1
|
Karim S, Alkreathy H, Khan MI. Untargeted metabolic profiling of high-dose methotrexate toxicity shows alteration in betaine metabolism. Drug Chem Toxicol 2024:1-9. [PMID: 38938090 DOI: 10.1080/01480545.2024.2369587] [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: 01/23/2024] [Accepted: 06/13/2024] [Indexed: 06/29/2024]
Abstract
Cardiotoxicity is a well-established adverse effect of several drugs across multiple therapeutic indications. It is particularly prevalent following anticancer therapy. In order to evaluate the changes in cellular metabolism associated with methotrexate cardiotoxicity, we treated Wistar rats with a single high dose of methotrexate (HDMTX), and after five days, the animals were sacrificed. We then analyzed the cardiotoxicity parameters in serum like Cardiac enzymes(CK-MB, Troponin T, ALP), Inflammatory markers (TNF-α and IL-6), oxidative stress markers (NO, NOX-2), histopathology and cardiac tissue with the goal of identifying a metabolic signature of cardiotoxicity using discovery-based metabolomics. The biochemical parameters for cardiac enzymes, oxidative stress and inflammatory markers showed a significant increase in all three categories in rats treated with HDMTX. These findings were mirrored in the histopathological analysis confirming cardiotoxicity due to HDMTX. The results showed a total of 95 metabolites that were found to be significantly (p < 0.05) modulated: either up- or downregulated in the HDMTX-treated group when compared with the control group. Using integrated pathway analysis we found these metabolites were associated with many important cardiac tissue metabolic pathways, such as the malate aspartate shuttle, taurine and hypotaurine metabolism, betaine metabolism, spermidine biosynthesis, and homocysteine degradation. Among them, L-arginine, homocysteine, and betaine were significantly upregulated, suggesting their possible association with cardiac tissue injury. Overall, we provided evidence for using untargeted metabolomics to identify novel metabolites associated with HDMTX cardiac toxicity.
Collapse
Affiliation(s)
- Shahid Karim
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Huda Alkreathy
- Department of Pharmacology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mohammad Imran Khan
- Research Center, King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia
| |
Collapse
|
2
|
Tlegenova Z, Balmagambetova S, Zholdin B, Kurmanalina G, Talipova I, Koyshybaev A, Sultanbekova G, Baspayeva M, Madinova S, Kubenova K, Amanova A, Tamadon A. Role of Clinical Risk Factors and B-Type Natriuretic Peptide in Assessing the Risk of Asymptomatic Cardiotoxicity in Breast Cancer Patients in Kazakhstan. Diagnostics (Basel) 2023; 13:3557. [PMID: 38066798 PMCID: PMC10706779 DOI: 10.3390/diagnostics13233557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 11/08/2023] [Accepted: 11/23/2023] [Indexed: 10/16/2024] Open
Abstract
The asymptomatic progression of chemotherapy-induced cardiotoxicity poses a significant risk to breast cancer patients. In the present single-center cohort study, a predictive model for evaluating the risk of cardiotoxicity during or by the end of chemotherapy was designed. The risk-prediction nomogram was delineated and assessed. In total, 34 patients out of 120 developed asymptomatic cardiotoxicity (28.3%). Of six explored biomarkers, only B-type natriuretic peptide showed a reliable pattern of incremental increase, revealing statistical significance between cardiotoxicity "+" and "-" groups by visit 4 or by the 9th month of monitoring (p 0.006). The following predictors were included in the model: age, hypertension, diabetes mellitus, baseline glomerular filtration rate, 6 min walk test measured at visit 4, BNP values at visit 4, left ventricular ejection fraction levels at visit 4, a total dose of radiotherapy received, and anthracycline cumulative doses. The model's AUC was 0.72 (95% CI 0.59; 0.86), evidencing the satisfactory predictive ability of the model; sensitivity 100% (95% CI 90.36; 100.0) at a specificity of 66.67% (95% CI 50.33; 79.79); PPV 54.1% [95% CI 47.13; 60.91]; PVN 100% [95% CI 94.64; 100.00]. The calibration plot showed satisfactory agreement between predicted and actual chances (p = 0.98). The designed model can be applied in settings lacking speckle tracking echocardiography.
Collapse
Affiliation(s)
- Zhenisgul Tlegenova
- Department of Internal Diseases-2, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (Z.T.); (B.Z.); (G.K.); (I.T.); (G.S.); (A.A.)
| | - Saule Balmagambetova
- Department of Oncology, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Bekbolat Zholdin
- Department of Internal Diseases-2, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (Z.T.); (B.Z.); (G.K.); (I.T.); (G.S.); (A.A.)
| | - Gulnara Kurmanalina
- Department of Internal Diseases-2, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (Z.T.); (B.Z.); (G.K.); (I.T.); (G.S.); (A.A.)
| | - Iliada Talipova
- Department of Internal Diseases-2, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (Z.T.); (B.Z.); (G.K.); (I.T.); (G.S.); (A.A.)
| | - Arip Koyshybaev
- Department of Oncology, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan;
| | - Gulmira Sultanbekova
- Department of Internal Diseases-2, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (Z.T.); (B.Z.); (G.K.); (I.T.); (G.S.); (A.A.)
| | - Mira Baspayeva
- Chemotherapy Division at University Medical Center, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (M.B.); (S.M.)
| | - Saule Madinova
- Chemotherapy Division at University Medical Center, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (M.B.); (S.M.)
| | - Kulparshan Kubenova
- Clinical Laboratory at University Medical Center, West Kazakhstan Marat Ospanov Medical University, Aktobe 030017, Kazakhstan;
| | - Aiganym Amanova
- Department of Internal Diseases-2, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan; (Z.T.); (B.Z.); (G.K.); (I.T.); (G.S.); (A.A.)
| | - Amin Tamadon
- Percia Vista R&D Co., Shiraz, Iran;
- Department for Scientific Work, West Kazakhstan Marat Ospanov Medical University, Aktobe 030019, Kazakhstan
| |
Collapse
|
3
|
Yarana C, Maneechote C, Khuanjing T, Ongnok B, Prathumsap N, Thanasrisuk S, Pattanapanyasat K, Chattipakorn SC, Chattipakorn N. Potential roles of 4HNE-adducted protein in serum extracellular vesicles as an early indicator of oxidative response against doxorubicin-induced cardiomyopathy in rats. Curr Res Toxicol 2023; 5:100134. [PMID: 37964944 PMCID: PMC10641738 DOI: 10.1016/j.crtox.2023.100134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Revised: 10/18/2023] [Accepted: 10/31/2023] [Indexed: 11/16/2023] Open
Abstract
Late-onset cardiomyopathy is becoming more common among cancer survivors, particularly those who received doxorubicin (DOXO) treatment. However, few clinically available cardiac biomarkers can predict an unfavorable cardiac outcome before cell death. Extracellular vesicles (EVs) are emerging as biomarkers for cardiovascular diseases and others. This study aimed to measure dynamic 4-hydroxynonenal (4HNE)-adducted protein levels in rats treated chronically with DOXO and examine their link with oxidative stress, antioxidant gene expression in cardiac tissues, and cardiac function. Twenty-two male Wistar rats were randomly assigned to receive intraperitoneal injection of normal saline (n = 8) or DOXO (3 mg/kg, 6 doses, n = 14). Before and after therapy, serum EVs and N-terminal pro-B-type natriuretic peptide (NT-proBNP) levels were determined. Tunable resistive pulse sensing was used to measure EV size and concentration. ELISA was used to assess 4HNE-adducted protein in EVs and cardiac tissues. Differential-display reverse transcription-PCR was used to quantitate cardiac Cat and Gpx1 gene expression. Potential correlations between 4HNE-adducted protein levels in EVs, cardiac oxidative stress, antioxidant gene expression, and cardiac function were determined. DOXO-treated rats showed more serum EV 4HNE-adducted protein than NSS-treated rats at day 9 and later endpoints, whereas NT-proBNP levels were not different between groups. Moreover, on day 9, surviving rats' EVs had higher levels of 4HNE-adducted protein, and these correlated positively with concentrations of heart tissue 4HNE adduction and copy numbers of Cat and Gpx1, while at endpoint correlated negatively with cardiac functions. Therefore, 4HNE-adducted protein in serum EVs could be an early, minimally invasive biomarker of the oxidative response and cardiac function in DOXO-induced cardiomyopathy.
Collapse
Affiliation(s)
- Chontida Yarana
- Center for Research Innovation and Biomedical Informatics, Faculty of Medical Technology, Mahidol University, 999 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thawatchai Khuanjing
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Benjamin Ongnok
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nanthip Prathumsap
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirasa Thanasrisuk
- Faculty of Medical Technology, Mahidol University, 999 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Kovit Pattanapanyasat
- Center of Excellence for Microparticle and Exosome in Diseases, Department of Research and Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Siriporn C. Chattipakorn
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| |
Collapse
|
4
|
Liu C, Chen H, Guo S, Liu Q, Chen Z, Huang H, Zhao Q, Li L, Cen H, Jiang Z, Luo Q, Chen X, Zhao J, Chen W, Yang PC, Wang L. Anti-breast cancer-induced cardiomyopathy: Mechanisms and future directions. Biomed Pharmacother 2023; 166:115373. [PMID: 37647693 DOI: 10.1016/j.biopha.2023.115373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/14/2023] [Accepted: 08/19/2023] [Indexed: 09/01/2023] Open
Abstract
With the progression of tumor treatment, the 5-year survival rate of breast cancer is close to 90%. Cardiovascular toxicity caused by chemotherapy has become a vital factor affecting the survival of patients with breast cancer. Anthracyclines, such as doxorubicin, are still some of the most effective chemotherapeutic agents, but their resulting cardiotoxicity is generally considered to be progressive and irreversible. In addition to anthracyclines, platinum- and alkyl-based antitumor drugs also demonstrate certain cardiotoxic effects. Targeted drugs have always been considered a relatively safe option. However, in recent years, some random clinical trials have observed the occurrence of subclinical cardiotoxicity in targeted antitumor drug users, which may be related to the effects of targeted drugs on the angiotensin converting enzyme, angiotensin receptor and β receptor. The use of angiotensin-converting enzyme inhibitors, angiotensin II receptor blockers and beta-blockers may prevent clinical cardiotoxicity. This article reviews the toxicity and mechanisms of current clinical anti-breast cancer drugs and proposes strategies for preventing cardiovascular toxicity to provide recommendations for the clinical prevention and treatment of chemotherapy-related cardiomyopathy.
Collapse
Affiliation(s)
- Chunping Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China; Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510080, Guangdong Province, China; State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, China
| | - Huiqi Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Sien Guo
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Qiaojing Liu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Zhijun Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Haiding Huang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Qi Zhao
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, Guangdong Province, China
| | - Longmei Li
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Huan Cen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Zebo Jiang
- Guangdong Provincial Key Laboratory of Biomedical Imaging and Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai 519000, Guangdong Province, China
| | - Qiyuan Luo
- Health Science Center, Shenzhen University, Shenzhen 518060, Guangdong Province, China
| | - Xiaoling Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Jiaxiong Zhao
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Wensheng Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Phillip C Yang
- Cardiovascular Stem Cell (Yang) Laboratory, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Lei Wang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China; Department of Cardiovascular Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China.
| |
Collapse
|
5
|
Vasbinder A, Cheng RK, Heckbert SR, Thompson H, Zaslavksy O, Chlebowski RT, Shadyab AH, Johnson L, Wactawski-Wende J, Wells G, Yung R, Martin LW, Paskett ED, Reding K. Chronic Oxidative Stress as a Marker of Long-term Radiation-Induced Cardiovascular Outcomes in Breast Cancer. J Cardiovasc Transl Res 2023; 16:403-413. [PMID: 36178659 PMCID: PMC10060433 DOI: 10.1007/s12265-022-10320-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/06/2022] [Indexed: 10/14/2022]
Abstract
While biomarkers have been proposed to identify individuals at risk for radiation-induced cardiovascular disease (RICVD), little is known about long-term associations with cardiac events. We examined associations of biomarkers of oxidative stress (myeloperoxidase, growth differentiation factor-15, 8-hydroxy-2'-deoxyguanosine [8-OH-dG], placental growth factor), cardiac injury (troponin I, cystatin-C), inflammation (interleukin-6, C-reactive protein), and myocardial fibrosis (transforming growth factor-ß) with long-term RICVD in breast cancer (BC) survivors. We conducted a nested case-control study within the Women's Health Initiative of postmenopausal women with incident BC stages I-III, who received radiation and had pre- and post-BC diagnosis serum samples. Cases (n = 55) were defined as developing incident, physician-adjudicated myocardial infarction, coronary heart disease death, other CVD death, heart failure, or stroke after BC. Cases were matched to three controls (n = 158). After adjustment, a higher 8-OH-dG ratio was significantly associated with an elevated long-term risk of RICVD, suggesting oxidative DNA damage may be a putative pathway for RICVD.
Collapse
Affiliation(s)
- Alexi Vasbinder
- Department of Biobehavioral Nursing and Health Informatics, School of Nursing, University of Washington, Seattle, USA
| | - Richard K Cheng
- Division of Cardiology, Department of Medicine, University of Washington Medical Center, Seattle, USA
| | - Susan R Heckbert
- Department of Epidemiology, School of Public Health, University of Washington, Seattle, USA
| | - Hilaire Thompson
- Department of Biobehavioral Nursing and Health Informatics, School of Nursing, University of Washington, Seattle, USA
| | - Oleg Zaslavksy
- Department of Biobehavioral Nursing and Health Informatics, School of Nursing, University of Washington, Seattle, USA
| | - Rowan T Chlebowski
- Lundquist Institute for Biomedical Innovation at Harbor-UCLA Medical Center, Torrance, USA
| | - Aladdin H Shadyab
- Herbert Wertheim School of Public Health and Human Longevity Science, University of California, San Diego, USA
| | - Lisa Johnson
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, USA
| | - Jean Wactawski-Wende
- Department of Epidemiology and Environmental Health, School of Public Health and Health Professions, University of Buffalo, Buffalo, USA
| | - Gretchen Wells
- Department of Internal Medicine, College of Medicine, University of Kentucky, Lexington-Fayette, USA
| | - Rachel Yung
- Division of Medical Oncology, University of Washington, Seattle, USA
| | - Lisa Warsinger Martin
- Division of Cardiology, School of Medicine and Health Sciences, George Washington University, Seattle, USA
| | - Electra D Paskett
- Comprehensive Cancer Center, Department of Medicine, The Ohio State University, Columbus, USA
| | - Kerryn Reding
- Department of Biobehavioral Nursing and Health Informatics, School of Nursing, University of Washington, Seattle, USA.
| |
Collapse
|
6
|
Sharifkazemi M, Elahi M, Sayad M. Case report: Early acute myocarditis after radiation therapy for breast cancer: A case presentation and review of literature. Front Cardiovasc Med 2023; 10:1020082. [PMID: 37153473 PMCID: PMC10154576 DOI: 10.3389/fcvm.2023.1020082] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 03/24/2023] [Indexed: 05/09/2023] Open
Abstract
Breast cancer is the most commonly diagnosed cancer in women worldwide, and with the increased survival of patients by novel treatments, the frequency of complications of cancer treatments rises. Radiotherapy, especially on the chest wall, can damage different cardiac structures. Radiotherapy-induced cardiomyopathy mainly occurs over 10 years after breast cancer treatment; however, there is a gap in the literature on acute myocarditis following radiotherapy. Here, we present a 54-year-old woman who developed acute myocarditis shortly after 25 sessions of radiotherapy with 50 Gy of radiation, successfully diagnosed with the use of speckle tracking echocardiography (STE) and cardiac magnetic resonance (CMR), and responded to the medical treatment with relative clinical improvement until the final follow-up. This case suggests the necessity of detailed examination of patients after radiotherapy, not only for chronic occurrence of cardiomyopathy but also for acute myocarditis. Although STE and CMR resulted in accurate diagnosis, in this case, further studies are required to determine the diagnostic accuracy of these two imaging methods compared with other imaging modalities in such patients and investigate the best diagnostic tool and therapeutic approach for these patients.
Collapse
Affiliation(s)
- Mohammadbagher Sharifkazemi
- Department of Cardiology, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
- Correspondence: Mohammadbagher Sharifkazemi
| | - Mahsa Elahi
- Radiation Oncology Department, Nemazee Hospital, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Masoud Sayad
- Cardio-Oncology Department, Rajaie Heart Hospital, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
7
|
Podlesnikar T, Berlot B, Dolenc J, Goričar K, Marinko T. Radiotherapy-Induced Cardiotoxicity: The Role of Multimodality Cardiovascular Imaging. Front Cardiovasc Med 2022; 9:887705. [PMID: 35966531 PMCID: PMC9366112 DOI: 10.3389/fcvm.2022.887705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 05/05/2022] [Indexed: 12/04/2022] Open
Abstract
Radiotherapy (RT) is one of the pillars of cancer therapy. High-dose radiation exposure on the thorax is mainly used in the context of adjuvant RT after breast surgery, in lung and esophageal cancer, and as a complement to systemic treatment in lymphoma. Due to the anatomical proximity, the heart inevitably receives some radiation that can result in acute and chronic cardiotoxicity, leading to heart failure, coronary artery disease, pericardial and valvular heart disease. Current evidence suggests there is no safe radiation dose to the heart, which poses a need for early recognition of RT-induced cardiac injury to initiate cardioprotective treatment and prevent further damage. Multimodality cardiac imaging provides a powerful tool to screen for structural and functional abnormalities secondary to RT. Left ventricular ejection fraction, preferably with three-dimensional echocardiography or cardiovascular magnetic resonance (CMR), and global longitudinal strain with speckle-tracking echocardiography are currently the key parameters to detect cardiotoxicity. However, several novel imaging parameters are tested in the ongoing clinical trials. CMR parametric imaging holds much promise as T1, T2 mapping and extracellular volume quantification allow us to monitor edema, inflammation and fibrosis, which are fundamental processes in RT-induced cardiotoxicity. Moreover, the association between serum biomarkers, genetic polymorphisms and the risk of developing cardiovascular disease after chest RT has been demonstrated, providing a platform for an integrative screening approach for cardiotoxicity. The present review summarizes contemporary evidence of RT-induced cardiac injury obtained from multimodality imaging—echocardiography, cardiovascular computed tomography, CMR and nuclear cardiology. Moreover, it identifies gaps in our current knowledge and highlights future perspectives to screen for RT-induced cardiotoxicity.
Collapse
Affiliation(s)
- Tomaž Podlesnikar
- Department of Cardiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Department of Cardiac Surgery, University Medical Centre Maribor, Maribor, Slovenia
- *Correspondence: Tomaž Podlesnikar,
| | - Boštjan Berlot
- Department of Cardiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Jure Dolenc
- Department of Cardiology, University Medical Centre Ljubljana, Ljubljana, Slovenia
| | - Katja Goričar
- Pharmacogenetics Laboratory, Institute of Biochemistry and Molecular Genetics, Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Tanja Marinko
- Department of Radiotherapy, Institute of Oncology Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| |
Collapse
|
8
|
Venetoclax Induces Cardiotoxicity through Modulation of Oxidative-Stress-Mediated Cardiac Inflammation and Apoptosis via NF-κB and BCL-2 Pathway. Int J Mol Sci 2022; 23:ijms23116260. [PMID: 35682939 PMCID: PMC9181135 DOI: 10.3390/ijms23116260] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 05/19/2022] [Accepted: 05/30/2022] [Indexed: 02/01/2023] Open
Abstract
Cardiovascular damage induced by anticancer therapy has become the main health problem after tumor elimination. Venetoclax (VTX) is a promising novel agent that has been proven to have a high efficacy in multiple hematological diseases, especially acute myeloid leukemia (AML) and chronic lymphocytic leukemia (CLL). Considering its mechanism of action, the possibility that VTX may cause cardiotoxicity cannot be ruled out. Therefore, this study was designed to investigate the toxic effect of VTX on the heart. Male Sprague-Dawley rats were randomly divided into three groups: control, low-dose VTX (50 mg/kg via oral gavage), and high-dose VTX (100 mg/kg via oral gavage). After 21 days, blood and tissue samples were collected for histopathological, biochemical, gene, and protein analyses. We demonstrated that VTX treatment resulted in cardiac damages as evidenced by major changes in histopathology and markedly elevated cardiac enzymes and hypertrophic genes markers. Moreover, we observed a drastic increase in oxidative stress, as well as inflammatory and apoptotic markers, with a remarkable decline in the levels of Bcl-2. To the best of our knowledge, this study is the first to report the cardiotoxic effect of VTX. Further experiments and future studies are strongly needed to comprehensively understand the cardiotoxic effect of VTX.
Collapse
|
9
|
Azimzadeh O, von Toerne C, Subramanian V, Sievert W, Multhoff G, Atkinson MJ, Tapio S. Data-Independent Acquisition Proteomics Reveals Long-Term Biomarkers in the Serum of C57BL/6J Mice Following Local High-Dose Heart Irradiation. Front Public Health 2021; 9:678856. [PMID: 34277544 PMCID: PMC8283568 DOI: 10.3389/fpubh.2021.678856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 05/18/2021] [Indexed: 12/23/2022] Open
Abstract
Background and Purpose: Cardiotoxicity is a well-known adverse effect of radiation therapy. Measurable abnormalities in the heart function indicate advanced and often irreversible heart damage. Therefore, early detection of cardiac toxicity is necessary to delay and alleviate the development of the disease. The present study investigated long-term serum proteome alterations following local heart irradiation using a mouse model with the aim to detect biomarkers of radiation-induced cardiac toxicity. Materials and Methods: Serum samples from C57BL/6J mice were collected 20 weeks after local heart irradiation with 8 or 16 Gy X-ray; the controls were sham-irradiated. The samples were analyzed by quantitative proteomics based on data-independent acquisition mass spectrometry. The proteomics data were further investigated using bioinformatics and ELISA. Results: The analysis showed radiation-induced changes in the level of several serum proteins involved in the acute phase response, inflammation, and cholesterol metabolism. We found significantly enhanced expression of proinflammatory cytokines (TNF-α, TGF-β, IL-1, and IL-6) in the serum of the irradiated mice. The level of free fatty acids, total cholesterol, low-density lipoprotein (LDL), and oxidized LDL was increased, whereas that of high-density lipoprotein was decreased by irradiation. Conclusions: This study provides information on systemic effects of heart irradiation. It elucidates a radiation fingerprint in the serum that may be used to elucidate adverse cardiac effects after radiation therapy.
Collapse
Affiliation(s)
- Omid Azimzadeh
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Section Radiation Biology, Federal Office for Radiation Protection, Oberschleissheim, Germany
| | - Christine von Toerne
- Research Unit Protein Science, Helmholtz Zentrum München - German Research Center for Environmental Health, Munich, Germany
| | - Vikram Subramanian
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| | - Wolfgang Sievert
- Department of Radiation Oncology, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Gabriele Multhoff
- Department of Radiation Oncology, Center for Translational Cancer Research (TranslaTUM), Campus Klinikum rechts der Isar, Technical University of Munich, Munich, Germany
| | - Michael J Atkinson
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Radiation Biology, Technical University of Munich, Munich, Germany
| | - Soile Tapio
- Institute of Radiation Biology, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany.,Institute for Biological and Medical Imaging, Helmholtz Zentrum München - German Research Center for Environmental Health, Neuherberg, Germany
| |
Collapse
|
10
|
Kim SR, Heo JI, Park JW, Kang CM, Kim KS. Radiation-induced lipoprotein-associated phospholipase A2 increases lysophosphatidylcholine and induces endothelial cell damage. Toxicology 2021; 458:152841. [PMID: 34216699 DOI: 10.1016/j.tox.2021.152841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2021] [Revised: 06/15/2021] [Accepted: 06/22/2021] [Indexed: 10/21/2022]
Abstract
The cardiotoxicity of various anticancer therapies, including radiotherapy, can lead to cardiovascular complications. These complications can range from damaging cardiac tissues within the irradiation field to increasing the long-term risks of developing heart failure, coronary artery disease, and myocardial infarction. We analyzed radiation-induced metabolites capable of mediating critical biological processes, such as inflammation, senescence, and apoptosis. Previously, by applying QTOF-MASS analysis to irradiated human fibroblasts, we identified that metabolite sets of lysophosphatidylcholine (LPC) were increased in these cells. In this study, radiation-induced LPC accumulation in human aortic endothelial cells (HAECs) increased reactive oxygen species (ROS) production and senescence-associated-beta-galactosidase staining, in addition to decreasing their tube-forming ability. Knockdown of lipoprotein-associated phospholipase A2 (Lp-PLA2) with small interfering RNA (siRNA) inhibited the increased LPC production induced by radiation, and reduced the radiation-induced cell damage produced by ROS and oxidized low-density lipoprotein (LDL). Lp-PLA2 depletion abolished the induction of proinflammatory factors, such as interleukin 1β, tumor necrosis factor-alpha, matrix metalloproteinase 2, and matrix metalloproteinase 9, as well as adhesion molecules, such as intercellular adhesion molecule 1 (ICAM-1) and E-selection. Likewise, we showed that Lp-PLA2 expression was upregulated in the vasculature of irradiated rat, resulting in increased LPC production and LDL oxidation. Our data demonstrate that radiation-induced LPC production is a potential risk factor for cardiotoxicity that is mediated by Lp-PLA2 activity, suggesting that LPC and Lp-PLA2 offer potential diagnostic and therapeutic approaches to cardiovascular damage during radiotherapy.
Collapse
Affiliation(s)
- So-Ra Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea; School of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34054, Republic of Korea
| | - Jong-Ik Heo
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Jeong-Woo Park
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea
| | - Chang-Mo Kang
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea; School of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34054, Republic of Korea.
| | - Kwang Seok Kim
- Division of Radiation Biomedical Research, Korea Institute of Radiological and Medical Sciences, Seoul, 01812, Republic of Korea; School of Radiological and Medico-Oncological Sciences, University of Science and Technology, Daejeon, 34054, Republic of Korea.
| |
Collapse
|
11
|
Ansund J, Mijwel S, Bolam KA, Altena R, Wengström Y, Rullman E, Rundqvist H. High intensity exercise during breast cancer chemotherapy - effects on long-term myocardial damage and physical capacity - data from the OptiTrain RCT. CARDIO-ONCOLOGY 2021; 7:7. [PMID: 33588948 PMCID: PMC7883413 DOI: 10.1186/s40959-021-00091-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/19/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND Adjuvant systemic breast cancer treatment improves disease specific outcomes, but also presents with cardiac toxicity. In this post-hoc exploratory analysis of the OptiTrain trial, the effects of exercise on cardiotoxicity were monitored by assessing fitness and biomarkers over the intervention and into survivorship. Methods; Women starting chemotherapy were randomized to 16-weeks of resistance and high-intensity interval training (RT-HIIT), moderate-intensity aerobic and high-intensity interval training (AT-HIIT), or usual care (UC). Outcome measures included plasma troponin-T (cTnT), Nt-pro-BNP and peak oxygen uptake (VO2peak), assessed at baseline, post-intervention, and at 1- and 2-years. RESULTS For this per-protocol analysis, 88 women met criteria for inclusion. Plasma cTnT increased in all groups post-intervention. At the 1-year follow-up, Nt-pro-BNP was lower in the exercise groups compared to UC. At 2-years there was a drop in VO2peak for patients with high cTnT and Nt-pro-BNP. Fewer patients in the RT-HIIT group fulfilled biomarker risk criteria compared to UC (OR 0.200; 95% CI = 0.055-0.734). CONCLUSIONS In this cohort, high-intensity exercise was associated with lower levels of NT-proBNP 1-year post-baseline, but not with cTnT directly after treatment completion. This may, together with the preserved VO2peak in patients with low levels of biomarkers, indicate a long-term cardioprotective effect of exercise. TRIAL REGISTRATION Clinicaltrials. govNCT02522260 , Registered 13th of august 2015 - Retrospectively Registered.
Collapse
Affiliation(s)
- Josefin Ansund
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Mijwel
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Kate A Bolam
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Renske Altena
- Cancer Theme, Karolinska University Hospital, Stockholm, Sweden.,Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Yvonne Wengström
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.,Cancer Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Eric Rullman
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden.,Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden
| | - Helene Rundqvist
- Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden. .,Unit of Clinical Physiology, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
12
|
Vaidya TR, Mody H, Franco YL, Brown A, Ait-Oudhia S. Multiscale and Translational Quantitative Systems Toxicology, Pharmacokinetic-Toxicodynamic Modeling Analysis for Assessment of Doxorubicin-Induced Cardiotoxicity. AAPS JOURNAL 2021; 23:18. [PMID: 33404976 DOI: 10.1208/s12248-020-00542-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/27/2020] [Indexed: 11/30/2022]
Abstract
Dose-dependent life-threatening doxorubicin-induced cardiotoxicity (DIC) is a major clinical challenge that needs to be addressed. Here, we developed an integrated multiscale and translational quantitative systems toxicology and pharmacokinetic-toxicodynamic (QST-PK/TD) model for optimization of doxorubicin dosing regimens for early monitoring and minimization of DIC. A QST model was established by exposing human cardiomyocytes, AC16 cells, to doxorubicin over a time course, and measuring the dynamics of intracellular signaling proteins, AC16 cell viability and released biomarkers of cardiomyocyte injury such as the B-type natriuretic peptide (BNP). Experiments were scaled up to a three-dimensional and dynamic (3DD) cell culture system to evaluate DIC under various dosing regimens. The PK determinants of doxorubicin influencing DIC were identified in vitro and then translated to the in vivo setting through hybrid physiologically based PK (PBPK)/TD models using preclinical- and clinical-level data extracted from literature. The developed cellular-level QST model captured well the observed dynamics of intracellular proteins, AC16 cell viability and BNP kinetics. In the 3DD setting, dose fractionation of doxorubicin displayed a significant reduction in cardiotoxicity compared to single intravenous doses with equal exposure, implying doxorubicin peak concentrations as the PK determinant for DIC. The in vivo hybrid PBPK/TD models captured well doxorubicin PK and DIC. Peak doxorubicin concentrations correlated well with acute DIC for dose-fractionated regimens, while maximum 48-h moving average concentrations correlated with DIC for dose-fractionated and long-term infusion regimens in vivo. The developed multiscale and translational QST-PK/TD modeling platform may serve as an in silico tool for assessment of early toxicity and/or efficacy of developmental drugs in vitro.
Collapse
Affiliation(s)
- Tanaya R Vaidya
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Florida, Orlando, USA
| | - Hardik Mody
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Florida, Orlando, USA
| | - Yesenia L Franco
- Center for Pharmacometrics and Systems Pharmacology, College of Pharmacy, University of Florida, Florida, Orlando, USA
| | - Ashley Brown
- Institute for Therapeutic Innovation Department of Medicine Institute for Therapeutic Innovation, Orlando, Florida, USA
| | | |
Collapse
|
13
|
Broberg AM, Geisler J, Tuohinen S, Skytta T, Hrafnkelsdóttir ÞJ, Nielsen KM, Hedayati E, Omland T, Offersen BV, Lyon AR, Gulati G. Prevention, Detection, and Management of Heart Failure in Patients Treated for Breast Cancer. Curr Heart Fail Rep 2020; 17:397-408. [PMID: 32979150 PMCID: PMC7683437 DOI: 10.1007/s11897-020-00486-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Long-term survival has increased significantly in breast cancer patients, and cardiovascular side effects are surpassing cancer-related mortality. We summarize risk factors, prevention strategies, detection, and management of cardiotoxicity, with focus on left ventricular dysfunction and heart failure, during breast cancer treatment. RECENT FINDINGS Baseline treatment of cardiovascular risk factors is recommended. Anthracycline and trastuzumab treatment constitute a substantial risk of developing cardiotoxicity. There is growing evidence that this can be treated with beta blockers and angiotensin antagonists. Early detection of cardiotoxicity with cardiac imaging and circulating cardiovascular biomarkers is currently evaluated in clinical trials. Chest wall irradiation accelerates atherosclerotic processes and induces fibrosis. Immune checkpoint inhibitors require consideration for surveillance due to a small risk of severe myocarditis. Cyclin-dependent kinases4/6 inhibitors, cyclophosphamide, taxanes, tyrosine kinase inhibitors, and endocrine therapy have a lower-risk profile for cardiotoxicity. Preventive and management strategies to counteract cancer treatment-related left ventricular dysfunction or heart failure in breast cancer patients should include a comprehensive cardiovascular risk assessment and individual clinical evaluation. This should include both patient and treatment-related factors. Further clinical trials especially on early detection, cardioprevention, and management are urgently needed.
Collapse
Affiliation(s)
- Agneta Månsson Broberg
- Department of Cardiology, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Jürgen Geisler
- Department of Oncology, Akershus University Hospital, Lørenskog & Institute of Clinical Medicine, University of Oslo, Campus AHUS, Lørenskog, Norway
| | - Suvi Tuohinen
- Heart and Lung Center, Helsinki University Hospital, Helsinki, Finland
| | - Tanja Skytta
- Department of Oncology, Tampere University Hospital, Tampere, Finland
| | - Þórdís Jóna Hrafnkelsdóttir
- Department of Cardiology, Landspitali University Hospital, Reykjavík, Iceland and Faculty of Medicine, University of Iceland, Reykjavík, Iceland
| | | | - Elham Hedayati
- Department of Oncology-Pathology, Karolinska Institute, Stockholm, Sweden
- Department of Breast Cancer, Sarcoma and Endocrine Tumors, Theme Cancer, Karolinska University Hospital, Stockholm, Sweden
| | - Torbjørn Omland
- Department of Cardiology, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Birgitte V. Offersen
- Department of Experimental Clinical Oncology & Department of Oncology, Aarhus University Hospital, Aarhus, Denmark
| | - Alexander R. Lyon
- Cardio-Oncology Service, Royal Brompton Hospital and Imperial College London, London, UK
| | - Geeta Gulati
- Department of Cardiology, Oslo University Hospital, Postbox 4950, Ullevål, Nydalen, 0424 Oslo, Norway
- Department of Research, Akershus University Hospital, Lørenskog and Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| |
Collapse
|
14
|
Pang L, Liu Z, Wei F, Cai C, Yang X. Improving cardiotoxicity prediction in cancer treatment: integration of conventional circulating biomarkers and novel exploratory tools. Arch Toxicol 2020; 95:791-805. [PMID: 33219404 DOI: 10.1007/s00204-020-02952-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 11/05/2020] [Indexed: 12/31/2022]
Abstract
Early detection strategies and improvements in cancer treatment have dramatically reduced the cancer mortality rate in the United States (US). However, cardiovascular (CV) side effects of cancer therapy are frequent among the 17 million cancer survivors in the US today, and cardiovascular disease (CVD) has become the second leading cause of morbidity and mortality among cancer survivors. Circulating biomarkers are ideal for detecting and monitoring CV side effects of cancer therapy. Here, we summarize the current state of clinical studies on conventional serum and plasma CVD biomarkers to detect and prevent cardiac injury during cancer treatment. We also review how novel exploratory tools such as genetic testing, human stem cell-derived cardiomyocytes, Omics technologies, and artificial intelligence can elucidate underlying molecular and genetic mechanisms of CV injury and to improve predicting cancer therapy-related cardiotoxicity (CTRC). Current regulatory requirements for biomarker qualifications are also addressed. We present generally applicable lessons learned from published studies, particularly on how to improve reproducibility. The combination of conventional circulating biomarkers and novel exploratory tools will pave the way for precision medicine and improve the clinical practice of prediction, detection, and management of CTRC.
Collapse
Affiliation(s)
- Li Pang
- Division of Systems Biology, National Center for Toxicological Research, US. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA.
| | - Zhichao Liu
- Division of Bioinformation and Biostatistics, National Center for Toxicological Research, US. Food and Drug Administration, Jefferson, AR, USA
| | - Feng Wei
- Department of Structural Heart Disease, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, People's Republic of China
| | - Chengzhong Cai
- Division of Systems Biology, National Center for Toxicological Research, US. Food and Drug Administration, 3900 NCTR Road, Jefferson, AR, 72079, USA
| | - Xi Yang
- Division of Pharmacology & Toxicology, Office of Cardiology, Hematology, Endocrinology, & Nephrology, Office of New Drug, Center for Drug Evaluation and Research, US. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|
15
|
Cardiac Monitoring for Thoracic Radiation Therapy: Survey of Practice Patterns in the United States. Am J Clin Oncol 2020; 43:249-256. [PMID: 31972567 DOI: 10.1097/coc.0000000000000666] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
OBJECTIVE The American Society of Clinical Oncology (ASCO) 2017 guidelines on cardiac monitoring during cancer treatments identified patients receiving thoracic radiation (TRT) ≥30 Gy (heart in field) at increased risk for developing radiation-induced heart disease (RIHD). ASCO encouraged clinicians to actively screen and monitor for baseline modifiable cardiac risk factors and therapy-induced cardiotoxicity in this high-risk population. Coronary artery calcium (CAC) is an independent risk factor for adverse cardiac events that can be mitigated with preventative medical therapy. It is unclear whether radiation oncologists (ROs) are aware of ASCO guidelines or the implications of CAC observed on computed tomographic scans. We report on practice patterns, perceptions, and experiences of cardiac monitoring for patients receiving definitive TRT, excluding breast patients. MATERIALS AND METHODS A 28-question survey was emailed to United States ROs 3 times from September 2018 to January 2019. RESULTS There were 162 respondents from 42 states, 51% in academic practice. Most ROs (81%) were not aware of the ASCO guidelines. Only 24% agreed with the guidelines, only 27% believed symptomatic RIHD could manifest within 2 years of TRT, and 69% thought there was a lack of strong evidence for type and timing of cardiac monitoring tests. If CAC was evident on computed tomographic scans, 40% took no further action to inform the patient or referring doctor. CONCLUSIONS This survey highlights a critical gap in knowledge about cardiac monitoring and potentially life-saving opportunities for preventive cardiac medical management. Future studies focusing on timing and detection of RIHD may elucidate the utility of cardiac monitoring for TRT patients.
Collapse
|
16
|
Zhou F, Niu L, Zhao M, Ni WX, Liu J. Real-time three-dimensional echocardiography predicts cardiotoxicity induced by postoperative chemotherapy in breast cancer patients. World J Clin Cases 2020; 8:2542-2553. [PMID: 32607331 PMCID: PMC7322441 DOI: 10.12998/wjcc.v8.i12.2542] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 04/01/2020] [Accepted: 06/02/2020] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND The anthracycline chemotherapeutic drugs are cardiotoxic. Studies have found some indicators related to cardiotoxicity. However, there is currently no accurate indicator that can predict cardiac toxicity early.
AIM To explore the diagnostic value of real-time three-dimensional echocardiography (RT3DE) in predicting cardiac toxicity in breast cancer patients undergoing chemotherapy.
METHODS Female breast cancer patients who underwent radical mastectomy and postoperative chemotherapy at the Affiliated Hanzhou First People’s Hospital, Zhejiang University School of Medicine were recruited. All patients were routinely administered with chemotherapy for four cycles (T1-T4) after surgery. Two-dimensional (2D) echocardiography, RT3DE, and serological examinations were performed after each cycle of chemotherapy. Patients were divided into a toxic group and a non-toxic group based on whether patients had Δ left ventricular ejection fraction > 10% after one year of chemotherapy. Repeated measurement analysis of variance was used to compare the changes in 2D echocardiographic indicators, serological indicators, and RT3DE indicators before and after chemotherapy. Multivariate logistic regression was used to identify independent predictive indicators for cardiac toxicity in postoperative chemotherapy patients. Receiver operating characteristics (ROC) curve analysis was performed to analyze the diagnostic value of potential indicators in the diagnosis of cardiotoxicity.
RESULTS A total of 107 female breast cancer patients were included in the study. T4 maximum peak velocity in early diastole (E peak)/mitral annulus lateral tissue Doppler (e' peak) (E/e'), serological indicators [T4 cardiac troponin I (cTnI) and T4 pro-brain natriuretic peptide (Pro-BNP)], T3 minimum left atrial volume (LAV), T4 LAVmin, T3 LAV before the start of the P wave (LAVprep), and T4 LAVprep in the toxicity group were significantly higher than those in the non-toxic group. Multivariate logistic regression found that T4 cTnI, T4 Pro-BNP, T3 LAVmin, T4 LAVmin, T3 LAVprep, and T4 LAVprep had potential predictive value for cardiac toxicity (P < 0.05). ROC results showed that T4 LAVmin had the highest accuracy for diagnosing cardiac toxicity [area under the curve (AUC) = 0.947; sensitivity = 78.57%; specificity = 94.62%], followed by T4 LAVprep (AUC = 0.899; sensitivity = 100%; specificity = 66.67%). The accuracies of LAVprep and LAVprep in predicting cardiac toxicity were higher than those of T3 LAVmin and T3 LAVprep.
CONCLUSION RT3DE of left atrial volume can be used to predict the cardiotoxicity caused by chemotherapy, and it is expected to guide the clinical adjustment of dose and schedule in time.
Collapse
Affiliation(s)
- Fang Zhou
- Department of Breast Surgery, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Lin Niu
- Department of Ultrasound, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Min Zhao
- Department of Ultrasound, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Wei-Xing Ni
- Department of Ultrasound, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| | - Jian Liu
- Department of Breast Surgery, Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou 310006, Zhejiang Province, China
| |
Collapse
|
17
|
Abstract
Purpose of Review Currently, cardiotoxicity is monitored through echocardiography or multigated acquisition scanning and is defined as 10% or higher LVEF reduction. The latter stage may represent irreversible myocardium injury and limits modification of therapeutic paradigms at earliest stages. To stratify patients for anthracycline-related heart failure, highly sensitive and molecularly specific probes capable of interrogating cardiac damage at the subcellular levels have been sought. Recent Findings PET tracers may provide noninvasive assessment of earliest changes within myocardium. These tracers are at nascent stages of development and belong primarily to (a) mitochondrial potential-targeted and (b) general ROS (reactive oxygen species)-targeted radiotracers. Given that electrochemical gradient changes at the mitochondrial membrane represent an upstream, and earliest event before triggering the production of the ROS and caspase activity in a biochemical cascade, the former category might offer interrogation of cardiotoxicity at earliest stages exemplified by PET imaging, using 18F-Mitophos and 68Ga-Galmydar in rodent models. Summary Both categories of radiotracers may provide tools for monitoring chemotherapy-induced cardiotoxicity and interrogating therapeutic efficacy of cardio-protectants.
Collapse
Affiliation(s)
- Jothilingam Sivapackiam
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, PO Box 8225, 510 S. Kingshighway Blvd, St. Louis, MO, 63110, USA
| | - Monica Sharma
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, PO Box 8225, 510 S. Kingshighway Blvd, St. Louis, MO, 63110, USA
| | - Thomas H Schindler
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, PO Box 8225, 510 S. Kingshighway Blvd, St. Louis, MO, 63110, USA.,Departments of Medicine, Cardiology and Nuclear Medicine, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Vijay Sharma
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, PO Box 8225, 510 S. Kingshighway Blvd, St. Louis, MO, 63110, USA. .,Department of Neurology, Washington University School of Medicine, St. Louis, MO, 63110, USA. .,Department of Biomedical Engineering, School of Engineering & Applied Science, Washington University, St. Louis, MO, 63105, USA.
| |
Collapse
|
18
|
Shati AA, Dallak M. Acylated Ghrelin Protects the Hearts of Rats from Doxorubicin-Induced Fas/FasL Apoptosis by Stimulating SERCA2a Mediated by Activation of PKA and Akt. Cardiovasc Toxicol 2020; 19:529-547. [PMID: 31093930 DOI: 10.1007/s12012-019-09527-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
This study investigated if the cardioprotective effect of acylated ghrelin (AG) against doxorubicin (DOX)-induced cardiac toxicity in rats involves inhibition of Fas/FasL-mediated cell death. It also investigated if such an effect is mediated by restoring Ca+2 homeostasis from the aspect of stimulation of SERCA2a receptors. Adult male Wistar rats were divided into 4 groups (20 rats/each) as control, control + AG, DOX, and DOX + AG. AG was co-administered to all rats consecutively for 35 days. In addition, isolated cardiomyocytes were cultured and treated with AG in the presence or absence of DOX with or without pre-incubation with [D-Lys3]-GHRP-6 (a AG receptor antagonist), VIII (]an Akt inhibitor), or KT-5720 (a PKA inhibitor). AG increased LVSP, dp/dtmax, and dp/dtmin in both control and DOX-treated animals and improved cardiac ultrastructural changes in DOX-treated rats. It also inhibited ROS in control rats and lowered LVEDP, intracellular levels of ROS and Ca2+, and activity of calcineurin in LVs of DOX-treated rats. Concomitantly, it inhibited LV NFAT-4 nuclear translocation and downregulated their protein levels of Fas and FasL. Mechanistically, in control or DOX-treated hearts or cells, AG upregulated the levels of SERCA2a and increased the activities of PKA and Akt, leading to increase phosphorylation of phospholamban at Ser16 and Thr17. All these effects were abolished by D-Lys3-GHRP-6, VIII, or KT-5720 and were independent of food intake or GH/IGF-1. In conclusion, AG cardioprotection against DOX involves inhibition of extrinsic cell death and restoring normal Ca+2 homeostasis.
Collapse
Affiliation(s)
- Ali A Shati
- Department of Biology, College of Science, King Khalid University, Abha, Saudi Arabia.
| | - M Dallak
- Department of Physiology, College of Medicine, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
19
|
Cardiotoxicity - the first cause of morbidity and mortality in pediatric patients survivors of acute lymphoblastic leukemia. REV ROMANA MED LAB 2020. [DOI: 10.2478/rrlm-2020-0007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Acute lymphoblastic leukemia is the most common hematological malignancy at pediatric age. Cardiotoxicity holds the first place among the causes of morbidity and mortality in these patients. Anthracyclines are cytostatic drugs frequently associated with cardiotoxicity. Early diagnosis of cardiac impairment during the treatment of pediatric patients is extremely important, both for modulating future chemotherapy and for administering cardioprotective agents. Long term monitoring after chemotherapy helps to identify the risk of late cardiotoxicity among cancer survivors. There are several biomarkers, already in use or still under study, which may represent an operator-independent alternative for echocardiography in the diagnosis of cardiotoxicity. In case of cardiac damage, the clinician has options for treating or limiting the progression, either with the use of already approved agents, such as Dexrazoxane, or by administrating other cardioprotective drugs. International experts are still attempting to establish the best algorithm for early detection of cardiotoxicity, as well as the most efficient treatment plan in case of already existing myocardial damage in these patients. We present a review on treatment-related cardiotoxicity, including mechanisms of development, useful biomarkers and treatment options, after carefully analyzing specialty literature.
Collapse
|
20
|
Meehan J, Gray M, Martínez-Pérez C, Kay C, Pang LY, Fraser JA, Poole AV, Kunkler IH, Langdon SP, Argyle D, Turnbull AK. Precision Medicine and the Role of Biomarkers of Radiotherapy Response in Breast Cancer. Front Oncol 2020; 10:628. [PMID: 32391281 PMCID: PMC7193869 DOI: 10.3389/fonc.2020.00628] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/06/2020] [Indexed: 12/24/2022] Open
Abstract
Radiotherapy remains an important treatment modality in nearly two thirds of all cancers, including the primary curative or palliative treatment of breast cancer. Unfortunately, largely due to tumor heterogeneity, tumor radiotherapy response rates can vary significantly, even between patients diagnosed with the same tumor type. Although in recent years significant technological advances have been made in the way radiation can be precisely delivered to tumors, it is proving more difficult to personalize radiotherapy regimens based on cancer biology. Biomarkers that provide prognostic or predictive information regarding a tumor's intrinsic radiosensitivity or its response to treatment could prove valuable in helping to personalize radiation dosing, enabling clinicians to make decisions between different treatment options whilst avoiding radiation-induced toxicity in patients unlikely to gain therapeutic benefit. Studies have investigated numerous ways in which both patient and tumor radiosensitivities can be assessed. Tumor molecular profiling has been used to develop radiosensitivity gene signatures, while the assessment of specific intracellular or secreted proteins, including circulating tumor cells, exosomes and DNA, has been performed to identify prognostic or predictive biomarkers of radiation response. Finally, the investigation of biomarkers related to radiation-induced toxicity could provide another means by which radiotherapy could become personalized. In this review, we discuss studies that have used these methods to identify or develop prognostic/predictive signatures of radiosensitivity, and how such assays could be used in the future as a means of providing personalized radiotherapy.
Collapse
Affiliation(s)
- James Meehan
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Mark Gray
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Carlos Martínez-Pérez
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Charlene Kay
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| | - Lisa Y Pang
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Jennifer A Fraser
- School of Applied Science, Sighthill Campus, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Amy V Poole
- School of Applied Science, Sighthill Campus, Edinburgh Napier University, Edinburgh, United Kingdom
| | - Ian H Kunkler
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - Simon P Langdon
- Cancer Research UK Edinburgh Centre and Division of Pathology Laboratories, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom
| | - David Argyle
- The Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, United Kingdom
| | - Arran K Turnbull
- Translational Oncology Research Group, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom.,Breast Cancer Now Edinburgh Research Team, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom
| |
Collapse
|
21
|
AlAsmari AF, Ali N, AlAsmari F, AlAnazi WA, AlShammari MA, Al-Harbi NO, Alhoshani A, As Sobeai HM, AlSwayyed M, AlAnazi MM, AlGhamdi NS. Liraglutide attenuates gefitinib-induced cardiotoxicity and promotes cardioprotection through the regulation of MAPK/NF-κB signaling pathways. Saudi Pharm J 2020; 28:509-518. [PMID: 32273812 PMCID: PMC7132601 DOI: 10.1016/j.jsps.2020.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Accepted: 03/03/2020] [Indexed: 12/15/2022] Open
Abstract
Gefitinib is an effective treatment for patients with locally advanced non-small cell lung cancer. However, it is associated with cardiotoxicity that can limit its clinical use. Liraglutide, a glucagon-like peptide 1 receptor agonist, showed potent cardioprotective effects with the mechanism is yet to be elucidated. Therefore, this study aimed to determine the efficiency of liraglutide in protecting the heart from damage induced by gefitinib. Adult male Wistar rats were randomly divided into control group, liraglutide group (200 µg/kg by i.p. injection), gefitinib group (30 mg/kg orally) and liraglutide plus gefitinib group. After 28 days, blood and tissue samples were collected for histopathological, biochemical, gene and protein analysis. We demonstrated that gefitinib treatment (30 mg/kg) resulted in cardiac damage as evidenced by histopathological studies. Furthermore, serum Creatine kinase-MB (CK-MB), N-terminal pro B-type natriuretic peptide (NT-proBNP) and cardiac Troponin-I (cTnI) were markedly elevated in gefitinib group. Pretreatment with liraglutide (200 µg/kg), however, restored the elevation in serum markers and diminished gefitinib-induced cardiac damage. Moreover, liraglutide improved the gene and protein levels of anti-oxidant (superoxide dismutase) and decreased the oxidative stress marker (NF-κB). Mechanistically, liraglutide offered protection through upregulation of the survival kinases (ERK1/2 and Akt) and downregulation of stress-activated kinases (JNK and P38). In this study, we provide evidence that liraglutide protects the heart from gefitinib-induced cardiac damage through its anti-oxidant property and through the activation of survival kinases.
Collapse
Key Words
- Akt, Protein kinase B
- Antioxidant
- CK-MB, Creatine kinase-MB
- Cardioprotection
- Cardiotoxicity
- EGFR, Epidermal growth factor receptor
- ERK1/2, Extracellular signal–regulated kinase 1/2
- GEF, Gefitinib
- GLP-1, Glucagon-like peptide-1
- Gefitinib
- JNK, C-Jun N-terminal kinase
- LDH, Lactate dehydrogenase
- LIRA, Liraglutide
- Liraglutide
- MAPK
- MAPK, Mitogen activated protein kinase
- NF-κB, Nuclear factor kappa B
- NT-proBNP, N-terminal pro B-type natriuretic peptide
- RTKIs, Receptor tyrosine kinases inhibitors
- RTKs, Receptor tyrosine kinases
- cTnI, cardiac Troponin-I
Collapse
Affiliation(s)
- Abdullah F AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nemat Ali
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Fawaz AlAsmari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Wael A AlAnazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Musaad A AlShammari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Ali Alhoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Homood M As Sobeai
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed AlSwayyed
- Department of Pathology, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
| | - Mohammed M AlAnazi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Nader S AlGhamdi
- Department of Pharmacy Services, Prince Mohammed Bin Abdulaziz Hospital, Riyadh 14214, Saudi Arabia
| |
Collapse
|
22
|
Tromp J, Boerman LM, Sama IE, Maass SWMC, Maduro JH, Hummel YM, Berger MY, de Bock GH, Gietema JA, Berendsen AJ, van der Meer P. Long-term survivors of early breast cancer treated with chemotherapy are characterized by a pro-inflammatory biomarker profile compared to matched controls. Eur J Heart Fail 2020; 22:1239-1246. [PMID: 32078215 PMCID: PMC7540448 DOI: 10.1002/ejhf.1758] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/24/2019] [Accepted: 01/17/2020] [Indexed: 12/25/2022] Open
Abstract
Background Chemo‐ and radiotherapy for breast cancer (BC) can lead to cardiotoxicity even years after the initial treatment. The pathophysiology behind these late cardiac effects is poorly understood. Therefore, we studied a large panel of biomarkers from different pathophysiological domains in long‐term BC survivors, and compared these to matched controls. Methods and results In total 91 biomarkers were measured in 688 subjects: 342 BC survivors stratified either to treatment with chemotherapy ± radiotherapy (n = 170) or radiotherapy alone (n = 172) and matched controls. Mean age was 59 ± 9 years and 65 ± 8 years for women treated with chemotherapy ± radiotherapy and radiotherapy alone, respectively, with a mean time since treatment of 11 ± 5.5 years. No biomarkers were differentially expressed in survivors treated with radiotherapy alone vs. controls (P for all >0.1). In sharp contrast, a total of 19 biomarkers were elevated, relative to controls, in BC survivors treated with chemotherapy ± radiotherapy after correction for multiple comparisons (P <0.05 for all). Network analysis revealed upregulation of pathways relating to collagen degradation and activation of matrix metalloproteinases. Furthermore, several inflammatory biomarkers including growth differentiation factor 15, monocyte chemoattractant protein 1, chemokine (C‐X‐C motif) ligand 16, tumour necrosis factor super family member 13b and proprotein convertase subtilisin/kexin type 9, elevated in survivors treated with chemotherapy, showed an independent association with lower left ventricular ejection fraction. Conclusion Breast cancer survivors treated with chemotherapy ± radiotherapy show a distinct biomarker profile associated with mild cardiac dysfunction even 10 years after treatment. These results suggest that an ongoing pro‐inflammatory state and activation of matrix metalloproteinases following initial treatment with chemotherapy might play a role in the observed cardiac dysfunction in late BC survivors.
Collapse
Affiliation(s)
- Jasper Tromp
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,National Heart Centre Singapore, Singapore, Singapore.,Duke-NUS Medical School, Singapore, Singapore
| | - Liselotte M Boerman
- Department of General Practice, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Iziah E Sama
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Saskia W M C Maass
- Department of General Practice, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - John H Maduro
- Department of Radiation Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Yoran M Hummel
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Marjolein Y Berger
- Department of General Practice, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Geertruida H de Bock
- Department of Epidemiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jourik A Gietema
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Annette J Berendsen
- Department of General Practice, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter van der Meer
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Gramatyka M, Sokół M. Radiation metabolomics in the quest of cardiotoxicity biomarkers: the review. Int J Radiat Biol 2020; 96:349-359. [PMID: 31976800 DOI: 10.1080/09553002.2020.1704299] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Purpose: Ionizing radiation is a risk factor to the whole organism, including the heart. Cardiac damage is considered to be a late effect of radiation exposure. While the acute cardiotoxicity of high doses is well characterized, the knowledge about nature and magnitude of the cardiac risk following lower doses exposure is incomplete. It has been shown that the cardiotoxic effects of radiation are source-, dose- and time-dependent. This paper provides an overview on these dependencies with regard to the molecular responses at the cellular and tissue levels. Main focus is put on the Nuclear Magnetic Resonance (NMR)-based and Mass Spectrometry (MS)-based metabolomic approaches in search of toxicity markers of relatively small doses of radiation.Conclusions: Available literature indicates that radiation exposure affects metabolites associated with: energy production, degradation of proteins and cell membranes, expression of proteins and stress response. Such effects are common for both animal and human studies. However, the specific metabolic response depends on several factors, including the examined organ. Radiation metabolomics can be used to explain the mechanisms of development of radiation-induced heart disease and to find an organ-specific biomarker of radiation exposure. The main aim of this review was to collect the information on the human cardiotoxicity biomarkers. In addition it also summarizes results of the studies on the metabolic responses to ionizing radiation for other organs, as well as the comparative data concerning animal studies.
Collapse
Affiliation(s)
- Michalina Gramatyka
- Department of Medical Physics, Maria Sklodowska-Curie Memorial Center and Institute of Oncology Gliwice Branch, Gliwice, Poland
| | - Maria Sokół
- Department of Medical Physics, Maria Sklodowska-Curie Memorial Center and Institute of Oncology Gliwice Branch, Gliwice, Poland
| |
Collapse
|
24
|
Shati AA. Doxorubicin-induces NFAT/Fas/FasL cardiac apoptosis in rats through activation of calcineurin and P38 MAPK and inhibition of mTOR signalling pathways. Clin Exp Pharmacol Physiol 2020; 47:660-676. [PMID: 31811646 DOI: 10.1111/1440-1681.13225] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 11/28/2019] [Accepted: 12/02/2019] [Indexed: 01/06/2023]
Abstract
This study investigated the role of NFAT/Fas/FasL axis in cardiomyocyte apoptosis following doxorubicin (DOX) treatment in rats and evaluated the involvement and regulation of all NFAT members in cardiac apoptosis. Forty adult male Wistar rats were divided equally into control or DOX-treated groups (15 mg/kg over 2 weeks). Cardiomyocytes were cultured and pre-incubated with various inhibitors and activators (10 μmol/L) prior to DOX exposure (1 μmol/L). In the left ventricles and cultured cells, DOX increased cytoplasmic protein levels of cytochrome C, Bax and increased the activities of caspase-8, caspase3, ERK1/2, JNK, and P38 mitogen-activated protein kinases (MAPKs), reducing levels of Bcl-2 and the activity of mTOR, and inducing cell death. In addition, DOX enhanced mRNA and protein levels of Fas and FasL. Furthermore, the nuclear and cytoplasmic levels of NFAT1 and nuclear accumulation of NFAT2-4were increased with DOX treatment. The inhibition of calcineurin with FK506 significantly inhibited the nuclear levels of NFAT2 and NFAT4 and the inhibition of P38 MAPK with SB203580 inhibited the nuclear and cytoplasmic accumulation of NFAT1. However, the activation of mTOR by IGF-1 significantly lowered NFAT3. In conclusion, NFAT/Fas/FasL-induced cell death in cardiac myocytes of DOX-treated rats is regulated, at least, by the activation of calcineurin and P38 MAPK and inhibition of mTOR.
Collapse
Affiliation(s)
- Ali A Shati
- Biology Department, College of Science, King Khalid University, Abha, Saudi Arabia
| |
Collapse
|
25
|
Belenkov YN, Ershov VI, Antyufeeva ON, Budanova DA, Kochkareva YB, Gadaev IY, Bochkarnikova OV, Sokolova IY, Kirichenko YY. [Assessment of the Role of Oxidative Stress Indicators and Early Markers of Myocardial Injury and Disfunction in Patients with Lymphoproliferative Diseases]. KARDIOLOGIIA 2019; 59:47-53. [PMID: 31397229 DOI: 10.18087/cardio.2019.8.n682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 07/24/2019] [Indexed: 06/10/2023]
Abstract
PURPOSE to study dynamics of indicators of oxidative stress and their role in development of cardiotoxicity in patients with lympho-proliferative diseases at the background of polychemotherapy. MATERIALS AND METHODS We included into this study 30 patients with newly detected Non-Hodgkin lymphomas. The control group comprised 15 healthy volunteers. For registration of dynamics of formation of oxygen active forms (OAF), we used highly sensitive chemiluminescence assay. The state of initial stage of lipoperoxidation OAF and radicals of organic compounds) was assessed by dynamics of the leukocyte chemiluminescence. The state of final stage of lipoperoxidation (formation of nonmetabolizing lipid hydroperoxides and other compounds) was assessed by the level of malonic dialdehyde. We also determined indicator of leukocyte chemiluminescence intensity (both basal and zymosan-stimulated). For assessment of the cardiovascular system functional state before and after chemotherapy we used electrocardiography (ECG), echocardiography (EchoCG) and 24-hour ECG monitoring. RESULTS The data obtained were indicative of increased generation of free radicals by leukocytes during polychemotherapy. After chemotherapy course we detected various types of cardiotoxicity. We noted substantial elevation of frequency of supraventricular and ventricular extrasystoles. There was direct correlation between rate of appearance of supraventricular extrasystoles and level of chemiluminescence of leukocytes (r=0.7; p=0.03). According to data of EchoCG although the left ventricular ejection fraction remained within the normal range during chemotherapy, there was a persistent tendency to its decrease (р<0.001). CONCLUSION In this study we for the first time in patients with Non-Hodgkin lymphomas detected an elevation of level of free radical reactions and lipid peroxidation with simultaneous lowering of antiperoxidative activity of blood plasma and their relation to development of cardiotoxic effects. The results obtained indicate to necessity of search for novel early markers of oxidative stress activation, myocardial injury and disfunction able to help to substantially decrease risk of development of cardiovascular complications during and after chemotherapy.
Collapse
Affiliation(s)
- Yu N Belenkov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - V I Ershov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - O N Antyufeeva
- Sechenov First Moscow State Medical University (Sechenov University)
| | - D A Budanova
- Sechenov First Moscow State Medical University (Sechenov University)
| | | | - I Yu Gadaev
- Sechenov First Moscow State Medical University (Sechenov University)
| | - O V Bochkarnikova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - I Ya Sokolova
- Sechenov First Moscow State Medical University (Sechenov University)
| | - Yu Yu Kirichenko
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
26
|
Zaher E, Fahmy E, Mahmoud K, El Kerm Y, Auf M. Assessment of the onset of radiation-induced cardiac damage after radiotherapy of breast cancer patients. ALEXANDRIA JOURNAL OF MEDICINE 2019. [DOI: 10.1016/j.ajme.2017.12.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Affiliation(s)
- Ebtsam Zaher
- Radiation Sciences Department, Medical Research Institute, Alexandria University, Egypt
| | - Enayat Fahmy
- Radiation Sciences Department, Medical Research Institute, Alexandria University, Egypt
| | - Kamal Mahmoud
- Clinical and Experimental Internal Medicine Department, Medical Research Institute, Alexandria University, Egypt
| | - Yasser El Kerm
- Cancer Management and Research Department, Medical Research Institute, Alexandria University, Egypt
| | - Mohammad Auf
- Demonstrator in 6th of October University, Egypt
| |
Collapse
|
27
|
Chalubinska-Fendler J, Graczyk L, Piotrowski G, Wyka K, Nowicka Z, Tomasik B, Fijuth J, Kozono D, Fendler W. Lipopolysaccharide-Binding Protein Is an Early Biomarker of Cardiac Function After Radiation Therapy for Breast Cancer. Int J Radiat Oncol Biol Phys 2019; 104:1074-1083. [PMID: 30991100 DOI: 10.1016/j.ijrobp.2019.04.002] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 03/27/2019] [Accepted: 04/07/2019] [Indexed: 12/28/2022]
Abstract
PURPOSE To evaluate the prognostic potential of lipopolysaccharide-binding protein (LBP) levels after breast cancer radiation therapy (RT) for incipient cardiac dysfunction. METHODS AND MATERIALS In this single-centered study, we prospectively enrolled female patients treated for left breast cancer. Healthy age- and sex-matched participants were recruited as controls. LBP levels, cardiac troponin T, N-terminal propeptide of the brain natriuretic peptide, fatty acid binding protein, and C-reactive protein were assessed at three timepoints-before RT, after the last RT fraction, and 1 month after the last fraction. Echocardiographic evaluation was done 3 to 3.75 years after RT. RESULTS We recruited 51 patients and 78 controls. Baseline LBP concentrations in the study group were significantly higher than in controls at baseline (P < .001), at 24 hours, and at 1 month after RT (P = .003 and P < .001, respectively). Other biomarkers (cardiac troponin T, N-terminal propeptide of the brain natriuretic peptide, fatty acid binding protein, and C-reactive protein) did not differ in any of the timepoints. Posttreatment LBP concentrations were significantly and positively correlated with heart- and lung-associated dose-volume histogram variables. Posttreatment and follow-up LBP levels correlated positively with the E/E' echocardiographic index reflective of the diastolic function. After adjustment for left anterior descending artery mean dose, left ventricle mean dose, mean heart dose, and type of surgery, LBP remained significantly correlated with E/E' when measured 24 hours after RT (beta = 0.41, P = .032) and 1 month after RT (beta = 0.43, P = .028). CONCLUSIONS Serum LBP concentrations correlate with diastolic function evaluated 3 years after the completion of RT, making LBP a potentially useful prognostic parameter.
Collapse
Affiliation(s)
| | | | - Grzegorz Piotrowski
- Department of Cardiology, N. Copernicus Provincial Multidisciplinary Centre of Oncology and Traumatology, Lodz, Poland; Institute of Health Science, University of Social Science, Lodz, Poland
| | - Krystyna Wyka
- Department of Pediatrics, Oncology, Hematology and Diabetology, Medical University of Lodz, Poland
| | - Zuzanna Nowicka
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Poland
| | - Bartlomiej Tomasik
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Poland; Department of Radiotherapy, Medical University of Lodz, Poland; Postgraduate School of Molecular Medicine, Medical University of Warsaw, Poland
| | - Jacek Fijuth
- Department of Radiotherapy, Medical University of Lodz, Poland
| | - David Kozono
- Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Wojciech Fendler
- Department of Biostatistics and Translational Medicine, Medical University of Lodz, Poland; Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts.
| |
Collapse
|
28
|
Rebeiz AG, Mahfoud Z, Abdul Fattah M, Saad A, Safar A, Bashshur ZF. Change in cardiac troponin T level after intravitreal anti-vascular endothelial growth factor treatment: Prospective pilot study. Eur J Ophthalmol 2019; 30:563-569. [DOI: 10.1177/1120672119832171] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Evaluate subclinical myocardial injury associated with intravitreal anti-vascular endothelial growth factor therapy by measuring serum high-sensitivity cardiac troponin T. Methods: This is a prospective pilot comparative study conducted at American University of Beirut Medical Center, Beirut, Lebanon. In total, 40 consecutive patients were randomized to receive either intravitreal bevacizumab or ranibizumab. Patients received three consecutive monthly injections of the assigned drug, then continued treatment as needed. Systemic concentrations of high-sensitivity cardiac troponin T and vascular endothelial growth factor were obtained at baseline, week 9, and week 24. Primary endpoint measure was change in high-sensitivity cardiac troponin T levels compared to baseline. Secondary endpoint measure was change in systemic vascular endothelial growth factor levels. Results: There was no significant difference in high-sensitivity cardiac troponin T levels over time ( p = 0.227) within each treatment group and no significant difference between treatments at any time point ( p = 0.276). There was a significant decrease in plasma vascular endothelial growth factor levels at week 9 ( p = 0.001) and week 24 ( p < 0.001) compared to baseline. In the ranibizumab group, vascular endothelial growth factor levels were not significantly different at weeks 9 and 24 compared to baseline ( p = 0.708 and p = 0.117, respectively). There was a significant association between the number of bevacizumab injections from weeks 8 to 24 and the decrease in vascular endothelial growth factor levels at week 24 ( R = −0.67, p = 0.032). This correlation was not observed in the ranibizumab group ( R = −0.341, p = 0.141). Conclusion: Repeated intravitreal bevacizumab or ranibizumab did not influence serum high-sensitivity cardiac troponin levels. Intravitreal bevacizumab but not ranibizumab lowered free-systemic vascular endothelial growth factor levels, which was observed in this study to be inversely related to the number of bevacizumab injections.
Collapse
Affiliation(s)
- Abdallah G Rebeiz
- Division of Cardiology, Department of Internal Medicine, American University of Beirut, Beirut, Lebanon
| | - Ziyad Mahfoud
- Department of Global and Public Health, Weill Cornell Medical College, Doha, Qatar
| | | | - Alain Saad
- Damien Gatinel’s Department of Ophthalmology, Rothschild Foundation, Paris, France
| | - Ammar Safar
- Department of Ophthalmology, American Hospital Dubai, Dubai, UAE
| | - Ziad F Bashshur
- Department of Ophthalmology, American University of Beirut, Beirut, Lebanon
- Vitreoretinal Service, Department of Ophthalmology, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
29
|
Banala VT, Urandur S, Sharma S, Sharma M, Shukla RP, Marwaha D, Gautam S, Dwivedi M, Mishra PR. Targeted co-delivery of the aldose reductase inhibitor epalrestat and chemotherapeutic doxorubicin via a redox-sensitive prodrug approach promotes synergistic tumor suppression. Biomater Sci 2019; 7:2889-2906. [DOI: 10.1039/c9bm00221a] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Redox responsive epalrestat prodrug micelles facilitate synergistic concentrations of doxorubicin with an advantage of CD44 down-regulation and reduced cardiotoxicity.
Collapse
Affiliation(s)
- Venkatesh Teja Banala
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Sandeep Urandur
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Shweta Sharma
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Ravi P. Shukla
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Monika Dwivedi
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics
- CSIR-Central Drug Research Institute Lucknow
- India
| |
Collapse
|
30
|
Portaluri M, Petruzzelli MF, Tramacere F, Andreassi MG. B-type natriuretic peptide plasma level in 5-year breast cancer survivors after radiotherapy. Int J Radiat Biol 2018; 95:201-206. [PMID: 30431375 DOI: 10.1080/09553002.2019.1542183] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
BACKGROUND Left-sided breast cancer patients treated with radiotherapy (RT) are at risk for late radiation-induced cardiovascular complications. AIM The aim of this study was to investigate the BNP plasma levels in long-term breast cancer survivors who received only RT as well to assess whether cardiac dose was associated with BNP values. METHODS Plasma samples for BNP measurement were repeated in 29 patients (63 ± 11 years) who were alive at 5 years after radiotherapy, free of heart disease and available to provide new blood sample. All patients had BNP measurements at baseline. The ΔBNP was measured to analyze the role of marker variations. No patients received chemotherapy. RESULTS The mean cardiac and ventricle dose were 2.1 ± 1.0 (range 0.02-4.5) Gy and 3.0 ± 1.7 (range 0.02-7.6), respectively. Median value of BNP was 47 pg/mL (interquartile ranges, 26-58.2 pg/mL) at baseline, and 34 pg/mL (interquartile ranges, 17.5-54 pg/mL) at 5 years after radiotherapy. There was no significantly different between two measurements (p = ns). Fifteen (52%) reported an improvement in BNP levels, 1 (3%) no changes and 13 (45%) reported a worsening. There was no correlation between ΔBNP and age (p = ns). When patients were stratified according to the median value of dose-volume data, ΔBNP was significantly higher in patients with increased cardiac Dmean (p = .02) and left ventricle Dmean (p = .009). CONCLUSION At 5 years after radiotherapy, median plasma BNP levels remained within the normal range, but the delta-BNP levels are directly related to the heart and ventricular dose received.
Collapse
|
31
|
Michel L, Rassaf T, Totzeck M. Biomarkers for the detection of apparent and subclinical cancer therapy-related cardiotoxicity. J Thorac Dis 2018; 10:S4282-S4295. [PMID: 30701097 DOI: 10.21037/jtd.2018.08.15] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Progress in cancer therapy over the past decades improved long-term survival but increased cancer therapy-related cardiotoxicity. Many novel treatment options have been implemented with yet incompletely characterized cardiovascular side effects including heart failure, coronary artery disease, arrhythmias, valvular disease, venous thromboembolism and myocarditis. Diagnosis of potential cardiotoxic side effects is essential for an optimal treatment but remains challenging. Cardiac biomarkers troponin and brain natriuretic peptide/N-terminal proBNP (BNP/NT-proBNP) have been extensively studied in heart failure and acute coronary syndromes. Emerging evidence implicates a significant role in the detection of cardiotoxicity and guidance of therapy in cancer patients. Elevated troponin or BNP/NT-proBNP levels were associated with increased all-cause mortality in cancer patients and have been shown to predict manifest heart failure. BNP/NT-proBNP may be useful for the prediction of cancer therapy-related heart failure and response to heart failure therapy in adult and pediatric cancer patients while troponin can indicate acute myocardial infarction in patients with cancer therapy-related risk for coronary artery disease. Furthermore, troponin may be used for the identification of immune checkpoint inhibitor-related myocarditis with very high sensitivity. Finally, even D-dimer levels have been shown to improve risk stratification and diagnosis in cancer-associated venous thromboembolism. This review aims to summarize the current knowledge about biomarkers in cancer therapy-related cardiotoxicity. We also outline possible clinical recommendations for the detection and treatment of subclinical and clinically apparent cardiotoxic effects using biomarkers.
Collapse
Affiliation(s)
- Lars Michel
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen, Germany
| | - Matthias Totzeck
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, Medical Faculty, University Hospital Essen, Essen, Germany
| |
Collapse
|
32
|
Corremans R, Adão R, De Keulenaer GW, Leite-Moreira AF, Brás-Silva C. Update on pathophysiology and preventive strategies of anthracycline-induced cardiotoxicity. Clin Exp Pharmacol Physiol 2018; 46:204-215. [DOI: 10.1111/1440-1681.13036] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2018] [Revised: 08/31/2018] [Accepted: 09/13/2018] [Indexed: 12/13/2022]
Affiliation(s)
| | - Rui Adão
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | | | - Adelino F. Leite-Moreira
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
| | - Carmen Brás-Silva
- Department of Surgery and Physiology; UnIC-Cardiovascular Research Centre; Faculty of Medicine; University of Porto; Porto Portugal
- Faculty of Nutrition and Food Sciences; University of Porto; Porto Portugal
| |
Collapse
|
33
|
Cancer and the Broken Heart: Complications and Implications of Therapy-Related Cardiotoxicity. JOURNAL OF INFUSION NURSING 2018; 41:229-240. [PMID: 29958259 DOI: 10.1097/nan.0000000000000285] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The growing number of adult long-term cancer survivors has expanded our knowledge of negative physiologic sequelae associated with curative therapies. Of note are the cardiovascular corollaries of chest radiotherapy and some commonly used chemotherapy agents. A contemporary understanding of risk factors has facilitated the development of guidelines for prevention and surveillance of cardiac compromise. The future holds promise with enhanced opportunities to use cardioprotectant drugs and biomarkers to prevent and detect early myocardial changes. Infusion therapy nurses must keep abreast of these developments to facilitate their roles as patient educators and advocates in the face of this toxicity's prevalence.
Collapse
|
34
|
Zhu Q, Kirova YM, Cao L, Arsene-Henry A, Chen J. Cardiotoxicity associated with radiotherapy in breast cancer: A question-based review with current literatures. Cancer Treat Rev 2018; 68:9-15. [DOI: 10.1016/j.ctrv.2018.03.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2017] [Revised: 03/07/2018] [Accepted: 03/08/2018] [Indexed: 12/25/2022]
|
35
|
New NO- and H2S-releasing doxorubicins as targeted therapy against chemoresistance in castration-resistant prostate cancer: in vitro and in vivo evaluations. Invest New Drugs 2018; 36:985-998. [PMID: 29607467 DOI: 10.1007/s10637-018-0590-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Accepted: 03/15/2018] [Indexed: 12/12/2022]
Abstract
Chemotherapy for castration-resistant prostate cancer (CRPC) is only temporarily effective due to the onset of chemoresistance. We investigated the efficacy of NO- and H2S-releasing doxorubicins (NitDox and H2SDox) in overcoming drug resistance and evaluated their safety. New and innovative NO- and H2S-releasing doxorubicins (NitDox and H2SDox) showed a good intracellular accumulation and high cytotoxic activity in vitro in an androgen-independent and doxorubicin-resistant DU-145 prostate cancer cell line. Nude mice were subcutaneously injected with 4*106 DU-145 cells and treated once a week for 3 weeks with 5 mg/kg doxorubicin, NitDox, H2SDox or vehicle, i.p. Animal weight, tumor volume, intra-tumoral drug accumulation, apoptosis and the presence of nitrotyrosine and sulfhydryl (SH) groups within the tumor, were evaluated. Cardiotoxicity was assessed by measuring troponin plasma levels and the left ventricular wall thickness. In vivo, NitDox and H2SDox accumulated inside the tumors, significantly reduced tumor volumes by 60%, increased the percentage of apoptotic cells in both the inner and the outer parts of the tumors and the presence of nitrotyrosine and SH groups. Doxorubicin treatment was associated with reduced body weight and cardiotoxicity. On the contrary, NitDox and H2SDox were well tolerated and had a better safety profile. Combining efficacy with reduced cardiovascular side effects, NitDox and H2SDox are promising novel therapeutic agents for reversing chemoresistance in CRCP.
Collapse
|
36
|
Chemotherapeutic-Induced Cardiovascular Dysfunction: Physiological Effects, Early Detection-The Role of Telomerase to Counteract Mitochondrial Defects and Oxidative Stress. Int J Mol Sci 2018. [PMID: 29534446 PMCID: PMC5877658 DOI: 10.3390/ijms19030797] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Although chemotherapeutics can be highly effective at targeting malignancies, their ability to trigger cardiovascular morbidity is clinically significant. Chemotherapy can adversely affect cardiovascular physiology, resulting in the development of cardiomyopathy, heart failure and microvascular defects. Specifically, anthracyclines are known to cause an excessive buildup of free radical species and mitochondrial DNA damage (mtDNA) that can lead to oxidative stress-induced cardiovascular apoptosis. Therefore, oncologists and cardiologists maintain a network of communication when dealing with patients during treatment in order to treat and prevent chemotherapy-induced cardiovascular damage; however, there is a need to discover more accurate biomarkers and therapeutics to combat and predict the onset of cardiovascular side effects. Telomerase, originally discovered to promote cellular proliferation, has recently emerged as a potential mechanism to counteract mitochondrial defects and restore healthy mitochondrial vascular phenotypes. This review details mechanisms currently used to assess cardiovascular damage, such as C-reactive protein (CRP) and troponin levels, while also unearthing recently researched biomarkers, including circulating mtDNA, telomere length and telomerase activity. Further, we explore a potential role of telomerase in the mitigation of mitochondrial reactive oxygen species and maintenance of mtDNA integrity. Telomerase activity presents a promising indicator for the early detection and treatment of chemotherapy-derived cardiac damage.
Collapse
|
37
|
Pharmacodynamic modeling of cardiac biomarkers in breast cancer patients treated with anthracycline and trastuzumab regimens. J Pharmacokinet Pharmacodyn 2018; 45:431-442. [PMID: 29429038 PMCID: PMC5953989 DOI: 10.1007/s10928-018-9579-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 02/07/2018] [Indexed: 01/24/2023]
Abstract
Trastuzumab is associated with cardiotoxicity, manifesting as a decrease of the left-ventricular ejection fraction (LVEF). Administration of anthracyclines prior to trastuzumab increases risk of cardiotoxicity. High-sensitive troponin T and N-terminal-pro-brain natriuretic peptide (NT-proBNP) are molecular markers that may allow earlier detection of drug-induced cardiotoxicity. In this analysis we aimed to quantify the kinetics and exposure–response relationships of LVEF, troponin T and NT-proBNP measurements, in patients receiving anthracycline and trastuzumab. Repeated measurements of LVEF, troponin T and NT-proBNP and dosing records of anthracyclines and trastuzumab were available from a previously published clinical trial. This trial included 206 evaluable patients with early breast cancer. Exposure to anthracycline and trastuzumab was simulated based on available dosing records and by using a kinetic-pharmacodynamic (K-PD) and a fixed pharmacokinetic (PK) model from literature, respectively. The change from baseline troponin T was described with a direct effect model, affected by simulated anthracycline concentrations, representing myocyte damage. The relationship between trastuzumab and LVEF was described by an indirect effect compartment model. The EC50 for LVEF decline was significantly affected by the maximum troponin T concentration after anthracycline treatment, explaining 15.1% of inter-individual variability. In this cohort, NT-proBNP changes could not be demonstrated to be related to anthracycline or trastuzumab treatment. Pharmacodynamic models for troponin T and LVEF were successfully developed, identifying maximum troponin T concentration after anthracycline treatment as a significant determinant for trastuzumab-induced LVEF decline. These models can help identify patients at risk of drug-induced cardiotoxicity and optimize cardiac monitoring strategies.
Collapse
|
38
|
Moazeni S, Cadeiras M, Yang EH, Deng MC, Nguyen KL. Anthracycline induced cardiotoxicity: biomarkers and "Omics" technology in the era of patient specific care. Clin Transl Med 2017; 6:17. [PMID: 28493232 PMCID: PMC5425361 DOI: 10.1186/s40169-017-0148-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2017] [Accepted: 05/04/2017] [Indexed: 12/17/2022] Open
Abstract
Anthracyclines are highly effective against a variety of malignancies. However, their dose-dependent cardiotoxic effects can potentially limit their use. In the past decade, serum biomarkers have been used to diagnose, monitor, predict, and prognosticate disease. Biomarkers such as cardiac troponin and natriuretic peptides have some predictive value, but still lack reliability in this patient population. Novel biomarkers such as galectin-3, soluble ST-2 proteins, myeloperoxidase, and fibrocytes are being explored as potential biomarkers to reliably predict the onset of cardiotoxicity. Leveraging multiomics technology to map highly sensitive biomarkers in an integrated approach through pattern deconvolution may better define those at highest risk of developing cardiotoxicity and further the goal of precision medicine. In this work, we aim to provide a brief overview of traditional serum biomarkers, summarize current investigations on novel circulating biomarkers, and discuss a systems-based approach to anthracycline-induced cardiotoxicity through "omics" technology.
Collapse
Affiliation(s)
- Shayan Moazeni
- Division of Cardiology, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA
- Division of Molecular Medicine, Department of Anesthesiology, David Geffen School of Medicine at UCLA, Los Angeles, CA USA
| | - Martin Cadeiras
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90024 USA
| | - Eric H. Yang
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90024 USA
| | - Mario C. Deng
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90024 USA
| | - Kim-Lien Nguyen
- Division of Cardiology, VA Greater Los Angeles Healthcare System, Los Angeles, CA USA
- Division of Cardiology, Department of Medicine, David Geffen School of Medicine at UCLA, 11301 Wilshire Blvd, MC 111E, Los Angeles, CA 90024 USA
| |
Collapse
|
39
|
Gulati G, Heck SL, Røsjø H, Ree AH, Hoffmann P, Hagve TA, Norseth J, Gravdehaug B, Steine K, Geisler J, Omland T. Neurohormonal Blockade and Circulating Cardiovascular Biomarkers During Anthracycline Therapy in Breast Cancer Patients: Results From the PRADA (Prevention of Cardiac Dysfunction During Adjuvant Breast Cancer Therapy) Study. J Am Heart Assoc 2017; 6:JAHA.117.006513. [PMID: 29118031 PMCID: PMC5721750 DOI: 10.1161/jaha.117.006513] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Anthracyclines are associated with cardiotoxic effects. Cardiovascular biomarkers may reflect myocardial injury, dysfunction, inflammation, and fibrosis and may precede and predict the development of left ventricular impairment. The aim of this study was to assess: (1) longitudinal change in circulating cardiovascular biomarkers, (2) the effect of metoprolol succinate and candesartan cilexetil on the biomarker response, and (3) the associations between on-treatment changes in biomarker concentrations and subsequent left ventricular dysfunction in patients with early breast cancer receiving anthracyclines. METHODS AND RESULTS This report encompasses 121 women included in the 2×2 factorial, placebo-controlled, double-blind PRADA (Prevention of Cardiac Dysfunction During Adjuvant Breast Cancer Therapy) trial with metoprolol and candesartan given concomitantly with anticancer therapy containing the anthracycline, epirubicin (total cumulative dose, 240-400 mg/m2). Cardiovascular magnetic resonance, echocardiography images, and circulating levels of biomarkers were obtained before and after anthracycline treatment. Cardiac troponins I and T, B-type natriuretic peptide, N-terminal pro-B-type natriuretic peptide, C-reactive protein, and galectin-3 increased during anthracycline therapy (all P<0.05). The troponin response was attenuated by metoprolol (P<0.05), but not candesartan. There was no association between change in biomarker concentrations and change in cardiac function during anthracycline therapy. CONCLUSIONS Treatment with contemporary anthracycline doses for early breast cancer is associated with increase in circulating cardiovascular biomarkers. This increase is, however, not associated with early decline in ventricular function. Beta-blockade may attenuate early myocardial injury, but whether this attenuation translates into reduced risk of developing ventricular dysfunction in the long term remains unclear. CLINICAL TRIAL REGISTRATION URL: http://www.clinicaltrial.gov. Unique identifier: NCT01434134.
Collapse
Affiliation(s)
- Geeta Gulati
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Norway
| | - Siri L Heck
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Norway
| | - Helge Røsjø
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Norway
| | - Anne H Ree
- Department of Oncology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Norway
| | - Pavel Hoffmann
- Department of Cardiology, Division of Cardiovascular and Pulmonary Diseases, Oslo University Hospital, Ullevål, Oslo, Norway
| | - Tor-Arne Hagve
- Institute of Clinical Medicine, University of Oslo, Norway.,Section for Medical Biochemistry, Division for Diagnostics and Technology, Akershus University Hospital, Lørenskog, Norway
| | - Jon Norseth
- Clinic for Medical Diagnostics, Vestre Viken Hospital Trust, Drammen, Norway
| | - Berit Gravdehaug
- Department of Breast and Endocrine Surgery, Division of Surgery, Akershus University Hospital, Lørenskog, Norway
| | - Kjetil Steine
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway.,Center for Heart Failure Research, University of Oslo, Norway
| | - Jürgen Geisler
- Department of Oncology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Norway
| | - Torbjørn Omland
- Department of Cardiology, Division of Medicine, Akershus University Hospital, Lørenskog, Norway .,Center for Heart Failure Research, University of Oslo, Norway
| |
Collapse
|
40
|
Yarana C, Carroll D, Chen J, Chaiswing L, Zhao Y, Noel T, Alstott M, Bae Y, Dressler EV, Moscow JA, Butterfield DA, Zhu H, St Clair DK. Extracellular Vesicles Released by Cardiomyocytes in a Doxorubicin-Induced Cardiac Injury Mouse Model Contain Protein Biomarkers of Early Cardiac Injury. Clin Cancer Res 2017; 24:1644-1653. [PMID: 29070527 DOI: 10.1158/1078-0432.ccr-17-2046] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 09/08/2017] [Accepted: 10/18/2017] [Indexed: 01/21/2023]
Abstract
Purpose: Cardiac injury is a major cause of death in cancer survivors, and biomarkers for it are detectable only after tissue injury has occurred. Extracellular vesicles (EV) remove toxic biomolecules from tissues and can be detected in the blood. Here, we evaluate the potential of using circulating EVs as early diagnostic markers for long-term cardiac injury.Experimental Design: Using a mouse model of doxorubicin (DOX)-induced cardiac injury, we quantified serum EVs, analyzed proteomes, measured oxidized protein levels in serum EVs released after DOX treatment, and investigated the alteration of EV content.Results: Treatment with DOX caused a significant increase in circulating EVs (DOX_EV) compared with saline-treated controls. DOX_EVs exhibited a higher level of 4-hydroxynonenal adducted proteins, a lipid peroxidation product linked to DOX-induced cardiotoxicity. Proteomic profiling of DOX_EVs revealed the distinctive presence of brain/heart, muscle, and liver isoforms of glycogen phosphorylase (GP), and their origins were verified to be heart, skeletal muscle, and liver, respectively. The presence of brain/heart GP (PYGB) in DOX_EVs correlated with a reduction of PYGB in heart, but not brain tissues. Manganese superoxide dismutase (MnSOD) overexpression, as well as pretreatment with cardioprotective agents and MnSOD mimetics, resulted in a reduction of EV-associated PYGB in mice treated with DOX. Kinetic studies indicated that EVs containing PYGB were released prior to the rise of cardiac troponin in the blood after DOX treatment, suggesting that PYGB is an early indicator of cardiac injury.Conclusions: EVs containing PYGB are an early and sensitive biomarker of cardiac injury. Clin Cancer Res; 24(7); 1644-53. ©2017 AACRSee related commentary by Zhu and Gius, p. 1516.
Collapse
Affiliation(s)
- Chontida Yarana
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky.,Faculty of Medical Technology, Mahidol University, Salaya, Thailand
| | - Dustin Carroll
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Jing Chen
- Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky
| | - Luksana Chaiswing
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Yanming Zhao
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Teresa Noel
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky
| | - Michael Alstott
- Markey Cancer Center, Redox Metabolism Shared Resource Facility, University of Kentucky, Lexington, Kentucky
| | - Younsoo Bae
- Department of Pharmaceutical Sciences, University of Kentucky, Lexington, Kentucky
| | - Emily V Dressler
- Division of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, North Carolina
| | - Jeffrey A Moscow
- Investigational Drug Branch, National Cancer Institute, Bethesda, Maryland
| | - D Allan Butterfield
- Markey Cancer Center, Redox Metabolism Shared Resource Facility, University of Kentucky, Lexington, Kentucky.,Department of Chemistry, University of Kentucky, Lexington, Kentucky
| | - Haining Zhu
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky.,Department of Molecular and Cellular Biochemistry, University of Kentucky, Lexington, Kentucky.,Markey Cancer Center, Redox Metabolism Shared Resource Facility, University of Kentucky, Lexington, Kentucky
| | - Daret K St Clair
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, Kentucky.
| |
Collapse
|
41
|
Cardiac Nonmyocyte Cell Functions and Crosstalks in Response to Cardiotoxic Drugs. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:1089359. [PMID: 29201269 PMCID: PMC5671742 DOI: 10.1155/2017/1089359] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 09/24/2017] [Indexed: 01/06/2023]
Abstract
The discovery of the molecular mechanisms involved in the cardiac responses to anticancer drugs represents the current goal of cardio-oncology research. The oxidative stress has a pivotal role in cardiotoxic responses, affecting the function of all types of cardiac cells, and their functional crosstalks. Generally, cardiomyocytes are the main target of research studies on cardiotoxicity, but recently the contribution of the other nonmyocyte cardiac cells is becoming of growing interest. This review deals with the role of oxidative stress, induced by anticancer drugs, in cardiac nonmyocyte cells (fibroblasts, vascular cells, and immune cells). The alterations of functional interplays among these cardiac cells are discussed, as well. These interesting recent findings increase the knowledge about cardiotoxicity and suggest new molecular targets for both diagnosis and therapy.
Collapse
|
42
|
Alrushaid S, Sayre CL, Yáñez JA, Forrest ML, Senadheera SN, Burczynski FJ, Löbenberg R, Davies NM. Pharmacokinetic and Toxicodynamic Characterization of a Novel Doxorubicin Derivative. Pharmaceutics 2017; 9:pharmaceutics9030035. [PMID: 28902176 PMCID: PMC5620576 DOI: 10.3390/pharmaceutics9030035] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2017] [Revised: 09/11/2017] [Accepted: 09/11/2017] [Indexed: 11/16/2022] Open
Abstract
Doxorubicin (Dox) is an effective anti-cancer medication with poor oral bioavailability and systemic toxicities. DoxQ was developed by conjugating Dox to the lymphatically absorbed antioxidant quercetin to improve Dox’s bioavailability and tolerability. The purpose of this study was to characterize the pharmacokinetics and safety of Dox after intravenous (IV) and oral (PO) administration of DoxQ or Dox (10 mg/kg) and investigate the intestinal lymphatic delivery of Dox after PO DoxQ administration in male Sprague–Dawley rats. Drug concentrations in serum, urine, and lymph were quantified by HPLC with fluorescence detection. DoxQ intact IV showed a 5-fold increase in the area under the curve (AUC)—18.6 ± 1.98 compared to 3.97 ± 0.71 μg * h/mL after Dox—and a significant reduction in the volume of distribution (Vss): 0.138 ± 0.015 versus 6.35 ± 1.06 L/kg. The fraction excreted unchanged in urine (fe) of IV DoxQ and Dox was ~5% and ~11%, respectively. Cumulative amounts of Dox in the mesenteric lymph fluid after oral DoxQ were twice as high as Dox in a mesenteric lymph duct cannulation rat model. Oral DoxQ increased AUC of Dox by ~1.5-fold compared to after oral Dox. Concentrations of β-N-Acetylglucosaminidase (NAG) but not cardiac troponin (cTnI) were lower after IV DoxQ than Dox. DoxQ altered the pharmacokinetic disposition of Dox, improved its renal safety and oral bioavailability, and is in part transported through intestinal lymphatics.
Collapse
Affiliation(s)
- Samaa Alrushaid
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| | - Casey L Sayre
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
- College of Pharmacy, Roseman University of Health Sciences, South Jordan, UT 84096, USA.
| | - Jaime A Yáñez
- YARI International Group, New Brunswick, NJ 08901 and INDETEC Corp., Lima, Peru.
| | - M Laird Forrest
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA.
| | - Sanjeewa N Senadheera
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA.
| | - Frank J Burczynski
- College of Pharmacy, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0T5, Canada.
| | - Raimar Löbenberg
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| | - Neal M Davies
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, AB T6G 2R3, Canada.
| |
Collapse
|
43
|
Rac1-mediated cardiac damage causes diastolic dysfunction in a mouse model of subacute doxorubicin-induced cardiotoxicity. Arch Toxicol 2017; 92:441-453. [PMID: 28710503 DOI: 10.1007/s00204-017-2017-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2017] [Accepted: 06/14/2017] [Indexed: 01/13/2023]
Abstract
The anticancer efficacy of anthracyclines is limited by congestive heart failure. Clinically established markers of early onset of cardiotoxicity following anthracycline treatment and preventive measures are missing. Although statins are reported to alleviate anthracycline-induced cardiotoxicity in vivo, the molecular mechanisms involved remain elusive. In vitro data point to Rac1 as major target of the cytoprotective statin effects. Here we investigated whether specific inhibition of Rac1 by NSC23766 is as effective as lovastatin in preventing subacute cardiotoxicity following doxorubicin treatment. C57BL/6 mice were treated over 3 weeks with multiple low doses of doxorubicin (6 × 3 mg/kg BW, i.p.) and the level of DNA damage, apoptosis and regenerative proliferation as well as pro-inflammatory, pro-fibrotic and oxidative stress responses were investigated. Moreover, heart function was monitored by echocardiography. Doxorubicin induced subacute cardiotoxicity which was reflected on the level of residual DNA damage, frequency of apoptotic and mitotic cells as well as elevated mRNA expression of markers of heart failure, remodeling and mitochondrial biogenesis. These molecular markers of cardiotoxicity were mitigated to a similar extent by co-treatment with either lovastatin (10 mg/kg BW, p.o.) or NSC23766 (5 mg/kg BW, i.p.) three times a week. Moreover, doxorubicin caused diastolic dysfunction as reflected by increased E-wave acceleration time (EAT), which again was prevented by pharmacological inhibition of Rac1. Inhibition of Rac1 signaling is of major relevance for the cardioprotective effects of lovastatin in the context of anthracycline-induced cardiotoxicity. Moreover, EAT is a useful marker of subacute cardiotoxicity caused by persisting harmful effects of doxorubicin.
Collapse
|
44
|
Dokmanovic M, King KE, Mohan N, Endo Y, Wu WJ. Cardiotoxicity of ErbB2-targeted therapies and its impact on drug development, a spotlight on trastuzumab. Expert Opin Drug Metab Toxicol 2017; 13:755-766. [PMID: 28571477 DOI: 10.1080/17425255.2017.1337746] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
INTRODUCTION Trastuzumab, a therapeutic monoclonal antibody directed against ErbB2, is often noted as a successful example of targeted therapy. Trastuzumab improved outcomes for many patients with ErbB2-positive breast and gastric cancers, however, cardiac side effects [e.g., left ventricular dysfunction and congestive heart failure (CHF)] were reported in the early phase clinical studies. This finding, subsequently corroborated by multiple clinical studies, raised concerns that the observed cardiotoxicity induced by trastuzumab might adversely impact the clinical development of other therapeutics targeting ErbB family members. Areas covered: In this review we summarize both basic research and clinical findings regarding trastuzumab-induced cardiotoxicity and assess if there has been an impact of trastuzumab-induced cardiotoxicity on the development of other agents targeting ErbB family members. Expert opinion: There are a number of scientific gaps that are critically important to address for the continued success of HER2-targeted agents. These include: 1) elucidating the molecular mechanisms contributing to cardiotoxicity; 2) developing relevant preclinical testing systems for predicting cardiotoxicity; 3) developing clinical strategies to identify patients at risk of cardiotoxicity; and 4) enhancing management of clinical symptoms of cardiotoxicity.
Collapse
Affiliation(s)
- Milos Dokmanovic
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Kathryn E King
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Nishant Mohan
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Yukinori Endo
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| | - Wen Jin Wu
- a Division of Biotechnology Review and Research I, Office of Biotechnology Products, Office of Pharmaceutical Quality, Center for Drug Evaluation and Research , U.S. Food and Drug Administration , Silver Spring , MD 20993 , USA
| |
Collapse
|
45
|
Radu LE, Beldiman A, Ghiorghiu I, Oprescu A, Arion C, Coliță A. The use of biomarkers in detecting subclinical cardiotoxicity in doxorubicin-based treatment for paediatric patients with acute lymphoblastic leukaemia. REV ROMANA MED LAB 2017. [DOI: 10.1515/rrlm-2017-0014] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Abstract
The international standard protocol for acute lymphoblastic leukaemia (ALL), the most common haemato-oncological pathology at paediatric age, uses anthracyclines as antitumor agents, potentially associated with early or late onset cardiac damage. Currently, echocardiography is the gold standard in the diagnosis of cardiotoxicity, but several biomarkers are evaluated as a possible replacement, pending more extensive clinical studies. We started a prospective study in order to determine the role of two biomarkers, troponin and heart-type fatty acid binding protein, in the evaluation of cardiotoxicity in children over one year of age, diagnosed with ALL. Between February 2015 and April 2016, 20 patients were enrolled and monitored at diagnosis, during chemotherapy and four months after the end of reinduction, through cardiac evaluation and dosing of those two markers in five different points of the treatment protocol. During the first year of follow-up, the patients did not develop clinical signs of cardiac damage, but the study showed a slight increase in troponin levels during chemotherapy, with the return to baseline value after treatment cessation, and also a correlation with the total dose of anthracyclines given to the patient. On the other hand, the second biomarker, heart-type fatty acid binding protein, did not seem to be useful in detecting subclinical cardiac damage in these patients.
Collapse
Affiliation(s)
- Letiția Elena Radu
- Paediatric Haematology and Oncology, Fundeni Clinical Institute, , Bucharest , Romania
| | | | - Ioana Ghiorghiu
- “Carol Davila” University of Medicine and Pharmacy, Fundeni Clinical Institute , Bucharest , Romania
- V. Gomoiu Children’s Hospital, , Bucharest , Romania
| | - Alina Oprescu
- Institute for Cardiovascular Diseases and Transplantation , Tirgu Mures , Romania
| | - Constantin Arion
- “Carol Davila” University of Medicine and Pharmacy, Fundeni Clinical Institute, , Bucharest , Romania
| | - Anca Coliță
- “Carol Davila” University of Medicine and Pharmacy, Fundeni Clinical Institute, , Bucharest , Romania
| |
Collapse
|
46
|
Pilot study to determine the feasibility of radiation therapy for dogs with right atrial masses and hemorrhagic pericardial effusion. J Vet Cardiol 2017; 19:132-143. [DOI: 10.1016/j.jvc.2016.12.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Revised: 12/07/2016] [Accepted: 12/12/2016] [Indexed: 11/18/2022]
|
47
|
Cuaron JJ, MacDonald SM, Cahlon O. Novel applications of proton therapy in breast carcinoma. Chin Clin Oncol 2017; 5:52. [PMID: 27558253 DOI: 10.21037/cco.2016.06.04] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 05/28/2016] [Indexed: 12/18/2022]
Abstract
This review will focus on the indications, clinical experience, and technical considerations of proton beam radiation therapy in the treatment of patients with breast cancer. For patients with early stage disease, proton therapy delivers less dose to non-target breast tissue for patients receiving partial breast irradiation (PBI) therapy, which may result in improved cosmesis but requires further investigation. For patients with locally advanced breast cancer requiring treatment to the regional lymph nodes, proton therapy allows for an improved dosimetric profile compared with conventional photon and electron techniques. Early clinical results demonstrate acceptable toxicity. The possible reduction in cardiopulmonary events as a result of reduced dose to organs at risk will be tested in a randomized control trial of protons vs. photons.
Collapse
Affiliation(s)
- John J Cuaron
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| | - Shannon M MacDonald
- Massachusetts General Hospital, Francis H. Burr Proton Therapy Center, Boston, MA 02114-7250, USA
| | - Oren Cahlon
- Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
48
|
Puppe J, van Ooyen D, Neise J, Thangarajah F, Eichler C, Krämer S, Pfister R, Mallmann P, Wirtz M, Michels G. Evaluation of QTc Interval Prolongation in Breast Cancer Patients after Treatment with Epirubicin, Cyclophosphamide, and Docetaxel and the Influence of Interobserver Variation. Breast Care (Basel) 2017; 12:40-44. [PMID: 28611540 DOI: 10.1159/000455065] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Chemotherapy with anthracyclines is associated with life-threatening electrocardiographic alterations including corrected QT (QTc) interval prolongation. PATIENTS AND METHODS In this study we assessed the effect of epirubicin, cyclophosphamide, and docetaxel (EC-Doc) on the QTc interval in 10 patients with early breast cancer. Cardiac toxicity was assessed with symptoms, transthoracic echocardiography, electrocardiography (ECG), and serum cardiac markers at baseline and after 4 cycles of EC and 4 cycles of docetaxel. To evaluate the influence of interobserver variation, the QTc interval was analyzed by a cardiologist, a gynecologist, and with automated ECG interpretation software. RESULTS There was a significant QTc prolongation after EC treatment independent of the investigator. In addition, a significant increase in N-terminal prohormone of brain natriuretic peptide (NT-proBNP) levels was noted after EC treatment. QTc prolongation and NT-proBNP levels normalized after docetaxel treatment. Other biochemical markers were within normal ranges. No clinically relevant effect on left ventricular ejection fraction was observed. CONCLUSION This prospective study demonstrated that EC treatment increases the QTc interval and NT-proBNP levels in women with early breast cancer. This effect was reversible and independent of docetaxel administration. Moreover, the treating physician can safely perform QTc interval evaluation as part of clinical routine independent of his/her specialty. Due to the small number of patients, further conclusions are limited at this point.
Collapse
Affiliation(s)
- Julian Puppe
- Department of Gynaecology and Obstetrics, University Hospital Cologne, Cologne, Germany.,CIO Center for Integrated Oncology Cologne/Bonn, Bonn, Germany
| | - Deborah van Ooyen
- Department of Gynaecology and Obstetrics, University Hospital Cologne, Cologne, Germany.,CIO Center for Integrated Oncology Cologne/Bonn, Bonn, Germany
| | - Jeanne Neise
- Department of Gynaecology and Obstetrics, University Hospital Cologne, Cologne, Germany.,CIO Center for Integrated Oncology Cologne/Bonn, Bonn, Germany
| | - Fabinshy Thangarajah
- Department of Gynaecology and Obstetrics, University Hospital Cologne, Cologne, Germany.,CIO Center for Integrated Oncology Cologne/Bonn, Bonn, Germany
| | | | - Stefan Krämer
- Department of Gynaecology and Obstetrics, University Hospital Cologne, Cologne, Germany.,CIO Center for Integrated Oncology Cologne/Bonn, Bonn, Germany
| | - Roman Pfister
- Department III of Internal Medicine, Heart Centre of the University of Cologne, Cologne, Germany
| | - Peter Mallmann
- Department of Gynaecology and Obstetrics, University Hospital Cologne, Cologne, Germany.,CIO Center for Integrated Oncology Cologne/Bonn, Bonn, Germany
| | - Marina Wirtz
- Department of Gynaecology and Obstetrics, University Hospital Cologne, Cologne, Germany.,CIO Center for Integrated Oncology Cologne/Bonn, Bonn, Germany
| | - Guido Michels
- Department III of Internal Medicine, Heart Centre of the University of Cologne, Cologne, Germany
| |
Collapse
|
49
|
Marlière S, Vautrin E, Saunier C, Chaikh A, Gabelle-Flandin I. [Radiation-related heart toxicity: Update in women]. Ann Cardiol Angeiol (Paris) 2016; 65:411-419. [PMID: 27842711 DOI: 10.1016/j.ancard.2016.10.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Breast cancer is a common diagnosis in women and thus women are at risk of radiation-induced heart disease, in particular during radiotherapy for left breast cancer and when the internal mammary chain is included. Rates of major cardiac events increase with younger age at the time of irradiation, diagnosis before 1990s, higher radiation doses, coexisting cardiovascular risk factors and adjuvant cardiotoxic chemotherapy. Radiation-induced heart disease comprises a spectrum of cardiac pathologies, including pericardial disease, cardiomyopathy, coronary artery disease and valvular disease. The cardiac injury can appear a long time after radiotherapy and can consist of complex lesions with poor prognosis. The disciplines of cardiology and oncology have increasingly recognized the benefits of collaborating in the care of cancer patients with cardiac disease, developing guidelines for the assessment and management of radiation-related cardiovascular disease. We could consider screening patients with previous chest radiation every 5 years with transthoracic echocardiography and functional imaging. However, prevention remains the primary goal, using cardiac sparing doses and avoidance techniques in radiotherapy to improve patient survival.
Collapse
Affiliation(s)
- S Marlière
- Clinique universitaire de cardiologie, centre hospitalier universitaire de Grenoble-Alpes, BP 217, 38043 Grenoble cedex 9, France.
| | - E Vautrin
- Clinique universitaire de cardiologie, centre hospitalier universitaire de Grenoble-Alpes, BP 217, 38043 Grenoble cedex 9, France
| | - C Saunier
- Clinique universitaire de cardiologie, centre hospitalier universitaire de Grenoble-Alpes, BP 217, 38043 Grenoble cedex 9, France
| | - A Chaikh
- Service de cancérologie-radiothérapie, centre hospitalier universitaire de Grenoble-Alpes, BP 217, 38043 Grenoble cedex 9, France
| | - I Gabelle-Flandin
- Service de cancérologie-radiothérapie, centre hospitalier universitaire de Grenoble-Alpes, BP 217, 38043 Grenoble cedex 9, France
| |
Collapse
|
50
|
Jacob S, Ferrières J. Breast cancer radiotherapy: A case of double jeopardy. Arch Cardiovasc Dis 2016; 109:587-590. [PMID: 27836785 DOI: 10.1016/j.acvd.2016.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2016] [Revised: 09/05/2016] [Accepted: 09/07/2016] [Indexed: 10/20/2022]
Affiliation(s)
- Sophie Jacob
- Institut de Radioprotection et de Sureté Nucléaire (IRSN), PRP-HOM, SRBE, LEPID, Laboratory of Epidemiology, 92262 Fontenay-aux-Roses, France.
| | - Jean Ferrières
- Department of Cardiology, Toulouse University School of Medicine, Rangueil Hospital, TSA 50032, 31059 Toulouse, France
| |
Collapse
|