1
|
Makihara K, Kongo K, Motomura K, Kimoto D, Yamamoto Y, Tanihata M, Yoshidome M, Matsumura T. Optimal follow-up duration of cardiac function tests in patients treated with trastuzumab: an analysis using the Japanese Adverse Drug Event Report (JADER) database. Int J Clin Oncol 2025; 30:886-892. [PMID: 40009314 DOI: 10.1007/s10147-025-02727-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
BACKGROUND One of the most serious adverse events associated with trastuzumab treatment is cardiac dysfunction, including congestive heart failure. Therefore, regular cardiac screening with echocardiography is commonly performed during trastuzumab treatment, although it is unclear for how long the patient will continue to be evaluated. We investigated the time to the occurrence of trastuzumab-induced cardiac dysfunction using the Japanese Adverse Drug Event Report (JADER) database. We examined the optimal duration of cardiac function evaluation in patients treated with trastuzumab. METHODS This study used data registered between April 2004 and September 2023 in the JADER database. We investigated the time to onset of cardiotoxicity in patients treated with trastuzumab, trastuzumab emtansine, or trastuzumab deruxtecan. We considered the time to exclude outliers detected using the Smirnov-Grubbs test as the optimal follow-up duration for cardiac function tests. RESULTS Of 868,478 patients who reported adverse drug events, 977 experienced cardiac dysfunctions among those treated with trastuzumab. A total of 375 patients were included in the analysis after excluding patients for whom the time to onset of cardiotoxicity was unknown or those who experienced cardiac dysfunction after receiving trastuzumab followed by anthracycline. The median time to cardiotoxicity was 4.5 months (range 0-100 months). However, ≥ 19 months after the start of trastuzumab administration was detected as an outlier in the target population (P = 0.036). CONCLUSION The duration of regular follow-up of cardiac function using echocardiography during anti-HER2 therapy can be 18 months from the start of treatment.
Collapse
Affiliation(s)
- Katsuya Makihara
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan.
| | - Keisuke Kongo
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan
| | - Kayo Motomura
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan
| | - Daiki Kimoto
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan
| | - Yukako Yamamoto
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan
| | - Misato Tanihata
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan
| | - Mieko Yoshidome
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan
| | - Tomokazu Matsumura
- Department of Pharmacy, Yodogawa Christian Hospital, 1-7-50 Kunijima, Higashi Yodogawa-Ku, Osaka, 533-0024, Japan
| |
Collapse
|
2
|
Zhang X, Yin Y, Yu Q, Chen X, Cheng Y. Review of the clinical status of cardiotoxicity of HER-2 positive breast cancer targeted therapeutic drugs. Front Oncol 2025; 14:1492203. [PMID: 39991185 PMCID: PMC11842234 DOI: 10.3389/fonc.2024.1492203] [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: 09/06/2024] [Accepted: 12/30/2024] [Indexed: 02/25/2025] Open
Abstract
Breast cancer is a major health challenge for women worldwide, and human epidermal growth factor receptor 2 (HER-2)-positive breast cancers have a relatively high incidence and are highly aggressive. Targeted therapeutic agents, represented by trastuzumab, have been effective in improving the survival rate of HER-2-positive breast cancer patients. However, in clinical applications, this type of targeted drugs exhibits varying degrees of cardiotoxicity, and the mechanism of their cardiotoxicity is currently unclear. In this paper, we classify them into three categories: monoclonal antibodies (mAbs), small-molecule tyrosine kinase inhibitors (TKIs), and antibody-drug conjugate (ADCs). We list the evidence of cardiotoxicity for various drugs based on current clinical trials and summarize their corresponding epidemiological profiles. We also discuss the regulation of cardiotoxicity from three perspectives: clinical biomarkers of cardiotoxicity, permissive cardiotoxicity, and the current status of cardiotoxicity regulation.
Collapse
Affiliation(s)
- Xiang Zhang
- Department of Cardiology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yulian Yin
- Department of Breast Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qiuting Yu
- Department of Cardiology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xinlin Chen
- Hospital Administration Office, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yiqin Cheng
- Department of Breast Surgery, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Kettana KM, El-Haggar SM, Alm El-Din MA, El-Afify DR. Possible protective effect of rosuvastatin in chemotherapy-induced cardiotoxicity in HER2 positive breast cancer patients: a randomized controlled trial. Med Oncol 2024; 41:196. [PMID: 38977536 PMCID: PMC11230999 DOI: 10.1007/s12032-024-02426-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 06/10/2024] [Indexed: 07/10/2024]
Abstract
Cardiotoxicity is a side effect of chemotherapy in human epidermal growth factor receptor 2 (HER2) positive breast cancer patients receiving both anthracyclines and trastuzumab. We looked for a possible protective effect of rosuvastatin against chemotherapy-induced cardiotoxicity. Methods: 50 newly diagnosed HER2 positive breast cancer patients were randomly allocated into two groups: 25patients in each. Group 1(control group) received doxorubicin for 4 cycles (3 months) followed by trastuzumab adjuvant therapy. Group 2 (treatment group) received doxorubicin for 4 cycles (3 months) followed by trastuzumab adjuvant therapy and 20 mg of oral rosuvastatin 24 h before the first cycle of chemotherapy and once daily for the rest of the follow-up period (6 months). Transthoracic echocardiography was done, and blood samples were collected for patients 24 h before the initiation of therapy, after 3 months and after 6 months to assess serum levels of high sensitivity cardiac troponin I (hs-cTnI), Myeloperoxidase (MPO), Interleukin-6 (IL-6) and Alanine aminotransferase (ALT). The study was retrospectively registered in Clinical Trials.gov in April 2022. Its ID is NCT05338723. Compared to control group, Rosuvastatin-treated group had a significantly lower decline in LVEF after 3 months and after 6 months. They had significantly lower Hs-cTnI and IL-6 after 3 months and after 6 months, and significantly lower MPO after 6 months. Four patients in control group experienced cardiotoxicity while no one in rosuvastatin-treated group. Rosuvastatin attenuated cardiotoxicity, so it is a promising protective agent against chemotherapy-induced cardiotoxicity.
Collapse
Affiliation(s)
- Khlood M Kettana
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt.
| | - Sahar M El-Haggar
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Mohamed A Alm El-Din
- Clinical Oncology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dalia R El-Afify
- Clinical Pharmacy Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| |
Collapse
|
4
|
Wernhart S, Rassaf T. Relevance of Cardiovascular Exercise in Cancer and Cancer Therapy-Related Cardiac Dysfunction. Curr Heart Fail Rep 2024; 21:238-251. [PMID: 38696059 PMCID: PMC11090948 DOI: 10.1007/s11897-024-00662-0] [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] [Accepted: 04/03/2024] [Indexed: 05/14/2024]
Abstract
PURPOSE OF THE REVIEW Cancer therapy-related cardiac dysfunction (CTRCD) has been identified as a threat to overall and cancer-related survival. Although aerobic exercise training (AET) has been shown to improve cardiorespiratory fitness (CRF), the relationship between specific exercise regimens and cancer survival, heart failure development, and reduction of CTRCD is unclear. In this review, we discuss the impact of AET on molecular pathways and the current literature of sports in the field of cardio-oncology. RECENT FINDINGS Cardio-oncological exercise trials have focused on variations of AET intensity by using moderate continuous and high intensity interval training, which are applicable, safe, and effective approaches to improve CRF. AET increases CRF, reduces cardiovascular morbidity and heart failure hospitalization and should thus be implemented as an adjunct to standard cancer therapy, although its long-term effect on CTRCD remains unknown. Despite modulating diverse molecular pathways, it remains unknown which exercise regimen, including variations of AET duration and frequency, is most suited to facilitate peripheral and central adaptations to exercise and improve survival in cancer patients.
Collapse
Affiliation(s)
- Simon Wernhart
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart- and Vascular Center, University Hospital Essen, University Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany
| |
Collapse
|
5
|
Jiang J, Liu B, Hothi SS. Herceptin-Mediated Cardiotoxicity: Assessment by Cardiovascular Magnetic Resonance. Cardiol Res Pract 2022; 2022:1910841. [PMID: 35265371 PMCID: PMC8898877 DOI: 10.1155/2022/1910841] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 10/12/2021] [Accepted: 01/20/2022] [Indexed: 02/07/2023] Open
Abstract
Herceptin (trastuzumab) is a recombinant, humanized, monoclonal antibody that targets the human epidermal growth factor receptor 2 (HER2) and is used in the treatment of HER2-positive breast and gastric cancers. However, it carries a risk of cardiotoxicity, manifesting as left ventricular (LV) systolic dysfunction, conventionally assessed for by transthoracic echocardiography. Clinical surveillance of cardiac function and discontinuation of trastuzumab at an early stage of LV systolic dysfunction allow for the timely initiation of heart failure drug therapies that can result in the rapid recovery of cardiac function in most patients. Often considered the reference standard for the noninvasive assessment of cardiac volume and function, cardiac magnetic resonance (CMR) imaging has superior reproducibility and accuracy compared to other noninvasive imaging modalities. However, due to limited availability, it is not routinely used in the serial assessment of cardiac function in patients receiving trastuzumab. In this article, we review the diagnostic and prognostic role of CMR in trastuzumab-mediated cardiotoxicity.
Collapse
Affiliation(s)
- Jin Jiang
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
| | - Boyang Liu
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Sandeep S Hothi
- Heart and Lung Centre, New Cross Hospital, Wolverhampton, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
6
|
Bohdan M, Kowalczys A, Mickiewicz A, Gruchała M, Lewicka E. Cancer Therapy-Related Cardiovascular Complications in Clinical Practice: Current Perspectives. J Clin Med 2021; 10:1647. [PMID: 33924543 PMCID: PMC8069381 DOI: 10.3390/jcm10081647] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 04/02/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022] Open
Abstract
Cardiovascular (CV) diseases and cancer are the leading causes of death in Europe and the United States. Both diseases have extensive overlap and share common risk factors, symptoms, and outcomes. As the number of patients with both cancer and CV diseases continues to rise, the field of cardio-oncology is gaining increased attention. A frequent problem during anti-cancer treatment is cardiotoxicity caused by the side-effects of chemo-, immuno-, targeted, and radiation therapies. This problem may manifest as acute coronary syndrome, myocarditis, arrhythmias, or heart failure. Modern cardio-oncology spans many different research areas. While some researchers focus on treating patients that have already developed cardiotoxicity, others aim to identify new methods for preventing cardiotoxicity before, during, and after anti-cancer therapy. Both groups share the common understanding that regular monitoring of cancer patients is the basis for optimal medical treatment. Optimal treatment can only be achieved through close cooperation between cardiologists and oncologists. This review summarizes the current views on cardio-oncology and discusses the cardiotoxicities associated with commonly used chemotherapeutics.
Collapse
Affiliation(s)
- Michał Bohdan
- First Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (A.M.); (M.G.)
| | - Anna Kowalczys
- First Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (A.M.); (M.G.)
| | - Agnieszka Mickiewicz
- First Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (A.M.); (M.G.)
| | - Marcin Gruchała
- First Department of Cardiology, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (A.K.); (A.M.); (M.G.)
| | - Ewa Lewicka
- Department of Cardiology and Electrotherapy, Medical University of Gdańsk, 80-211 Gdańsk, Poland;
| |
Collapse
|
7
|
Wu Q, Bai B, Tian C, Li D, Yu H, Song B, Li B, Chu X. The Molecular Mechanisms of Cardiotoxicity Induced by HER2, VEGF, and Tyrosine Kinase Inhibitors: an Updated Review. Cardiovasc Drugs Ther 2021; 36:511-524. [PMID: 33847848 DOI: 10.1007/s10557-021-07181-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/31/2021] [Indexed: 02/07/2023]
Abstract
AIM In recent decades, there has been a revolutionary decrease in cancer-related mortality and an increase in survival due to the introduction of novel targeted drugs. Nevertheless, drugs targeting human epidermal growth factor receptor 2 (HER-2), angiogenesis, and other tyrosine kinases also come with unexpected cardiac side effects, including heart failure, hypertension, arterial thrombosis, and arrhythmias, and have mechanisms that are unlike those of classic chemotherapeutic agents. In addition, it is challenging to address some problems, as the existing guidelines need to be more specific, and further large-scale clinical trials and experimental studies are required to confirm the benefit of administering cardioprotective agents to patients treated with targeted therapies. Therefore, an improved understanding of cardiotoxicity becomes increasingly important to minimize the pernicious effects and maximize the beneficial effects of targeted agents. METHODS "Cardiotoxicity", "targeted drugs", "HER2", "trastuzumab", "angiogenesis inhibitor", "VEGF inhibitor" and "tyrosine kinase inhibitors" are used as keywords for article searches. RESULTS In this article, we report several targeted therapies that induce cardiotoxicity and update knowledge of the clinical evidence, molecular mechanisms, and management measures.
Collapse
Affiliation(s)
- Qinchao Wu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Baochen Bai
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Chao Tian
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Haichu Yu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Bingxue Song
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China
| | - Bing Li
- Department of Hematology, The Affiliated Hospital of Qingdao University, No.16 Jiangsu Road, Qingdao, Shandong, China.
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, No. 308 Ningxia Road, Qingdao, 266000, Shandong, China.
| | - Xianming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No. 59 Haier Road, Qingdao, 266100, Shandong, China.
- The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, Shandong, China.
| |
Collapse
|
8
|
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: 10] [Impact Index Per Article: 2.0] [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
|
9
|
Bouwer NI, Jager A, Liesting C, Kofflard MJM, Brugts JJ, Kitzen JJEM, Boersma E, Levin MD. Cardiac monitoring in HER2-positive patients on trastuzumab treatment: A review and implications for clinical practice. Breast 2020; 52:33-44. [PMID: 32361151 PMCID: PMC7375662 DOI: 10.1016/j.breast.2020.04.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 03/17/2020] [Accepted: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Trastuzumab prolongs progression-free and overall survival in patients with human epidermal growth factor receptor 2 (HER2) positive breast cancer. However, trastuzumab treatment is hampered by cardiotoxicity, defined as a left ventricular ejection fraction (LVEF) decline with a reported incidence ranging from 3 to 27% depending on variable factors. Early identification of patients at increased risk of trastuzumab-induced myocardial damage is of great importance to prevent deterioration to irreversible cardiotoxicity. Although current cardiac monitoring with multi gated acquisition (MUGA) scanning and/or conventional 2D-echocardiography (2DE) have a high availability, their reproducibility are modest, and more sensitive and reliable techniques are needed such as 3D-echocardiography (3DE) and speckle tracking echocardiography (STE). But which other diagnostic imaging modalities are available for patients before and during trastuzumab treatment? In addition, what is the optimal frequency and duration of cardiac monitoring? At last, which biomarker monitoring strategies are currently available for the identification of cardiotoxicity in patients treated with trastuzumab?
Collapse
Affiliation(s)
- Nathalie I Bouwer
- Department of Cardiology, Albert Schweitzer Hospital, Albert Schweitzerplaats 25, 3300 AK, Dordrecht, the Netherlands; Department of Internal Medicine, Albert Schweitzer Hospital, Albert Schweitzerplaats 25, 3300 AK, Dordrecht, the Netherlands
| | - Agnes Jager
- Department of Medical Oncology, Erasmus MC Cancer Institute, Dr. Molewaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - Crista Liesting
- Department of Cardiology, Albert Schweitzer Hospital, Albert Schweitzerplaats 25, 3300 AK, Dordrecht, the Netherlands
| | - Marcel J M Kofflard
- Department of Cardiology, Albert Schweitzer Hospital, Albert Schweitzerplaats 25, 3300 AK, Dordrecht, the Netherlands
| | - Jasper J Brugts
- Department of Cardiology, Erasmus MC Thoraxcenter, Dr. Molewaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - Jos J E M Kitzen
- Department of Internal Medicine, Albert Schweitzer Hospital, Albert Schweitzerplaats 25, 3300 AK, Dordrecht, the Netherlands
| | - Eric Boersma
- Department of Cardiology, Erasmus MC Thoraxcenter, Dr. Molewaterplein 40, 3000 CA, Rotterdam, the Netherlands
| | - Mark-David Levin
- Department of Internal Medicine, Albert Schweitzer Hospital, Albert Schweitzerplaats 25, 3300 AK, Dordrecht, the Netherlands.
| |
Collapse
|
10
|
Avila MS, Siqueira SRR, Ferreira SMA, Bocchi EA. Prevention and Treatment of Chemotherapy-Induced Cardiotoxicity. Methodist Debakey Cardiovasc J 2020; 15:267-273. [PMID: 31988687 DOI: 10.14797/mdcj-15-4-267] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Recent decades have seen an increase in survival rates for cancer patients, partially explained by earlier diagnoses and new chemotherapeutic agents. However, chemotherapy may be associated with adverse cardiovascular events, including hypertension and pulmonary hypertension, supraventricular and ventricular arrhythmias, cardiomyopathy, and other forms of cardiovascular disease. For patients, the benefits of chemotherapy may be partially obfuscated by deleterious effects on the cardiovascular system, resulting in a significant increase in morbidity and mortality. In this article, we review strategies for prevention and treatment of chemotherapy-related cardiotoxicity.
Collapse
Affiliation(s)
- Monica Samuel Avila
- HOSPITAL DAS CLINICAS DA FACULDADE DE MEDICINA DA UNIVERSIDADE DE SAO PAULO, SAO PAULO, BRAZIL
| | | | | | - Edimar Alcides Bocchi
- HOSPITAL DAS CLINICAS DA FACULDADE DE MEDICINA DA UNIVERSIDADE DE SAO PAULO, SAO PAULO, BRAZIL
| |
Collapse
|
11
|
Makavos G, Ikonomidis I, Palios J, Rigopoulos A, Katogiannis K, Parissis J, Paraskevaidis I, Noutsias M. Cardiac imaging in cardiotoxicity: a focus on clinical practice. Heart Fail Rev 2020; 26:1175-1187. [PMID: 32306221 DOI: 10.1007/s10741-020-09952-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Cancer therapeutics induced cardiotoxicity has emerged as an important factor of long-term adverse cardiovascular outcomes in survivors of various malignant diseases. Early detection of myocardial injury in the setting of cancer treatment is important for the initiation of targeted cardioprotective therapy, in order to prevent irreversible cardiac dysfunction and heart failure, while not withholding a potentially life-saving cancer therapy. Cardiac imaging techniques including echocardiography, cardiac magnetic resonance, and nuclear cardiac imaging are the main tools for the identification of cardiotoxicity. There is also growing evidence for the detection of subclinical cardiac dysfunction in cancer patients by speckle tracking echocardiography. In this review article, we focus on current and emerging data regarding the role of cardiac imaging for the detection of changes in myocardial function related with cancer treatment in clinical practice.
Collapse
Affiliation(s)
- George Makavos
- 2nd Department of Cardiology, "Attikon" Hospital, National and Kapodistrian University of Athens Medical School, Rimini 1, Haidari, 12462, Athens, Greece.
| | - Ignatios Ikonomidis
- 2nd Department of Cardiology, "Attikon" Hospital, National and Kapodistrian University of Athens Medical School, Rimini 1, Haidari, 12462, Athens, Greece
| | - John Palios
- 2nd Department of Cardiology, "Attikon" Hospital, National and Kapodistrian University of Athens Medical School, Rimini 1, Haidari, 12462, Athens, Greece
| | - Angelos Rigopoulos
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| | - Konstantinos Katogiannis
- 2nd Department of Cardiology, "Attikon" Hospital, National and Kapodistrian University of Athens Medical School, Rimini 1, Haidari, 12462, Athens, Greece
| | - John Parissis
- 2nd Department of Cardiology, "Attikon" Hospital, National and Kapodistrian University of Athens Medical School, Rimini 1, Haidari, 12462, Athens, Greece
| | - Ioannis Paraskevaidis
- Department of Clinical Therapeutics, "Alexandra" Hospital, National and Kapodistrian University of Athens Medical School, Athens, Greece
| | - Michel Noutsias
- Mid-German Heart Center, Department of Internal Medicine III, Division of Cardiology, Angiology and Intensive Medical Care, University Hospital Halle, Martin-Luther-University Halle-Wittenberg, Ernst-Grube-Strasse 40, 06120, Halle (Saale), Germany
| |
Collapse
|
12
|
ARNIs: balancing “the good and the bad” of neuroendocrine response to HF. Clin Res Cardiol 2019; 109:599-610. [DOI: 10.1007/s00392-019-01547-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 09/10/2019] [Indexed: 10/26/2022]
|