R‐04 patients were invited to enroll in the HRQOL substudy, with questionnaires administered before randomization, after completion of chemoradiotherapy, and 1‐year after surgery. HRQOL measures included: Functional Assessment of Cancer Therapy for colorectal cancer (FACT‐C); Short Form‐36v.2 Vitality scale; a treatment‐specific symptom scale; and the FACT neurotoxicity scale. A 5‐year postsurgery assessment was added to the protocol in 2012. Mixed‐effects models examined neoadjuvant therapy treatment effects in the 1‐year sample and models that explored associations of host factors and treatment impact on 5‐year HRQOL. Results A total of 1373 patients completed baseline HRQOL and at least one additional assessment. The average age was 58 years (range, 23–85 years), male (68%), and 59% Stage II. There were no statistically significant differences in HRQOL outcomes by treatment arm, but HRQOL worsened from baseline to postneoadjuvant chemoradiotherapy, with statistically significant effect sizes changes ranging from 0.6 (Vitality) to 0.9 (FACT‐C Trial Outcome Index). Neurotoxicity was greater in the oxaliplatin‐treated groups. Obese/overweight patients had statistically significantly worse FACT‐C Trial Outcome Index scores than did underweight/normal weight groups. At 5 years, younger patients and those with normal baseline weight had statistically significantly better physical function scores and older patients had better mental health outcomes. Conclusions HRQOL did not differ across the four R‐04 treatment arms; however, host factors explained significant variation in posttreatment HRQOL. ClinicalTrials.gov: NCT00058474 (https://ClinicalTrials.gov/ct2/show/NCT00058474). Lay summary This article reports on the health‐related quality of life (HRQOL) outcomes of patients treated with four different chemotherapy regimens combined with radiation in rectal cancer patients before definitive surgical treatment. There were no significant differences in HRQOL by treatment regimen, but all patients experienced decreased vitality (energy) and physical functioning. By 1 year after treatment, most patients had returned to pretreatment vitality and physical functioning, with the exception of increased neurotoxicity. In a subsample of patients assessed at 5 years after surgery, physical function was better in those who at pretreatment were younger, normal weight, and had better performance status. Mental function was better in those who at pretreatment were older and had better performance status.
.
Collapse
Affiliation(s)
- Patricia A Ganz
- UCLA Jonsson Comprehensive Cancer Center at UCLA, Los Angeles, California, USA.,Health Policy & Management, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Ron D Hays
- Health Policy & Management, UCLA Fielding School of Public Health, Los Angeles, California, USA.,Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Karen L Spritzer
- Division of General Internal Medicine and Health Services Research, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - André Rogatko
- Cedars-Sinai Health System, Los Angeles, California, USA
| | - Clifford Y Ko
- Health Policy & Management, UCLA Fielding School of Public Health, Los Angeles, California, USA
| | - Linda H Colangelo
- NRG Oncology Statistics and Data Management Center, Pittsburgh, Pennsylvania, USA.,The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Amit Arora
- Kaiser Permanente NCI Community Oncology Research Program, Fremont, California, USA
| | - Judith O Hopkins
- Novant Health (Forsyth Medical) Cancer Institute/Southeast Clinical Oncology Research (SCOR) NCORP, Winston-Salem, North Carolina, USA
| | - Terry L Evans
- The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Greg Yothers
- NRG Oncology Statistics and Data Management Center, Pittsburgh, Pennsylvania, USA.,The University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
7
|
Sheibani M, Azizi Y, Shayan M, Nezamoleslami S, Eslami F, Farjoo MH, Dehpour AR. Doxorubicin-Induced Cardiotoxicity: An Overview on Pre-clinical Therapeutic Approaches. Cardiovasc Toxicol 2022; 22:292-310. [DOI: 10.1007/s12012-022-09721-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/12/2022] [Indexed: 12/20/2022]
|
8
|
Mensah SA, Nersesyan AA, Ebong EE. Endothelial Glycocalyx-Mediated Intercellular Interactions: Mechanisms and Implications for Atherosclerosis and Cancer Metastasis. Cardiovasc Eng Technol 2020; 12:72-90. [PMID: 33000443 PMCID: PMC7904750 DOI: 10.1007/s13239-020-00487-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 09/11/2020] [Indexed: 12/21/2022]
Abstract
Purpose The endothelial glycocalyx (GCX) plays a critical role in the health of the vascular system. Degradation of the GCX has been implicated in the onset of diseases like atherosclerosis and cancer because it disrupts endothelial cell (EC) function that is meant to protect from atherosclerosis and cancer. Examples of such EC function include interendothelial cell communication via gap junctions and receptor-mediated interactions between endothelial and tumor cells. This review focuses on GCX-dependent regulation of these intercellular interactions in healthy and diseased states. The ultimate goal is to build new knowledge that can be applied to developing GCX regeneration strategies that can control intercellular interaction in order to combat the progression of diseases such as atherosclerosis and cancer. Methods In vitro and in vivo studies were conducted to determine the baseline expression of GCX in physiologically relevant conditions. Chemical and mechanical GCX degradation approaches were employed to degrade the GCX. The impact of intact versus degraded GCX on intercellular interactions was assessed using cytochemistry, histochemistry, a Lucifer yellow dye transfer assay, and confocal, intravital, and scanning electron microscopy techniques. Results Relevant to atherosclerosis, we found that GCX stability determines the expression and functionality of Cx43 in gap junction-mediated EC-to-EC communication. Relevant to cancer metastasis, we found that destabilizing the GCX through either disturbed flow-induced or enzyme induced GCX degradation results in increased E-selectin receptor-mediated EC-tumor cell interactions. Conclusion Our findings lay a foundation for future endothelial GCX-targeted therapy, to control intercellular interactions and limit the progression of atherosclerosis and cancer.
Collapse
Affiliation(s)
- Solomon A Mensah
- Department of Bioengineering, Northeastern University, Boston, MA, USA.,Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, USA
| | - Alina A Nersesyan
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Eno E Ebong
- Department of Bioengineering, Northeastern University, Boston, MA, USA. .,Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, 335 Interdisciplinary Science and Engineering Complex, Boston, MA, 02115, USA. .,Department of Neuroscience, Albert Einstein College of Medicine, New York, NY, USA.
| |
Collapse
|
9
|
Risk of heart failure after systemic treatment for early breast cancer: results of a cohort study. Breast Cancer Res Treat 2020; 185:205-214. [PMID: 32964358 DOI: 10.1007/s10549-020-05930-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 09/05/2020] [Indexed: 12/31/2022]
Abstract
PURPOSE Anthracyclines and trastuzumab can increase the risk of heart failure (HF), but long-term cardiotoxicity data in breast cancer (BC) patients treated at younger ages are limited. Furthermore, it is unknown whether aromatase inhibitors are associated with HF risk. METHODS HF risk was studied in a multicenter cohort of BC survivors treated during 2000-2009, at age < 61 years. Information on treatment and cardiovascular disease incidence was collected through medical records, general practitioners and cardiologists. Analyses included multivariable Cox regression and cumulative incidence curves. RESULTS In total, 10,209 women with a median age at BC diagnosis of 50.3 years and a median follow-up of 8.9 years were enrolled in the study. Anthracycline-based chemotherapy was associated with HF (hazard ratio [HR] 2.18, 95% confidence interval [CI] 1.41-3.39) and risk increased with increasing cumulative anthracycline dose. For trastuzumab, HF risk was highest within the first 2 years after treatment (HR0-2 years: 13.06, 95% CI 5.70-29.92) and decreased thereafter (HR2-4 years: 4.84, 95% CI 1.99-11.75 and HR≥4 years: 0.64, 95% CI 0.23-1.81). The 10-year cumulative incidence of HF was 4.8% (95% CI 3.2-6.8) among patients treated with anthracyclines and trastuzumab. One-third of patients who developed HF after trastuzumab had long-term impaired cardiac function. Patients treated with aromatase inhibitors alone also had higher HF risk (HR 2.18, 95% CI 1.24-3.82) compared to patients not receiving endocrine therapy. CONCLUSIONS Our results stress the importance of considering anthracycline-free regimens in BC patients who need trastuzumab-containing treatment. The association between aromatase inhibitors and HF needs confirmation.
Collapse
|
10
|
Angier HE, Marino M, Springer RJ, Schmidt TD, Huguet N, DeVoe JE. The Affordable Care Act improved health insurance coverage and cardiovascular-related screening rates for cancer survivors seen in community health centers. Cancer 2020; 126:3303-3311. [PMID: 32294251 PMCID: PMC7340351 DOI: 10.1002/cncr.32900] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 03/20/2020] [Accepted: 03/23/2020] [Indexed: 02/05/2023]
Abstract
Background This study assessed the impact of Affordable Care Act (ACA) Medicaid expansion on health insurance rates and receipt of cardiovascular‐related preventive screenings (body mass index, glycated hemoglobin [HbA1c], low‐density lipoproteins, and blood pressure) for cancer survivors seen in community health centers (CHCs). Methods This study identified cancer survivors aged 19 to 64 years with at least 3 CHC visits in 13 states from the Accelerating Data Value Across a National Community Health Center Network (ADVANCE). Via inverse probability of treatment weighting multilevel multinomial modeling, insurance rates before and after the ACA were estimated by whether a patient lived in a state that expanded Medicaid, and changes between a pre‐ACA time period and 2 post‐ACA time periods were assessed. Results The weighted estimated sample size included 409 cancer survivors in nonexpansion states and 2650 in expansion states. In expansion states, the proportion of uninsured cancer survivors decreased significantly from 20.3% in 2012‐2013 to 4.5%in 2016‐2017, and the proportion of those with Medicaid coverage increased significantly from 38.8% to 55.6%. In nonexpansion states, there was a small decrease in uninsurance rates (from 33.6% in 2012‐2013 to 22.5% in 2016‐2017). Cardiovascular‐related preventive screening rates increased over time in both expansion and nonexpansion states: HbA1c rates nearly doubled from the pre‐ACA period (2012‐2013) to the post‐ACA period (2016‐2017) in expansion states (from 7.2% to 12.8%) and nonexpansion states (from 9.3% to 16.8%). Conclusions This study found a substantial decline in uninsured visits among cancer survivors in Medicaid expansion states. Yet, 1 in 5 cancer survivors living in a state that did not expand Medicaid remained uninsured. Several ACA provisions likely worked together to increase cardiovascular‐related preventive screening rates for cancer survivors seen in CHCs. The Affordable Care Act (ACA) provides coverage options for cancer survivors seen in community health centers, especially in states that have expanded Medicaid; unfortunately, 1 in 5 cancer survivors living in a state that has not expanded Medicaid coverage eligibility remains uninsured. The ACA Medicaid expansion provision change, likely in tandem with other ACA changes, has also contributed to modest improvements in rates of cardiovascular‐related screenings for cancer survivors.
Collapse
Affiliation(s)
- Heather E Angier
- Family Medicine, Oregon Health & Science University, Portland, Oregon
| | - Miguel Marino
- Family Medicine, Oregon Health & Science University, Portland, Oregon
| | - Rachel J Springer
- Family Medicine, Oregon Health & Science University, Portland, Oregon
| | | | - Nathalie Huguet
- Family Medicine, Oregon Health & Science University, Portland, Oregon
| | - Jennifer E DeVoe
- Family Medicine, Oregon Health & Science University, Portland, Oregon
| |
Collapse
|
11
|
Boekel NB, Duane FK, Jacobse JN, Hauptmann M, Schaapveld M, Sonke GS, Gietema JA, Hooning MJ, Seynaeve CM, Maas AH, Darby SC, Aleman BM, Taylor CW, van Leeuwen FE. Heart failure after treatment for breast cancer. Eur J Heart Fail 2020; 22:366-374. [PMID: 31721395 PMCID: PMC7137787 DOI: 10.1002/ejhf.1620] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 05/10/2019] [Accepted: 08/23/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND We aimed to develop dose-response relationships for heart failure (HF) following radiation and anthracyclines in breast cancer treatment, and to assess HF associations with trastuzumab and endocrine therapies. METHODS AND RESULTS A case-control study was performed within a cohort of breast cancer survivors treated during 1980-2009. Cases (n = 102) had HF as first cardiovascular diagnosis and were matched 1:3 on age and date of diagnosis. Individual cardiac radiation doses were estimated, and anthracycline doses and use of trastuzumab and endocrine therapy were abstracted from oncology notes. For HF cases who received radiotherapy, the estimated median mean heart dose (MHD) was 6.8 Gy [interquartile range (IQR) 0.9-13.7]. MHD was not associated with HF risk overall [excess rate ratio (ERR) = 1%/Gy, 95% confidence interval (CI) -2 to 10]. In patients treated with anthracyclines, exposure of ≥20% of the heart to ≥20 Gy was associated with a rate ratio of 5.7 (95% CI 1.7-21.7) compared to <10% exposed to ≥20 Gy. For cases who received radiotherapy, median cumulative anthracycline dose was 247 mg/m2 (IQR 240-319). A dose-dependent increase was observed after anthracycline without trastuzumab (ERR = 1.5% per mg/m2 , 95% CI 0.5-4.1). After anthracycline and trastuzumab, the rate ratio was 34.9 (95% CI 11.1-110.1) compared to no chemotherapy. CONCLUSIONS In absence of anthracyclines, breast cancer radiotherapy was not associated with increased HF risk. Strongly elevated HF risks were observed after treatment with anthracyclines and also after treatment with trastuzumab. The benefits of these systemic treatments usually exceed the risks of HF, but our results emphasize the need to support ongoing efforts to evaluate preventative strategies.
Collapse
Affiliation(s)
- Naomi B. Boekel
- EpidemiologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Fran K. Duane
- Medical Research Council Population Health Research UnitUniversity of OxfordOxfordUK
- Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | - Judy N. Jacobse
- EpidemiologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | | | | | - Gabe S. Sonke
- Medical OncologyNetherlands Cancer InstituteAmsterdamThe Netherlands
| | - Jourik A. Gietema
- Medical OncologyUniversity Medical Center GroningenGroningenThe Netherlands
| | | | | | - Angela H.E.M. Maas
- Department of CardiologyRadboud University Medical CentreNijmegenThe Netherlands
| | - Sarah C. Darby
- Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | | | - Carolyn W. Taylor
- Nuffield Department of Population HealthUniversity of OxfordOxfordUK
| | | |
Collapse
|
12
|
Jacobse JN, Steggink LC, Sonke GS, Schaapveld M, Hummel YM, Steenbruggen TG, Lefrandt JD, Nuver J, Crijns APG, Aleman BMP, van der Meer P, Gietema JA, van Leeuwen FE. Myocardial dysfunction in long-term breast cancer survivors treated at ages 40-50 years. Eur J Heart Fail 2019; 22:338-346. [PMID: 31696625 PMCID: PMC7077738 DOI: 10.1002/ejhf.1610] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2019] [Revised: 07/04/2019] [Accepted: 08/11/2019] [Indexed: 12/12/2022] Open
Abstract
Aims Anthracyclines increase heart failure (HF) risk, but the long‐term prevalence of myocardial dysfunction in young breast cancer (BC) survivors is unknown. Early measures of left ventricular myocardial dysfunction are needed to identify BC patients at risk of symptomatic HF. Methods and results Within an established cohort, we studied markers for myocardial dysfunction among 569 women, who were 5–7 years (n = 277) or 10–12 years (n = 292) after BC treatment at ages 40–50 years. Left ventricular ejection fraction (LVEF) and global longitudinal strain (GLS) were assessed by echocardiography. N‐terminal pro‐brain natriuretic peptide (NT‐proBNP) was measured in serum. Associations between patient‐related and treatment‐related risk factors and myocardial dysfunction were evaluated using linear and logistic regression. Median ages at BC diagnosis and cardiac assessment were 46.7 and 55.5 years, respectively. Anthracycline‐treated patients (n = 313), compared to the no‐anthracycline group (n = 256), more often had decreased LVEF (10% vs. 4%), impaired GLS (34% vs. 27%) and elevated NT‐proBNP (23% vs. 8%). GLS and LVEF declined in a linear fashion with increasing cumulative anthracycline dose (GLS: +0.23 and LVEF: −0.40 per cycle of 60 mg/m2; P < 0.001) and GLS was worse for patients with left breast irradiation. The risk of NT‐proBNP >125 ng/L was highest for patients who received 241–300 mg/m2 anthracycline dose compared to the no‐anthracycline group (odds ratio: 3.30, 95% confidence interval: 1.83–5.96). Conclusion Impaired GLS and increased NT‐proBNP levels are present in a substantial proportion of young BC survivors treated with anthracyclines. Whether this will lead to future cardiac disease needs to be evaluated by longitudinal assessment.
Collapse
Affiliation(s)
- Judy N Jacobse
- Department of Epidemiology & Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Lars C Steggink
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Gabe S Sonke
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michael Schaapveld
- Department of Epidemiology & Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Yoran M Hummel
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Tessa G Steenbruggen
- Department of Medical Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Joop D Lefrandt
- Department of Vascular Medicine, University Medical Center Groningen, Groningen, The Netherlands
| | - Janine Nuver
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Anne P G Crijns
- Department of Radiation Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Berthe M P Aleman
- Department of Radiation Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University Medical Center Groningen, Groningen, The Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University Medical Center Groningen, Groningen, The Netherlands
| | - Flora E van Leeuwen
- Department of Epidemiology & Biostatistics, Netherlands Cancer Institute, Amsterdam, The Netherlands
| |
Collapse
|
13
|
The Evolving Design of NIH-Funded Cardio-Oncology Studies to Address Cancer Treatment-Related Cardiovascular Toxicity. JACC: CARDIOONCOLOGY 2019; 1:105-113. [PMID: 32529192 PMCID: PMC7288847 DOI: 10.1016/j.jaccao.2019.08.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Cardiovascular (CV) toxicity from cancer therapy is a significant and growing concern. Conventional oncology clinical trial designs focused solely on cancer treatment efficacy have not provided sufficient information on both CV risk factors and outcomes. Similarly, traditional CV trials evaluating standard interventions typically exclude cancer patients, particularly those actively receiving cancer therapy. Neither trial type simultaneously evaluates the balance between CV toxicity and cancer outcomes; however, there is increasing collaboration among oncologists and cardiologists to design new cardio-oncology trials that address this important need. In this review, we detail 5 ongoing, oncology-based trials with integrated CV endpoints. Key design features include: 1) a careful assessment of CV risk factors and disease before, during, and after cancer therapy with standardized collection of clinical imaging, functional, and biomarker data; 2) an introduction of cardioprotective interventions at various timepoints in cancer therapy; 3) a balance of the risk of subclinical CV injury with the need for ongoing cancer treatment; and 4) an understanding of the time profile for development of clinically apparent CV toxicity. Additional critical priorities in cardio-oncology clinical research include harmonization of data collection and definitions for all physician- and patient-reported exposures and outcomes. Prospective assessment of CV risk factors before, during, and after cancer treatment. Longitudinal monitoring of CV function with standardized review of CV imaging and functional and biomarker endpoints for evidence of subclinical cardiotoxicity. Consideration for the timing of the introduction of the cardioprotective strategy. Need to balance the delivery of cancer treatment with the risk of CV injury. Long-term follow-up beyond cancer treatment intervention to determine clinical cardiotoxicity outcomes. Rigorous collection of cancer and CV endpoints to answer questions about the impact of CV events on the delivery of cancer treatment and the long-term patient outcomes.
Collapse
|
14
|
Hochheiser L, Hornberger J, Turner M, Lyman GH. Multi-gene assays: effect on chemotherapy use, toxicity and cost in estrogen receptor-positive early stage breast cancer. J Comp Eff Res 2019; 8:289-304. [DOI: 10.2217/cer-2018-0137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Aim: To assess multi-gene assay (MGA) effects on chemotherapy use, toxicities, recurrences, and costs in estrogen receptor-positive early breast cancer. Methods: Meta-analysis performed using data from public databases. Results: Studies included 12,202 women. Relative to no testing, chemotherapy use was higher with 12-gene and 70-gene and lower with PAM50 (commercial) and 21-gene MGAs. Overall, 1643 distant recurrences occurred with no testing, declining by 231 (21-gene), 121 (70-gene), 54 (12-gene) and 94 (PAM50); only the 21-gene assay resulted in no risk of increasing the number of distant recurrences. Relative to ‘no testing’, total cost of care declined only with 21-gene MGA. Conclusion: MGAs differ in chemotherapy use and related outcomes for women with estrogen receptor-positive early breast cancer.
Collapse
Affiliation(s)
- Lou Hochheiser
- Professor Emeritus, Department of Family Practice, University of Vermont, Burlington, VT 83001, USA
| | | | | | - Gary H Lyman
- Fred Hutchinson Cancer Center & The University of Washington, Seattle, WA 98109, USA
| |
Collapse
|
15
|
Chang L, Weiner LS, Hartman SJ, Horvath S, Jeste D, Mischel PS, Kado DM. Breast cancer treatment and its effects on aging. J Geriatr Oncol 2019; 10:346-355. [PMID: 30078714 PMCID: PMC7062379 DOI: 10.1016/j.jgo.2018.07.010] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/11/2018] [Accepted: 07/16/2018] [Indexed: 10/28/2022]
Abstract
Breast cancer is the most common cancer of women in the United States. It is also proving to be one of the most treatable. Early detection, surgical intervention, therapeutic radiation, cytotoxic chemotherapies and molecularly targeted agents are transforming the lives of patients with breast cancer, markedly improving their survival. Although current breast cancer treatments are largely successful in producing cancer remission and extending lifespan, there is concern that these treatments may have long lasting detrimental effects on cancer survivors, in part, through their impact on non-tumor cells. Presently, the impact of breast cancer treatment on normal cells, its impact on cellular function and its effect on the overall function of the individual are incompletely understood. In particular, it is unclear whether breast cancer and/or its treatments are associated with an accelerated aging phenotype. In this review, we consider breast cancer survivorship from the perspective of accelerated aging, and discuss the evidence suggesting that women treated for breast cancer may suffer from an increased rate of physical and cognitive decline that likely corresponds with underlying vulnerabilities of genome instability, epigenetic changes, and cellular senescence.
Collapse
Affiliation(s)
- Leslie Chang
- Departments of Family Medicine & Public Health, School of Medicine, University of California, San Diego, United States; Department of Internal Medicine, School of Medicine University of California, San Diego, United States
| | - Lauren S Weiner
- Departments of Family Medicine & Public Health, School of Medicine, University of California, San Diego, United States; University of California San Diego, Moores Cancer Center, La Jolla, CA, United States
| | - Sheri J Hartman
- Departments of Family Medicine & Public Health, School of Medicine, University of California, San Diego, United States; University of California San Diego, Moores Cancer Center, La Jolla, CA, United States
| | - Steve Horvath
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, United States; Department of Biostatistics, Fielding School of Public Health, University of California, Los Angeles, United States
| | - Dilip Jeste
- Departments of Psychiatry & Neuroscience, University of California, San Diego, United States; Sam and Rose Stein Institute for Research on Aging, United States
| | - Paul S Mischel
- Department of Pathology, School of Medicine, University of California, San Diego, United States; Ludwig Institute for Cancer Research, University of California, San Diego, United States
| | - Deborah M Kado
- Departments of Family Medicine & Public Health, School of Medicine, University of California, San Diego, United States; Department of Internal Medicine, School of Medicine University of California, San Diego, United States; Sam and Rose Stein Institute for Research on Aging, United States.
| |
Collapse
|
16
|
Boekel NB, Jacobse JN, Schaapveld M, Hooning MJ, Gietema JA, Duane FK, Taylor CW, Darby SC, Hauptmann M, Seynaeve CM, Baaijens MHA, Sonke GS, Rutgers EJT, Russell NS, Aleman BMP, van Leeuwen FE. Cardiovascular disease incidence after internal mammary chain irradiation and anthracycline-based chemotherapy for breast cancer. Br J Cancer 2018; 119:408-418. [PMID: 30065254 PMCID: PMC6133926 DOI: 10.1038/s41416-018-0159-x] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 04/14/2018] [Accepted: 06/07/2018] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Improved breast cancer (BC) survival and evidence showing beneficial effects of internal mammary chain (IMC) irradiation underscore the importance of studying late cardiovascular effects of BC treatment. METHODS We assessed cardiovascular disease (CVD) incidence in 14,645 Dutch BC patients aged <62 years, treated during 1970-2009. Analyses included proportional hazards models and general population comparisons. RESULTS CVD rate-ratio for left-versus-right breast irradiation without IMC was 1.11 (95% CI 0.93-1.32). Compared to right-sided breast irradiation only, IMC irradiation (interquartile range mean heart doses 9-17 Gy) was associated with increases in CVD rate overall, ischaemic heart disease (IHD), heart failure (HF) and valvular heart disease (hazard ratios (HRs): 1.6-2.4). IHD risk remained increased until at least 20 years after treatment. Anthracycline-based chemotherapy was associated with an increased HF rate (HR = 4.18, 95% CI 3.07-5.69), emerging <5 years and remaining increased at least 10-15 years after treatment. IMC irradiation combined with anthracycline-based chemotherapy was associated with substantially increased HF rate (HR = 9.23 95% CI 6.01-14.18), compared to neither IMC irradiation nor anthracycline-based chemotherapy. CONCLUSIONS Women treated with anthracycline-based chemotherapy and IMC irradiation (in an older era) with considerable mean heart dose exposure have substantially increased incidence of several CVDs. Screening may be appropriate for some BC patient groups.
Collapse
Affiliation(s)
- Naomi B Boekel
- Epidemiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Judy N Jacobse
- Epidemiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Michael Schaapveld
- Epidemiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Maartje J Hooning
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| | - Jourik A Gietema
- Medical Oncology, University Medical Center Groningen, Hanzeplein 1, 9213 GZ, Groningen, The Netherlands
| | - Frances K Duane
- Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
- Medical Research Council Population Health Research Unit, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Carolyn W Taylor
- Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Sarah C Darby
- Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, UK
| | - Michael Hauptmann
- Epidemiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Caroline M Seynaeve
- Department of Medical Oncology, Erasmus MC - Cancer Institute, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| | - Margreet H A Baaijens
- Radiation Oncology, Erasmus MC - Cancer Institute, Groene Hilledijk 301, 3075 EA, Rotterdam, The Netherlands
| | - Gabe S Sonke
- Medical Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Emiel J T Rutgers
- Surgery, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Nicola S Russell
- Radiation Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Berthe M P Aleman
- Radiation Oncology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands
| | - Flora E van Leeuwen
- Epidemiology, Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX, Amsterdam, The Netherlands.
| |
Collapse
|
17
|
Govender J, Loos B, Marais E, Engelbrecht AM. Melatonin improves cardiac and mitochondrial function during doxorubicin-induced cardiotoxicity: A possible role for peroxisome proliferator-activated receptor gamma coactivator 1-alpha and sirtuin activity? Toxicol Appl Pharmacol 2018; 358:86-101. [PMID: 29966675 DOI: 10.1016/j.taap.2018.06.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 06/27/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023]
Abstract
Mitochondrial dysfunction is a central element in the development of doxorubicin (DXR)-induced cardiotoxicity. In this context, melatonin is known to influence mitochondrial homeostasis and function. This study aimed to investigate the effects of melatonin on cardiac function, tumor growth, mitochondrial fission and fusion, PGC1-α and sirtuin activity in an acute model of DXR-induced cardiotoxicity. During the in vitro study, H9c2 rat cardiomyoblasts were pre-treated with melatonin (10 μM, 24 h) followed by DXR exposure (3 μM, 24 h). Following treatment, cellular ATP levels and mitochondrial morphology were assessed. In the in vivo study, female Sprague Dawley rats (16 weeks old), were inoculated with a LA7 rat mammary tumor cell line and tumors were measure daily. Animals were injected with DXR (3 × 4 mg/kg) and/or received melatonin (6 mg/kg) for 14 days in their drinking water. Rat hearts were used to conduct isolated heart perfusions to assess cardiac function and thereafter, heart tissue was used for immunoblot analysis. DXR treatment increased cell death and mitochondrial fission which were reduced with melatonin treatment. Cardiac output increased in rats treated with DXR + melatonin compared to DXR-treated rats. Tumor volumes was significantly reduced in DXR + melatonin-treated rats on Day 8 in comparison to DXR-treated rats. Furthermore, DXR + melatonin treatment increased cellular ATP levels, PGC1-α and SIRT1 expression which was attenuated by DXR treatment. These results indicate that melatonin treatment confers a dual cardio-protective and oncostatic effect by improving mitochondrial function and cardiac function whilst simultaneously retarding tumor growth during DXR-induced cardiotoxicity.
Collapse
Affiliation(s)
- Jenelle Govender
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa.
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| | - Erna Marais
- Department of Medical Physiology, Faculty of Medicine, Stellenbosch University, Tygerberg Campus, 7505, South Africa
| | - Anna-Mart Engelbrecht
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch 7600, South Africa
| |
Collapse
|
18
|
Ganz PA, Romond EH, Cecchini RS, Rastogi P, Geyer CE, Swain SM, Jeong JH, Fehrenbacher L, Gross HM, Brufsky AM, Flynn PJ, Wahl TA, Seay TE, Wade JL, Biggs DD, Atkins JN, Polikoff J, Zapas JL, Mamounas EP, Wolmark N. Long-Term Follow-Up of Cardiac Function and Quality of Life for Patients in NSABP Protocol B-31/NRG Oncology: A Randomized Trial Comparing the Safety and Efficacy of Doxorubicin and Cyclophosphamide (AC) Followed by Paclitaxel With AC Followed by Paclitaxel and Trastuzumab in Patients With Node-Positive Breast Cancer With Tumors Overexpressing Human Epidermal Growth Factor Receptor 2. J Clin Oncol 2017; 35:3942-3948. [PMID: 29072977 DOI: 10.1200/jco.2017.74.1165] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose Early cardiac toxicity is a risk associated with adjuvant chemotherapy plus trastuzumab. However, objective measures of cardiac function and health-related quality of life are lacking in long-term follow-up of patients who remain cancer free after completion of adjuvant treatment. Patients and Methods Patients in NSABP Protocol B-31 received anthracycline and taxane chemotherapy with or without trastuzumab for adjuvant treatment of node-positive, human epidermal growth factor receptor 2-positive early-stage breast cancer. A long-term follow-up assessment was undertaken for patients who were alive and disease free, which included measurement of left ventricular ejection fraction by multigated acquisition scan along with patient-reported outcomes using the Duke Activity Status Index (DASI), the Medical Outcomes Study questionnaire, and a review of current medications and comorbid conditions. Results At a median follow-up of 8.8 years among eligible participants, five (4.5%) of 110 in the control group and 10 (3.4%) of 297 in the trastuzumab group had a > 10% decline in left ventricular ejection fraction from baseline to a value < 50%. Lower DASI scores correlated with age and use of medications for hypertension, cardiac conditions, diabetes, and hyperlipidemia, but not with whether patients had received trastuzumab. Conclusion In patients without underlying cardiac disease at baseline, the addition of trastuzumab to adjuvant anthracycline and taxane-based chemotherapy does not result in long-term worsening of cardiac function, cardiac symptoms, or health-related quality of life. The DASI questionnaire may provide a simple and useful tool for monitoring patient-reported changes that reflect cardiac function.
Collapse
Affiliation(s)
- Patricia A Ganz
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Edward H Romond
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Reena S Cecchini
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Priya Rastogi
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Charles E Geyer
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Sandra M Swain
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Jong-Hyeon Jeong
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Louis Fehrenbacher
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Howard M Gross
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Adam M Brufsky
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Patrick J Flynn
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Tanya A Wahl
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Thomas E Seay
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - James L Wade
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - David D Biggs
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - James N Atkins
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Jonathan Polikoff
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - John L Zapas
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Eleftherios P Mamounas
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| | - Norman Wolmark
- All authors: National Surgical Adjuvant Breast and Bowel Project/NRG Oncology; Reena S. Cecchini, Priya Rastogi, Jong-Hyeon Jeong, and Adam M. Brufsky, University of Pittsburgh; Priya Rastogi and Adam M. Brufsky, Magee-Womens Hospital; Norman Wolmark, Allegheny Health Network Cancer Institute, Pittsburgh, PA; Patricia A. Ganz, University of California at Los Angeles, Los Angeles; Louis Fehrenbacher, Kaiser Permanente, Vallejo; Jonathan Polikoff, Kaiser Permanente, San Marco, CA; Edward H. Romond, University of Kentucky, Lexington, KY; Charles E. Geyer Jr, Virginia Commonwealth University, Richmond, VA; Sandra M. Swain and John L. Zapas, MedStar Washington Hospital Center, Washington, DC; Howard M. Gross, Dayton National Cancer Institute Community Oncology Research Program (NCORP), Dayton, OH; Patrick J. Flynn, Metro-Minnesota Community Clinical Oncology Program (CCOP), Woodbury, MN; Tanya A. Wahl, Fred Hutchinson Cancer Research Center, Seattle, WA; Thomas E. Seay, Atlanta Regional CCOP, Atlanta, GA; James L. Wade III, Heartland NCORP, Decatur, IL; David D. Biggs, Christiana Care Health System, Newark, DE; James N. Atkins, Southeast Clinical Oncology Research Consortium NCORP, Goldsboro, NC; and Eleftherios P. Mamounas, Orlando Health, Orlando, FL
| |
Collapse
|
19
|
Boerman LM, Maass SW, van der Meer P, Gietema JA, Maduro JH, Hummel YM, Berger MY, de Bock GH, Berendsen AJ. Long-term outcome of cardiac function in a population-based cohort of breast cancer survivors: A cross-sectional study. Eur J Cancer 2017; 81:56-65. [DOI: 10.1016/j.ejca.2017.05.013] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2017] [Revised: 04/28/2017] [Accepted: 05/06/2017] [Indexed: 12/28/2022]
|
20
|
Armenian SH, Lacchetti C, Barac A, Carver J, Constine LS, Denduluri N, Dent S, Douglas PS, Durand JB, Ewer M, Fabian C, Hudson M, Jessup M, Jones LW, Ky B, Mayer EL, Moslehi J, Oeffinger K, Ray K, Ruddy K, Lenihan D. Prevention and Monitoring of Cardiac Dysfunction in Survivors of Adult Cancers: American Society of Clinical Oncology Clinical Practice Guideline. J Clin Oncol 2017; 35:893-911. [DOI: 10.1200/jco.2016.70.5400] [Citation(s) in RCA: 652] [Impact Index Per Article: 93.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Purpose Cardiac dysfunction is a serious adverse effect of certain cancer-directed therapies that can interfere with the efficacy of treatment, decrease quality of life, or impact the actual survival of the patient with cancer. The purpose of this effort was to develop recommendations for prevention and monitoring of cardiac dysfunction in survivors of adult-onset cancers. Methods Recommendations were developed by an expert panel with multidisciplinary representation using a systematic review (1996 to 2016) of meta-analyses, randomized clinical trials, observational studies, and clinical experience. Study quality was assessed using established methods, per study design. The guideline recommendations were crafted in part using the Guidelines Into Decision Support methodology. Results A total of 104 studies met eligibility criteria and compose the evidentiary basis for the recommendations. The strength of the recommendations in these guidelines is based on the quality, amount, and consistency of the evidence and the balance between benefits and harms. Recommendations It is important for health care providers to initiate the discussion regarding the potential for cardiac dysfunction in individuals in whom the risk is sufficiently high before beginning therapy. Certain higher risk populations of survivors of cancer may benefit from prevention and screening strategies implemented during cancer-directed therapies. Clinical suspicion for cardiac disease should be high and threshold for cardiac evaluation should be low in any survivor who has received potentially cardiotoxic therapy. For certain higher risk survivors of cancer, routine surveillance with cardiac imaging may be warranted after completion of cancer-directed therapy, so that appropriate interventions can be initiated to halt or even reverse the progression of cardiac dysfunction.
Collapse
Affiliation(s)
- Saro H. Armenian
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Christina Lacchetti
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Ana Barac
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Joseph Carver
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Louis S. Constine
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Neelima Denduluri
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Susan Dent
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Pamela S. Douglas
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Jean-Bernard Durand
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Michael Ewer
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Carol Fabian
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Melissa Hudson
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Mariell Jessup
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Lee W. Jones
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Bonnie Ky
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Erica L. Mayer
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Javid Moslehi
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Kevin Oeffinger
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Katharine Ray
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Kathryn Ruddy
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| | - Daniel Lenihan
- Saro H. Armenian, City of Hope, Duarte, CA; Christina Lacchetti, American Society of Clinical Oncology, Alexandria; Neelima Denduluri, Virginia Cancer Specialists, Arlington, VA; Ana Barac, Medstar Heart Institute, Medstar Washington Hospital Center, Washington, DC; Joseph Carver and Mariell Jessup, University of Pennsylvania; Bonnie Ky, Hospital of the University of Pennsylvania, Philadelphia, PA; Louis S. Constine, University of Rochester Medical Center, Rochester; Lee W. Jones and Kevin Oeffinger,
| |
Collapse
|
21
|
Unger JM, Till C, Thompson IM, Tangen CM, Goodman PJ, Wright JD, Barlow WE, Ramsey SD, Minasian LM, Hershman DL. Long-term Consequences of Finasteride vs Placebo in the Prostate Cancer Prevention Trial. J Natl Cancer Inst 2016; 108:djw168. [PMID: 27565902 DOI: 10.1093/jnci/djw168] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2015] [Accepted: 05/31/2016] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Finasteride has been found to reduce the risk of low-grade prostate cancer but to have no impact on overall survival. The long-term adverse and beneficial consequences of finasteride have not been examined. METHODS We used a linkage between data from the Prostate Cancer Prevention Trial (PCPT) and Medicare claims. Patients were examined by randomized study arm (finasteride vs placebo for 7 years) for long-term consequences of the intervention, including cardiac, endocrine, and sexual dysfunction, depression, diabetes, and benign prostatic hyperplasia (BPH)-related events. To examine time to events, we used cumulative incidence and Cox regression, adjusting for covariates. All statistical tests were two-sided. RESULTS A total of 13 935 of 18 880 participants (73.8%) in the PCPT were linked to Medicare claims, with median Medicare follow-up assessment time of 16 years from trial registration. There were no differences between finasteride and placebo participants with respect to important baseline factors or amount of Medicare follow-up assessment time. Finasteride patients had a 10% higher risk of new claims for depression (hazard ratio [HR] = 1.10, 95% confidence interval [CI] = 1.01 to 1.19, P = .04) and a 6% lower risk of procedures for BPH-related events (primarily lower urinary tract symptoms; HR = 0.94, 95% CI = 0.89 to 1.00, P = .03). No other differences were found in rates of long-term consequences of intervention in the two study arms. CONCLUSIONS Finasteride use is associated with reduced need for procedures for relief of BPH-related events and a modest increase in depression. Overall, there is little need to worry about long-term noncancer consequences of finasteride use in those who use it for treatment of symptomatic BPH, hair growth, or prevention of cancer.
Collapse
Affiliation(s)
- Joseph M Unger
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Cathee Till
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Ian M Thompson
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Catherine M Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Phyllis J Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Jason D Wright
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - William E Barlow
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Scott D Ramsey
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Lori M Minasian
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| | - Dawn L Hershman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA (JMU, CT, CMT, PJG, WEB); Cancer Therapy and Research Center, University of Texas Health Science Center at San Antonio, San Antonio, TX (IMTJr); Columbia University, New York, NY (JDW, DLH); Fred Hutchinson Cancer Research Center, Seattle, WA (SDR); Division of Cancer Prevention, U.S. National Cancer Institute, Rockville, MD (LMM)
| |
Collapse
|
22
|
Gavila J, Seguí MÁ, Calvo L, López T, Alonso JJ, Farto M, Sánchez-de la Rosa R. Evaluation and management of chemotherapy-induced cardiotoxicity in breast cancer: a Delphi study. Clin Transl Oncol 2016; 19:91-104. [PMID: 27101413 PMCID: PMC5215075 DOI: 10.1007/s12094-016-1508-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/25/2016] [Indexed: 12/03/2022]
Abstract
Purpose While much progress has been made in the treatment of breast cancer, cardiac complications resulting from therapy remain a significant concern. Both anthracyclines and novel targeted agents can inflict cardiac damage. The present study aimed to evaluate the difference between what it is currently done and what standards of care should be used to minimizing and managing cardiac toxicity in breast cancer survivors. Methods A two-round multicenter Delphi study was carried out. The panel consisted of 100 oncologists who were asked to define the elected therapies for breast cancer patients, the clinical definition and patterns of cancer drug-derived cardiac toxicity, and those protocols focused on early detection and monitoring of cardiovascular outcomes. Results Experts agreed a more recent definition of cardiotoxicity. Around 38 % of patients with early-stage disease, and 51.3 % cases with advanced metastatic breast cancer had preexisting risk factors for cardiotoxicity. Among risk factors, cumulative dose of anthracycline ≥450 mg/m2 and its combination with other anticancer drugs, and a preexisting cardiovascular disease were considered the best predictors of cardiotoxicity. Echocardiography and radionuclide ventriculography have been the proposed methods for monitoring changes in cardiac structure and function. Breast cancer is generally treated with anthracyclines (80 %), so that the panel strongly stated about the need to plan a strategy to managing cardiotoxicity. A decline of left ventricular ejection fraction (LVEF) >10 %, to an LVEF value <53 % was suggested as a criterion for changing the dose schedule of anthracyclines, or suspending the treatment of chemotherapy plus trastuzumab until the normalization of the left ventricular function. The use of liposomal anthracyclines was strongly suggested as a treatment option for breast cancer patients. Conclusions The present report is the first to produce a set of statements on the prevention, evaluation and monitoring of chemotherapy-induced cardiac toxicity in breast cancer patients.
Collapse
Affiliation(s)
- J Gavila
- Servicio de Oncología Médica, Fundación Instituto Valenciano de Oncología, Calle del Profesor Beltrán Bàguena, 8, 46009, Valencia, Spain.
| | - M Á Seguí
- Servicio de Oncología Médica, Corporació Sanitaria ParcTaulí, Barcelona, Spain
| | - L Calvo
- Servicio de Oncología Médica, Complejo Hospitalario Universitario A Coruña, A Coruña, Spain
| | - T López
- Servicio de Cardiología, Hospital Universitario La Paz, Madrid, Spain
| | - J J Alonso
- Servicio de Cardiología, Hospital Universitario de Getafe, Madrid, Spain
| | - M Farto
- Medical Department, TEVA Pharma, Madrid, Spain
| | | |
Collapse
|
23
|
Ahmed SH, Moussa Sherif DE, Fouad Y, Kelany M, Abdel-Rahman O. Principles of a risk evaluation and mitigation strategy (REMS) for breast cancer patients receiving potentially cardiotoxic adjuvant treatments. Expert Opin Drug Saf 2016; 15:911-23. [DOI: 10.1517/14740338.2016.1170115] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
24
|
Abstract
Long term survival of childhood cancers is now more than 70%. Anthracyclines, including doxorubicin, are some of the most efficacious anticancer drugs available. However, its use as a chemotherapeutic agent is severely hindered by its dose-limiting toxicities. Most notably observed is cardiotoxicity, but other organ systems are also degraded by doxorubicin use. Despite the years of its use and the amount of information written about this drug, an understanding of its cellular mechanisms is not fully appreciated. The mechanisms by which doxorubicin induces cytotoxicity in target cancer cells have given insight about how the drug damages cardiomyocytes. The major mechanisms of doxorubicin actions are thought to be as an oxidant generator and as an inhibitor of topoisomerase 2. However, other signaling pathways are also invoked with significant consequences for the cardiomyocyte. Further the interaction between oxidant generation and topoisomerase function has only recently been appreciated and the consequences of this interaction are still not fully understood. The unfortunate consequences of doxorubicin within cardiomyocytes have promoted the search for new drugs and methods that can prevent or reverse the damage caused to the heart after treatment in cancer patients. Alternative protocols have lessened the impact on newly diagnosed cancer patients. However the years of doxorubicin use have generated a need for monitoring the onset of cardiotoxicity as well as understanding its potential long-term consequences. Although a fairly clear understanding of the short-term pathologic mechanisms of doxorubicin actions has been achieved, the long-term mechanisms of doxorubicin induced heart failure remain to be carefully delineated.
Collapse
Affiliation(s)
| | - John G. Edwards
- Corresponding author at: Department of Physiology, New York Medical College, 15 Dana Road, Valhalla, NY, United States.Department of PhysiologyNew York Medical College15 Dana RoadValhallaNYUnited States
| |
Collapse
|
25
|
Armenian SH, Xu L, Ky B, Sun C, Farol LT, Pal SK, Douglas PS, Bhatia S, Chao C. Cardiovascular Disease Among Survivors of Adult-Onset Cancer: A Community-Based Retrospective Cohort Study. J Clin Oncol 2016; 34:1122-30. [PMID: 26834065 DOI: 10.1200/jco.2015.64.0409] [Citation(s) in RCA: 349] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
PURPOSE Cardiovascular diseases (CVDs), including ischemic heart disease, stroke, and heart failure, are well-established late effects of therapy in survivors of childhood and young adult (< 40 years at diagnosis) cancers; less is known regarding CVD in long-term survivors of adult-onset (≥ 40 years) cancer. METHODS A retrospective cohort study design was used to describe the magnitude of CVD risk in 36,232 ≥ 2-year survivors of adult-onset cancer compared with matched (age, sex, and residential ZIP code) noncancer controls (n = 73,545) within a large integrated managed care organization. Multivariable regression was used to examine the impact of cardiovascular risk factors (CVRFs; hypertension, diabetes, dyslipidemia) on long-term CVD risk in cancer survivors. RESULTS Survivors of multiple myeloma (incidence rate ratio [IRR], 1.70; P < .01), carcinoma of the lung/bronchus (IRR, 1.58; P < .01), non-Hodgkin lymphoma (IRR, 1.41; P < .01), and breast cancer (IRR, 1.13; P < .01) had significantly higher CVD risk when compared with noncancer controls. Conversely, prostate cancer survivors had a lower CVD risk (IRR, 0.89; P < .01) compared with controls. Cancer survivors with two or more CVRFs had the highest risk of CVD when compared with noncancer controls with less than two CVRFs (IRR, 1.83 to 2.59; P < .01). Eight-year overall survival was significantly worse among cancer survivors who developed CVD (60%) when compared with cancer survivors without CVD (81%; P < .01). CONCLUSION The magnitude of subsequent CVD risk varies according to cancer subtype and by the presence of CVRFs. Overall survival in survivors who develop CVD is poor, emphasizing the need for targeted prevention strategies for individuals at highest risk of developing CVD.
Collapse
Affiliation(s)
- Saro H Armenian
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL.
| | - Lanfang Xu
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| | - Bonnie Ky
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| | - Canlan Sun
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| | - Leonardo T Farol
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| | - Sumanta Kumar Pal
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| | - Pamela S Douglas
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| | - Smita Bhatia
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| | - Chun Chao
- Saro H. Armenian, Canlan Sun, and Sumanta Kumar Pal, City of Hope Comprehensive Cancer Center, Duarte; Lanfang Xu and Chun Chao, Kaiser Permanente Southern California, Pasadena; Leonardo T. Farol, City of Hope-Kaiser Permanente, Los Angeles, CA; Bonnie Ky, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA; Pamela S. Douglas, Duke Clinical Research Institute, Duke University, Durham, NC; and Smita Bhatia, University of Alabama at Birmingham, Birmingham, AL
| |
Collapse
|
26
|
Hertz DL, Caram MV, Kidwell KM, Thibert JN, Gersch C, Seewald NJ, Smerage J, Rubenfire M, Henry NL, Cooney KA, Leja M, Griggs JJ, Rae JM. Evidence for association of SNPs in ABCB1 and CBR3, but not RAC2, NCF4, SLC28A3 or TOP2B, with chronic cardiotoxicity in a cohort of breast cancer patients treated with anthracyclines. Pharmacogenomics 2016; 17:231-40. [PMID: 26799497 PMCID: PMC5558515 DOI: 10.2217/pgs.15.162] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022] Open
Abstract
AIMS Validation of associations for SNPs in RAC2, NCF4 and SLC28A3, identification of a novel association with a TOP2B SNP and screening 23 SNPs putatively relevant to anthracycline-induced cardiotoxicity. PATIENTS & METHODS A total of 166 breast cancer patients treated with doxorubicin underwent echocardiogram, including 19 cases with systolic dysfunction (ejection fraction <55%) and 147 controls. Four high priority SNPs were tested in the primary analysis, with appropriate statistical correction, and 23 additional SNPs were screened in an uncorrected secondary analysis. RESULTS Previously reported associations for RAC2, NCF4 and SLC28A3 could not be validated and a novel association with TOP2B was not discovered in this cohort (all p > 0.05), likely due to inadequate power. Two SNPs were identified in the uncorrected secondary analysis including a protective SNP in ABCB1 (3435C>T, p = 0.049) and a risk allele in CBR3 (V244M, p = 0.012). CONCLUSION The associations reported in prior publications and those discovered in this secondary analysis require further replication in independent cohorts.
Collapse
Affiliation(s)
- Daniel L Hertz
- Department of Clinical Pharmacy, University of Michigan College of Pharmacy, Ann Arbor, MI 48109, USA
| | - Megan V Caram
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kelley M Kidwell
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jacklyn N Thibert
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Christina Gersch
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Nicholas J Seewald
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, USA
| | - Jeffrey Smerage
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Melvyn Rubenfire
- Department of Internal Medicine, Division of Cardiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - N Lynn Henry
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Kathleen A Cooney
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Urology, University of Michigan Health System, Ann Arbor, MI 48109, USA
| | - Monika Leja
- Department of Internal Medicine, Division of Cardiology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - Jennifer J Griggs
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| | - James M Rae
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, MI 48109, USA
| |
Collapse
|
27
|
Abstract
Introduction Breast cancer in young women (<50 years) has been associated with an increased risk of recurrence and decreased survival compared with patients older than 50. The objective of this analysis was to determine, from a large database of patients with early-stage breast cancer, if the Recurrence Score® result (Oncotype DX®, Genomic Health, Inc, Redwood City, CA, USA) provided clinically meaningful differences in predicted risk of recurrence in younger—compared with older—patients. Methods Tumor samples from patients with estrogen receptor (ER)-positive breast cancers that were successfully processed in the Genomic Health central lab between June 2004 and December 2013 for Recurrence Score and quantitative gene expression of ER, progesterone receptor (PR), and Her/2neu, were included. Descriptive statistics were used to describe the distribution of scores by age group: <40, 40–49, 50–59, 60–69, and ≥70 years, nodal status, and histologic subtype. Results Specimens from 394,031 patients [3.3% (n = 13,029) aged <40 years; 15.6% (n = 61,643) aged ≥70 years] were included; 81.6% of patients had invasive ductal carcinoma. Nodal status was specified for 362,001 patients (87.0% negative). Median Recurrence Score results were similar across risk groups. Low (<18)- and high (≥31)- risk Recurrence Score results were seen in 58.5% and 8.5% of patients, respectively. A greater proportion of patients aged <40 (14.1%) than ≥70 (8.8%) years had a high-risk score. ER expression increased as a function of age and PR single-gene and invasion gene group expression were similar across age groups. Conclusion These data indicate that in patients with ER-positive breast cancer, age alone does not reflect the underlying individual tumor biology, suggesting that the Recurrence Score result may add potentially useful information for personalized treatment decisions. Funding Genomic Health, Inc. Electronic supplementary material The online version of this article (doi:10.1007/s12325-015-0268-3) contains supplementary material, which is available to authorized users.
Collapse
|
28
|
Turner N, Biganzoli L, Di Leo A. Continued value of adjuvant anthracyclines as treatment for early breast cancer. Lancet Oncol 2015; 16:e362-9. [PMID: 26149888 DOI: 10.1016/s1470-2045(15)00079-0] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2014] [Revised: 11/18/2014] [Accepted: 11/18/2014] [Indexed: 01/15/2023]
Abstract
Anthracyclines are frequently used in the adjuvant treatment of early-stage breast cancer. However, with the increasing use of other active drugs--mainly taxanes and trastuzumab in HER2-positive disease--coupled with concerns about anthracycline-associated toxic effects, there is debate about whether anthracyclines are still needed. Three major factors should be taken into consideration with the investigation of the role of anthracyclines in management of early breast cancer; specifically, the proven efficacy of anthracyclines in breast cancer, the absence of superiority of non-anthracycline-based chemotherapy over anthracycline-taxane regimens, and the low risk of toxic effects associated with the cumulative doses of anthracyclines used in contemporary regimens. The risks remain substantially outweighed by the benefits of treatment with anthracyclines, and thus, they maintain an important role in adjuvant treatment of breast cancer, particularly in women with high-risk disease.
Collapse
Affiliation(s)
- Natalie Turner
- Sandro Pitigliani Medical Oncology Department, Prato Hospital, Istituto Toscano Tumori, Prato, Italy
| | - Laura Biganzoli
- Sandro Pitigliani Medical Oncology Department, Prato Hospital, Istituto Toscano Tumori, Prato, Italy
| | - Angelo Di Leo
- Sandro Pitigliani Medical Oncology Department, Prato Hospital, Istituto Toscano Tumori, Prato, Italy.
| |
Collapse
|
29
|
Caram MEV, Guo C, Leja M, Smerage J, Henry NL, Giacherio D, Rubenfire M, Schott A, Davis M, Hayes DF, Van Poznak C, Cooney KA, Hertz DL, Banerjee M, Griggs JJ. Doxorubicin-induced cardiac dysfunction in unselected patients with a history of early-stage breast cancer. Breast Cancer Res Treat 2015; 152:163-172. [PMID: 26050157 DOI: 10.1007/s10549-015-3454-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2015] [Accepted: 05/28/2015] [Indexed: 10/23/2022]
Abstract
Cardiomyopathy is a known complication of anthracycline-based adjuvant chemotherapy and is more commonly reported in population-based studies of breast cancer survivors than in clinical trials. This study prospectively evaluated the prevalence of elevated cardiac biomarkers in unselected patients who had been treated with doxorubicin for early-stage breast cancer and the prevalence of reduced LVEF in patients with an elevated biomarker. All participants underwent an examination, symptom inventory, medical record review, and biomarker analysis for BNP, troponin, and plasma and urine NT-proBNP. Patients who had one or more elevated biomarkers were referred for echocardiogram; systolic dysfunction was defined as LVEF less than 55 %. Multivariable logistic regression was used to determine the associations between age, BMI, cumulative dose of doxorubicin, diabetes, hypertension, and left-sided radiation therapy and the risk of reduced LVEF. Among the 269 patients who underwent lab testing (mean age 56 years, mean time since completion of doxorubicin-based chemotherapy 6 years), 192 (72 %) had one or more elevated biomarker. Among the 166 patients who completed an echocardiogram, 11.5 % had a LVEF < 55 %. After adjusting for covariates known to affect cardiac function, multivariable logistic regression revealed plasma NT-proBNP to be the only measured cardiac biomarker associated with systolic dysfunction. There is a relationship between NT-proBNP and the frequency of reduced LVEF in women treated with doxorubicin for curative intent; further study of NT-proBNP as a potential biomarker for subclinical cardiac dysfunction after exposure to anthracyclines is warranted.
Collapse
Affiliation(s)
- Megan E V Caram
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States.
| | - Cui Guo
- Department of Biostatistics, University of Michigan School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, United States
| | - Monika Leja
- Department of Internal Medicine, Division of Cardiology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive; Cardiovascular Center, Ann Arbor, MI, 48109, United States
| | - Jeffrey Smerage
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States
| | - N Lynn Henry
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States
| | - Donald Giacherio
- Department of Pathology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States
| | - Melvyn Rubenfire
- Department of Internal Medicine, Division of Cardiology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive; Cardiovascular Center, Ann Arbor, MI, 48109, United States
| | - Anne Schott
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States
| | - Melinda Davis
- Department of Internal Medicine, Division of Cardiology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive; Cardiovascular Center, Ann Arbor, MI, 48109, United States
| | - Daniel F Hayes
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States
| | - Catherine Van Poznak
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States
| | - Kathleen A Cooney
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States.,Department of Urology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States
| | - Daniel L Hertz
- Department of Pharmacology, College of Pharmacy, University of Michigan , 428 Church Street, Ann Arbor, MI, 48104-1065, United States
| | - Mousumi Banerjee
- Department of Biostatistics, University of Michigan School of Public Health, University of Michigan, 1415 Washington Heights, Ann Arbor, MI, 48109, United States
| | - Jennifer J Griggs
- Department of Internal Medicine, Division of Hematology/Oncology, University of Michigan Medical School, University of Michigan, 1500 E. Medical Center Drive, Ann Arbor, MI, 48109, United States.,Department of Health Management and Policy, University of Michigan School of Public Health, 1415 Washington Heights, Ann Arbor, 48109, United States
| |
Collapse
|
30
|
Hurria A, Muss H. Special Issues in Older Women with Breast Cancer. IMPROVING OUTCOMES FOR BREAST CANCER SURVIVORS 2015; 862:23-37. [DOI: 10.1007/978-3-319-16366-6_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
31
|
Reed SC, Partridge AH, Nekhlyudov L. Shared Medical Appointments in Cancer Survivorship Care: A Review of the Literature. J Oncol Pract 2014; 11:6-11. [PMID: 25424650 DOI: 10.1200/jop.2014.001411] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
PURPOSE The physical and psychological effects of a cancer diagnosis and treatment on an individual may be significant and require appropriate monitoring and management. Furthermore, attention to preventive care and comorbid medical conditions is critical. Innovative approaches are needed to provide quality care to this growing population. METHODS We reviewed the literature evaluating shared medical appointments (SMAs) in noncancer and cancer care settings. Supported by a conceptual framework and adhering to the goals of patient-centered communication, we propose how SMAs may be used in clinical practice to enhance survivorship care. RESULTS The literature suggests that SMAs are an effective model in noncancer settings, showing improved blood pressure and health-related quality of life, among others. Although evidence for SMAs in cancer care is limited, there is significant potential for improved patient outcomes and costs. In particular, SMAs may provide an opportunity to address medical and psychological needs while creating a structure for enhanced communication. CONCLUSION SMAs may offer an innovative care model for cancer survivors and their providers. Implementation and evaluation of SMAs in the care of cancer survivors is warranted.
Collapse
Affiliation(s)
- Sarah C Reed
- Betty Irene Moore School of Nursing, University of California Davis, Davis, CA; Dana-Farber Cancer Institute-Harvard Medical School; and Harvard Vanguard Medical Associates, Boston, MA
| | - Ann H Partridge
- Betty Irene Moore School of Nursing, University of California Davis, Davis, CA; Dana-Farber Cancer Institute-Harvard Medical School; and Harvard Vanguard Medical Associates, Boston, MA
| | - Larissa Nekhlyudov
- Betty Irene Moore School of Nursing, University of California Davis, Davis, CA; Dana-Farber Cancer Institute-Harvard Medical School; and Harvard Vanguard Medical Associates, Boston, MA
| |
Collapse
|
32
|
Govender J, Loos B, Marais E, Engelbrecht AM. Mitochondrial catastrophe during doxorubicin-induced cardiotoxicity: a review of the protective role of melatonin. J Pineal Res 2014; 57:367-80. [PMID: 25230823 DOI: 10.1111/jpi.12176] [Citation(s) in RCA: 120] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2014] [Accepted: 09/12/2014] [Indexed: 12/18/2022]
Abstract
Anthracyclines, such as doxorubicin, are among the most valuable treatments for various cancers, but their clinical use is limited due to detrimental side effects such as cardiotoxicity. Doxorubicin-induced cardiotoxicity is emerging as a critical issue among cancer survivors and is an area of much significance to the field of cardio-oncology. Abnormalities in mitochondrial functions such as defects in the respiratory chain, decreased adenosine triphosphate production, mitochondrial DNA damage, modulation of mitochondrial sirtuin activity and free radical formation have all been suggested as the primary causative factors in the pathogenesis of doxorubicin-induced cardiotoxicity. Melatonin is a potent antioxidant, is nontoxic, and has been shown to influence mitochondrial homeostasis and function. Although a number of studies support the mitochondrial protective role of melatonin, the exact mechanisms by which melatonin confers mitochondrial protection in the context of doxorubicin-induced cardiotoxicity remain to be elucidated. This review focuses on the role of melatonin on doxorubicin-induced bioenergetic failure, free radical generation, and cell death. A further aim is to highlight other mitochondrial parameters such as mitophagy, autophagy, mitochondrial fission and fusion, and mitochondrial sirtuin activity, which lack evidence to support the role of melatonin in the context of cardiotoxicity.
Collapse
Affiliation(s)
- Jenelle Govender
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | | | | | | |
Collapse
|
33
|
Crozier JA, Swaika A, Moreno-Aspitia A. Adjuvant chemotherapy in breast cancer: To use or not to use, the anthracyclines. World J Clin Oncol 2014; 5:529-538. [PMID: 25114866 PMCID: PMC4127622 DOI: 10.5306/wjco.v5.i3.529] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 05/14/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Breast cancer continues to be one of the leading causes of cancer mortality in the world. The treatment generally involves multiple modalities including surgery, radiation and/or chemotherapy. Anthracyclines, one of the first chemotherapeutic agents introduced in the 1960s, has been the backbone for the last 30 years and has been used extensively so far. However, the cardiac toxicity and the concern for secondary hematological malignancy has always been a challenge. A better understanding of the tumor biology, role of Her2 expression and the discovery of trastuzumab and other anti-Her 2 agents along with other effective novel therapeutic options, have revolutionized the treatment for breast cancer. The role of anthracyclines has come under close scrutiny, especially in the adjuvant setting for patients with early stage breast cancer and those with low or intermediate risk of disease recurrence. Recent studies have highlighted such a shift in the use of anthracyclines in both the academic and community clinical practice. However, in patients with a high risk of relapse, anthracyclines still hold promise. Ongoing clinical trials are underway to further define the role of anthracyclines in such a patient population. This review highlights the development, clinical utility, limitations and potential future use of anthracyclines in the adjuvant setting for patients with breast cancer. We consulted PubMed, Scopus, MEDLINE, ASCO annual symposium abstracts, and http://clinicaltrials.gov/ for the purpose of this review.
Collapse
|
34
|
Hill DA, Horick NK, Isaacs C, Domchek SM, Tomlinson GE, Lowery JT, Kinney AY, Berg JS, Edwards KL, Moorman PG, Plon SE, Strong LC, Ziogas A, Griffin CA, Kasten CH, Finkelstein DM. Long-term risk of medical conditions associated with breast cancer treatment. Breast Cancer Res Treat 2014; 145:233-43. [PMID: 24696430 DOI: 10.1007/s10549-014-2928-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2014] [Accepted: 03/18/2014] [Indexed: 11/30/2022]
Abstract
Early and late effects of cancer treatment are of increasing concern with growing survivor populations, but relevant data are sparse. We sought to determine the prevalence and hazard ratio of such effects in breast cancer cases. Women with invasive breast cancer and women with no cancer history recruited for a cancer research cohort completed a mailed questionnaire at a median of 10 years post-diagnosis or matched reference year (for the women without cancer). Reported medical conditions including lymphedema, osteopenia, osteoporosis, and heart disease (congestive heart failure, myocardial infarction, coronary heart disease) were assessed in relation to breast cancer therapy and time since diagnosis using Cox regression. The proportion of women currently receiving treatment for these conditions was calculated. Study participants included 2,535 women with breast cancer and 2,428 women without cancer (response rates 66.0 % and 50.4 %, respectively) Women with breast cancer had an increased risk of lymphedema (Hazard ratio (HR) 8.6; 95 % confidence interval (CI) 6.3-11.6), osteopenia (HR 2.1; 95 % CI 1.8-2.4), and osteoporosis (HR 1.5; 95 % CI 1.2-1.9) but not heart disease, compared to women without cancer Hazard ratios varied by treatment and time since diagnosis. Overall, 49.3 % of breast cancer cases reported at least one medical condition, and at 10 or more years post-diagnosis, 37.7 % were currently receiving condition-related treatment. Responses from survivors a decade following cancer diagnosis demonstrate substantial treatment-related morbidity, and emphasize the need for continued medical surveillance and follow-up care into the second decade post-diagnosis.
Collapse
Affiliation(s)
- Deirdre A Hill
- Department of Internal Medicine and Cancer Research and Treatment Center, University of New Mexico, MSC 10-5550, Albuquerque, NM, 87131-0001, USA,
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Steingart RM, Yadav N, Manrique C, Carver JR, Liu J. Cancer Survivorship: Cardiotoxic Therapy in the Adult Cancer Patient; Cardiac Outcomes With Recommendations for Patient Management. Semin Oncol 2013; 40:690-708. [DOI: 10.1053/j.seminoncol.2013.09.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
36
|
Bhatia S, Blanco JG, Landier W, Relling MV. Reply to C.D. Atkins. J Clin Oncol 2012. [DOI: 10.1200/jco.2012.44.8688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
37
|
Jain S, Wei J, Mitrani LR, Bishopric NH. Auto-acetylation stabilizes p300 in cardiac myocytes during acute oxidative stress, promoting STAT3 accumulation and cell survival. Breast Cancer Res Treat 2012; 135:103-14. [PMID: 22562121 DOI: 10.1007/s10549-012-2069-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 04/14/2012] [Indexed: 12/15/2022]
Abstract
The nuclear acetyltransferase p300 is rapidly and stably induced in the heart during hemodynamic stress, but the mechanism of this induction is unknown. To determine the role of oxidative stress in p300 induction, we exposed neonatal rat cardiac myocytes to doxorubicin (DOX, 1 μM) or its vehicle, and monitored p300 protein content and stability for 24 h. Levels of p300 rose substantially within 1 h and remained elevated for at least 24 h, while p300 transcript levels declined. In the presence of cycloheximide, the estimated half-life of p300 in control cells was approximately 4.5 h, typical of an immediate-early response protein. DOX treatment prolonged p300 t(1/2) to >24 h, indicating that the sharp rise in p300 levels was attributable to rapid protein stabilization. p300 stabilization was entirely due to an increase in acetylated p300 species with greatly enhanced resistance to proteasomal degradation. The half-life of p300 was dependent on its acetyltransferase activity, falling in the presence of p300 inhibitors curcumin and anacardic acid, and increasing with histone deacetylase (HDAC) inhibition. At the same time, acetyl-STAT3, phospho-STAT3-(Tyr 705) and -(Ser 727) increased, together with a prolongation of STAT3 half-life. SiRNA-mediated p300 knockdown abrogated all of these effects, and strongly enhanced DOX-mediated myocyte apoptosis. We conclude that DOX induces an acute amplification of p300 levels through auto-acetylation and stabilization. In turn, elevated p300 provides a key defense against acute oxidative stress in cardiac myocytes by acetylation, activation, and stabilization of STAT3. Our results suggest that HDAC inhibitors could potentially reduce acute anthracycline-mediated cardiotoxicity by promoting p300 auto-acetylation.
Collapse
Affiliation(s)
- Sumit Jain
- Departments of Molecular and Cellular Pharmacology, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | | | | | | |
Collapse
|
38
|
Mitochondria death/survival signaling pathways in cardiotoxicity induced by anthracyclines and anticancer-targeted therapies. Biochem Res Int 2012; 2012:951539. [PMID: 22482055 PMCID: PMC3318211 DOI: 10.1155/2012/951539] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/04/2012] [Accepted: 01/09/2012] [Indexed: 01/27/2023] Open
Abstract
Anthracyclines remain the cornerstone of treatment in many malignancies but these agents have a cumulative dose relationship with cardiotoxicity. Development of cardiomyopathy and congestive heart failure induced by anthracyclines are typically dose-dependent, irreversible, and cumulative. Although past studies of cardiotoxicity have focused on anthracyclines, more recently interest has turned to anticancer drugs that target many proteins kinases, such as tyrosine kinases. An attractive model to explain the mechanism of this cardiotoxicity could be myocyte loss through cell death pathways. Inhibition of mitochondrial transition permeability is a valuable tool to prevent doxorubicin-induced cardiotoxicity. In response to anthracycline treatment, activation of several protein kinases, neuregulin/ErbB2 signaling, and transcriptional factors modify mitochondrial functions that determine cell death or survival through the modulation of mitochondrial membrane permeability. Cellular response to anthracyclines is also modulated by a myriad of transcriptional factors that influence cell fate. Several novel targeted chemotherapeutic agents have been associated with a small but worrying risk of left ventricular dysfunction. Agents such as trastuzumab and tyrosine kinase inhibitors can lead to cardiotoxicity that is fundamentally different from that caused by anthracyclines, whereas biological effects converge to the mitochondria as a critical target.
Collapse
|
39
|
Yood MU, Wells KE, Alford SH, Dakki H, Beiderbeck AB, Hurria A, Gross CP, Oliveria SA. Cardiovascular outcomes in women with advanced breast cancer exposed to chemotherapy. Pharmacoepidemiol Drug Saf 2012; 21:818-27. [PMID: 22419528 DOI: 10.1002/pds.3239] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2011] [Revised: 01/26/2012] [Accepted: 01/27/2012] [Indexed: 11/06/2022]
Abstract
PURPOSE To quantify incidence of cardiovascular outcomes in patients with advanced breast cancer receiving cardiotoxic and non-cardiotoxic chemotherapy. METHODS This study identified all women at a Midwestern health system with initial diagnosis of American Joint Commission on Cancer Stage III/IV breast cancer (1995-2003) and random sample of 50 women initially diagnosed with Stage I/II who progressed to Stage III/IV. The rate of new cardiovascular outcomes (heart failure, dysrhythmia, and ischemia events) for cardiotoxic (anthracycline or trastuzumab) and non-cardiotoxic agents was calculated. RESULTS Of 315 patients, 90.5% (n = 285) received systemic cancer therapy; 67.7% (n = 193) received cardiotoxic drugs. Older patients were less likely to receive cardiotoxic agents (86.4%, ≤59 years vs. 31.9%, 70+ years). Adjusting for age, race, stage, surgery/radiation, estrogen receptor/progesterone receptor status, and diagnosis year, rate of new cardiac events was higher in patients exposed to cardiotoxic drugs compared with those exposed to non-cardiotoxic drugs (adjusted hazard ratio = 2.5, 95%CI = 0.9-7.2). Patients with cardiac event history (relative risk = 3.2, 95%CI = 2.0-5.1) and those with heart failure history (relative risk = 5.9, 95%CI = 2.4-14.6) were more likely to receive non-cardiotoxic treatment. Heart failure events occurred steadily over time; after 3 years of follow-up, 16% exposed to cardiotoxic drugs experienced an event, and 8% of those exposed to non-cardiotoxic drugs experienced an event. CONCLUSIONS Patients with cardiac comorbidity are less likely to receive cardiotoxic agents. Use of cardiotoxic agents is common; treatment is related to patient and tumor characteristics and is associated with substantial risk of cardiotoxicity that persists during patients' remaining lifespan.
Collapse
|
40
|
Bardia A, Arieas ET, Zhang Z, Defilippis A, Tarpinian K, Jeter S, Nguyen A, Henry NL, Flockhart DA, Hayes DF, Hayden J, Storniolo AM, Armstrong DK, Davidson NE, Fetting J, Ouyang P, Wolff AC, Blumenthal RS, Ashen MD, Stearns V. Comparison of breast cancer recurrence risk and cardiovascular disease incidence risk among postmenopausal women with breast cancer. Breast Cancer Res Treat 2011; 131:907-14. [PMID: 22042368 DOI: 10.1007/s10549-011-1843-1] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Accepted: 10/15/2011] [Indexed: 12/21/2022]
Abstract
The majority of breast cancers are diagnosed in postmenopausal women. Competing comorbidities, particularly cardiovascular disease (CVD), should be considered when individualizing adjuvant therapies for these women. We compared the 10-year predicted breast cancer recurrence risk with CVD risk among postmenopausal women with hormone receptor-positive (HR+), non-metastatic breast cancer. CVD risk factor data were prospectively collected from postmenopausal women with stage I-III, HR+ breast cancer initiating adjuvant aromatase inhibitor therapy. We compared predicted 10-year CVD risk, including the composite index heart age, computed from modified Framingham risk score, with predicted 10-year risk of breast cancer recurrence using Adjuvant! Online. We created multivariable logistic regression models to estimate the odds ratios (OR) and 95% confidence intervals (CI) for greater CVD risk than breast cancer recurrence risk. Among 415 women, mean age and heart age were 60 and 67 years, respectively. Overall, 43% of women had a predicted 10-year CVD risk equivalent to breast cancer recurrence risk and 37% had CVD risk higher than breast cancer recurrence risk. Predicted CVD risk was higher than breast cancer recurrence risk for stage I disease (OR: 6.1, 95% CI: 3.4-11.2) or heart age >65 (OR: 12.4, 95% CI: 7.0-22.6). The majority of postmenopausal women with HR+ early breast cancer had a predicted 10-year CVD risk that was equivalent to or higher than breast cancer recurrence risk. Physicians should weigh competing risks and offer early screening and cardiac prevention strategies for women at a greater risk for CVD.
Collapse
Affiliation(s)
- Aditya Bardia
- Breast Cancer Program, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins School of Medicine, Bunting-Blaustein Cancer Research Building 1, Room 144, 1650 Orleans Street, Baltimore, MD 21231, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Palesh O, Demark-Wahnefried W, Mustian K, Minasian L, Rowland J, Sprod L, Janelsins M, Peppone L, Sloan J, Engquist KB, Jones L, Buist D, Paskett ED. Conducting cancer control and survivorship research via cooperative groups: a report from the American Society of Preventive Oncology. Cancer Epidemiol Biomarkers Prev 2011; 20:1050-5. [PMID: 21502540 DOI: 10.1158/1055-9965.epi-11-0176] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
As the number of cancer survivors expands, the need for cancer control and survivorship research becomes increasingly important. The National Cancer Institute (NCI) Cooperative Groups may offer a viable platform to perform such research. Observational, preventive, and behavioral research can often be performed within the cooperative group setting, especially if resources needed for evaluation are fairly simple, if protocols are easily implemented within the typical clinical setting, and if interventions are well standardized. Some protocols are better suited to cooperative groups than are others, and there are advantages and disadvantages to conducting survivorship research within the cooperative group setting. Behavioral researchers currently involved in cooperative groups, as well as program staff within the NCI, can serve as sources of information for those wishing to pursue symptom management and survivorship studies within the clinical trial setting. The structure of the cooperative groups is currently changing, but going forward, survivorship is bound to be a topic of interest and one that perhaps may be more easily addressed using the proposed more centralized structure.
Collapse
Affiliation(s)
- Oxana Palesh
- Department of Psychiatry & Behavioral Sciences, Stanford University, Stanford, CA 94305-5718, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Abstract
Although important advances have been made in curing childhood cancer in the last several decades, long-term survivors face considerable morbidity and mortality because of late effects from their initial anticancer therapy. By 30 years after treatment, the cumulative mortality from treatment-related medical illness actually exceeds that of mortality from cancer recurrence. Cardiovascular disease, in particular, is a leading threat to the well-being of adult survivors of childhood cancers. Unfortunately, the mechanisms of these late cardiac effects are understudied and poorly understood. This article reviews cardiotoxicity associated with 2 major anticancer regimens used in treating childhood cancer patients: anthracycline treatment and radiation therapy. The known pathophysiology and clinical cardiac risk factors that further predispose these patients to late-onset cardiac events are discussed. Basic and translational research is urgently needed to clarify pathophysiologic mechanisms of late cardiac effects and to develop therapies to improve both long-term survival and quality of life of adults cured of pediatric cancers.
Collapse
Affiliation(s)
- Ming Hui Chen
- From the Departments of Cardiology (M.H.C., S.D.C.) and Medicine (L.D.), Children's Hospital Boston; Department of Medicine (M.H.C.), Divisions of Women's Health and Cardiovascular Medicine, Brigham and Women's Hospital; Departments of Medical Oncology (M.H.C.) and Pediatric Oncology (L.D.), Dana-Farber Cancer Institute; and the Departments of Medicine (M.H.C.) and Pediatrics (S.D.C., L.D.), Harvard Medical School, Boston, MA
| | - Steven D. Colan
- From the Departments of Cardiology (M.H.C., S.D.C.) and Medicine (L.D.), Children's Hospital Boston; Department of Medicine (M.H.C.), Divisions of Women's Health and Cardiovascular Medicine, Brigham and Women's Hospital; Departments of Medical Oncology (M.H.C.) and Pediatric Oncology (L.D.), Dana-Farber Cancer Institute; and the Departments of Medicine (M.H.C.) and Pediatrics (S.D.C., L.D.), Harvard Medical School, Boston, MA
| | - Lisa Diller
- From the Departments of Cardiology (M.H.C., S.D.C.) and Medicine (L.D.), Children's Hospital Boston; Department of Medicine (M.H.C.), Divisions of Women's Health and Cardiovascular Medicine, Brigham and Women's Hospital; Departments of Medical Oncology (M.H.C.) and Pediatric Oncology (L.D.), Dana-Farber Cancer Institute; and the Departments of Medicine (M.H.C.) and Pediatrics (S.D.C., L.D.), Harvard Medical School, Boston, MA
| |
Collapse
|
43
|
Azim HA, de Azambuja E, Colozza M, Bines J, Piccart MJ. Long-term toxic effects of adjuvant chemotherapy in breast cancer. Ann Oncol 2011; 22:1939-1947. [PMID: 21289366 DOI: 10.1093/annonc/mdq683] [Citation(s) in RCA: 179] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Breast cancer is the most common malignant tumor affecting women. Adjuvant systemic therapies have been shown to have a significant impact on reducing the risk for breast cancer recurrence and overall mortality. Chemotherapy remains an important and frequently used treatment option in the adjuvant setting, and the associated short-term adverse events are very well described. However, there is insufficient information regarding the long-term sequelae of most chemotherapeutic agents. In this review, we describe different potential long-term adverse events associated with adjuvant chemotherapy in breast cancer, with a particular focus on long-term cardiac toxicity, secondary leukemia, cognitive function, and neurotoxicity. In addition, we discuss the effect of adjuvant chemotherapy on fertility and sexual function of young breast cancer patients. These adverse events are frequently overshadowed by the well-demonstrated clinical efficacy and/or reassuring short-term safety profiles of the different chemotherapy regimens commonly used today. We believe that a proper understanding and appreciation of these adverse events will enable us to refine our strategies for managing breast cancer. The fact that adjuvant chemotherapy is often given to patients who might not really need it urges us to consider the whole spectrum of chemotherapy risks versus benefits to maximize benefit without compromising quality of life.
Collapse
Affiliation(s)
- H A Azim
- Department of Medical Oncology, Jules Bordet Institute, Brussels, Belgium
| | - E de Azambuja
- Department of Medical Oncology, Jules Bordet Institute, Brussels, Belgium
| | - M Colozza
- Department of Oncology, Terni Hospital, Terni, Italy
| | - J Bines
- Department of Clinical Oncology, University Hospital of Clementino Fraga Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - M J Piccart
- Department of Medical Oncology, Jules Bordet Institute, Brussels, Belgium.
| |
Collapse
|
44
|
DE BOER RH, CHAN A, TRAN B, WILCKEN N. Use of non-anthracycline regimens in early stage breast cancer in Australia. Asia Pac J Clin Oncol 2010; 7:4-10. [DOI: 10.1111/j.1743-7563.2010.01353.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
45
|
Senkus E, Jassem J. Cardiovascular effects of systemic cancer treatment. Cancer Treat Rev 2010; 37:300-11. [PMID: 21126826 DOI: 10.1016/j.ctrv.2010.11.001] [Citation(s) in RCA: 138] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2010] [Revised: 11/05/2010] [Accepted: 11/09/2010] [Indexed: 01/07/2023]
Abstract
Many methods of systemic anticancer treatment have detrimental effects on the cardiovascular system, thus limiting the possibility of further therapy, worsening patients' quality of life and increasing mortality. The best recognized and most clinically relevant is the cardiotoxicity of anthracyclines. Other cytotoxic drugs associated with significant risk of cardiovascular complications include alkylating agents, 5-fluorouracil and paclitaxel. Cardiovascular adverse effects are also associated with the use of targeted therapies, such as trastuzumab, bevacizumab and tyrosine kinase inhibitors, and some of the drugs used in the treatment of hematological malignancies, such as all-trans-retinoic acid and arsenic trioxide. The most serious cardiac complication of anticancer therapy is congestive heart failure, associated predominantly with the use of anthracyclines, trastuzumab and high-dose cyclophosphamide. Myocardial ischemia is mainly caused by antimetabolite and interferon alpha treatment. Other adverse effects may include hypotension, hypertension, arrhythmias and conduction disorders, edema, pericarditis and thrombo-embolic complications. The aim of this review is to summarize and critically analyze the available evidence on the cardiovascular toxicity of systemic anticancer therapies, with particular attention to the recently recognized adverse effects of targeted therapies.
Collapse
Affiliation(s)
- Elżbieta Senkus
- Department of Oncology and Radiotherapy, Medical University of Gdańsk, Dębinki 7, 80-211 Gdańsk, Poland.
| | | |
Collapse
|
46
|
Abstract
Although the introduction of novel therapies and drug combinations has improved the prognosis of metastatic breast cancer, the disease remains incurable. Increased knowledge of the biology and the molecular alterations in breast cancer has facilitated the design of targeted therapies. These agents include receptor and nonreceptor tyrosine kinase inhibitors (epidermal growth factor receptor family), intracellular signaling pathways (phosphatidylinositol-3-kinase, AKT, mammalian target of rapamycin) angiogenesis inhibitors and agents that interfere with DNA repair (poly(ADP-ribose) polymerase inhibitors). In the present review, we present the most promising studies of these new targeted therapies and novel combinations of targeted therapies with cytotoxic agents.
Collapse
Affiliation(s)
- Ricardo H Alvarez
- Department of Breast Medical Oncology, The University of Texas M.D. Anderson Cancer Center, 1155 Herman P, Pressler, CPB5.3458, Houston, TX 77030-3721, USA.
| |
Collapse
|
47
|
Chen T, Xu T, Li Y, Liang C, Chen J, Lu Y, Wu Z, Wu S. Risk of cardiac dysfunction with trastuzumab in breast cancer patients: a meta-analysis. Cancer Treat Rev 2010; 37:312-20. [PMID: 20952131 DOI: 10.1016/j.ctrv.2010.09.001] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2010] [Revised: 09/01/2010] [Accepted: 09/14/2010] [Indexed: 11/17/2022]
Abstract
BACKGROUND Trastuzumab is used widely for the treatment of early and advanced breast cancer. However, concerns have arisen regarding its cardiac toxicity. We did a systematic review and meta-analysis of published randomized controlled trials (RCTs) to assess the overall risk of cardiac dysfunction associated with trastuzumab treatment. METHODS We searched PubMed and Web of Science (January 1966-July 2009) and American Society of Clinical Oncology conferences held (January 2000-July 2009) for relevant articles and abstracts. Summary incidence rates, relative risks (RRs), and 95% confident intervals (CIs) were calculated using a fixed-effects or random-effects model. RESULTS 11,882 patients from 10 RCTs were included for analysis. The incidences of LVEF decrease and congestive heart failure (CHF) were 7.5% (95% CI 4.2-13.1) and 1.9% (95% CI 1.0-3.8) among patients receiving trastuzumab. Trastuzumab significantly increased the risk of LVEF decrease (RR = 2.13, 95% CI, 1.31-3.49; p = 0.003). In addition, it significantly increased the risk of CHF (RR = 4.19, 95% CI 2.73-6.42; p < 0.00001). The increased risk of CHF was observed in patients with early stage (RR = 4.05, 95% CI 2.49-6.58; p < 0.00001) as well as metastatic disease (RR = 4.75, 95% CI 1.93-11.71; p = 0.0007). Furthermore, trastuzumab significantly increased the risk of CHF (RR = 4.27, 95% CI 2.75-6.61, p < 0.00001) in patients receiving anthracycline-based chemotherapy, but not in patients receiving non-anthracycline chemotherapy (RR = 2.42, 95% CI 0.36-16.19, p = 0.36). CONCLUSION The addition of trastuzumab to anthracycline-based chemotherapy significantly increase the risk of cardiac dysfunction in breast cancer patients. Further studies are recommended for non-anthracycline chemotherapy.
Collapse
Affiliation(s)
- Tao Chen
- Department of Cardiology, Changzheng Hospital, Second Military Medical University, Shanghai 200433, China
| | | | | | | | | | | | | | | |
Collapse
|
48
|
Thomson CA, Thompson PA, Wright-Bea J, Nardi E, Frey GR, Stopeck A. Metabolic syndrome and elevated C-reactive protein in breast cancer survivors on adjuvant hormone therapy. J Womens Health (Larchmt) 2010; 18:2041-7. [PMID: 20044868 DOI: 10.1089/jwh.2009.1365] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
AIMS As the efficacy of treatment for breast cancer has improved, particularly with the use of antiestrogenic therapies, there is an increasing population of long-term breast cancer survivors who seeks care with unique health issues. These patients may be at increased risk for cardiovascular disease (CVD) resulting from excess adiposity and treatment effects. Metabolic syndrome (MetS) and elevated C-reactive protein (CRP), two predictors of CVD, have not been fully evaluated in overweight breast cancer survivors on hormone-modulating agents. METHODS Anthropometric measures, including weight, height, waist and hip circumferences; clinical laboratory assessments, including lipids, glucose, glycoslyated hemoglobin (HbA1c), insulin, and high sensitivity CRP; and body composition and blood pressure (BP) were collected from overweight breast cancer survivors (n=42). Select measures were used to derive MetS using the National Cholesterol Education Program Adult Treatment Panel III (NCEP-ATP III) diagnostic criteria. RESULTS Participants had a mean body weight of 83.8 kg and body mass index (BMI) of 31.4 kg/m2. Mean fasting glucose (98+/-12.9 mg/dL), HbA1c (6.0+/-0.5 mg/dL), cholesterol (199+/-33.7 mg/dL), and insulin (16+/-3.2 mg/dL) were all at the upper end of the normal range. MetS was diagnosed in 54.8% of overweight postmenopausal breast cancer survivors. CRP was moderately or severely elevated in 90.5% of the population (mean of 5.1+/-5.3 mg/dL). CONCLUSIONS In our sample, overweight breast cancer survivors commonly have MetS and elevated CRP that place them at increased risk for cardiovascular and other metabolic diseases. If replicated in a larger sample, this warrants close medical monitoring to prevent and reduce morbidity and mortality unrelated to breast cancer.
Collapse
Affiliation(s)
- Cynthia A Thomson
- Department of Nutritional Sciences, University of Arizona, Tucson, Arizona 85724, USA.
| | | | | | | | | | | |
Collapse
|
49
|
Abstract
The author argues for the view that the clinical role of the anthracyclines will persist in the adjuvant therapy of breast cancer for some time to come.
Collapse
Affiliation(s)
- Kathleen I Pritchard
- Sunnybrook Health Sciences Centre, 2075 Bayview Ave., Toronto, Ontario M4N 3M5, Canada.
| |
Collapse
|
50
|
Late effects of adjuvant systemic therapies in women diagnosed with breast cancer at a young age. Breast 2009; 18 Suppl 3:S135-6. [DOI: 10.1016/s0960-9776(09)70288-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
|